Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
Immun Ageing ; 18(1): 28, 2021 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-34130717

RESUMEN

MicroRNAs (miRNAs) are regulatory noncoding RNAs important for many aspects of cellular processes including cell differentiation and proliferation. Functions of numerous miRNAs have been identified in T cells, with miR-181a regulating T cell activation thresholds during thymic T cell development and during activation of peripheral T cells. Intriguingly, miR-181a is implicated in defective antiviral and vaccine responses in older individuals, as its expression declines in naïve T cells with increasing age. Here, we review the pathways that are regulated by miR-181a and that explain the unique role of miR-181a in T cell development, T cell activation and antiviral T cell responses. These studies provide a framework for understanding how a decline in miR-181a expression in T cells could contribute to age-related defects in adaptive immunity. We furthermore review the mechanisms that cause the age-related decline in miR-181a expression and discuss the potential of restoring miR-181a expression or targeting miR-181a-regulated pathways to improve impaired T cell responses in older individuals.

2.
J Biol Chem ; 288(12): 8726-8736, 2013 Mar 22.
Artículo en Inglés | MEDLINE | ID: mdl-23378536

RESUMEN

Disturbance of homeostasis at endoplasmic reticulum (ER) causes stress to cells that in turn triggers an adaptive signaling pathway termed unfolded protein response for the purpose of restoring normal cellular physiology or initiating signaling events leading to apoptosis. Identification of those genes that are involved in the unfolded protein response-mediated apoptotic signaling pathway would be valuable toward elucidating the molecular mechanism underlying the relationship between ER stress and apoptosis. We initiated a genetic screen by using the retroviral insertion mutation system to search for genes whose inactivation confers resistance to apoptosis induction by staurosporine. Using this approach, RING finger protein 13 (RNF13) was identified. Interestingly, RNF13 is highly enriched in ER. RNF13 knockdown cells are resistant to apoptosis and JNK activation triggered by ER stress. Conversely, overexpression of RNF13 induces JNK activation and caspase-dependent apoptosis. The RING and transmembrane domains of RNF13 are both required for its effects on JNK activation and apoptosis. Moreover, systematic analysis of the involvement of individual signaling components in the ER stress pathway using knockdown approach reveals that RNF13 acts upstream of the IRE1α-TRAF2 signaling axis for JNK activation and apoptosis. Finally, RNF13 co-immunoprecipitates with IRE1α, and the intact RING domain is also required for mediating its interaction. Together, our data support a model that RNF13 is a critical mediator for facilitating ER stress-induced apoptosis through the activation of the IRE1α-TRAF2-JNK signaling pathway.


Asunto(s)
Apoptosis , Estrés del Retículo Endoplásmico , Endorribonucleasas/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Ubiquitina-Proteína Ligasas/fisiología , Animales , Proteínas Reguladoras de la Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/metabolismo , Proteínas Reguladoras de la Apoptosis/fisiología , Línea Celular , Chlorocebus aethiops , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Retículo Endoplásmico/metabolismo , Activación Enzimática , Humanos , Fosforilación , Unión Proteica , Procesamiento Proteico-Postraduccional , Dominios RING Finger , Empalme del ARN , ARN Mensajero/genética , ARN Mensajero/metabolismo , Factores de Transcripción del Factor Regulador X , Transducción de Señal , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo
3.
Blood ; 120(11): 2330-9, 2012 Sep 13.
Artículo en Inglés | MEDLINE | ID: mdl-22718841

RESUMEN

CD146 is a novel endothelial biomarker and plays an essential role in angiogenesis; however, its role in the molecular mechanism underlying angiogenesis remains poorly understood. In the present study, we show that CD146 interacts directly with VEGFR-2 on endothelial cells and at the molecular level and identify the structural basis of CD146 binding to VEGFR-2. In addition, we show that CD146 is required in VEGF-induced VEGFR-2 phosphorylation, AKT/p38 MAPKs/NF-κB activation, and thus promotion of endothelial cell migration and microvascular formation. Furthermore, we show that anti-CD146 AA98 or CD146 siRNA abrogates all VEGFR-2 activation induced by VEGF. An in vivo angiogenesis assay showed that VEGF-promoted microvascular formation was impaired in the endothelial conditional knockout of CD146 (CD146(EC-KO)). Our animal experiments demonstrated that anti-CD146 (AA98) and anti-VEGF (bevacizumab) have an additive inhibitory effect on xenografted human pancreatic and melanoma tumors. The results of the present study suggest that CD146 is a new coreceptor for VEGFR-2 and is therefore a promising target for blocking tumor-related angiogenesis.


Asunto(s)
Endotelio Vascular/metabolismo , Neovascularización Patológica/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , Animales , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Antígeno CD146/química , Antígeno CD146/genética , Antígeno CD146/metabolismo , Línea Celular Tumoral , Células Cultivadas , Endotelio Vascular/efectos de los fármacos , Femenino , Humanos , Ratones , Ratones Noqueados , Ratones Desnudos , Terapia Molecular Dirigida , Proteínas Mutantes/antagonistas & inhibidores , Proteínas Mutantes/metabolismo , Neovascularización Patológica/tratamiento farmacológico , Fosforilación/efectos de los fármacos , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Interferencia de ARN , ARN Interferente Pequeño , Proteínas Recombinantes/antagonistas & inhibidores , Proteínas Recombinantes/metabolismo , Organismos Libres de Patógenos Específicos , Receptor 2 de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores
4.
Front Immunol ; 13: 911132, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35572522

RESUMEN

[This corrects the article DOI: 10.3389/fimmu.2020.585168.].

5.
EBioMedicine ; 76: 103825, 2022 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-35085847

RESUMEN

BACKGROUND: Rheumatoid arthritis (RA) is an inflammatory disease that manifests as a preclinical stage of systemic autoimmunity followed by chronic progressive synovitis. Disease-associated genetic SNP variants predominantly map to non-coding, regulatory regions of functional importance in CD4 T cells, implicating these cells as key regulators. A better understanding of the epigenome of CD4 T cells holds the promise of providing information on the interaction between genetic susceptibility and exogenous factors. METHODS: We mapped regions of chromatin accessibility using ATAC-seq in peripheral CD4 T cell subsets of patients with RA (n=18) and compared them to T cells from patients with psoriatic arthritis (n=11) and age-matched healthy controls (n=10). Transcripts of selected genes were quantified using qPCR. FINDINGS: RA-associated epigenetic signatures were identified that in part overlapped between central and effector memory CD4 T cells and that were to a lesser extent already present in naïve cells. Sites more accessible in RA were highly enriched for the motif of the transcription factor (TF) CTCF suggesting differences in the three-dimensional chromatin structure. Unexpectedly, sites with reduced chromatin accessibility were enriched for motifs of TFs pertinent for T cell function. Most strikingly, super-enhancers encompassing RA-associated SNPs were less accessible. Analysis of selected transcripts and published DNA methylation patterns were consistent with this finding. The preferential loss in accessibility at these super-enhancers was seen in patients with high and low disease activity and on a variety of immunosuppressive treatment modalities. INTERPRETATION: Disease-associated genes are epigenetically less poised to respond in CD4 T cells from patients with established RA. FUNDING: This work was supported by I01 BX001669 from the Veterans Administration.


Asunto(s)
Artritis Reumatoide , Linfocitos T CD4-Positivos , Artritis Reumatoide/genética , Cromatina/genética , Metilación de ADN , Humanos , Secuencias Reguladoras de Ácidos Nucleicos
6.
Nat Cardiovasc Res ; 1(3): 253-262, 2022 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-35990913

RESUMEN

The pleiotropic benefits of statins may result from their impact on vascular inflammation. The molecular process underlying this phenomenon is not fully elucidated. Here, RNA sequencing designed to investigate gene expression patterns following CD47-SIRPα inhibition identifies a link between statins, efferocytosis, and vascular inflammation. In vivo and in vitro studies provide evidence that statins augment programmed cell removal by inhibiting the nuclear translocation of NFκB1 p50 and suppressing the expression of the critical 'don't eat me' molecule, CD47. Statins amplify the phagocytic capacity of macrophages, and thus the anti-atherosclerotic effects of CD47-SIRPα blockade, in an additive manner. Analyses of clinical biobank specimens suggest a similar link between statins and CD47 expression in humans, highlighting the potential translational implications. Taken together, our findings identify efferocytosis and CD47 as pivotal mediators of statin pleiotropy. In turn, statins amplify the anti-atherosclerotic effects of pro-phagocytic therapies independently of any lipid-lowering effect.

7.
Cardiovasc Res ; 118(13): 2792-2804, 2022 10 21.
Artículo en Inglés | MEDLINE | ID: mdl-34849613

RESUMEN

AIMS: De-differentiation and activation of pro-inflammatory pathways are key transitions vascular smooth muscle cells (SMCs) make during atherogenesis. Here, we explored the upstream regulators of this 'atherogenic transition'. METHODS AND RESULTS: Genome-wide sequencing studies, including Assay for Transposase-Accessible Chromatin using sequencing and RNA-seq, were performed on cells isolated from both murine SMC-lineage-tracing models of atherosclerosis and human atherosclerotic lesions. At the bulk level, alterations in chromatin accessibility were associated with the atherogenic transitioning of lesional SMCs, especially in relation to genes that govern differentiation status and complement-dependent inflammation. Using computational biology, we observed that a transcription factor previously related to coronary artery disease, Activating transcription factor 3 (ATF3), was predicted to be an upstream regulator of genes altered during the transition. At the single-cell level, our results indicated that ATF3 is a key repressor of SMC transitioning towards the subset of cells that promote vascular inflammation by activating the complement cascade. The expression of ATF3 and complement component C3 was negatively correlated in SMCs from human atherosclerotic lesions, suggesting translational relevance. Phenome-wide association studies indicated that genetic variation that results in reduced expression of ATF3 is correlated with an increased risk for atherosclerosis, and the expression of ATF3 was significantly down-regulated in humans with advanced vascular disease. CONCLUSION: Our study indicates that the plasticity of atherosclerotic SMCs may in part be explained by dynamic changes in their chromatin architecture, which in turn may contribute to their maladaptive response to inflammation-induced stress.


Asunto(s)
Aterosclerosis , Músculo Liso Vascular , Humanos , Ratones , Animales , Músculo Liso Vascular/metabolismo , Cromatina/genética , Factor de Transcripción Activador 3/genética , Factor de Transcripción Activador 3/metabolismo , Miocitos del Músculo Liso/metabolismo , Aterosclerosis/metabolismo , Inflamación/metabolismo
8.
NPJ Aging Mech Dis ; 7(1): 4, 2021 Feb 08.
Artículo en Inglés | MEDLINE | ID: mdl-33558531

RESUMEN

MicroRNAs play an important role in the regulation of T cell development, activation, and differentiation. One of the most abundant microRNAs in lymphocytes is miR-181a, which controls T cell receptor (TCR) activation thresholds in thymic selection as well as in peripheral T cell responses. We previously found that miR-181a levels decline in T cells in the elderly. In this study, we identified TCF1 as a transcriptional regulator of pri-miR-181a. A decline in TCF1 levels in old individuals accounted for the reduced miR-181a expression impairing TCR signaling. Inhibition of GSK3ß restored expression of miR-181a by inducing TCF1 in T cells from old adults. GSK3ß inhibition enhanced TCR signaling to increase downstream expression of activation markers and production of IL-2. The effect involved the upregulation of miR-181a and the inhibition of DUSP6 expression. Thus, inhibition of GSK3ß can restore responses of old T cells by inducing miR-181a expression through TCF1.

9.
Nat Commun ; 12(1): 907, 2021 02 10.
Artículo en Inglés | MEDLINE | ID: mdl-33568645

RESUMEN

Rheumatoid arthritis (RA) and psoriatic arthritis (PsA) are two distinct autoimmune diseases that manifest with chronic synovial inflammation. Here, we show that CD4+ T cells from patients with RA and PsA have increased expression of the pore-forming calcium channel component ORAI3, thereby increasing the activity of the arachidonic acid-regulated calcium-selective (ARC) channel and making T cells sensitive to arachidonic acid. A similar increase does not occur in T cells from patients with systemic lupus erythematosus. Increased ORAI3 transcription in RA and PsA T cells is caused by reduced IKAROS expression, a transcriptional repressor of the ORAI3 promoter. Stimulation of the ARC channel with arachidonic acid induces not only a calcium influx, but also the phosphorylation of components of the T cell receptor signaling cascade. In a human synovium chimeric mouse model, silencing ORAI3 expression in adoptively transferred T cells from patients with RA attenuates tissue inflammation, while adoptive transfer of T cells from healthy individuals with reduced expression of IKAROS induces synovitis. We propose that increased ARC activity due to reduced IKAROS expression makes T cells more responsive and contributes to chronic inflammation in RA and PsA.


Asunto(s)
Ácido Araquidónico/inmunología , Artritis Reumatoide/inmunología , Linfocitos T CD4-Positivos/inmunología , Membrana Sinovial/inmunología , Anciano , Artritis Psoriásica/genética , Artritis Psoriásica/inmunología , Artritis Reumatoide/genética , Calcio/inmunología , Canales de Calcio/genética , Canales de Calcio/inmunología , Señalización del Calcio , Femenino , Humanos , Masculino , Persona de Mediana Edad
10.
J Clin Invest ; 131(11)2021 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-34060486

RESUMEN

With increasing age, individuals are more vulnerable to viral infections such as with influenza or the SARS-CoV-2 virus. One age-associated defect in human T cells is the reduced expression of miR-181a. miR-181ab1 deficiency in peripheral murine T cells causes delayed viral clearance after infection, resembling human immune aging. Here we show that naive T cells from older individuals as well as miR-181ab1-deficient murine T cells develop excessive replication stress after activation, due to reduced histone expression and delayed S-phase cell cycle progression. Reduced histone expression was caused by the miR-181a target SIRT1 that directly repressed transcription of histone genes by binding to their promoters and reducing histone acetylation. Inhibition of SIRT1 activity or SIRT1 silencing increased histone expression, restored cell cycle progression, diminished the replication-stress response, and reduced the production of inflammatory mediators in replicating T cells from old individuals. Correspondingly, treatment with SIRT1 inhibitors improved viral clearance in mice with miR-181a-deficient T cells after LCMV infection. In conclusion, SIRT1 inhibition may be beneficial to treat systemic viral infection in older individuals by targeting antigen-specific T cells that develop replication stress due to miR-181a deficiency.


Asunto(s)
COVID-19/inmunología , Senescencia Celular/inmunología , Histonas/deficiencia , MicroARNs/inmunología , SARS-CoV-2/inmunología , Linfocitos T/inmunología , Animales , COVID-19/genética , Senescencia Celular/genética , Femenino , Histonas/inmunología , Humanos , Masculino , Ratones Noqueados , MicroARNs/genética , SARS-CoV-2/genética , Sirtuina 1/genética , Sirtuina 1/inmunología
11.
Am J Physiol Gastrointest Liver Physiol ; 298(5): G784-94, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20224008

RESUMEN

The ability of Salmonella typhimurium to enter intestinal epithelial cells constitutes a crucial step in pathogenesis. Salmonella invasion of the intestinal epithelium requires bacterial type three secretion system. Type three secretion system is a transport device that injects virulence proteins, called effectors, to paralyze or reprogram the eukaryotic cells. Avirulence factor for Salmonella (AvrA) is a Salmonella effector that inhibits the host's inflammatory responses. The mechanism by which AvrA modulates host cell signaling is not entirely clear. p53 is situated at the crossroads of a network of signaling pathways that are essential for genotoxic and nongenotoxic stress responses. We hypothesized that Salmonella infection activates the p53 pathway. We demonstrated that Salmonella infection increased p53 acetylation. Cells infected with AvrA-sufficient Salmonella have increased p53 acetylation, whereas cells infected with AvrA-deficient Salmonella have less p53 acetylation. In a cell-free system, AvrA possessed acetyltransferase activity and used p53 as a substrate. AvrA expression increased p53 transcriptional activity and induced cell cycle arrest. HCT116 p53-/- cells had less inflammatory responses. In a mouse model of Salmonella infection, intestinal epithelial p53 acetylation was increased by AvrA expression. Our studies provide novel mechanistic evidence that Salmonella modulates the p53 pathway during intestinal inflammation and infection.


Asunto(s)
Proteínas Bacterianas/metabolismo , Mucosa Intestinal/metabolismo , Infecciones por Salmonella/fisiopatología , Salmonella typhimurium , Proteína p53 Supresora de Tumor/metabolismo , Acetilación , Animales , Ciclo Celular/efectos de los fármacos , Células Cultivadas , Femenino , Expresión Génica/efectos de los fármacos , Células HeLa , Humanos , Mucosa Intestinal/citología , Ratones , Factor de Necrosis Tumoral alfa/farmacología
12.
Front Immunol ; 11: 585168, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33262764

RESUMEN

Healthy immune aging is in part determined by how well the sizes of naïve T cell compartments are being maintained with advancing age. Throughout adult life, replenishment largely derives from homeostatic proliferation of existing naïve and memory T cell populations. However, while the subpopulation composition of CD4 T cells is relatively stable, the CD8 T cell compartment undergoes more drastic changes with loss of naïve CD8 T cells and accumulation of effector T cells, suggesting that CD4 T cells are more resilient to resist age-associated changes. To determine the epigenetic basis for these differences in behaviors, we compared chromatin accessibility maps of CD4 and CD8 T cell subsets from young and old individuals and related the results to the expressed transcriptome. The dominant age-associated signatures resembled hallmarks of differentiation, which were more pronounced for CD8 naïve and memory than the corresponding CD4 T cell subsets, indicating that CD8 T cells are less able to keep cellular quiescence upon homeostatic proliferation. In parallel, CD8 T cells from old adults, irrespective of their differentiation state, displayed greater reduced accessibility to genes of basic cell biological function, including genes encoding ribosomal proteins. One possible mechanism is the reduced expression of the transcription factors YY1 and NRF1. Our data suggest that chromatin accessibility signatures can be identified that distinguish CD4 and CD8 T cells from old adults and that may confer the higher resilience of CD4 T cells to aging.


Asunto(s)
Envejecimiento/inmunología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Inmunosenescencia/inmunología , Adulto , Anciano , Envejecimiento/genética , Epigénesis Genética/genética , Epigénesis Genética/inmunología , Femenino , Humanos , Memoria Inmunológica/genética , Memoria Inmunológica/inmunología , Inmunosenescencia/genética , Masculino , Transcriptoma/genética , Transcriptoma/inmunología
13.
Biochim Biophys Acta ; 1780(12): 1383-7, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18708124

RESUMEN

Severe acute respiratory syndrome (SARS) coronavirus (CoV) spread from China to more than 30 countries, causing severe outbreaks of atypical pneumonia and over 800 deaths worldwide. CoV primarily infects the upper respiratory and gastrointestinal tract; however, SARS-CoV has a unique pathogenesis because it infects both the upper and lower respiratory tracts and leads to human respiratory diseases. SARS-CoV genome has shown containing 14 open reading frames (ORFs) and 8 of them encode novel proteins. Previous reports show that overexpression of ORF-3a, ORF-3b and ORF-7a induce apoptosis. In this report, we demonstrate that overexpression of ORF-6 also induces apoptosis and that Caspase-3 inhibitor and JNK inhibitor block ORF-6 induced apoptosis. Importantly, the protein level of ER chaperon protein, GRP94, was up-regulated when ORF-6 was overexpressed. All these data suggest that ORF-6 induces apoptosis via Caspase-3 mediated, ER stress and JNK-dependent pathways.


Asunto(s)
Apoptosis , Caspasa 3/fisiología , Retículo Endoplásmico/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/fisiología , Proteínas Virales/biosíntesis , Animales , Chlorocebus aethiops , Activación Enzimática , Proteínas HSP70 de Choque Térmico/biosíntesis , Humanos , Proteínas Quinasas JNK Activadas por Mitógenos/antagonistas & inhibidores , Proteínas de la Membrana/biosíntesis , Chaperonas Moleculares/biosíntesis , Oligopéptidos/farmacología , Regulación hacia Arriba , Proteínas de la Matriz Viral/antagonistas & inhibidores , Proteínas de la Matriz Viral/fisiología , Proteínas Virales/antagonistas & inhibidores
14.
Aging Cell ; 18(4): e12957, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31264370

RESUMEN

With reduced thymic activity, the population of naïve T cells in humans is maintained by homeostatic proliferation throughout adult life. In young adults, naïve CD4 T cells have enormous proliferative potential and plasticity to differentiate into different lineages. Here, we explored whether naïve CD4 T-cell aging is associated with a partial loss of this unbiased multipotency. We find that naïve CD4 T cells from older individuals have developed a propensity to develop into TH9 cells. Two major mechanisms contribute to this predisposition. First, responsiveness to transforming growth factor ß (TGFß) stimulation is enhanced with age due to an upregulation of the TGFßR3 receptor that results in increased expression of the transcription factor PU.1. Secondly, aged naïve CD4 T cells display altered transcription factor profiles in response to T-cell receptor stimulation, including enhanced expression of BATF and IRF4 and reduced expression of ID3 and BCL6. These transcription factors are involved in TH9 differentiation as well as IL9 transcription suggesting that the aging-associated changes in the transcription factor profile favor TH9 commitment.


Asunto(s)
Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/metabolismo , Linfocitos T CD4-Positivos/metabolismo , Diferenciación Celular/genética , Proteínas Inhibidoras de la Diferenciación/metabolismo , Factores Reguladores del Interferón/metabolismo , Proteínas de Neoplasias/metabolismo , Proteínas Proto-Oncogénicas c-bcl-6/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Transactivadores/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/genética , Donantes de Sangre , Femenino , Células HEK293 , Voluntarios Sanos , Humanos , Proteínas Inhibidoras de la Diferenciación/genética , Factores Reguladores del Interferón/genética , Interleucina-9/metabolismo , Masculino , Persona de Mediana Edad , Proteínas de Neoplasias/genética , Proteoglicanos/genética , Proteoglicanos/metabolismo , Proteínas Proto-Oncogénicas c-bcl-6/genética , Receptores de Antígenos de Linfocitos T/metabolismo , Receptores de Factores de Crecimiento Transformadores beta/genética , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Linfocitos T Colaboradores-Inductores/metabolismo , Transfección , Adulto Joven
15.
Nat Commun ; 9(1): 3060, 2018 08 03.
Artículo en Inglés | MEDLINE | ID: mdl-30076309

RESUMEN

MicroRNAs have emerged as key regulators in T cell development, activation, and differentiation, with miR-181a having a prominent function. By targeting several signaling pathways, miR-181a is an important rheostat controlling T cell receptor (TCR) activation thresholds in thymic selection as well as peripheral T cell responses. A decline in miR-181a expression, due to reduced transcription of pri-miR-181a, accounts for T cell activation defects that occur with older age. Here we examine the transcriptional regulation of miR-181a expression and find a putative pri-miR-181a enhancer around position 198,904,300 on chromosome 1, which is regulated by a transcription factor complex including YY1. The decline in miR-181a expression correlates with reduced transcription of YY1 in older individuals. Partial silencing of YY1 in T cells from young individuals reproduces the signaling defects seen in older T cells. In conclusion, YY1 controls TCR signaling by upregulating miR-181a and dampening negative feedback loops mediated by miR-181a targets.


Asunto(s)
MicroARNs/metabolismo , Linfocitos T/citología , Adulto , Anciano , Anciano de 80 o más Años , Diferenciación Celular/genética , Senescencia Celular/genética , Cromosomas Humanos Par 1 , Células HEK293 , Humanos , Activación de Linfocitos , MicroARNs/genética , Transducción de Señal , Linfocitos T/metabolismo , Regulación hacia Arriba , Factor de Transcripción YY1/metabolismo , Adulto Joven
16.
Sci Immunol ; 2(8)2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-28439570

RESUMEN

The efficacy of the adaptive immune response declines dramatically with age, but the cell-intrinsic mechanisms driving immune aging in humans remain poorly understood. Immune aging is characterized by a loss of self-renewing naïve cells and the accumulation of differentiated but dysfunctional cells within the CD8 T cell compartment. Using ATAC-seq, we inferred the transcription factor binding activities correlated with naive and central and effector memory CD8 T cell states in young adults. Integrating our results with RNA-seq, we identified transcription networks associated with CD8 T cell differentiation, with prominent roles implicated for BATF, ETS1, Eomes, and Sp1. Extending our analysis to aged humans, we found that the differences between the memory and naive subsets were largely preserved across age, but that naive and central memory cells from older individuals exhibited a shift toward more differentiated patterns of chromatin openness. Additionally, aged naive cells displayed a loss in chromatin accessibility at gene promoters, largely associated with a decrease in NRF1 binding. This shift was implicated in a marked drop-off in the ability of the aged naive cells to transcribe respiratory chain genes, which may explain the reduced capacity of oxidative phosphorylation in older naïve cells. Our findings identify BATF- and NRF1-driven gene regulation as potential targets for delaying CD8 T cell aging and restoring function.

17.
Oncotarget ; 7(26): 40704-40718, 2016 Jun 28.
Artículo en Inglés | MEDLINE | ID: mdl-27302922

RESUMEN

Cancer stemness drives tumor progression and drug resistance, representing a challenge to cancer eradication. Compelling evidence indicates that cancer cells can reenter the stem cell state due to the reprogramming of self-renewal machinery. Here, we show that CD146 knockdown induces stem cell properties in colorectal cancer (CRC) cells through activating canonical Wnt signaling. shRNA-mediated CD146 knockdown in CRC cells facilitates tumor initiation in serial xenotransplantation experiments. Moreover, upon CD146 knockdown, CRC cells show elevated expression of specific cancer stem cell (CSC) markers, increased sphere and clone formation as well as drug resistance in vitro. Mechanistically, our findings provide evidence that CD146 expression negatively correlates with canonical Wnt/ß-catenin activity in CRC cell lines and primary CRC specimens. Knockdown of CD146 results in inhibition of NF-κB/p65-initiated GSK-3ß expression, subsequently promoting nuclear translocation and activation of ß-catenin, and as a consequence restoring stem cell phenotypes in differentiated CRC cells. Together, our data strongly suggest that CD146 functions as a suppressor of tumorigenesis and cancer stemness in CRC through inactivating the canonical Wnt/ß-catenin cascade. Our findings provide important insights into stem cell plasticity and the multifunctional role of CD146 in CRC progression.


Asunto(s)
Neoplasias Colorrectales/metabolismo , Vía de Señalización Wnt , Transporte Activo de Núcleo Celular , Adulto , Animales , Biomarcadores de Tumor/metabolismo , Antígeno CD146/metabolismo , Línea Celular Tumoral , Neoplasias Colorrectales/patología , Progresión de la Enfermedad , Femenino , Regulación Neoplásica de la Expresión Génica , Glucógeno Sintasa Quinasa 3/metabolismo , Humanos , Masculino , Ratones , Ratones Endogámicos NOD , Ratones SCID , Persona de Mediana Edad , Trasplante de Neoplasias , Células Madre Neoplásicas/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Fenotipo , ARN Interferente Pequeño/metabolismo
18.
Cell Rep ; 14(5): 1218-1231, 2016 Feb 09.
Artículo en Inglés | MEDLINE | ID: mdl-26832412

RESUMEN

In an immune response, CD4(+) T cells expand into effector T cells and then contract to survive as long-lived memory cells. To identify age-associated defects in memory cell formation, we profiled activated CD4(+) T cells and found an increased induction of the ATPase CD39 with age. CD39(+) CD4(+) T cells resembled effector T cells with signs of metabolic stress and high susceptibility to undergo apoptosis. Pharmacological inhibition of ATPase activity dampened effector cell differentiation and improved survival, suggesting that CD39 activity influences T cell fate. Individuals carrying a low-expressing CD39 variant responded better to vaccination with an increase in vaccine-specific memory T cells. Increased inducibility of CD39 after activation may contribute to the impaired vaccine response with age.


Asunto(s)
Envejecimiento/inmunología , Antígenos CD/metabolismo , Apirasa/metabolismo , Linfocitos T CD4-Positivos/citología , Linfocitos T CD4-Positivos/inmunología , Activación de Linfocitos/inmunología , Adenosina Trifosfatasas/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Apoptosis , Proliferación Celular , Supervivencia Celular , Células Clonales , Humanos , Persona de Mediana Edad , Estrés Fisiológico , Adulto Joven
19.
Cell Res ; 25(3): 275-87, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25656845

RESUMEN

Angiogenesis, a process that newly-formed blood vessels sprout from pre-existing ones, is vital for vertebrate development and adult homeostasis. Previous studies have demonstrated that the neuronal guidance molecule netrin-1 participates in angiogenesis and morphogenesis of the vascular system. Netrin-1 exhibits dual activities in angiogenesis: either promoting or inhibiting angiogenesis. The anti-angiogenic activity of netrin-1 is mediated by UNC5B receptor. However, how netrin-1 promotes angiogenesis remained unclear. Here we report that CD146, an endothelial transmembrane protein of the immunoglobulin superfamily, is a receptor for netrin-1. Netrin-1 binds to CD146 with high affinity, inducing endothelial cell activation and downstream signaling in a CD146-dependent manner. Conditional knockout of the cd146 gene in the murine endothelium or disruption of netrin-CD146 interaction by a specific anti-CD146 antibody blocks or reduces netrin-1-induced angiogenesis. In zebrafish embryos, downregulating either netrin-1a or CD146 results in vascular defects with striking similarity. Moreover, knocking down CD146 blocks ectopic vascular sprouting induced by netrin-1 overexpression. Together, our data uncover CD146 as a previously unknown receptor for netrin-1 and also reveal a functional ligand for CD146 in angiogenesis, demonstrating the involvement of netrin-CD146 signaling in angiogenesis during vertebrate development.


Asunto(s)
Neovascularización Fisiológica/fisiología , Factores de Crecimiento Nervioso/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Animales , Antígeno CD146/genética , Antígeno CD146/metabolismo , Línea Celular , Movimiento Celular , Proliferación Celular , Células HEK293 , Células Endoteliales de la Vena Umbilical Humana/fisiología , Humanos , Ratones , Ratones Transgénicos , Neovascularización Fisiológica/genética , Factores de Crecimiento Nervioso/genética , Receptores de Netrina , Netrina-1 , Unión Proteica , Interferencia de ARN , ARN Interferente Pequeño , Receptores de Superficie Celular/genética , Proteínas Supresoras de Tumor/genética , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , Pez Cebra , Proteínas de Pez Cebra
20.
J Proteomics ; 103: 153-69, 2014 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-24704855

RESUMEN

The cell adhesion molecule CD146 is a novel inducer of epithelial-mesenchymal transition (EMT), which was associated with triple-negative breast cancer (TNBC). To gain insights into the complex networks that mediate CD146-induced EMT in breast cancers, we conducted a triple Stable Isotope Labeling with Amino Acids in Cell Culture (SILAC), to analyze whole cell protein profiles of MCF-7 cells that had undergone gradual EMT upon CD146 expression from moderate to high levels. In this study, we identified 2293 proteins in total, of which 103 exhibited changes in protein abundance that correlated with CD146 expression levels, revealing extensive morphological and biochemical changes associated with EMT. Ingenuity Pathway Analysis (IPA) showed that estrogen receptor (ER) was the most significantly inhibited transcription regulator during CD146-induced EMT. Functional assays further revealed that ER-α expression was repressed in cells undergoing CD146-induced EMT, whereas re-expression of ER-α abolished their migratory and invasive behavior. Lastly, we found that ER-α mediated its effects on CD146-induced EMT via repression of the key EMT transcriptional factor Slug. Our study revealed the molecular details of the complex signaling networks during CD146-induced EMT, and provided important clues for future exploration of the mechanisms underlying the association between CD146 and TNBC as observed in the clinic. BIOLOGICAL SIGNIFICANCE: This study used a proteomics screen to reveal molecular changes mediated by CD146-induced epithelial-mesenchymal transition (EMT) in breast cancer cells. Estrogen receptor (ER) was found to be the most significantly inhibited transcription regulator, which mediated its effects on CD146-induced EMT via repression of the transcriptional factor Slug. Elucidation of protein interaction networks and signal networks generated from 103 significantly changed proteins would facilitate future investigation into the mechanisms underlying CD146 induced-EMT in breast cancers.


Asunto(s)
Transición Epitelial-Mesenquimal , Receptor alfa de Estrógeno/fisiología , Neoplasias de la Mama Triple Negativas/fisiopatología , Antígeno CD146/fisiología , Línea Celular Tumoral , Transición Epitelial-Mesenquimal/efectos de los fármacos , Receptor alfa de Estrógeno/biosíntesis , Femenino , Humanos , Marcaje Isotópico , ARN Mensajero/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA