Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 82
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Nat Immunol ; 18(1): 54-63, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27721430

RESUMEN

Genes and pathways in which inactivation dampens tissue inflammation present new opportunities for understanding the pathogenesis of common human inflammatory diseases, including inflammatory bowel disease, rheumatoid arthritis and multiple sclerosis. We identified a mutation in the gene encoding the deubiquitination enzyme USP15 (Usp15L749R) that protected mice against both experimental cerebral malaria (ECM) induced by Plasmodium berghei and experimental autoimmune encephalomyelitis (EAE). Combining immunophenotyping and RNA sequencing in brain (ECM) and spinal cord (EAE) revealed that Usp15L749R-associated resistance to neuroinflammation was linked to dampened type I interferon responses in situ. In hematopoietic cells and in resident brain cells, USP15 was coexpressed with, and functionally acted together with the E3 ubiquitin ligase TRIM25 to positively regulate type I interferon responses and to promote pathogenesis during neuroinflammation. The USP15-TRIM25 dyad might be a potential target for intervention in acute or chronic states of neuroinflammation.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Encefalomielitis Autoinmune Experimental/inmunología , Malaria Cerebral/inmunología , Inflamación Neurogénica/inmunología , Factores de Transcripción/metabolismo , Proteasas Ubiquitina-Específicas/metabolismo , Animales , Proteínas de Unión al ADN/genética , Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Células HEK293 , Humanos , Inmunidad Innata , Interferón Tipo I/metabolismo , Malaria Cerebral/tratamiento farmacológico , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Transgénicos , Terapia Molecular Dirigida , Glicoproteína Mielina-Oligodendrócito/inmunología , Inflamación Neurogénica/tratamiento farmacológico , Fragmentos de Péptidos/inmunología , Plasmodium berghei/inmunología , Factores de Transcripción/genética , Proteasas Ubiquitina-Específicas/genética
3.
Int J Mol Sci ; 24(8)2023 Apr 13.
Artículo en Inglés | MEDLINE | ID: mdl-37108368

RESUMEN

Aggressive tumors evade cytotoxic T lymphocytes by suppressing MHC class-I (MHC-I) expression that also compromises tumor responsiveness to immunotherapy. MHC-I defects strongly correlate to defective expression of NLRC5, the transcriptional activator of MHC-I and antigen processing genes. In poorly immunogenic B16 melanoma cells, restoring NLRC5 expression induces MHC-I and elicits antitumor immunity, raising the possibility of using NLRC5 for tumor immunotherapy. As the clinical application of NLRC5 is constrained by its large size, we examined whether a smaller NLRC5-CIITA fusion protein, dubbed NLRC5-superactivator (NLRC5-SA) as it retains the ability to induce MHC-I, could be used for tumor growth control. We show that stable NLRC5-SA expression in mouse and human cancer cells upregulates MHC-I expression. B16 melanoma and EL4 lymphoma tumors expressing NLRC5-SA are controlled as efficiently as those expressing full-length NLRC5 (NLRC5-FL). Comparison of MHC-I-associated peptides (MAPs) eluted from EL4 cells expressing NLRC5-FL or NLRC5-SA and analyzed by mass spectrometry revealed that both NLRC5 constructs expanded the MAP repertoire, which showed considerable overlap but also included a substantial proportion of distinct peptides. Thus, we propose that NLRC5-SA, with its ability to increase tumor immunogenicity and promote tumor growth control, could overcome the limitations of NLRC5-FL for translational immunotherapy applications.


Asunto(s)
Regulación de la Expresión Génica , Melanoma Experimental , Humanos , Animales , Ratones , Melanoma Experimental/genética , Melanoma Experimental/terapia , Genes MHC Clase I , Antígenos de Histocompatibilidad Clase I , Presentación de Antígeno , Péptidos y Proteínas de Señalización Intracelular/genética
4.
PLoS Biol ; 17(9): e3000451, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31525189

RESUMEN

Nucleotide-binding, leucine-rich repeat containing X1 (NLRX1) is a mitochondria-located innate immune sensor that inhibits major pro-inflammatory pathways such as type I interferon and nuclear factor-κB signaling. We generated a novel, spontaneous, and rapidly progressing mouse model of multiple sclerosis (MS) by crossing myelin-specific T-cell receptor (TCR) transgenic mice with Nlrx1-/- mice. About half of the resulting progeny developed spontaneous experimental autoimmune encephalomyelitis (spEAE), which was associated with severe demyelination and inflammation in the central nervous system (CNS). Using lymphocyte-deficient mice and a series of adoptive transfer experiments, we demonstrate that genetic susceptibility to EAE lies within the innate immune compartment. We show that NLRX1 inhibits the subclinical stages of microglial activation and prevents the generation of neurotoxic astrocytes that induce neuronal and oligodendrocyte death in vitro. Moreover, we discovered several mutations within NLRX1 that run in MS-affected families. In summary, our findings highlight the importance of NLRX1 in controlling the early stages of CNS inflammation and preventing the onset of spontaneous autoimmunity.


Asunto(s)
Encefalomielitis Autoinmune Experimental/etiología , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/fisiología , Adulto , Animales , Astrocitos/fisiología , Estudios de Casos y Controles , Sistema Nervioso Central/patología , Codón sin Sentido , Enfermedades Desmielinizantes , Encefalomielitis Autoinmune Experimental/patología , Femenino , Humanos , Inmunidad Innata , Masculino , Ratones Transgénicos , Persona de Mediana Edad , Mutación Missense , Adulto Joven
5.
Int J Mol Sci ; 23(22)2022 Nov 16.
Artículo en Inglés | MEDLINE | ID: mdl-36430600

RESUMEN

Naïve CD8+ T lymphocytes exposed to certain inflammatory cytokines undergo proliferation and display increased sensitivity to antigens. Such 'cytokine priming' can promote the activation of potentially autoreactive and antitumor CD8+ T cells by weak tissue antigens and tumor antigens. To elucidate the molecular mechanisms of cytokine priming, naïve PMEL-1 TCR transgenic CD8+ T lymphocytes were stimulated with IL-15 and IL-21, and chromatin accessibility was assessed using the assay for transposase-accessible chromatin (ATAC) sequencing. PMEL-1 cells stimulated by the cognate antigenic peptide mgp10025-33 served as controls. Cytokine-primed cells showed a limited number of opening and closing chromatin accessibility peaks compared to antigen-stimulated cells. However, the ATACseq peaks in cytokine-primed cells substantially overlapped with those of antigen-stimulated cells and mapped to several genes implicated in T cell signaling, activation, effector differentiation, negative regulation and exhaustion. Nonetheless, the expression of most of these genes was remarkably different between cytokine-primed and antigen-stimulated cells. In addition, cytokine priming impacted the expression of several genes following antigen stimulation in a synergistic or antagonistic manner. Our findings indicate that chromatin accessibility changes in cytokine-primed naïve CD8+ T cells not only underlie their increased antigen responsiveness but may also enhance their functional fitness by reducing exhaustion without compromising regulatory controls.


Asunto(s)
Linfocitos T CD8-positivos , Citocinas , Citocinas/metabolismo , Cromatina/genética , Cromatina/metabolismo , Activación de Linfocitos , Antígenos/metabolismo
6.
Int J Mol Sci ; 22(4)2021 Feb 17.
Artículo en Inglés | MEDLINE | ID: mdl-33671123

RESUMEN

The immune system constantly monitors the emergence of cancerous cells and eliminates them. CD8+ cytotoxic T lymphocytes (CTLs), which kill tumor cells and provide antitumor immunity, select their targets by recognizing tumor antigenic peptides presented by MHC class-I (MHC-I) molecules. Cancer cells circumvent immune surveillance using diverse strategies. A key mechanism of cancer immune evasion is downregulation of MHC-I and key proteins of the antigen processing and presentation machinery (APM). Even though impaired MHC-I expression in cancers is well-known, reversing the MHC-I defects remains the least advanced area of tumor immunology. The discoveries that NLRC5 is the key transcriptional activator of MHC-I and APM genes, and genetic lesions and epigenetic modifications of NLRC5 are the most common cause of MHC-I defects in cancers, have raised the hopes for restoring MHC-I expression. Here, we provide an overview of cancer immunity mediated by CD8+ T cells and the functions of NLRC5 in MHC-I antigen presentation pathways. We describe the impressive advances made in understanding the regulation of NLRC5 expression, the data supporting the antitumor functions of NLRC5 and a few reports that argue for a pro-tumorigenic role. Finally, we explore the possible avenues of exploiting NLRC5 for cancer immunotherapy.


Asunto(s)
Genes MHC Clase I , Inmunoterapia , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Neoplasias/inmunología , Neoplasias/terapia , Transactivadores/metabolismo , Animales , Antígenos de Neoplasias/metabolismo , Humanos
7.
Cytokine ; 136: 155256, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32866898

RESUMEN

The COVID-19 pandemic has rapidly spread around the world with significant morbidity and mortality in a subset of patients including the elderly. The poorer outcomes are associated with 'cytokine storm-like' immune responses, otherwise referred to as 'hyperinflammation'. While most of the infected individuals show minimal or no symptoms and recover spontaneously, a small proportion of the patients exhibit severe symptoms characterized by extreme dyspnea and low tissue oxygen levels, with extensive damage to the lungs referred to as acute respiratory distress symptom (ARDS). The consensus is that the hyperinflammatory response of the host is akin to the cytokine storm observed during sepsis and is the major cause of death. Uncertainties remain on the factors that lead to hyperinflammatory response in some but not all individuals. Hyperinflammation is a common feature in different viral infections such as dengue where existing low-titer antibodies to the virus enhances the infection in immune cells through a process called antibody-dependent enhancement or ADE. ADE has been reported following vaccination or secondary infections with other corona, Ebola and dengue virus. Detailed analysis has shown that antibodies to any viral epitope can induce ADE when present in sub-optimal titers or is of low affinity. In this review we will discuss ADE in the context of dengue and coronavirus infections including Covid-19.


Asunto(s)
Acrecentamiento Dependiente de Anticuerpo/inmunología , Infecciones por Coronavirus/inmunología , Infecciones por Coronavirus/veterinaria , Peritonitis Infecciosa Felina/inmunología , Inflamación/patología , Pandemias/veterinaria , Neumonía Viral/inmunología , Neumonía Viral/veterinaria , Dengue Grave/inmunología , Animales , COVID-19 , Gatos , Citocinas/metabolismo
8.
Cytokine ; 136: 155258, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32919253

RESUMEN

Interleukin-15 (IL-15) is a member of the IL-2 family of cytokines, which use receptor complexes containing the common gamma (γc) chain for signaling. IL-15 plays important roles in innate and adaptative immune responses and is implicated in the pathogenesis of several immune diseases. The IL-15 receptor consists of 3 subunits namely, the ligand-binding IL-15Rα chain, the ß chain (also used by IL-2) and the γc chain. IL-15 uses a unique signaling pathway whereby IL-15 associates with IL-15Rα during biosynthesis, and this complex is 'trans-presented' to responder cells that expresses the IL-2/15Rßγc receptor complex. IL-15 is subject to post-transcriptional and post-translational regulation, and evidence also suggests that IL-15 cis-signaling can occur under certain conditions. IL-15 has been implicated in the pathology of various autoimmune diseases such as rheumatoid arthritis, autoimmune diabetes, inflammatory bowel disease, coeliac disease and psoriasis. Studies with pre-clinical models have shown the beneficial effects of targeting IL-15 signaling in autoimmunity. Unlike therapies targeting other cytokines, anti-IL-15 therapies have not yet been successful in humans. We discuss the complexities of IL-15 signaling in autoimmunity and explore potential immunotherapeutic approaches to target the IL-15 signaling pathway.


Asunto(s)
Enfermedades Autoinmunes/inmunología , Autoinmunidad , Interleucina-15/inmunología , Transducción de Señal/inmunología , Animales , Humanos
9.
BMC Cancer ; 20(1): 774, 2020 Aug 17.
Artículo en Inglés | MEDLINE | ID: mdl-32807134

RESUMEN

BACKGROUND: SOCS1 and SOCS3 genes are considered tumor suppressors in hepatocellular carcinoma (HCC) due to frequent epigenetic repression. Consistent with this notion, mice lacking SOCS1 or SOCS3 show increased susceptibility to diethylnitrosamine (DEN)-induced HCC. As SOCS1 and SOCS3 are important regulators of cytokine and growth factor signaling, their loss could activate oncogenic signaling pathways. Therefore, we examined the correlation between SOCS1/SOCS3 and key oncogenic signaling pathway genes as well as their prognostic significance in HCC. METHODS: The Cancer Genome Atlas dataset on HCC comprising clinical and transcriptomic data was retrieved from the cBioportal platform. The correlation between the expression of SOCS1 or SOCS3 and oncogenic pathway genes was evaluated using the GraphPad PRISM software. The inversely correlated genes were assessed for their impact on patient survival using the UALCAN platform and their expression quantified in the regenerating livers and DEN-induced HCC tissues of mice lacking Socs1 or Socs3. Finally, the Cox proportional hazards model was used to evaluate the predictive potential of SOCS1 and SOCS3 when combined with the genes of select oncogenic signaling pathways. RESULTS: SOCS1 expression was comparable between HCC and adjacent normal tissues, yet higher SOCS1 expression predicted favorable prognosis. In contrast, SOCS3 expression was significantly low in HCC, yet it lacked predictive potential. The correlation between SOCS1 or SOCS3 expression and key genes of the cell cycle, receptor tyrosine kinase, growth factor and MAPK signaling pathways were mostly positive than negative. Among the negatively correlated genes, only a few showed elevated expression in HCC and predicted survival. Many PI3K pathway genes showed mutual exclusivity with SOCS1 and/or SOCS3 and displayed independent predictive ability. Among genes that negatively correlated with SOCS1 and/or SOCS3, only CDK2 and AURKA showed corresponding modulations in the regenerating livers and DEN-induced tumors of hepatocyte-specific Socs1 or Socs3 deficient mice and predicted patient survival. The Cox proportional hazards model identified the combinations of SOCS1 or SOCS3 with CXCL8 and DAB2 as highly predictive. CONCLUSIONS: SOCS1 expression in HCC has an independent prognostic value whereas SOCS3 expression does not. The predictive potential of SOCS1 expression is increased when combined with other oncogenic signaling pathway genes.


Asunto(s)
Biomarcadores de Tumor/genética , Carcinoma Hepatocelular/genética , Neoplasias Hepáticas/genética , Proteína 1 Supresora de la Señalización de Citocinas/genética , Proteína 3 Supresora de la Señalización de Citocinas/genética , Animales , Biomarcadores de Tumor/metabolismo , Carcinoma Hepatocelular/mortalidad , Carcinoma Hepatocelular/patología , Línea Celular Tumoral , Conjuntos de Datos como Asunto , Dietilnitrosamina/toxicidad , Regulación Neoplásica de la Expresión Génica , Hepatocitos , Humanos , Estimación de Kaplan-Meier , Neoplasias Hepáticas/mortalidad , Neoplasias Hepáticas/patología , Neoplasias Hepáticas Experimentales/genética , Neoplasias Hepáticas Experimentales/patología , Masculino , Ratones Noqueados , Valor Predictivo de las Pruebas , Pronóstico , RNA-Seq , Transducción de Señal/genética , Proteína 1 Supresora de la Señalización de Citocinas/metabolismo , Proteína 3 Supresora de la Señalización de Citocinas/metabolismo
10.
Cytokine ; 124: 154608, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-30563739

RESUMEN

This special issue on 'Cytokines in Liver Diseases' was inspired by many talks and presentations on liver cancer during the 2nd Aegean Conference on Cytokine Signaling in Cancer (ACCSC) held at Heraklion, Crete, Greece on May 30-June 04, 2017 (Cytokine 2018, 108: 225-231). The liver is the biggest blood filtration and detoxification unit, and is a vital metabolic organ. Being constantly exposed to potentially harmful dietary chemicals, drugs, alcohol abuse and pathogens, the liver displays an extraordinary capacity to repair tissue damage and to regenerate. Moreover, only a healthy liver can provide the vast majority of plasma proteins, plus serving as a key organ for body homeostasis and metabolic fitness. Occasionally, the liver may have to deal with chronic damage inflicted by hepatotropic infections such as hepatitis viruses and metabolic derangements caused by obesity and the consequent metabolic syndrome. Overwhelming the natural defenses of the liver can compromise its vital functions and this can lead to more severe liver disease such as fibrosis that may progress towards cirrhosis and eventually to hepatocellular carcinoma (HCC). However, in obesity, a worldwide crisis that has been developing during the last few decades, HCC can develop in the fatty liver bypassing the fibrosis and cirrhosis stages. With the availability of effective therapies and vaccination strategies for hepatitis viruses, over nutrition has become the biggest new threat for a healthy liver. Cytokines and chemokines play a key role in the initiation and perpetuation of acute and chronic injury to the liver, and thus they contribute to most liver pathologies. The topic of cytokines in liver diseases is so vast that it cannot be adequately covered in this special issue. However, we have attempted to provide a glimpse of hot topics through comprehensive reviews and a few accompanying original articles on key research areas.


Asunto(s)
Carcinoma Hepatocelular/metabolismo , Citocinas/metabolismo , Hepatopatías/metabolismo , Hígado/metabolismo , Hígado Graso/genética , Hígado Graso/metabolismo , Hígado Graso/virología , Hepatitis Viral Humana/metabolismo , Humanos , Inmunidad Innata , Hígado/inmunología , Cirrosis Hepática/genética , Cirrosis Hepática/inmunología , Cirrosis Hepática/metabolismo , Cirrosis Hepática/virología , Neoplasias Hepáticas/metabolismo
11.
Cytokine ; 124: 154501, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-30097285

RESUMEN

The hepatic fibrogenic response is a protective mechanism activated by hepatocyte damage and is resolved upon elimination of the cause. However, persistent injuries cause liver fibrosis (LF) to evolve into cirrhosis, which promotes the development of hepatocellular carcinoma (HCC). Development of efficient treatments for LF requires better understanding the underlying molecular pathogenic mechanisms. The loss of suppressor of cytokine signaling 1 (SOCS1) expression promotes LF and HCC in human and mice, but the underlying mechanisms remain unclear. SOCS1 is a key regulator of immune cell activation. To investigate the anti-fibrogenic functions of SOCS1 in hepatocytes and macrophages, we generated mice lacking SOCS1 in hepatocytes (Socs1fl/flAlbCre) or macrophages (Socs1fl/flLysMCre) and evaluated hepatic fibrogenic response to carbon tetrachloride (CCl4). Socs1fl/flAlbCre and Socs1fl/flLysMCre mice showed severe LF characterized by increased collagen deposition, hydroxyproline content, myofibroblast accumulation along with elevated expression of Acta2 and Col1a1 genes. CCl4 treatment triggered significant damage to hepatocytes in Socs1fl/flAlbCre mice but not in Socs1fl/flLysMCre mice. In both mice CCl4 treatment reduced the expression of Mmp2 and increased the expression of Timp1. SOCS1 deficiency in hepatocytes or macrophages did not affect Il6, Tnfa or Tgfb, but diminished Infg and augmented Pdgfb expression. Both Socs1fl/flAlbCre and Socs1fl/flLysMCre livers showed increased mononuclear cell infiltration accompanied by elevated Ccl2 expression. Our findings show that SOCS1 exerts non-redundant functions in hepatocytes and macrophages to regulate the hepatic fibrogenic response possibly through limiting hepatocyte damage and the inflammatory response of macrophages, and support the idea of exploiting SOCS1 in LF treatment.


Asunto(s)
Hepatocitos/metabolismo , Cirrosis Hepática/metabolismo , Hígado/fisiopatología , Macrófagos/metabolismo , Proteína 1 Supresora de la Señalización de Citocinas/metabolismo , Actinas/genética , Actinas/metabolismo , Animales , Tetracloruro de Carbono/toxicidad , Quimiocina CCL2/metabolismo , Colágeno Tipo I/genética , Colágeno Tipo I/metabolismo , Cadena alfa 1 del Colágeno Tipo I , Citocinas/metabolismo , Regulación de la Expresión Génica/genética , Inflamación/metabolismo , Leucocitos/metabolismo , Hígado/citología , Hígado/efectos de los fármacos , Hígado/metabolismo , Cirrosis Hepática/inducido químicamente , Cirrosis Hepática/enzimología , Cirrosis Hepática/patología , Linfocinas/genética , Linfocinas/metabolismo , Metaloproteinasa 2 de la Matriz/genética , Metaloproteinasa 2 de la Matriz/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Noqueados , Miofibroblastos/citología , Miofibroblastos/efectos de los fármacos , Factor de Crecimiento Derivado de Plaquetas/genética , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Albúmina Sérica/genética , Albúmina Sérica/metabolismo , Proteína 1 Supresora de la Señalización de Citocinas/genética , Inhibidor Tisular de Metaloproteinasa-1/genética , Inhibidor Tisular de Metaloproteinasa-1/metabolismo
12.
Cytokine ; 121: 154733, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31154249

RESUMEN

The extraordinary capacity of the liver to regenerate following injury is dependent on coordinated and regulated actions of cytokines and growth factors. Whereas hepatocyte growth factor (HGF) and epidermal growth factor (EGF) are direct mitogens to hepatocytes, inflammatory cytokines such as TNFα and IL-6 also play essential roles in the liver regeneration process. These cytokines and growth factors activate different signaling pathways in a sequential manner to elicit hepatocyte proliferation. The kinetics and magnitude of these hepatocyte-activating stimuli are tightly regulated to ensure restoration of a functional liver mass without causing uncontrolled cell proliferation. Hepatocyte proliferation can become deregulated under conditions of chronic inflammation, leading to accumulation of genetic aberrations and eventual neoplastic transformation. Among the control mechanisms that regulate hepatocyte proliferation, negative feedback inhibition by the 'suppressor of cytokine signaling (SOCS)' family proteins SOCS1 and SOCS3 play crucial roles in attenuating cytokine and growth factor signaling. Loss of SOCS1 or SOCS3 in the mouse liver increases the rate of liver regeneration and renders hepatocytes susceptible to neoplastic transformation. The frequent epigenetic repression of the SOCS1 and SOCS3 genes in hepatocellular carcinoma has stimulated research in understanding the growth regulatory mechanisms of SOCS1 and SOCS3 in hepatocytes. Whereas SOCS3 is implicated in regulating JAK-STAT signaling induced by IL-6 and attenuating EGFR signaling, SOCS1 is crucial for the regulation of HGF signaling. These two proteins also module the functions of certain key proteins that control the cell cycle. In this review, we discuss the current understanding of the functions of SOCS1 and SOCS3 in controlling hepatocyte proliferation, and its implications to liver health and disease.


Asunto(s)
Hepatocitos/citología , Hepatocitos/metabolismo , Proteínas Supresoras de la Señalización de Citocinas/metabolismo , Secuencia de Aminoácidos , Animales , Proliferación Celular , Transformación Celular Neoplásica/patología , Humanos , Modelos Biológicos , Proteínas Supresoras de la Señalización de Citocinas/química
13.
Cytokine ; 108: 225-231, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29102683

RESUMEN

Cytokines and chemokines are intricately connected to cancer initiation, progression and metastasis as well as to innate and adaptive host defense mechanisms against transformed cells. The Aegean Conference on Cytokine Signaling in Cancer (ACCSC) aims to bring together researchers in this highly targeted area of cancer research in a lovely and relaxing Greek-Mediterranean backdrop to discuss latest developments. Being small in size with about one hundred participants, this conference fosters scientific and social interactions among established and emerging scientists in clinical and basic research in diverse fields of oncology, biochemistry, biophysics, genetics and immunology. The 2nd ACCSC held at Heraklion on the Greek island of Crete was organized by Serge Fuchs (University of Pennsylvania), Mathias Muller (University of Veterinary Medicine Vienna), Leonidas Platanias (Northwestern University, Chicago) and Belinda Parker (La Trobe University, Melbourne) between May 30 and June 04, 2017, was a great success in every single aspect of a high level scientific meeting. Signaling within cancer cells as well as in stromal and immune cells is the common thread of this conference series. An outline of the research topics discussed at this conference is presented here to emphasize its high quality and to stimulate interest among cytokine researchers to participate in future ACCSC meetings.


Asunto(s)
Citocinas/inmunología , Neoplasias/inmunología , Transducción de Señal , Grecia , Humanos
14.
Curr Top Microbiol Immunol ; 410: 159-189, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28900678

RESUMEN

SOCS1 is a negative feedback regulator of cytokine and growth factor receptor signaling, and plays an indispensable role in attenuating interferon gamma signaling. Studies on SOCS1-deficient mice have established a crucial role for SOCS1 in regulating CD8+ T cell homeostasis. In the thymus, SOCS1 prevents thymocytes that had failed positive selection from surviving and expanding, ensures negative selection and prevents inappropriate developmental skewing toward the CD8 lineage. In the periphery, SOCS1 not only controls production of T cell stimulatory cytokines but also attenuates the sensitivity of CD8+ T cells to synergistic cytokine stimulation and antigen non-specific activation. As cytokine stimulation of CD8+ T lymphocytes increases their sensitivity to low affinity TCR ligands, SOCS1 likely contributes to peripheral T cell tolerance by putting brakes on aberrant T cell activation driven by inflammatory cytokines. In addition, SOCS1 is critical to maintain the stability of T regulatory cells and control their plasticity to become pathogenic Th17 and Th1 cells under the harmful influence of inflammatory cytokines. SOCS1 also regulates T cell activation by dendritic cells via modulating their generation, maturation, antigen presentation, costimulatory signaling, and cytokine production. The above control mechanisms of SOCS1 on T cells, T regulatory cells and dendritic cells collectively contribute to immunological tolerance and prevent autoimmune manifestation. On other hand, silencing SOCS1 in dendritic cells or CD8+ T cells stimulates efficient antitumor immunity. Thus, even though SOCS1 is not a cell surface checkpoint inhibitor, its regulatory functions on T cell responses qualify SOCS1as a "non-classical" checkpoint blocker. SOCS1 also functions as a tumor suppressor in cancer cells by regulating oncogenic signal transduction pathways. The loss of SOCS1 expression observed in many tumors may have an impact on classical checkpoint pathways. The potential to exploit SOCS1 to treat inflammatory/autoimmune diseases and elicit antitumor immunity is discussed.


Asunto(s)
Autoinmunidad , Proteína 1 Supresora de la Señalización de Citocinas , Proteínas Supresoras de la Señalización de Citocinas , Animales , Humanos , Ratones , Ratones Noqueados , Proteína 1 Supresora de la Señalización de Citocinas/fisiología , Linfocitos T
15.
BMC Cancer ; 17(1): 157, 2017 02 24.
Artículo en Inglés | MEDLINE | ID: mdl-28235401

RESUMEN

BACKGROUND: Suppressor of cytokine signaling 1 (SOCS1) is considered a tumor suppressor due to frequent epigenetic and micro-RNA-mediated repression of its gene expression in diverse cancers. In prostate cancer (PCa), elevated expression of miR-30d that targets SOCS1 mRNA is associated with increased risk of disease recurrence. SOCS1 can mediate its tumor suppressor functions by diverse mechanisms such as inhibiting the JAK-STAT signaling pathway, promoting the tumor suppressor functions of p53, attenuating MET receptor tyrosine kinase signaling and blocking the oncogenic potential of the cell cycle inhibitor p21CIP1 (p21). Here, we studied the expression of SOCS1 and the downstream targets of its putative tumor suppressor functions (p53, MET and p21) in human PCa specimens to evaluate their significance as markers of disease prognosis. METHODS: Tissue microarrays were constructed of 78 archived prostatectomy specimens that were grouped according to the recommendations of the International Society of Urological Pathology (ISUP) based on the Gleason patterns. SOCS1, p53, MET and p21 protein expression were evaluated by immunohistochemical staining alongside the common prostate cancer-related markers Ki67, prostein and androgen receptor. Statistical correlations between the staining intensities of these markers and ISUP grade groups, local invasion or lymph node metastasis were evaluated. RESULTS: SOCS1 showed diffuse staining in the prostatic epithelium. SOCS1 staining intensity correlated inversely with the ISUP grade groups (ρ = -0.4687, p <0.0001) and Ki67 (ρ = -0.2444, p = 0.031), and positively with prostein (ρ = 0.3511, p = 0.0016). Changes in SOCS1 levels did not significantly associate with those of p53, MET or p21. However, p21 positively correlated with androgen receptor expression (ρ = -0.1388, p = 0.0003). A subset of patients with regional lymph node metastasis, although small in number, showed reduced SOCS1 expression and increased expression of MET and p21. CONCLUSIONS: Our findings suggest that evaluating SOCS1 and p21 protein expression in prostatectomy specimens may have a prognostic value in identifying the aggressive disease. Hence, prospective studies with larger numbers of metastatic PCa specimens incorporating clinical correlates such as disease-free and overall survival are warranted.


Asunto(s)
Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Regulación Neoplásica de la Expresión Génica , Neoplasias de la Próstata/metabolismo , Transducción de Señal , Proteína 1 Supresora de la Señalización de Citocinas/genética , Genes Supresores de Tumor , Humanos , Masculino , Pronóstico , Neoplasias de la Próstata/diagnóstico , Neoplasias de la Próstata/genética , Proteína 1 Supresora de la Señalización de Citocinas/metabolismo
16.
Mol Cell ; 36(5): 754-67, 2009 Dec 11.
Artículo en Inglés | MEDLINE | ID: mdl-20005840

RESUMEN

SOCS1 is lost in many human tumors, but its tumor suppression activities are not well understood. We report that SOCS1 is required for transcriptional activity, DNA binding, and serine 15 phosphorylation of p53 in the context of STAT5 signaling. In agreement, inactivation of SOCS1 disabled p53-dependent senescence in response to oncogenic STAT5A and radiation-induced apoptosis in T cells. In addition, SOCS1 was sufficient to induce p53-dependent senescence in fibroblasts. The mechanism of activation of p53 by SOCS1 involved a direct interaction between the SH2 domain of SOCS1 and the N-terminal transactivation domain of p53, while the C-terminal domain of SOCS1 containing the SOCS Box mediated interaction with the DNA damage-regulated kinases ATM/ATR. Also, SOCS1 colocalized with ATM at DNA damage foci induced by oncogenic STAT5A. Collectively, these results add another component to the p53 and DNA damage networks and reveal a mechanism by which SOCS1 functions as a tumor suppressor.


Asunto(s)
Senescencia Celular , Proteínas Supresoras de la Señalización de Citocinas/fisiología , Proteína p53 Supresora de Tumor/metabolismo , Animales , Proteínas de la Ataxia Telangiectasia Mutada , Proteínas de Ciclo Celular/metabolismo , Línea Celular Tumoral , Núcleo Celular/metabolismo , Daño del ADN , Proteínas de Unión al ADN/metabolismo , Humanos , Ratones , Fosforilación , Proteínas Serina-Treonina Quinasas/metabolismo , Interferencia de ARN , Factor de Transcripción STAT5/metabolismo , Transducción de Señal , Proteína 1 Supresora de la Señalización de Citocinas , Proteínas Supresoras de la Señalización de Citocinas/análisis , Proteínas Supresoras de la Señalización de Citocinas/metabolismo , Proteínas Supresoras de Tumor/metabolismo
17.
Immun Ageing ; 14: 19, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28855949

RESUMEN

In an effort to understand the mechanisms underlying the high prevalence of gastrointestinal tract disorders in old age, we investigated the expression of intestinal antimicrobial peptides in the terminal small intestine of aged mice. Our results show that old mice have reduced transcript levels of ileal α-defensins and lysozyme, two important types of intestinal antimicrobial peptides produced by Paneth cells. In contrast, expression of the C-type lectins Reg3b and Reg3g, as well as ß-defensin 1, angiogenin 4 and Relmb, which are made by several epithelial cell types, was significantly upregulated in aged animals suggesting an ongoing response to epithelial distress. Those changes in antimicrobial peptide gene expression associated with histological damage of the ileal epithelium and subtle modifications in the composition of the commensal microbiota. Our findings suggest that dysregulation of antimicrobial peptides expression is a feature of homeostasis disruption in the aged intestine and may contribute to geriatric gastrointestinal dysfunction.

18.
Cytokine ; 85: 83-91, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27300756

RESUMEN

IL-21 promotes autoimmune type-1 diabetes (T1D) in NOD mice by facilitating CD4(+) T cell help to CD8(+) T cells. IL-21 also enables autoreactive CD8(+) T cells to respond to weak TCR ligands and induce T1D. Here, we assessed whether IL-21 is essential for T1D induction in a mouse model where the disease can occur independently of CD4 help. In this model, which expresses lymphocytic choriomeningitis virus (LCMV) glycoprotein (GP) antigen under the rat insulin promoter (RIP-GP), LCMV infection activates CD8(+) T cells reactive to the GP-derived GP33 peptide that attack pancreatic islets and cause T1D. We show that IL-21 deficiency in RIP-GP mice did not impair T1D induction by LCMV expressing the wildtype GP33 peptide. Surprisingly, LCMV-L6F, expressing a weak peptide mimic of GP33, induced T1D more efficiently in Il21(-/-)RIP-GP mice than in controls. However, LCMV-C4Y expressing a very weak peptide mimic of GP33 did not induce T1D in Il21(-/-) mice, but T cells from the infected mice caused disease in lymphopenic RIP-GP mice upon adoptive transfer. Using Nur77(GFP) reporter mice, we show that CD8(+) T cells from Il21(-/-) mice expressing the GP33-specific transgenic P14 TCR showed increased reactivity towards low affinity TCR ligands. Collectively, our findings show that IL-21 is not always required for T1D induction by autoreactive CD8(+) T cells, and suggest that IL-21 may play an important role in regulating CD8(+) T cell reactivity towards low affinity TCR ligands.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Interleucinas/inmunología , Receptores de Antígenos de Linfocitos T/inmunología , Traslado Adoptivo/métodos , Animales , Antígenos Virales/inmunología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/virología , Diabetes Mellitus Tipo 1/inmunología , Diabetes Mellitus Tipo 1/virología , Insulina/inmunología , Islotes Pancreáticos/inmunología , Islotes Pancreáticos/virología , Ligandos , Activación de Linfocitos/inmunología , Recuento de Linfocitos/métodos , Coriomeningitis Linfocítica/inmunología , Coriomeningitis Linfocítica/virología , Virus de la Coriomeningitis Linfocítica/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones Transgénicos/inmunología , Regiones Promotoras Genéticas/inmunología , Ratas
19.
Cytokine ; 82: 125-39, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-26822708

RESUMEN

Hepatocyte growth factor (HGF) signaling via the MET receptor is essential for embryonic development and tissue repair. On the other hand, deregulated MET signaling promotes tumor progression in diverse types of cancers. Even though oncogenic MET signaling remains the major research focus, the HGF-MET axis has also been implicated in diverse aspects of immune cell development and functions. In the presence of other hematopoietic growth factors, HGF promotes the development of erythroid, myeloid and lymphoid lineage cells and thrombocytes. In monocytes and macrophages responding to inflammatory stimuli, induction of autocrine HGF-MET signaling can contribute to tissue repair via stimulating anti-inflammatory cytokine production. HGF-MET signaling can also modulate adaptive immune response by facilitating the migration of Langerhans cells and dendritic cells to draining lymph nodes. However, MET signaling has also been shown to induce tolerogenic dendritic cells in mouse models of graft-versus-host disease and experimental autoimmune encephalomyelitis. HGF-MET axis is also implicated in promoting thymopoiesis and the survival and migration of B lymphocytes. Recent studies have shown that MET signaling induces cardiotropism in activated T lymphocytes. Further understanding of the HGF-MET axis in the immune system would allow its therapeutic manipulation to improve immune cell reconstitution, restore immune homeostasis and to treat immuno-inflammatory diseases.


Asunto(s)
Linfocitos B/inmunología , Factor de Crecimiento de Hepatocito/inmunología , Proteínas Proto-Oncogénicas c-met/inmunología , Transducción de Señal/inmunología , Linfocitos T/inmunología , Inmunidad Adaptativa , Animales , Movimiento Celular/inmunología , Encefalomielitis Autoinmune Experimental/inmunología , Humanos , Ratones
20.
Cytokine ; 82: 58-69, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-26748724

RESUMEN

Suppressor of cytokine signaling 1 (SOCS1) is an indispensable regulator of IFNγ signaling and has been implicated in the regulation of liver fibrosis. However, it is not known whether SOCS1 mediates its anti-fibrotic functions in the liver directly, or via modulating IFNγ, which has been implicated in attenuating hepatic fibrosis. Additionally, it is possible that SOCS1 controls liver fibrosis by regulating hepatic stellate cells (HSC), a key player in fibrogenic response. While the activation pathways of HSCs have been well characterized, the regulatory mechanisms are not yet clear. The goals of this study were to dissociate IFNγ-dependent and SOCS1-mediated regulation of hepatic fibrogenic response, and to elucidate the regulatory functions of SOCS1 in HSC activation. Liver fibrosis was induced in Socs1(-/-)Ifng(-/-) mice with dimethylnitrosamine or carbon tetrachloride. Ifng(-/-) and C57BL/6 mice served as controls. Following fibrogenic treatments, Socs1(-/-)Ifng(-/-) mice showed elevated serum ALT levels and increased liver fibrosis compared to Ifng(-/-) mice. The latter group showed higher ALT levels and fibrosis than C57BL/6 controls. The livers of SOCS1-deficient mice showed bridging fibrosis, which was associated with increased accumulation of myofibroblasts and abundant collagen deposition. SOCS1-deficient livers showed increased expression of genes coding for smooth muscle actin, collagen, and enzymes involved in remodeling the extracellular matrix, namely matrix metalloproteinases and tissue inhibitor of metalloproteinases. Primary HSCs from SOCS1-deficient mice showed increased proliferation in response to growth factors such as HGF, EGF and PDGF, and the fibrotic livers of SOCS1-deficient mice showed increased expression of the Pdgfb gene. Taken together, these data indicate that SOCS1 controls liver fibrosis independently of IFNγ and that part of this regulation may occur via regulating HSC proliferation and limiting growth factor availability.


Asunto(s)
Factor de Crecimiento Epidérmico/inmunología , Factor de Crecimiento de Hepatocito/inmunología , Interferón gamma/inmunología , Cirrosis Hepática/inmunología , Factor de Crecimiento Derivado de Plaquetas/inmunología , Transducción de Señal/inmunología , Proteína 1 Supresora de la Señalización de Citocinas/inmunología , Animales , Factor de Crecimiento Epidérmico/genética , Factor de Crecimiento de Hepatocito/genética , Interferón gamma/genética , Cirrosis Hepática/genética , Cirrosis Hepática/patología , Ratones , Ratones Noqueados , Factor de Crecimiento Derivado de Plaquetas/genética , Transducción de Señal/genética , Proteína 1 Supresora de la Señalización de Citocinas/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA