Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Más filtros

Base de datos
Tipo del documento
Intervalo de año de publicación
1.
Nat Commun ; 11(1): 3653, 2020 07 21.
Artículo en Inglés | MEDLINE | ID: mdl-32694534

RESUMEN

The vasculature represents a highly plastic compartment, capable of switching from a quiescent to an active proliferative state during angiogenesis. Metabolic reprogramming in endothelial cells (ECs) thereby is crucial to cover the increasing cellular energy demand under growth conditions. Here we assess the impact of mitochondrial bioenergetics on neovascularisation, by deleting cox10 gene encoding an assembly factor of cytochrome c oxidase (COX) specifically in mouse ECs, providing a model for vasculature-restricted respiratory deficiency. We show that EC-specific cox10 ablation results in deficient vascular development causing embryonic lethality. In adult mice induction of EC-specific cox10 gene deletion produces no overt phenotype. However, the angiogenic capacity of COX-deficient ECs is severely compromised under energetically demanding conditions, as revealed by significantly delayed wound-healing and impaired tumour growth. We provide genetic evidence for a requirement of mitochondrial respiration in vascular endothelial cells for neoangiogenesis during development, tissue repair and cancer.


Asunto(s)
Mitocondrias/metabolismo , Neoplasias/patología , Neovascularización Patológica/patología , Neovascularización Fisiológica , Cicatrización de Heridas/fisiología , Adenosina Trifosfato/metabolismo , Transferasas Alquil y Aril/genética , Transferasas Alquil y Aril/metabolismo , Animales , Línea Celular Tumoral/trasplante , Respiración de la Célula , Modelos Animales de Enfermedad , Embrión de Mamíferos , Desarrollo Embrionario/fisiología , Células Endoteliales/fisiología , Endotelio Vascular/citología , Endotelio Vascular/fisiología , Femenino , Técnicas de Inactivación de Genes , Humanos , Masculino , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Transgénicos , Mitocondrias/genética , Neoplasias/irrigación sanguínea , Fosforilación Oxidativa
2.
J Control Release ; 223: 165-177, 2016 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-26742942
4.
Cell Death Differ ; 21(11): 1721-32, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24971483

RESUMEN

Hepatocellular carcinoma (HCC) usually develops in the context of chronic hepatitis triggered by viruses or toxic substances causing hepatocyte death, inflammation and compensatory proliferation of liver cells. Death receptors of the TNFR superfamily regulate cell death and inflammation and are implicated in liver disease and cancer. Liver parenchymal cell-specific ablation of NEMO/IKKγ, a subunit of the IκB kinase (IKK) complex that is essential for the activation of canonical NF-κB signalling, sensitized hepatocytes to apoptosis and caused the spontaneous development of chronic hepatitis and HCC in mice. Here we show that hepatitis and HCC development in NEMO(LPC-KO) mice is triggered by death receptor-independent FADD-mediated hepatocyte apoptosis. TNF deficiency in all cells or conditional LPC-specific ablation of TNFR1, Fas or TRAIL-R did not prevent hepatocyte apoptosis, hepatitis and HCC development in NEMO(LPC-KO) mice. To address potential functional redundancies between death receptors we generated and analysed NEMO(LPC-KO) mice with combined LPC-specific deficiency of TNFR1, Fas and TRAIL-R and found that also simultaneous lack of all three death receptors did not prevent hepatocyte apoptosis, chronic hepatitis and HCC development. However, LPC-specific combined deficiency in TNFR1, Fas and TRAIL-R protected the NEMO-deficient liver from LPS-induced liver failure, showing that different mechanisms trigger spontaneous and LPS-induced hepatocyte apoptosis in NEMO(LPC-KO) mice. In addition, NK cell depletion did not prevent liver damage and hepatitis. Moreover, NEMO(LPC-KO) mice crossed into a RAG-1-deficient genetic background-developed hepatitis and HCC. Collectively, these results show that the spontaneous development of hepatocyte apoptosis, chronic hepatitis and HCC in NEMO(LPC-KO) mice occurs independently of death receptor signalling, NK cells and B and T lymphocytes, arguing against an immunological trigger as the critical stimulus driving hepatocarcinogenesis in this model.


Asunto(s)
Apoptosis/fisiología , Carcinoma Hepatocelular/etiología , Proteína de Dominio de Muerte Asociada a Fas/metabolismo , Hepatitis/etiología , Quinasa I-kappa B/metabolismo , Péptidos y Proteínas de Señalización Intracelular/genética , Neoplasias Hepáticas/etiología , Receptores de Muerte Celular/metabolismo , Animales , Carcinoma Hepatocelular/metabolismo , Hepatitis/inmunología , Hepatitis/metabolismo , Neoplasias Hepáticas/metabolismo , Ratones , Ratones Noqueados , FN-kappa B/metabolismo , Receptores Tipo I de Factores de Necrosis Tumoral/metabolismo
5.
Cell Death Differ ; 18(12): 1845-53, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21566665

RESUMEN

The ubiquitin-editing enzyme A20 (tumor necrosis factor-α-induced protein 3) serves as a critical brake on nuclear factor κB (NF-κB) signaling. In humans, polymorphisms in or near the A20 gene are associated with several inflammatory disorders, including psoriasis. We show here that epidermis-specific A20-knockout mice (A20(EKO)) develop keratinocyte hyperproliferation, but no signs of skin inflammation, such as immune cell infiltration. However, A20(EKO) mice clearly developed ectodermal organ abnormalities, including disheveled hair, longer nails and sebocyte hyperplasia. This phenotype resembles that of mice overexpressing ectodysplasin-A1 (EDA-A1) or the ectodysplasin receptor (EDAR), suggesting that A20 negatively controls EDAR signaling. We found that A20 inhibited EDAR-induced NF-κB signaling independent from its de-ubiquitinating activity. In addition, A20 expression was induced by EDA-A1 in embryonic skin explants, in which its expression was confined to the hair placodes, known to be the site of EDAR expression. In summary, our data indicate that EDAR-induced NF-κB levels are controlled by A20, which functions as a negative feedback regulator, to assure proper skin homeostasis and epidermal appendage development.


Asunto(s)
Cisteína Endopeptidasas/genética , Epidermis/fisiología , Homeostasis , Péptidos y Proteínas de Señalización Intracelular/genética , Queratinocitos/metabolismo , FN-kappa B/metabolismo , Animales , Cisteína Endopeptidasas/metabolismo , Cisteína Endopeptidasas/fisiología , Ectodisplasinas/farmacología , Ectodisplasinas/fisiología , Receptor Edar/agonistas , Receptor Edar/antagonistas & inhibidores , Receptor Edar/metabolismo , Epidermis/patología , Retroalimentación Fisiológica , Genes Reporteros , Células HEK293 , Cabello/anomalías , Cabello/embriología , Humanos , Hiperplasia , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Péptidos y Proteínas de Señalización Intracelular/fisiología , Queratinocitos/fisiología , Antígeno Ki-67/metabolismo , Luciferasas/biosíntesis , Luciferasas/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fenotipo , Técnicas de Cultivo de Tejidos , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa , Factor de Necrosis Tumoral alfa/farmacología , Factor de Necrosis Tumoral alfa/fisiología
6.
Mucosal Immunol ; 1 Suppl 1: S54-7, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19079232

RESUMEN

The intestinal epithelium regulates the interaction between commensal bacteria and the immune system not only by providing a physical barrier but also by expressing a number of immunoregulatory and antimicrobial peptides. Recent studies showed that inhibition of IkappaB kinase (IKK)/nuclear factor-kappaB (NF-kappaB) signaling specifically in intestinal epithelial cells in vivo causes the spontaneous development of intestinal inflammation in mice. These findings identify IKK/NF-kappaB signaling in intestinal epithelial cells as an important factor for the maintenance of epithelial integrity and immune homeostasis in the gut.


Asunto(s)
Células Epiteliales/metabolismo , Homeostasis/inmunología , Quinasa I-kappa B/metabolismo , Mucosa Intestinal/metabolismo , Intestinos/inmunología , FN-kappa B/metabolismo , Transducción de Señal/inmunología , Animales , Células Epiteliales/citología , Humanos , Intestinos/citología
7.
J Gen Virol ; 89(Pt 6): 1545-1550, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18474572

RESUMEN

Prions induce highly typical histopathological changes including cell death, spongiosis and activation of glia, yet the molecular pathways leading to neurodegeneration remain elusive. Following prion infection, enhanced nuclear factor-kappaB (NF-kappaB) activity in the brain parallels the first pathological changes. The NF-kappaB pathway is essential for proliferation, regulation of apoptosis and immune responses involving induction of inflammation. The IkappaB kinase (IKK) signalosome is crucial for NF-kappaB signalling, consisting of the catalytic IKKalpha/IKKbeta subunits and the regulatory IKKgamma subunit. This study investigated the impact of NF-kappaB signalling on prion disease in mouse models with a central nervous system (CNS)-restricted elimination of IKKbeta or IKKgamma in nearly all neuroectodermal cells, including neurons, astrocytes and oligodendrocytes, and in mice containing a non-phosphorylatable IKKalpha subunit (IKKalpha AA/AA). In contrast to previously published data, the observed results showed no evidence supporting the hypothesis that impaired NF-kappaB signalling in the CNS impacts on prion pathogenesis.


Asunto(s)
FN-kappa B/metabolismo , Proteínas PrPSc/metabolismo , Scrapie/metabolismo , Animales , Encéfalo/patología , Quinasa I-kappa B/genética , Ratones , Ratones Transgénicos , FN-kappa B/genética , Proteínas PrPSc/patogenicidad , Scrapie/patología , Transducción de Señal , Virulencia
8.
Br J Dermatol ; 155(1): 62-6, 2006 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16792753

RESUMEN

BACKGROUND: The expression of calcium-binding S100 molecules organized within the epidermal differentiation complex on chromosome 1q21 is disturbed in hyperproliferative skin diseases such as psoriasis. OBJECTIVES: We studied whether serum levels of S100 proteins A8 (S100A8) and A9 (S100A9) are elevated in psoriasis, correlated their amounts with disease activity and identified potential cellular sources. METHODS: Serum obtained from psoriasis patients or from healthy individuals was studied for S100A8 and S100A9 levels by enzyme-linked immunosorbent assay. Data were correlated to disease activity as reflected by the Psoriasis Area and Severity Index (PASI). Cellular sources of S100A8 and S100A9 were identified by in situ hybridization and immunohistochemistry of lesional psoriatic and nonlesional, nonpsoriatic skin. RESULTS: A significant increase of S100A8/S100A9 serum levels was found in patients with psoriasis compared with healthy controls. Grading the patients into two groups of severity, individuals with a PASI of <15 showed serum levels of 705+/-120 ng mL-1 (mean+/-SEM, n=18), those with a PASI of >or=15 showed levels of 1315+/-150 ng mL-1 (n=32) while controls presented with 365+/-50 ng mL-1. Performing in situ hybridization of lesional psoriatic skin we detected a dramatic induction of both S100A8 and S100A9 mRNA and protein primarily in the suprabasal layers of the epidermis while expression was negligible in nonlesional, nonpsoriatic interfollicular epidermis. CONCLUSIONS: Our data demonstrate that hyperproliferation and abnormal differentiation of psoriatic skin is associated with a massive upregulation and secretion of S100A8 and S100A9, suggesting not only a prominent role of these molecules during intracellular calcium-dependent signalling but also implying distinct extracellular functions.


Asunto(s)
Calgranulina A/sangre , Calgranulina B/sangre , Epidermis/patología , Queratinocitos/patología , Psoriasis/sangre , Psoriasis/patología , Enfermedad Aguda , Adulto , Anciano , Biomarcadores/sangre , Calgranulina A/análisis , Calgranulina B/análisis , Estudios de Casos y Controles , Diferenciación Celular , Ensayo de Inmunoadsorción Enzimática , Epidermis/química , Femenino , Humanos , Inmunohistoquímica/métodos , Hibridación in Situ/métodos , Queratinocitos/química , Masculino , Persona de Mediana Edad , Regulación hacia Arriba
9.
Cell Death Differ ; 13(5): 861-72, 2006 May.
Artículo en Inglés | MEDLINE | ID: mdl-16470223

RESUMEN

Studies in transgenic and knockout mice have made a major contribution to our current understanding of the physiological functions of the NF-kappaB signalling cascade. The generation and analysis of mice with targeted modifications of individual components of the NF-kappaB pathway tremendously advanced our knowledge of the roles of the NF-kappaB proteins themselves, and also of the many activators and negative regulators of NF-kappaB. These studies have highlighted the complexity of the NF-kappaB system, by revealing the multiple interactions, redundancies, but also diverse functions, performed by the different molecules participating in the regulation of NF-kappaB signalling. Furthermore, inhibition or enforced activation of NF-kappaB in transgenic mice has uncovered the critical roles that NF-kappaB plays in the pathogenesis of various diseases such as liver failure, diabetes and cancer.


Asunto(s)
Ratones Noqueados/metabolismo , Ratones Transgénicos/metabolismo , FN-kappa B/metabolismo , Activación Transcripcional , Animales , Genoma , Hígado/metabolismo , Hepatopatías/metabolismo , Ratones , Modelos Biológicos , Transducción de Señal , Linfocitos T/metabolismo
11.
Mol Cell ; 5(6): 981-92, 2000 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-10911992

RESUMEN

Disruption of the X-linked gene encoding NF-kappa B essential modulator (NEMO) produces male embryonic lethality, completely blocks NF-kappa B activation by proinflammatory cytokines, and interferes with the generation and/or persistence of lymphocytes. Heterozygous female mice develop patchy skin lesions with massive granulocyte infiltration and hyperproliferation and increased apoptosis of keratinocytes. Diseased animals present severe growth retardation and early mortality. Surviving mice recover almost completely, presumably through clearing the skin of NEMO-deficient keratinocytes. Male lethality and strikingly similar skin lesions in heterozygous females are hallmarks of the human genetic disorder incontinentia pigmenti (IP). Together with the recent discovery that mutations in the human NEMO gene cause IP, our results indicate that we have created a mouse model for that disease.


Asunto(s)
Incontinencia Pigmentaria/genética , Proteínas Serina-Treonina Quinasas/fisiología , Animales , Apoptosis , División Celular , Células Cultivadas , Quimiotaxis de Leucocito , Citocinas/metabolismo , Citocinas/farmacología , Modelos Animales de Enfermedad , Femenino , Marcación de Gen , Heterocigoto , Humanos , Hiperpigmentación , Quinasa I-kappa B , Incontinencia Pigmentaria/embriología , Incontinencia Pigmentaria/enzimología , Incontinencia Pigmentaria/patología , Queratinocitos/enzimología , Queratinocitos/patología , Hígado/embriología , Hígado/patología , Masculino , Melaninas/metabolismo , Ratones , Ratones Noqueados , FN-kappa B/genética , FN-kappa B/metabolismo , Óxido Nítrico Sintasa/genética , Óxido Nítrico Sintasa/metabolismo , Óxido Nítrico Sintasa de Tipo II , Proteínas Serina-Treonina Quinasas/deficiencia , Proteínas Serina-Treonina Quinasas/genética , Piel/embriología , Piel/enzimología , Piel/metabolismo , Piel/patología , Regulación hacia Arriba
12.
Cell Immunol ; 201(1): 33-41, 2000 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-10805971

RESUMEN

The development and function of secondary lymphoid tissue require signaling by tumor necrosis factor and lymphotoxins. Mice deficient in LTbetaR show defective organogenesis of lymph nodes and Peyer's patches and a severely disturbed splenic architecture. In contrast, TNF or p55TNF-R deficiency does not affect the organogenesis of peripheral lymphoid organs but interferes with the formation of B cell follicles and the appearance of FDC networks and germinal centers in all secondary lymphoid organs. Based on these differences, we have previously hypothesized that the role of TNF in lymphoid structure is distinct from that of LT and restricted in regulating cellular interactions that allow the differentiation and/or correct positioning of FDCs. In the present study we show that, in addition to the defects in follicular structure, TNF or p55TNF-R knockout mice exhibit defects in the formation of the macrophage populations and of the sinus lining cells of the splenic marginal zone. Interestingly, a large number of dendritic-shaped cells stained with FDC-specific markers and able to trap immune complexes are retained within the defective marginal zone of TNF and p55TNF-R knockout spleens. We conclude that the primary defect in the lymphoid phenotype of TNF or p55TNF-R knockout mice is the failure of FDC precursors to migrate through the disorganized marginal sinus and to home properly into the splenic follicular areas where they would promote the formation of B cell follicles and germinal centers.


Asunto(s)
Antígenos CD/fisiología , Movimiento Celular/fisiología , Células Dendríticas Foliculares/fisiología , Receptores del Factor de Necrosis Tumoral/fisiología , Factor de Necrosis Tumoral alfa/fisiología , Animales , Antígenos CD/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores del Factor de Necrosis Tumoral/genética , Receptores Tipo I de Factores de Necrosis Tumoral , Bazo/citología , Células Madre/fisiología , Factor de Necrosis Tumoral alfa/genética
13.
J Virol ; 74(7): 3338-44, 2000 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-10708451

RESUMEN

In most peripheral infections of rodents and sheep with scrapie, infectivity is found first in lymphoid tissues and later in the central nervous system (CNS). Cells within the germinal centers (GCs) of the spleen and lymph nodes are important sites of extraneural replication, from which infection is likely to spread to the CNS along peripheral nerves. Here, using immunodeficient mice, we investigate the identity of the cells in the spleen that are important for disease propagation. Despite possessing functional T and B lymphocytes, tumor necrosis factor alpha-deficient (TNF-alpha(-/-)) mice lack GCs and follicular dendritic cell (FDC) networks in lymphoid tissues. In contrast, lymphoid tissues of interleukin-6-deficient (IL-6(-/-)) mice possess FDC networks but have impaired GCs. When the CNSs of TNF-alpha(-/-), IL-6(-/-), and wild-type mice were directly challenged with the ME7 scrapie strain, 100% of the mice were susceptible, developing disease after closely similar incubation periods. However, when challenged peripherally (intraperitoneally), most TNF-alpha(-/-) mice failed to develop scrapie up to 503 days postinjection. All wild-type and IL-6(-/-) mice succumbed to disease approximately 300 days after the peripheral challenge. High levels of scrapie infection and the disease-specific isomer of the prion protein, PrP(Sc), were detectable in spleens from challenged wild-type and IL-6(-/-) mice but not from TNF-alpha(-/-) mice. Histopathological analysis of spleen tissue demonstrated heavy PrP accumulations in direct association with FDCs in challenged wild-type and IL-6(-/-) mice. No PrP(Sc) accumulation was detected in spleens from TNF-alpha(-/-) mice. We conclude that, for the ME7 scrapie strain, mature FDCs are critical for replication in lymphoid tissues and that in their absence, neuroinvasion following peripheral challenge is impaired.


Asunto(s)
Interleucina-6/genética , Scrapie/genética , Factor de Necrosis Tumoral alfa/genética , Animales , Predisposición Genética a la Enfermedad , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas PrPSc/metabolismo , Proteínas PrPSc/patogenicidad
14.
Nat Med ; 5(7): 828-31, 1999 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-10395330

RESUMEN

Given the associations between chronic inflammation and epithelial cancer, we studied susceptibility to skin carcinogenesis in mice deficient for the pro-inflammatory cytokine TNF-alpha (refs. 5,6). TNF-alpha(-/-) mice were resistant to development of benign and malignant skin tumors, whether induced by initiation with DMBA and promotion with TPA or by repeated dosing with DMBA. TNF-alpha(-/-) mice developed 5-10% the number of tumors developed by wild-type mice during initiation/promotion and 25% of those in wild-type mice after repeated carcinogen treatment. TNF-alpha could influence tumor and stromal cells during tumor development. The early stages of TPA promotion are characterized by keratinocyte hyperproliferation and inflammation. These were diminished in TNF-alpha(-/-) mice. TNF-alpha was extensively induced in the epidermis, but not the dermis, in TPA-treated wild-type skin, indicating that dermal inflammation is controlled by keratinocyte TNF-alpha production. Deletion of a TNF-alpha inducible chemokine also conferred some resistance to skin tumor development. TNF-alpha has little influence on later stages of carcinogenesis, as tumors in wild-type and TNF-alpha(-/-) mice had similar rates of malignant progression. These data provide evidence that a pro-inflammatory cytokine is required for de novo carcinogenesis and that TNF-alpha is important to the early stages of tumor promotion. Strategies that neutralize TNF-alpha production may be useful in cancer treatment and prevention.


Asunto(s)
Inmunidad Innata/genética , Neoplasias Cutáneas/inmunología , Factor de Necrosis Tumoral alfa/deficiencia , 9,10-Dimetil-1,2-benzantraceno/toxicidad , Animales , Cruzamientos Genéticos , Femenino , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Endogámicos , Ratones Noqueados , Invasividad Neoplásica , Estadificación de Neoplasias , Piel/efectos de los fármacos , Piel/patología , Neoplasias Cutáneas/inducido químicamente , Neoplasias Cutáneas/patología , Acetato de Tetradecanoilforbol/toxicidad , Factores de Tiempo , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/fisiología
15.
J Biol Chem ; 274(21): 14786-90, 1999 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-10329676

RESUMEN

Syndecan-4 is a unique member of the syndecan gene family that has the ability to bind and activate protein kinase C-alpha. Whereas increased syndecan-4 levels have been noted in ischemic hearts, little is known regarding the regulation of its expression. To investigate the role of cardiac myocytes in induction of syndecan-4 expression, human endothelial cells (ECV304) were exposed to a medium conditioned by primary mouse cardiac myocytes or H9c2 cells. The medium conditioned by hypoxic but not normal myocytes was able to induce syndecan-4 expression in ECV cells. Western analysis of the conditioned medium demonstrated an increased presence of tumor necrosis factor-alpha (TNF-alpha) in the medium conditioned by hypoxic but not normal myoblasts. Primary cardiac myocytes collected from the wild type C57/129 but not the homozygous TNF-alpha-/- knockout mice were able to induce syndecan-4 expression in ECV cells when cultured under hypoxic conditions. In vitro studies demonstrated that TNF-alpha induced endothelial cell syndecan-4 expression by both increasing syndecan-4 gene expression in an NF-kappaB-dependent manner and by prolonging syndecan-4 mRNA half-life. We conclude that TNF-alpha is the principal factor produced by the ischemic myocytes that is responsible for induction of endothelial cell syndecan-4 expression and that this requires both transcriptional and posttranscriptional mechanisms.


Asunto(s)
Glicoproteínas de Membrana/biosíntesis , Miocardio/citología , Proteoglicanos/biosíntesis , Animales , Hipoxia de la Célula , Medios de Cultivo Condicionados , Endotelio/fisiología , Humanos , Glicoproteínas de Membrana/genética , Ratones , Proteoglicanos/genética , Sindecano-4 , Factor de Necrosis Tumoral alfa/fisiología
16.
Immunity ; 10(3): 387-98, 1999 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-10204494

RESUMEN

We addressed the impact of deleting TNF AU-rich elements (ARE) from the mouse genome on the regulation of TNF biosynthesis and the physiology of the host. Absence of the ARE affected mechanisms responsible for TNF mRNA destabilization and translational repression in hemopoietic and stromal cells. In stimulated conditions, TNF ARE were required both for the alleviation and reinforcement of message destabilization and translational silencing. Moreover, the mutant mRNA was no longer responsive to translational modulation by the p38 and JNK kinases, demonstrating that TNF ARE are targets for these signals. Development of two specific pathologies in mutant mice, i.e., chronic inflammatory arthritis and Crohn's-like inflammatory bowel disease, suggests that defective function of ARE may be etiopathogenic for the development of analogous human pathologies.


Asunto(s)
Artritis/inmunología , Artritis/patología , Enfermedad de Crohn/inmunología , Enfermedad de Crohn/patología , Proteínas Quinasas Activadas por Mitógenos , Factor de Necrosis Tumoral alfa/biosíntesis , Regiones no Traducidas 3'/genética , Regiones no Traducidas 3'/inmunología , Animales , Antígenos CD/fisiología , Artritis/etiología , Artritis/genética , Linfocitos B/citología , Linfocitos B/inmunología , Proteínas Quinasas Dependientes de Calcio-Calmodulina/fisiología , Diferenciación Celular/inmunología , Enfermedad de Crohn/etiología , Enfermedad de Crohn/genética , Cruzamientos Genéticos , Fibroblastos/metabolismo , Trastornos del Crecimiento/genética , Trastornos del Crecimiento/inmunología , Trastornos del Crecimiento/mortalidad , Proteínas Quinasas JNK Activadas por Mitógenos , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos CBA , Ratones Transgénicos , ARN Mensajero/metabolismo , Receptores del Factor de Necrosis Tumoral/fisiología , Receptores Tipo II del Factor de Necrosis Tumoral , Transducción de Señal/genética , Transducción de Señal/inmunología , Membrana Sinovial/metabolismo , Linfocitos T/citología , Linfocitos T/inmunología , Factor de Necrosis Tumoral alfa/deficiencia , Factor de Necrosis Tumoral alfa/genética , Proteínas Quinasas p38 Activadas por Mitógenos
17.
Eur J Immunol ; 29(3): 774-80, 1999 03.
Artículo en Inglés | MEDLINE | ID: mdl-10092079

RESUMEN

Experimental autoimmune encephalomyelitis (EAE) induction in TNF gene-targeted mice has resulted in conflicting reports in part due to the strong association of TNF with the MHC locus. To define the participation of TNF in EAE development, we back-crossed TNF-deficient mice (H-2b) into the SJL/J strain and directly compared them to H-2b congenic SJL or inbred SJL/J mice. Induction of EAE with myelin basic protein (MBP) revealed that H-2b congenic SJL mice are fully susceptible, indicating that the H-2b haplotype does not affect disease susceptibility. Using H-2b congenic SJL mice we show here that TNF deficiency modifies the normal course of EAE by considerably delaying the onset for approximately 5 days, suggesting that TNF is required for the normal initiation of MBP-induced EAE. However, TNF-deficient mice eventually developed severe EAE with perivascular inflammation and primary demyelination similar to wild-type controls, indicating that TNF is not essential during these processes. Taken together, these results indicate that although TNF is not required for the progression of MBP-induced EAE, it contributes positively by advancing the onset of disease.


Asunto(s)
Encefalomielitis Autoinmune Experimental/inmunología , Factor de Necrosis Tumoral alfa/inmunología , Animales , Sistema Nervioso Central , Enfermedades Desmielinizantes , Susceptibilidad a Enfermedades , Femenino , Antígenos H-2/inmunología , Incidencia , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína Básica de Mielina/inmunología , Factor de Necrosis Tumoral alfa/genética
18.
Clin Exp Immunol ; 115(1): 42-55, 1999 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-9933419

RESUMEN

Immunosuppressive therapy with methotrexate (MTX) has been established as effective treatment for patients with rheumatoid arthritis. To analyse the therapeutic potential and mechanisms of action of MTX, we determined serum cytokine levels and cytokine production by splenic T cells and macrophages in untreated and MTX-treated mice. Furthermore, we assessed the role of MTX in a murine model of experimental arthritis induced by collagen type II (CIA). MTX reduced spontaneous and IL-15-induced tumour necrosis factor (TNF) production by splenic T cells but not by macrophages from healthy mice in vitro in a dose-dependent manner. In contrast, interferon-gamma (IFN-gamma) production was less strikingly reduced and IL-4 production was virtually unaffected. In addition, treatment of healthy mice with MTX in vivo led to reduced TNF serum levels and diminished TNF production by splenic T cells and macrophages. Intraperitoneal administration of MTX prior to the onset of arthritis completely prevented clinical and pathological signs of CIA. This was associated with a striking reduction of TNF production by spleen cells from MTX-treated mice. The role of TNF in MTX-mediated effects on cytokine production was further underlined by the finding that MTX effects on IFN-gamma production were augmented in TNF-transgenic mice but abrogated in mice in which the TNF-alpha gene had been inactivated by homologous recombination. Thus, MTX specifically modulates spontaneous and IL-15-induced TNF-alpha production in mice and prevents experimental murine CIA. These data suggest that TNF production by T cells is an important target of MTX and may serve as a basis to understand and further analyse MTX-mediated mechanisms of immunosuppression in patients with RA.


Asunto(s)
Artritis Experimental/metabolismo , Colágeno , Citocinas/biosíntesis , Inmunosupresores/farmacología , Macrófagos/metabolismo , Metotrexato/farmacología , Linfocitos T/metabolismo , Animales , Artritis Experimental/inducido químicamente , Artritis Experimental/prevención & control , Linfocitos T CD4-Positivos/metabolismo , Interferón gamma/biosíntesis , Interleucina-15/farmacología , Interleucina-4/biosíntesis , Interleucina-6/biosíntesis , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos DBA , Ratones Noqueados , Ratones Transgénicos , Bazo/citología , Bazo/metabolismo , Factor de Necrosis Tumoral alfa/biosíntesis
19.
Am J Pathol ; 153(3): 801-13, 1998 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-9736029

RESUMEN

The scientific dogma that multiple sclerosis (MS) is a disease caused by a single pathogenic mechanism has been challenged recently by the heterogeneity observed in MS lesions and the realization that not all patterns of demyelination can be modeled by autoimmune-triggered mechanisms. To evaluate the contribution of local tumor necrosis factor (TNF) ligand/receptor signaling pathways to MS immunopathogenesis we have analyzed disease pathology in central nervous system-expressing TNF transgenic mice, with or without p55 or p75TNF receptors, using combined in situ terminal deoxynucleotidyl transferase-mediated dUTP-biotin nick-end labeling and cell identification techniques. We demonstrate that local production of TNF by central nervous system glia potently and selectively induces oligodendrocyte apoptosis and myelin vacuolation in the context of an intact blood-brain barrier and absence of immune cell infiltration into the central nervous system parenchyma. Interestingly, primary demyelination then develops in a classical manner in the presence of large numbers of recruited phagocytic macrophages, possibly the result of concomitant pro-inflammatory effects of TNF in the central nervous system, and lesions progress into acute or chronic MS-type plaques with axonal damage, focal blood-brain barrier disruption, and considerable oligodendrocyte loss. Both the cytotoxic and inflammatory effects of TNF were abrogated in mice genetically deficient for the p55TNF receptor demonstrating a dominant role for p55TNF receptor-signaling pathways in TNF-mediated pathology. These results demonstrate that aberrant local TNF/p55TNF receptor signaling in the central nervous system can have a potentially major role in the aetiopathogenesis of MS demyelination, particularly in MS subtypes in which oligodendrocyte death is a primary pathological feature, and provide new models for studying the basic mechanisms underlying oligodendrocyte and myelin loss.


Asunto(s)
Antígenos CD/metabolismo , Apoptosis , Encéfalo/patología , Esclerosis Múltiple/etiología , Oligodendroglía/patología , Receptores del Factor de Necrosis Tumoral/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Animales , Antígenos CD/genética , Encéfalo/metabolismo , Enfermedad Crónica , Femenino , Técnica del Anticuerpo Fluorescente Indirecta , Técnicas para Inmunoenzimas , Hibridación in Situ , Masculino , Ratones , Ratones Noqueados , Ratones Transgénicos , Microscopía Confocal , Esclerosis Múltiple/metabolismo , Esclerosis Múltiple/patología , Oligodendroglía/metabolismo , Receptores del Factor de Necrosis Tumoral/genética , Receptores Tipo I de Factores de Necrosis Tumoral , Médula Espinal/metabolismo , Médula Espinal/patología , Factor de Necrosis Tumoral alfa/genética
20.
J Exp Med ; 188(4): 745-54, 1998 Aug 17.
Artículo en Inglés | MEDLINE | ID: mdl-9705956

RESUMEN

Lymphotoxin (LT)alpha knockout mice, as well as double LTalpha/tumor necrosis factor (TNF) knockout mice, show a severe splenic disorganization with nonsegregating T/B cell zones and complete absence of primary B cell follicles, follicular dendritic cell (FDC) networks, and germinal centers. In contrast, as shown previously and confirmed in this study, LTbeta-deficient mice show much more conserved T/B cell areas and a reduced but preserved capacity to form germinal centers and FDC networks. We show here that similar to the splenic phenotype of LTbeta-deficient mice, complementation of LTalpha knockout mice with TNF-expressing transgenes leads to a p55 TNF receptor-dependent restoration of B/T cell zone segregation and a partial preservation of primary B cell follicles, FDC networks, and germinal centers. Notably, upon lipopolysaccharide challenge, LTalpha knockout mice fail to produce physiological levels of TNF both in peritoneal macrophage supernatants and in their serum, indicating a coinciding deficiency in TNF expression. These findings suggest that defective TNF expression contributes to the complex phenotype of the LTalpha knockout mice, and uncover a predominant role for TNF and its p55 TNF receptor in supporting, even in the absence of LTalpha, the development and maintenance of splenic B cell follicles, FDC networks, and germinal centers.


Asunto(s)
Linfocitos B/fisiología , Linfotoxina-alfa/fisiología , Bazo/fisiología , Linfocitos T/fisiología , Factor de Necrosis Tumoral alfa/fisiología , Animales , Antígenos CD/genética , Antígenos CD/fisiología , Centro Germinal , Humanos , Linfotoxina-alfa/genética , Ratones , Ratones Noqueados , Receptores del Factor de Necrosis Tumoral/genética , Receptores del Factor de Necrosis Tumoral/fisiología , Receptores Tipo I de Factores de Necrosis Tumoral , Bazo/citología , Transgenes , Factor de Necrosis Tumoral alfa/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA