Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
bioRxiv ; 2024 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-38826303

RESUMEN

2',3'-cyclic nucleotide 3'-phosphodiesterase (CNPase) is an abundant constituent of central nervous system non-compact myelin, frequently used as a marker antigen for myelinating cells. The catalytic activity of CNPase, the 3'-hydrolysis of 2',3'-cyclic nucleotides, is well characterised in vitro, but the in vivo function of CNPase remains unclear. CNPase interacts with the actin cytoskeleton to counteract the developmental closure of cytoplasmic channels that travel through compact myelin; its enzymatic activity may be involved in adenosine metabolism and RNA degradation. We developed a set of high-affinity nanobodies recognizing the phosphodiesterase domain of CNPase, and the crystal structures of each complex show that the five nanobodies have distinct epitopes. One of the nanobodies bound deep into the CNPase active site and acted as an inhibitor. Moreover, the nanobodies were characterised in imaging applications and as intrabodies, expressed in mammalian cells, such as primary oligodendrocytes. Fluorescently labelled nanobodies functioned in imaging of teased nerve fibers and whole brain tissue sections, as well as super-resolution microscopy. These anti-CNPase nanobodies provide new tools for structural and functional biology of myelination, including high-resolution imaging of nerve tissue.

2.
Cells ; 12(18)2023 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-37759513

RESUMEN

Our previous work demonstrated that vitamin D (VitD) reduces experimental autoimmune encephalomyelitis (EAE) disease severity in wild-type (WT) but not in T cell-specific glucocorticoid (GC) receptor (GR)-deficient (GRlck) mice. This study aimed to investigate the interplay between the GR- and VitD receptor (VDR) signaling. In vivo, we confirmed the involvement of the GR in the VitD-induced effects in EAE using WT and GRlck mice. Furthermore, we observed that VitD-enhanced T cell apoptosis and T regulatory cell differentiation are diminished in vitro in CD3+ T cells of GRlck but not WT mice. Mechanistically, VitD does not appear to signal directly via the GR, as it does not bind to the GR, does not induce its nuclear translocation, and does not modulate the expression of two GR-induced genes. However, we observed that VitD enhances VDR protein expression in CD3+ T cells from WT but not GRlck mice in vitro, that the GR and the VDR spatially co-localize after VitD treatment, and that VitD does not modulate the expression of two VDR-induced genes in the absence of the GR. Our data suggest that a functional GR, specifically in T cells, is required for the VDR to signal appropriately to mediate the therapeutic effects of VitD.


Asunto(s)
Encefalomielitis Autoinmune Experimental , Receptores de Glucocorticoides , Ratones , Animales , Receptores de Glucocorticoides/metabolismo , Receptores de Calcitriol/metabolismo , Vitamina D/farmacología , Glucocorticoides/farmacología , Transducción de Señal , Vitaminas
3.
Sci Signal ; 14(709): eabe3800, 2021 Nov 16.
Artículo en Inglés | MEDLINE | ID: mdl-34784249

RESUMEN

The formation of Ca2+ microdomains during T cell activation is initiated by the production of nicotinic acid adenine dinucleotide phosphate (NAADP) from its reduced form NAADPH. The reverse reaction­NAADP to NAADPH­is catalyzed by glucose 6-phosphate dehydrogenase (G6PD). Here, we identified NADPH oxidases NOX and DUOX as NAADP-forming enzymes that convert NAADPH to NAADP under physiological conditions in vitro. T cells express NOX1, NOX2, and, to a minor extent, DUOX1 and DUOX2. Local and global Ca2+ signaling were decreased in mouse T cells with double knockout of Duoxa1 and Duoxa2 but not with knockout of Nox1 or Nox2. Ca2+ microdomains in the first 15 s upon T cell activation were significantly decreased in Duox2−/− but not in Duox1−/− T cells, whereas both DUOX1 and DUOX2 were required for global Ca2+ signaling between 4 and 12 min after stimulation. Our findings suggest that a DUOX2- and G6PD-catalyzed redox cycle rapidly produces and degrades NAADP through NAADPH as an inactive intermediate.


Asunto(s)
Señalización del Calcio , Oxidasas Duales , Activación de Linfocitos , NADPH Oxidasas , NADP/biosíntesis , Linfocitos T , Animales , Oxidasas Duales/genética , Células HEK293 , Humanos , Células Jurkat , Ratones Noqueados , NADP/análogos & derivados , NADPH Oxidasas/genética , Linfocitos T/enzimología
4.
Sci Signal ; 14(675)2021 03 23.
Artículo en Inglés | MEDLINE | ID: mdl-33758062

RESUMEN

NAADP-evoked Ca2+ release through type 1 ryanodine receptors (RYR1) is a major mechanism underlying the earliest signals in T cell activation, which are the formation of Ca2+ microdomains. In our characterization of the molecular machinery underlying NAADP action, we identified an NAADP-binding protein, called hematological and neurological expressed 1-like protein (HN1L) [also known as Jupiter microtubule-associated homolog 2 (JPT2)]. Gene deletion of Hn1l/Jpt2 in human Jurkat and primary rat T cells resulted in decreased numbers of initial Ca2+ microdomains and delayed the onset and decreased the amplitude of global Ca2+ signaling. Photoaffinity labeling demonstrated direct binding of NAADP to recombinant HN1L/JPT2. T cell receptor/CD3-dependent coprecipitation of HN1L/JPT2 with RYRs and colocalization of these proteins suggest that HN1L/JPT2 connects NAADP formation with the activation of RYR channels within the first seconds of T cell activation. Thus, HN1L/JPT2 enables NAADP to activate Ca2+ release from the endoplasmic reticulum through RYR.


Asunto(s)
Calcio/metabolismo , Microdominios de Membrana/metabolismo , Proteínas Asociadas a Microtúbulos/metabolismo , NADP/análogos & derivados , Animales , Complejo CD3/metabolismo , Señalización del Calcio , Retículo Endoplásmico/metabolismo , Humanos , Células Jurkat , Activación de Linfocitos , Proteínas Asociadas a Microtúbulos/genética , NADP/metabolismo , Unión Proteica , Ratas , Receptores de Antígenos de Linfocitos T/metabolismo , Canal Liberador de Calcio Receptor de Rianodina/metabolismo , Linfocitos T/metabolismo
5.
Nature ; 567(7749): E15, 2019 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-30867589

RESUMEN

In this Article, owing to an error during the production process, the y-axis label of Fig. 2c should read "Number of Tß-syn cells" rather than "Number of T1ß-syn cells" and the left and right panels of Fig. 4 should be labelled 'a' and 'b', respectively. These errors have been corrected online.

6.
Nature ; 566(7745): 503-508, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30787438

RESUMEN

The grey matter is a central target of pathological processes in neurodegenerative disorders such as Parkinson's and Alzheimer's diseases. The grey matter is often also affected in multiple sclerosis, an autoimmune disease of the central nervous system. The mechanisms that underlie grey matter inflammation and degeneration in multiple sclerosis are not well understood. Here we show that, in Lewis rats, T cells directed against the neuronal protein ß-synuclein specifically invade the grey matter and that this is accompanied by the presentation of multifaceted clinical disease. The expression pattern of ß-synuclein induces the local activation of these T cells and, therefore, determined inflammatory priming of the tissue and targeted recruitment of immune cells. The resulting inflammation led to significant changes in the grey matter, which ranged from gliosis and neuronal destruction to brain atrophy. In humans, ß-synuclein-specific T cells were enriched in patients with chronic-progressive multiple sclerosis. These findings reveal a previously unrecognized role of ß-synuclein in provoking T-cell-mediated pathology of the central nervous system.


Asunto(s)
Sustancia Gris/inmunología , Sustancia Gris/patología , Esclerosis Múltiple Crónica Progresiva/inmunología , Esclerosis Múltiple Crónica Progresiva/patología , Linfocitos T/inmunología , Sinucleína beta/inmunología , Animales , Encéfalo/patología , Movimiento Celular/inmunología , Femenino , Regulación de la Expresión Génica , Gliosis/patología , Humanos , Inflamación/inmunología , Inflamación/patología , Activación de Linfocitos , Recuento de Linfocitos , Masculino , Esclerosis Múltiple Crónica Progresiva/sangre , Enfermedades Neurodegenerativas/inmunología , Enfermedades Neurodegenerativas/patología , Neuronas/patología , Ratas , Ratas Endogámicas Lew , Linfocitos T/metabolismo , Linfocitos T/patología , Sinucleína beta/análisis , Sinucleína beta/genética , Sinucleína beta/metabolismo
7.
Sci Signal ; 11(561)2018 12 18.
Artículo en Inglés | MEDLINE | ID: mdl-30563862

RESUMEN

The earliest intracellular signals that occur after T cell activation are local, subsecond Ca2+ microdomains. Here, we identified a Ca2+ entry component involved in Ca2+ microdomain formation in both unstimulated and stimulated T cells. In unstimulated T cells, spontaneously generated small Ca2+ microdomains required ORAI1, STIM1, and STIM2. Super-resolution microscopy of unstimulated T cells identified a circular subplasmalemmal region with a diameter of about 300 nm with preformed patches of colocalized ORAI1, ryanodine receptors (RYRs), and STIM1. Preformed complexes of STIM1 and ORAI1 in unstimulated cells were confirmed by coimmunoprecipitation and Förster resonance energy transfer studies. Furthermore, within the first second after T cell receptor (TCR) stimulation, the number of Ca2+ microdomains increased in the subplasmalemmal space, an effect that required ORAI1, STIM2, RYR1, and the Ca2+ mobilizing second messenger NAADP (nicotinic acid adenine dinucleotide phosphate). These results indicate that preformed clusters of STIM and ORAI1 enable local Ca2+ entry events in unstimulated cells. Upon TCR activation, NAADP-evoked Ca2+ release through RYR1, in coordination with Ca2+ entry through ORAI1 and STIM, rapidly increases the number of Ca2+ microdomains, thereby initiating spread of Ca2+ signals deeper into the cytoplasm to promote full T cell activation.


Asunto(s)
Calcio/metabolismo , Activación de Linfocitos , Proteína ORAI1/metabolismo , Canal Liberador de Calcio Receptor de Rianodina/metabolismo , Molécula de Interacción Estromal 1/metabolismo , Molécula de Interacción Estromal 2/metabolismo , Linfocitos T/citología , Animales , Señalización del Calcio , Membrana Celular , Células Cultivadas , Femenino , Transferencia Resonante de Energía de Fluorescencia , Masculino , Microdominios de Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Linfocitos T/inmunología , Linfocitos T/metabolismo
8.
Neurobiol Dis ; 102: 60-69, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28235673

RESUMEN

Laquinimod is currently being tested as a therapeutic drug in multiple sclerosis. However, its exact mechanism of action is still under investigation. Tracking of fluorescently-tagged encephalitogenic T cells during experimental autoimmune encephalomyelitis (EAE), an animal model for multiple sclerosis, revealed that laquinimod significantly reduces the invasion of pathogenic effector T cells into the CNS tissue. T-cell activation, differentiation and amplification within secondary lymphoid organs after immunization with myelin antigen, their migratory capacity and re-activation within the nervous tissue were either only mildly affected or remained unchanged. Instead, laquinimod directly impacted the functionality of the CNS vasculature. The expression of tight junction proteins p120 and ZO-1 in human brain endothelial cells was up-regulated upon laquinimod treatment, resulting in a significant increase in the transendothelial electrical resistance of confluent monolayers of brain endothelial cells. Similarly, expression of the adhesion molecule activated leukocyte cell adhesion molecule (ALCAM) and inflammatory chemokines CCL2 and IP-10 was suppressed, leading to a significant reduction in the migration of memory TH1 and TH17 lymphocytes across the blood brain barrier (BBB). Our data indicate that laquinimod exerts its therapeutic effects by tightening the BBB and limiting parenchymal invasion of effector T cells, thereby reducing CNS damage.


Asunto(s)
Barrera Hematoencefálica/efectos de los fármacos , Barrera Hematoencefálica/metabolismo , Fármacos Neuroprotectores/farmacología , Quinolonas/farmacología , Adulto , Animales , Permeabilidad Capilar/efectos de los fármacos , Permeabilidad Capilar/fisiología , Células Cultivadas , Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Encefalomielitis Autoinmune Experimental/metabolismo , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Femenino , Humanos , Linfocitos/efectos de los fármacos , Linfocitos/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Esclerosis Múltiple Recurrente-Remitente/tratamiento farmacológico , Esclerosis Múltiple Recurrente-Remitente/metabolismo , Ratas Endogámicas Lew , Linfocitos T/efectos de los fármacos , Linfocitos T/metabolismo , Adulto Joven
9.
Cell Calcium ; 64: 118-129, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28126314

RESUMEN

Antigenic activation is a central process in T-cell biology essential for efficient protection of the host from infections and tumors by the adaptive immune system. Furthermore, this process is of paramount importance for the initiation of autoimmunity. Many insights into the mechanisms of T-cell activation have been gained from real-time studies. The development of 2-photon microscopy transferred the focus of T cell activation to in vivo research and the analysis of the highly dynamic and complex T-cell response in the physiologic context of an animal model. In the last 15 years, real-time analysis of T-cell activation has progressed from a descriptive characterization of T-cell locomotion and visualization of T-cell contacts with putative antigen-presenting cells in ex vivo explants toward true intravital imaging using more functionally informative indicators of TCR-driven signaling to spot and quantify productive T-cell-APC interactions in situ. In this review we will briefly summarize and discuss current approaches to the real-time analysis of T-cell activation in vivo and their impact on our understanding of T cell function under homeostatic and pathological conditions.


Asunto(s)
Sistemas de Computación , Enfermedad , Salud , Microscopía Intravital/métodos , Activación de Linfocitos , Linfocitos T/inmunología , Animales , Humanos
10.
Oncotarget ; 7(30): 46862-46870, 2016 Jul 26.
Artículo en Inglés | MEDLINE | ID: mdl-27409835

RESUMEN

The p53-inducible cell cycle regulator 14-3-3σ exhibits tumor suppressive functions and is highly expressed in differentiating layers of the epidermis and hair follicles. 14-3-3σ/SFN/stratifin is frequently silenced in human epithelial cancers, and experimental down-regulation of 14-3-3σ expression immortalizes primary human keratinocytes. In the repeated-epilation (ER) mouse model, a heterozygous nonsense mutation of 14-3-3σ causes repeated hair-loss, hyper-proliferative epidermis, and spontaneous development of papillomas and squamous cell carcinomas in aging mice. Therefore, loss of 14-3-3σ function might contribute to epithelial tumor development. Here, we generated mice with loxP sites surrounding the single 14-3-3σ exon which allowed Cre-mediated deletion of the gene. 14-3-3σ-deficient mice are viable, but demonstrate a permanently disheveled fur. However, histological analyses of the skin did not reveal obvious defects in the hair follicles or the epidermis. Deletion of 14-3-3σ did not enhance spontaneous epidermal tumor development, whereas it increased the frequency and size of DMBA/TPA-induced papillomas. In conclusion, 14-3-3σ is dispensable for normal epidermal homeostasis but critical for suppression of chemically-induced skin carcinogenesis. In addition, these results suggest that the ER mutation of 14-3-3σ is not equivalent to loss of 14-3-3σ, but may represent a gain-of-function variant, which does not reflect the organismal function of wild-type 14-3-3σ.


Asunto(s)
Proteínas 14-3-3/genética , Carcinogénesis/genética , Epidermis/patología , Folículo Piloso/patología , Papiloma/genética , Neoplasias Cutáneas/genética , Proteínas 14-3-3/metabolismo , 9,10-Dimetil-1,2-benzantraceno/toxicidad , Animales , Carcinogénesis/inducido químicamente , Diferenciación Celular , Proliferación Celular , Codón sin Sentido , Modelos Animales de Enfermedad , Regulación hacia Abajo , Células Epidérmicas , Exones/genética , Femenino , Mutación con Ganancia de Función , Eliminación de Gen , Heterocigoto , Inmunohistoquímica , Integrasas/genética , Queratinocitos/patología , Masculino , Ratones , Papiloma/inducido químicamente , Papiloma/mortalidad , Papiloma/patología , Neoplasias Cutáneas/inducido químicamente , Neoplasias Cutáneas/mortalidad , Neoplasias Cutáneas/patología , Acetato de Tetradecanoilforbol/toxicidad
11.
Nature ; 530(7590): 349-53, 2016 Feb 18.
Artículo en Inglés | MEDLINE | ID: mdl-26863192

RESUMEN

In multiple sclerosis, brain-reactive T cells invade the central nervous system (CNS) and induce a self-destructive inflammatory process. T-cell infiltrates are not only found within the parenchyma and the meninges, but also in the cerebrospinal fluid (CSF) that bathes the entire CNS tissue. How the T cells reach the CSF, their functionality, and whether they traffic between the CSF and other CNS compartments remains hypothetical. Here we show that effector T cells enter the CSF from the leptomeninges during Lewis rat experimental autoimmune encephalomyelitis (EAE), a model of multiple sclerosis. While moving through the three-dimensional leptomeningeal network of collagen fibres in a random Brownian walk, T cells were flushed from the surface by the flow of the CSF. The detached cells displayed significantly lower activation levels compared to T cells from the leptomeninges and CNS parenchyma. However, they did not represent a specialized non-pathogenic cellular sub-fraction, as their gene expression profile strongly resembled that of tissue-derived T cells and they fully retained their encephalitogenic potential. T-cell detachment from the leptomeninges was counteracted by integrins VLA-4 and LFA-1 binding to their respective ligands produced by resident macrophages. Chemokine signalling via CCR5/CXCR3 and antigenic stimulation of T cells in contact with the leptomeningeal macrophages enforced their adhesiveness. T cells floating in the CSF were able to reattach to the leptomeninges through steps reminiscent of vascular adhesion in CNS blood vessels, and invade the parenchyma. The molecular/cellular conditions for T-cell reattachment were the same as the requirements for detachment from the leptomeningeal milieu. Our data indicate that the leptomeninges represent a checkpoint at which activated T cells are licensed to enter the CNS parenchyma and non-activated T cells are preferentially released into the CSF, from where they can reach areas of antigen availability and tissue damage.


Asunto(s)
Movimiento Celular , Líquido Cefalorraquídeo/citología , Encefalomielitis Autoinmune Experimental/patología , Meninges/patología , Esclerosis Múltiple/patología , Linfocitos T/patología , Traslado Adoptivo , Animales , Adhesión Celular , Líquido Cefalorraquídeo/inmunología , Quimiocinas/metabolismo , Plexo Coroideo , Colágeno/metabolismo , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/inmunología , Femenino , Integrina alfa4beta1/metabolismo , Activación de Linfocitos , Antígeno-1 Asociado a Función de Linfocito/metabolismo , Macrófagos/inmunología , Macrófagos/metabolismo , Masculino , Meninges/inmunología , Esclerosis Múltiple/inmunología , Ratas , Ratas Endogámicas Lew , Receptores CCR5/metabolismo , Receptores CXCR3/metabolismo , Linfocitos T/inmunología
13.
J Clin Invest ; 124(4): 1853-67, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24642471

RESUMEN

Members of the miR-34 family are induced by the tumor suppressor p53 and are known to inhibit epithelial-to-mesenchymal transition (EMT) and therefore presumably suppress the early phases of metastasis. Here, we determined that exposure of human colorectal cancer (CRC) cells to the cytokine IL-6 activates the oncogenic STAT3 transcription factor, which directly represses the MIR34A gene via a conserved STAT3-binding site in the first intron. Repression of MIR34A was required for IL-6-induced EMT and invasion. Furthermore, we identified the IL-6 receptor (IL-6R), which mediates IL-6-dependent STAT3 activation, as a conserved, direct miR-34a target. The resulting IL-6R/STAT3/miR-34a feedback loop was present in primary colorectal tumors as well as CRC, breast, and prostate cancer cell lines and associated with a mesenchymal phenotype. An active IL-6R/STAT3/miR-34a loop was necessary for EMT, invasion, and metastasis of CRC cell lines and was associated with nodal and distant metastasis in CRC patient samples. p53 activation in CRC cells interfered with IL-6-induced invasion and migration via miR-34a-dependent downregulation of IL6R expression. In Mir34a-deficient mice, colitis-associated intestinal tumors displayed upregulation of p-STAT3, IL-6R, and SNAIL and progressed to invasive carcinomas, which was not observed in WT animals. Collectively, our data indicate that p53-dependent expression of miR-34a suppresses tumor progression by inhibiting a IL-6R/STAT3/miR-34a feedback loop.


Asunto(s)
Neoplasias Colorrectales/genética , Neoplasias Colorrectales/metabolismo , MicroARNs/genética , MicroARNs/metabolismo , Receptores de Interleucina-6/genética , Receptores de Interleucina-6/metabolismo , Factor de Transcripción STAT3/metabolismo , Animales , Línea Celular Tumoral , Neoplasias Colorrectales/patología , Modelos Animales de Enfermedad , Transición Epitelial-Mesenquimal , Retroalimentación Fisiológica , Femenino , Humanos , Masculino , Ratones , Ratones Noqueados , Invasividad Neoplásica , Metástasis de la Neoplasia , Factores de Transcripción de la Familia Snail , Factores de Transcripción/metabolismo
14.
Nat Med ; 19(6): 784-90, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23624600

RESUMEN

Multiple sclerosis is an autoimmune disease of the central nervous system (CNS) that is initiated when self-reactive T cells enter the brain and become locally activated after encountering their specific nervous antigens. When and where the disease-relevant antigen encounters occur is unclear. Here we combined fluorescently labeled nuclear factor of activated T cells (NFAT) with histone protein H2B to create a broadly applicable molecular sensor for intravital imaging of T cell activation. In experimental autoimmune encephalomyelitis, an animal model for multiple sclerosis, we report that effector T cells entering the CNS become activated after short contacts with leptomeningeal phagocytes. During established disease, the activation process is extended to the depth of the CNS parenchyma, where the cells form contacts with microglia and recruited phagocytes. We show that it is the activation processes during the preclinical phase rather than during established disease that are essential for the intensity and duration of the disease bout.


Asunto(s)
Autoinmunidad , Encéfalo/inmunología , Histonas/fisiología , Factores de Transcripción NFATC/fisiología , Linfocitos T/inmunología , Animales , Técnicas Biosensibles , Encefalomielitis Autoinmune Experimental/inmunología , Fluorescencia , Activación de Linfocitos , Ratones , Ratones Endogámicos C57BL , Transporte de Proteínas , Ratas , Ratas Endogámicas Lew , Transducción de Señal
15.
Nature ; 488(7413): 675-9, 2012 Aug 30.
Artículo en Inglés | MEDLINE | ID: mdl-22914092

RESUMEN

The blood­brain barrier (BBB) and the environment of the central nervous system (CNS) guard the nervous tissue from peripheral immune cells. In the autoimmune disease multiple sclerosis, myelin-reactive T-cell blasts are thought to transgress the BBB and create a pro-inflammatory environment in the CNS, thereby making possible a second autoimmune attack that starts from the leptomeningeal vessels and progresses into the parenchyma. Using a Lewis rat model of experimental autoimmune encephalomyelitis, we show here that contrary to the expectations of this concept, T-cell blasts do not efficiently enter the CNS and are not required to prepare the BBB for immune-cell recruitment. Instead, intravenously transferred T-cell blasts gain the capacity to enter the CNS after residing transiently within the lung tissues. Inside the lung tissues, they move along and within the airways to bronchus-associated lymphoid tissues and lung-draining mediastinal lymph nodes before they enter the blood circulation from where they reach the CNS. Effector T cells transferred directly into the airways showed a similar migratory pattern and retained their full pathogenicity. On their way the T cells fundamentally reprogrammed their gene-expression profile, characterized by downregulation of their activation program and upregulation of cellular locomotion molecules together with chemokine and adhesion receptors. The adhesion receptors include ninjurin 1, which participates in T-cell intravascular crawling on cerebral blood vessels. We detected that the lung constitutes a niche not only for activated T cells but also for resting myelin-reactive memory T cells. After local stimulation in the lung, these cells strongly proliferate and, after assuming migratory properties, enter the CNS and induce paralytic disease. The lung could therefore contribute to the activation of potentially autoaggressive T cells and their transition to a migratory mode as a prerequisite to entering their target tissues and inducing autoimmune disease.


Asunto(s)
Encéfalo/patología , Movimiento Celular , Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/patología , Pulmón/patología , Linfocitos T/patología , Traslado Adoptivo , Animales , Autoinmunidad/inmunología , Barrera Hematoencefálica/inmunología , Encéfalo/citología , Encéfalo/inmunología , Moléculas de Adhesión Celular Neuronal/metabolismo , Circulación Cerebrovascular , Modelos Animales de Enfermedad , Perfilación de la Expresión Génica , Memoria Inmunológica , Pulmón/citología , Pulmón/inmunología , Activación de Linfocitos , Vaina de Mielina/inmunología , Factores de Crecimiento Nervioso/metabolismo , Ratas , Ratas Endogámicas Lew , Linfocitos T/citología , Linfocitos T/inmunología , Linfocitos T/metabolismo
16.
Cell Cycle ; 7(16): 2591-600, 2008 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-18719384

RESUMEN

Recently, we and others identified the microRNA miR-34a as a target of the tumor suppressor gene product p53. Ectopic miR-34a induces a G(1) cell cycle arrest, senescence and apoptosis. Here we report that miR-34a expression is silenced in several types of cancer due to aberrant CpG methylation of its promoter. 19 out of 24 (79.1%) primary prostate carcinomas displayed CpG methylation of the miR-34a promoter and concomitant loss of miR-34a expression. CpG methylation of the miR-34a promoter was also detected in breast (6/24; 25%), lung (7/24; 29.1%), colon (3/23; 13%), kidney (3/14; 21.4%), bladder (2/6; 33.3%) and pancreatic (3/19; 15.7%) carcinoma cell lines, as well as in melanoma cell lines (19/44; 43.2%) and primary melanoma (20/32 samples; 62.5%). Silencing of miR-34a was dominant over its transactivation by p53 after DNA damage. Re-expression of miR-34a in prostate and pancreas carcinoma cell lines induced senescence and cell cycle arrest at least in part by targeting CDK6. These results show that miR-34a represents a tumor suppressor gene which is inactivated by CpG methylation and subsequent transcriptional silencing in a broad range of tumors.


Asunto(s)
Islas de CpG , Metilación de ADN , Regulación Neoplásica de la Expresión Génica , Silenciador del Gen , MicroARNs/genética , Neoplasias/genética , Animales , Línea Celular Tumoral , Quinasa 6 Dependiente de la Ciclina/metabolismo , Humanos , Ratones , Regiones Promotoras Genéticas , Proteína p53 Supresora de Tumor/metabolismo
17.
Cell Cycle ; 6(13): 1586-93, 2007 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-17554199

RESUMEN

In a genome-wide screen for microRNAs regulated by the transcription factor encoded by the p53 tumor suppressor gene we found that after p53-activation the abundance of thirty-four miRNAs was significantly increased, whereas sixteen miRNAs were suppressed. The induction of miR-34a was most pronounced among all differential regulations. Also expression of the primary miR-34a transcript was induced after p53 activation and by DNA damage in a p53-dependent manner. p53 occupied an evolutionarily conserved binding site proximal to the first non-coding exon of miR-34a. Ectopic miR-34a induced apoptosis and a cell cycle arrest in the G1-phase, thereby suppressing tumor cell proliferation. Other p53-induced miRNAs identified here may also have tumor suppressive potential as they are known to suppress the anti-apoptotic factor Bcl2 (miR-15a/16) and the oncogenes RAS and HMGA2 (let-7a). Our results for the first time directly integrate the regulation of miRNA expression into the transcriptional network regulated by p53. siRNAs corresponding to p53-induced miRNAs may have potential as cancer therapeutic agents as RNA interference based therapies are currently emerging.


Asunto(s)
Apoptosis/genética , Fase G1/genética , Regulación de la Expresión Génica , MicroARNs/genética , MicroARNs/fisiología , Proteína p53 Supresora de Tumor/fisiología , Secuencia de Bases , Mapeo Cromosómico , Daño del ADN/genética , Humanos , Datos de Secuencia Molecular , Análisis de Secuencia de ARN , Células Tumorales Cultivadas
18.
Cell Cycle ; 6(3): 339-52, 2007 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-17297307

RESUMEN

Here we show that the human BubR1 and MAD2 genes, which encode inhibitors of the anaphase promoting complex (APC/C), are directly activated by the oncogenic transcription factor c-MYC via E-box sequences in their first introns. In colorectal cancer biopsies elevated expression of c-MYC correlated with increased MAD2 levels. Activation of a conditional c-MYC allele delayed progression through mitosis in pro-metaphase in a MAD2- and BubR1-dependent manner. A fraction of the daughter cells derived from extended mitotic events underwent synchronous apoptosis, which was in part mediated by BubR1. Furthermore, c-MYC activation resulted in CIN (chromosomal instability) in the diploid MIN (microsatellite instability) cell line DLD-1 and further enhanced CIN in the aneuploid CIN-line MCF7. Unexpectedly, c-MYC-induced CIN was independent of c-MYC-induced BubR1/MAD2 expression and mitotic delay. Therefore, c-MYC-induced CIN may be caused be alternative pathways. We observed that activation of c-MYC induced DNA double-strand breaks, as evidenced by formation of gamma-H2AX foci, which colocalized with foci of active DNA replication. Furthermore, c-MYC activation resulted in mitotic chromosomes exhibiting DNA damage. Therefore, oncogenic deregulation of c-MYC prevents repair of replication-stress induced DNA lesions in the G(2)-phase. We suggest that the c-MYC-mediated persistence of DNA lesions throughout mitosis leads to chromosomal missegregation and underlies c-MYC-induced CIN. The effects of deregulated c-MYC on progression through mitosis described here may have important implications for the origin of chromosomal instability in many tumor types and the sensitivity towards cancer therapeutic agents targeting DNA or the mitotic spindle.


Asunto(s)
Proteínas de Unión al Calcio/genética , Proteínas de Ciclo Celular/genética , Inestabilidad Cromosómica , Daño del ADN , Histonas/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Proteínas Proto-Oncogénicas c-myc/metabolismo , Proteínas Represoras/genética , Anafase/genética , Anafase/fisiología , Apoptosis/genética , Proteínas de Unión al Calcio/metabolismo , Proteínas de Ciclo Celular/metabolismo , Línea Celular Tumoral , Cromatina/genética , Cromatina/metabolismo , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/metabolismo , Elementos E-Box , Etopósido/metabolismo , Genes myc , Histonas/genética , Humanos , Intrones/genética , Proteínas Mad2 , Mitosis/genética , Prometafase , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Represoras/metabolismo , Activación Transcripcional
19.
Semin Cancer Biol ; 16(3): 214-24, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16698281

RESUMEN

The 14-3-3sigma gene is a direct target of the p53 tumor suppressor and its product inhibits cell cycle progression. Recently, a proteomic analysis revealed that 14-3-3sigma regulates additional cellular processes relevant to carcinogenesis, as migration and MAP-kinase signalling. The expression of 14-3-3sigma is down-regulated by CpG methylation in several types of human cancer, among them prostate, lung, breast and several types of skin cancer. The epigenetic inactivation of 14-3-3sigma occurs at an early stage of tumor development and may allow evasion from senescence and promote genomic instability. In the future the detection of CpG methylation of 14-3-3sigma may be used for diagnostic and prognostic purposes.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Exonucleasas/metabolismo , Silenciador del Gen , Proteínas de Neoplasias/metabolismo , Neoplasias/genética , Transducción de Señal/fisiología , Proteína p53 Supresora de Tumor/metabolismo , Proteínas 14-3-3 , Animales , Metilación de ADN , Modelos Animales de Enfermedad , Epigénesis Genética , Exorribonucleasas , Femenino , Histona Desacetilasas/metabolismo , Humanos , Masculino , Neoplasias/metabolismo , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/metabolismo , Unión Proteica
20.
Cell Res ; 15(4): 237-46, 2005 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15857578

RESUMEN

Cancer cells show characteristic alterations in DNA methylation patterns. Aberrant CpG methylation of specific promoters results in inactivation of tumor suppressor genes and therefore plays an important role in carcinogenesis. The p53-regulated gene 14-3-3sigma undergoes frequent epigenetic silencing in several types of cancer, including carcinoma of the breast, prostate, and skin, suggesting that the loss of 14-3-3sigma expression may be causally involved in tumor progression. Functional studies demonstrated that 14-3-3sigma is involved in cell-cycle control and prevents the accumulation of chromosomal damage. The recent identification of novel 14-3-3sigma-associated proteins by a targeted proteomics approach implies that 14-3-3sigma regulates diverse cellular processes, which may become deregulated after silencing of 14-3-3sigma expression in cancer cells.


Asunto(s)
Biomarcadores de Tumor/genética , Biomarcadores de Tumor/fisiología , Exonucleasas/genética , Exonucleasas/fisiología , Silenciador del Gen , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/fisiología , Neoplasias/genética , Proteínas 14-3-3 , Islas de CpG/genética , Daño del ADN/genética , Metilación de ADN , Progresión de la Enfermedad , Exorribonucleasas , Regulación Neoplásica de la Expresión Génica , Genes Supresores de Tumor/fisiología , Humanos , Regiones Promotoras Genéticas/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA