Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
J Med Virol ; 96(2): e29455, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38323709

RESUMEN

Severe acute respiratory coronavirus 2 (SARS-CoV-2) causes neurological disease in the peripheral and central nervous system (PNS and CNS, respectively) of some patients. It is not clear whether SARS-CoV-2 infection or the subsequent immune response are the key factors that cause neurological disease. Here, we addressed this question by infecting human induced pluripotent stem cell-derived CNS and PNS neurons with SARS-CoV-2. SARS-CoV-2 infected a low number of CNS neurons and did not elicit a robust innate immune response. On the contrary, SARS-CoV-2 infected a higher number of PNS neurons. This resulted in expression of interferon (IFN) λ1, several IFN-stimulated genes and proinflammatory cytokines. The PNS neurons also displayed alterations characteristic of neuronal damage, as increased levels of sterile alpha and Toll/interleukin receptor motif-containing protein 1, amyloid precursor protein and α-synuclein, and lower levels of cytoskeletal proteins. Interestingly, blockade of the Janus kinase and signal transducer and activator of transcription pathway by Ruxolitinib did not increase SARS-CoV-2 infection, but reduced neuronal damage, suggesting that an exacerbated neuronal innate immune response contributes to pathogenesis in the PNS. Our results provide a basis to study coronavirus disease 2019 (COVID-19) related neuronal pathology and to test future preventive or therapeutic strategies.


Asunto(s)
COVID-19 , Células Madre Pluripotentes Inducidas , Humanos , SARS-CoV-2 , Inmunidad Innata , Neuronas
2.
J Virol ; 94(20)2020 09 29.
Artículo en Inglés | MEDLINE | ID: mdl-32669337

RESUMEN

During primary infection, herpes simplex virus 2 (HSV-2) replicates in epithelial cells and enters neurites to infect neurons of the peripheral nervous system. Growth factors and attractive and repulsive directional cues influence neurite outgrowth and neuronal survival. We hypothesized that HSV-2 modulates the activity of such cues to increase neurite outgrowth. To test this hypothesis, we exposed sensory neurons to nerve growth factor (NGF) and mock- or HSV-2-infected HEK-293T cells, since they express repellents of neurite outgrowth. We show that HEK-293T cells secrete factors that inhibit neurite outgrowth, while infection with HSV-2 strains MS and 333 reduces this repelling phenotype, increasing neurite numbers. The HSV-2-mediated restoration of neurite outgrowth required the activity of NGF. In the absence of infection, however, NGF did not overcome the repulsion mediated by HEK-293T cells. We previously showed that recombinant, soluble glycoprotein G of HSV-2 (rSgG2) binds and enhances NGF activity, increasing neurite outgrowth. However, the effect of gG2 during infection has not been investigated. Therefore, we addressed whether gG2 contributes to overcoming neurite outgrowth repulsion. To do so, we generated viruses lacking gG2 expression and complemented them by exogenous expression of gG2. Overall, our results suggest that HSV-2 infection of nonneuronal cells reduces their repelling effect on neurite outgrowth in an NGF-dependent manner. gG2 contributed to this phenotype, but it was not the only factor. The enhanced neurite outgrowth may facilitate HSV-2 spread from epithelial cells into neurons expressing NGF receptors and increase HSV-2-mediated pathogenesis.IMPORTANCE Herpes simplex virus 2 (HSV-2) is a prevalent human pathogen that establishes lifelong latency in neurons of the peripheral nervous system. Colonization of neurons is required for HSV-2 persistence and pathogenesis. The viral and cellular factors required for efficient infection of neurons are not fully understood. We show here that nonneuronal cells repel neurite outgrowth of sensory neurons, while HSV-2 infection overcomes this inhibition and, rather, stimulates neurite outgrowth. HSV-2 glycoprotein G and nerve growth factor contribute to this phenotype, which may attract neurites to sites of infection and facilitate virus spread to neurons. Understanding the mechanisms that modulate neurite outgrowth and facilitate HSV-2 infection of neurons might foster the development of therapeutics to reduce HSV-2 colonization of the nervous system and provide insights on neurite outgrowth and regeneration.


Asunto(s)
Herpes Genital/metabolismo , Herpesvirus Humano 2/metabolismo , Factor de Crecimiento Nervioso/metabolismo , Neuritas , Animales , Línea Celular Tumoral , Chlorocebus aethiops , Células HEK293 , Herpesvirus Humano 2/patogenicidad , Humanos , Ratones , Ratones Endogámicos BALB C , Neuritas/metabolismo , Neuritas/virología , Células Vero
3.
PLoS Pathog ; 13(5): e1006346, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-28542541

RESUMEN

Varicella zoster virus (VZV) is a highly prevalent human pathogen that establishes latency in neurons of the peripheral nervous system. Primary infection causes varicella whereas reactivation results in zoster, which is often followed by chronic pain in adults. Following infection of epithelial cells in the respiratory tract, VZV spreads within the host by hijacking leukocytes, including T cells, in the tonsils and other regional lymph nodes, and modifying their activity. In spite of its importance in pathogenesis, the mechanism of dissemination remains poorly understood. Here we addressed the influence of VZV on leukocyte migration and found that the purified recombinant soluble ectodomain of VZV glycoprotein C (rSgC) binds chemokines with high affinity. Functional experiments show that VZV rSgC potentiates chemokine activity, enhancing the migration of monocyte and T cell lines and, most importantly, human tonsillar leukocytes at low chemokine concentrations. Binding and potentiation of chemokine activity occurs through the C-terminal part of gC ectodomain, containing predicted immunoglobulin-like domains. The mechanism of action of VZV rSgC requires interaction with the chemokine and signalling through the chemokine receptor. Finally, we show that VZV viral particles enhance chemokine-dependent T cell migration and that gC is partially required for this activity. We propose that VZV gC activity facilitates the recruitment and subsequent infection of leukocytes and thereby enhances VZV systemic dissemination in humans.


Asunto(s)
Varicela/virología , Herpes Zóster/virología , Herpesvirus Humano 3/genética , Leucocitos/fisiología , Proteínas del Envoltorio Viral/genética , Animales , Línea Celular , Movimiento Celular , Quimiocinas/metabolismo , Varicela/inmunología , Drosophila melanogaster , Células Epiteliales/virología , Genes Reporteros , Herpes Zóster/inmunología , Herpesvirus Humano 3/inmunología , Herpesvirus Humano 3/fisiología , Interacciones Huésped-Patógeno , Humanos , Mutación , Tonsila Palatina/virología , Dominios Proteicos , Linfocitos T/virología , Proteínas del Envoltorio Viral/inmunología , Proteínas del Envoltorio Viral/metabolismo , Proteínas Virales/genética , Proteínas Virales/metabolismo , Virión
4.
mBio ; 15(2): e0330823, 2024 Feb 14.
Artículo en Inglés | MEDLINE | ID: mdl-38275838

RESUMEN

The highly prevalent herpes simplex virus type 1 (HSV-1) causes a range of diseases, including cold sores, blinding keratitis, and life-threatening encephalitis. HSV-1 initially replicates in epithelial cells, enters the peripheral nervous system via neurites, and establishes lifelong infection in the neuronal cell bodies. Neurites are highly dynamic structures that grow or retract in response to attractive or repulsive cues, respectively. Here, we show that infection with HSV-1, but not with a mutant virus lacking glycoprotein G (gG), reduced the repulsive effect of epithelial cells on neurite outgrowth and facilitated HSV-1 invasion of neurons. HSV-1 gG was required and sufficient to induce neurite outgrowth by modifying the protein composition of extracellular vesicles, increasing the amount of neurotrophic and neuroprotective proteins, including galectin-1. Antibodies directed against galectin-1 neutralized the capacity of extracellular vesicles released from HSV-1-infected cells to promote neurite outgrowth. Our study provides new insights into the neurotropism of HSV-1 and identifies a viral protein that modifies the protein composition of extracellular vesicles to stimulate neurite outgrowth and invasion of the nervous system.IMPORTANCEHerpes simplex virus type 1 (HSV-1) must infect neurites (or nerve endings) to establish a chronic infection in neurons. Neurites are highly dynamic structures that retract or grow in the presence of repulsive or attractive proteins. Some of these proteins are released by epithelial cells in extracellular vesicles and act upon interaction with their receptor present on neurites. We show here that HSV-1 infection of epithelial cells modulated their effect on neurites, increasing neurite growth. Mechanistically, HSV-1 glycoprotein G (gG) modifies the protein composition of extracellular vesicles released by epithelial cells, increasing the amount of attractive proteins that enhance neurite outgrowth and facilitate neuronal infection. These results could inform of therapeutic strategies to block HSV-1 induction of neurite outgrowth and, thereby, neuronal infection.


Asunto(s)
Enfermedades Transmisibles , Vesículas Extracelulares , Herpes Simple , Herpesvirus Humano 1 , Humanos , Herpesvirus Humano 1/fisiología , Galectina 1/metabolismo , Vesículas Extracelulares/metabolismo , Proyección Neuronal , Glicoproteínas/metabolismo
5.
Nat Commun ; 15(1): 5318, 2024 Jun 22.
Artículo en Inglés | MEDLINE | ID: mdl-38909022

RESUMEN

During primary varicella zoster virus (VZV) infection, infected lymphocytes drive primary viremia, causing systemic dissemination throughout the host, including the skin. This results in cytokine expression, including interferons (IFNs), which partly limit infection. VZV also spreads from skin keratinocytes to lymphocytes prior to secondary viremia. It is not clear how VZV achieves this while evading the cytokine response. Here, we show that VZV glycoprotein C (gC) binds IFN-γ and modifies its activity, increasing the expression of a subset of IFN-stimulated genes (ISGs), including intercellular adhesion molecule 1 (ICAM1), chemokines and immunomodulatory genes. The higher ICAM1 protein level at the plasma membrane of keratinocytes facilitates lymphocyte function-associated antigen 1-dependent T cell adhesion and expression of gC during infection increases VZV spread to peripheral blood mononuclear cells. This constitutes the discovery of a strategy to modulate IFN-γ activity, upregulating a subset of ISGs, promoting enhanced lymphocyte adhesion and virus spread.


Asunto(s)
Adhesión Celular , Herpesvirus Humano 3 , Molécula 1 de Adhesión Intercelular , Interferón gamma , Queratinocitos , Linfocitos T , Humanos , Interferón gamma/metabolismo , Interferón gamma/inmunología , Linfocitos T/inmunología , Linfocitos T/metabolismo , Linfocitos T/virología , Molécula 1 de Adhesión Intercelular/metabolismo , Molécula 1 de Adhesión Intercelular/genética , Queratinocitos/virología , Queratinocitos/metabolismo , Queratinocitos/inmunología , Herpesvirus Humano 3/fisiología , Infección por el Virus de la Varicela-Zóster/inmunología , Infección por el Virus de la Varicela-Zóster/virología , Leucocitos Mononucleares/virología , Leucocitos Mononucleares/metabolismo , Leucocitos Mononucleares/inmunología , Proteínas del Envoltorio Viral/metabolismo , Antígeno-1 Asociado a Función de Linfocito/metabolismo
6.
bioRxiv ; 2023 May 26.
Artículo en Inglés | MEDLINE | ID: mdl-37292914

RESUMEN

During primary infection, varicella zoster virus (VZV) infects epithelial cells in the respiratory lymphoid organs and mucosa. Subsequent infection of lymphocytes, T cells in particular, causes primary viremia allowing systemic spread throughout the host, including the skin. This results in the expression of cytokines, including interferons (IFNs) which partly limit primary infection. VZV also spreads from skin keratinocytes to lymphocytes prior to secondary viremia. How VZV infects lymphocytes from epithelial cells while evading the cytokine response has not been fully established. Here, we show that VZV glycoprotein C (gC) binds IFN-γ and modifies its activity. Transcriptomic analysis revealed that gC in combination with IFN-γ increased the expression of a small subset of IFN-stimulated genes (ISGs), including intercellular adhesion molecule 1 (ICAM1), as well as several chemokines and immunomodulatory genes. The higher ICAM1 protein level at the plasma membrane of epithelial cells resulted in lymphocyte function-associated antigen 1 (LFA-1)-dependent T cell adhesion. This gC activity required a stable interaction with IFN-γ and signalling through the IFN-γ receptor. Finally, the presence of gC during infection increased VZV spread from epithelial cells to peripheral blood mononuclear cells. This constitutes the discovery of a novel strategy to modulate the activity of IFN-γ, inducing the expression of a subset of ISGs, leading to enhanced T cell adhesion and virus spread.

7.
Cancer Discov ; 13(10): 2192-2211, 2023 10 05.
Artículo en Inglés | MEDLINE | ID: mdl-37489084

RESUMEN

In colorectal cancers, the tumor microenvironment plays a key role in prognosis and therapy efficacy. Patient-derived tumor organoids (PDTO) show enormous potential for preclinical testing; however, cultured tumor cells lose important characteristics, including the consensus molecular subtypes (CMS). To better reflect the cellular heterogeneity, we established the colorectal cancer organoid-stroma biobank of matched PDTOs and cancer-associated fibroblasts (CAF) from 30 patients. Context-specific phenotyping showed that xenotransplantation or coculture with CAFs improves the transcriptomic fidelity and instructs subtype-specific stromal gene expression. Furthermore, functional profiling in coculture exposed CMS4-specific therapeutic resistance to gefitinib and SN-38 and prognostic expression signatures. Chemogenomic library screening identified patient- and therapy-dependent mechanisms of stromal resistance including MET as a common target. Our results demonstrate that colorectal cancer phenotypes are encrypted in the cancer epithelium in a plastic fashion that strongly depends on the context. Consequently, CAFs are essential for a faithful representation of molecular subtypes and therapy responses ex vivo. SIGNIFICANCE: Systematic characterization of the organoid-stroma biobank provides a resource for context dependency in colorectal cancer. We demonstrate a colorectal cancer subtype memory of PDTOs that is independent of specific driver mutations. Our data underscore the importance of functional profiling in cocultures for improved preclinical testing and identification of stromal resistance mechanisms. This article is featured in Selected Articles from This Issue, p. 2109.


Asunto(s)
Fibroblastos Asociados al Cáncer , Neoplasias Colorrectales , Humanos , Bancos de Muestras Biológicas , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/metabolismo , Células Tumorales Cultivadas , Fibroblastos Asociados al Cáncer/metabolismo , Organoides/patología , Microambiente Tumoral/genética
8.
Chembiochem ; 13(3): 409-15, 2012 Feb 13.
Artículo en Inglés | MEDLINE | ID: mdl-22271561

RESUMEN

Elevated expression of interleukin-8 (IL-8) has been implicated in inflammatory diseases, in tumor growth, and in angiogenesis. The aim of this study was to identify natural or synthetic compounds that suppress IL-8 production in response to interleukin-1 (IL-1), the natural inflammatory stimulus of the IL-8 gene. We therefore developed an IL-1-inducible cell-based screening assay by stable integration of an IL-8 reporter gene into HeLa S3 cells. The screening of heterogeneous compound libraries revealed several compounds that displayed an inhibitory effect on the reporter gene expression. Following hit validation, we focused on the most efficient compound, spirangien A, and its chemical derivate spirangien M522. Detailed analysis shows that both compounds are potent inhibitors of the endogenous IL-8 gene transcription. Furthermore, both compounds decelerate the phosphorylation and degradation of IκBα, the key regulator of the IL-1-stimulated NF-κB signaling pathway. Our study has identified the two spirangiens A and M522 as potent inhibitors of IL-1/NF-κB-mediated IL-8 gene expression.


Asunto(s)
Acetales/farmacología , Ácidos Grasos Insaturados/farmacología , Interleucina-8/antagonistas & inhibidores , Myxococcales/química , Piranos/farmacología , Compuestos de Espiro/farmacología , Acetales/química , Relación Dosis-Respuesta a Droga , Ácidos Grasos Insaturados/química , Perfilación de la Expresión Génica , Células HeLa , Humanos , Proteínas I-kappa B/antagonistas & inhibidores , Proteínas I-kappa B/metabolismo , Interleucina-8/genética , Fosforilación/efectos de los fármacos , Piranos/química , ARN Mensajero/antagonistas & inhibidores , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Compuestos de Espiro/química , Transcripción Genética/efectos de los fármacos , Transcripción Genética/genética
9.
J Clin Immunol ; 31(1): 60-8, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20886283

RESUMEN

Interleukin-8 (IL-8) plays a central role in the pathogenesis of Helicobacter pylori infection. We used four different H. pylori strains isolated from patients with gastritis or duodenal ulcer disease to examine their differential effects on signaling pathways and IL-8 gene response in gastric epithelial cells. IL-8 mRNA level is elevated in response to high (100) multiplicity of infection (MOI) independent of cagA, vacA, and dupA gene characteristics. By lower MOIs (1 or 10), only cagA ( + ) strains significantly induce IL-8 gene expression. This is based on differential regulation of IL-8 promoter activity. Analysis of intracellular signaling pathways indicates that H. pylori clinical isolates induce IL-8 gene transcription through NF-κB p65, but by a MOI-dependent differential activation of MAPK pathways. Thus, the major virulence factors of H. pylori CagA, VacA, and DupA might play a minor role in the level of IL-8 gene response to a high bacterial load.


Asunto(s)
Carga Bacteriana , Regulación de la Expresión Génica , Helicobacter pylori/patogenicidad , Interleucina-8/metabolismo , Transducción de Señal , Factores de Virulencia/metabolismo , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Línea Celular , Úlcera Duodenal/inmunología , Úlcera Duodenal/microbiología , Úlcera Duodenal/fisiopatología , Células Epiteliales/inmunología , Células Epiteliales/microbiología , Gastritis/inmunología , Gastritis/microbiología , Gastritis/fisiopatología , Infecciones por Helicobacter/inmunología , Infecciones por Helicobacter/microbiología , Helicobacter pylori/clasificación , Helicobacter pylori/genética , Helicobacter pylori/aislamiento & purificación , Humanos , Interleucina-8/genética , Quinasas de Proteína Quinasa Activadas por Mitógenos/genética , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , FN-kappa B/genética , FN-kappa B/metabolismo , Regiones Promotoras Genéticas/genética , Estómago/citología , Estómago/inmunología , Estómago/microbiología , Transcripción Genética , Factores de Virulencia/genética
10.
Stem Cell Res ; 56: 102535, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34607262

RESUMEN

Somatosensory low threshold mechanoreceptors (LTMRs) sense innocuous mechanical forces, largely through specialized axon termini termed sensory nerve endings, where the mechanotransduction process initiates upon activation of mechanotransducers. In humans, a subset of sensory nerve endings is enlarged, forming bulb-like expansions, termed bulbous nerve endings. There is no in vitro human model to study these neuronal endings. Piezo2 is the main mechanotransducer found in LTMRs. Recent evidence shows that Piezo1, the other mechanotransducer considered absent in dorsal root ganglia (DRG), is expressed at low level in somatosensory neurons. We established a differentiation protocol to generate, from iPSC-derived neuronal precursor cells, human LTMR recapitulating bulbous sensory nerve endings and heterogeneous expression of Piezo1 and Piezo2. The derived neurons express LTMR-specific genes, convert mechanical stimuli into electrical signals and have specialized axon termini that morphologically resemble bulbous nerve endings. Piezo2 is concentrated within these enlarged axon termini. Some derived neurons express low level Piezo1, and a subset co-express both channels. Thus, we generated a unique, iPSCs-derived human model that can be used to investigate the physiology of bulbous sensory nerve endings, and the role of Piezo1 and 2 during mechanosensation.


Asunto(s)
Células Madre Pluripotentes Inducidas , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Canales Iónicos/genética , Canales Iónicos/metabolismo , Mecanorreceptores/metabolismo , Mecanotransducción Celular , Terminaciones Nerviosas/metabolismo , Células Receptoras Sensoriales/metabolismo
11.
Viruses ; 12(12)2020 12 12.
Artículo en Inglés | MEDLINE | ID: mdl-33322659

RESUMEN

Glycoprotein G (gG) from herpes simplex virus type 1 and 2 (HSV-1 and HSV-2, respectively) functions as a viral chemokine binding protein (vCKBP). Soluble recombinant forms of gG of HSV-1 and HSV-2 (SgG1 and SgG2, respectively) enhance chemokine-mediated leukocyte migration, in contrast to most known vCKBPs, including those from animal alpha-herpesviruses. Furthermore, both proteins bind to nerve growth factor (NGF), but only SgG2 enhances NGF-dependent neurite outgrowth. The basis and implications of this functional difference between the two proteins are still unknown. While gG1 and gG2 are positional homologues in the genome, they share very limited sequence homology. In fact, US4, the open reading frame encoding gG is the most divergent genetic locus between these viruses. Full-length gG1 and gG2 are type I transmembrane proteins located on the plasma membrane of infected cells and at the viral envelope. However, gG2 is larger than gG1 and is cleaved during protein maturation, secreting the N-terminal domain to the supernatant of infected cells, whereas gG1 is not. The enzyme involved in gG2 cleavage and the functional relevance of gG2 cleavage and secretion are unknown. We aim to identify the gG2 sequence required for cleavage to determine its functional role in future experiments. Our results prove the existence of at least two cleavage motifs in gG2 within the amino acid region 314-343. Transfer of this sequence to a fusion protein results in cleavage. Finally, we show that propeptide convertases like furin are responsible for gG2 cleavage.


Asunto(s)
Herpes Simple/virología , Herpesvirus Humano 2/fisiología , Dominios y Motivos de Interacción de Proteínas , Proteínas del Envoltorio Viral/metabolismo , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Animales , Línea Celular , Cromatografía Liquida , Expresión Génica , Genes Reporteros , Humanos , Espectrometría de Masas , Proteolisis
12.
J Clin Invest ; 130(10): 5461-5476, 2020 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-32663191

RESUMEN

Tumor immunosuppression is a limiting factor for successful cancer therapy. The lipid sphingosine-1-phosphate (S1P), which signals through 5 distinct G protein-coupled receptors (S1PR1-5), has emerged as an important regulator of carcinogenesis. However, the utility of targeting S1P in tumors is hindered by S1P's impact on immune cell trafficking. Here, we report that ablation of the immune cell-specific receptor S1PR4, which plays a minor role in immune cell trafficking, delayed tumor development and improved therapy success in murine models of mammary and colitis-associated colorectal cancer through increased CD8+ T cell abundance. Transcriptome analysis revealed that S1PR4 affected proliferation and survival of CD8+ T cells in a cell-intrinsic manner via the expression of Pik3ap1 and Lta4h. Accordingly, PIK3AP1 expression was connected to increased CD8+ T cell proliferation and clinical parameters in human breast and colon cancer. Our data indicate a so-far-unappreciated tumor-promoting role of S1P by restricting CD8+ T cell expansion via S1PR4.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Neoplasias Mamarias Experimentales/terapia , Receptores de Esfingosina-1-Fosfato/deficiencia , Receptores de Esfingosina-1-Fosfato/inmunología , Animales , Linfocitos T CD8-positivos/patología , Proliferación Celular/genética , Supervivencia Celular/genética , Supervivencia Celular/inmunología , Colitis/complicaciones , Colitis/inmunología , Colitis/patología , Neoplasias del Colon/etiología , Neoplasias del Colon/inmunología , Neoplasias del Colon/patología , Progresión de la Enfermedad , Femenino , Perfilación de la Expresión Génica , Técnicas de Inactivación de Genes , Humanos , Neoplasias Mamarias Experimentales/inmunología , Neoplasias Mamarias Experimentales/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores de Esfingosina-1-Fosfato/genética , Microambiente Tumoral/genética , Microambiente Tumoral/inmunología
13.
J Exp Med ; 216(6): 1234-1243, 2019 06 03.
Artículo en Inglés | MEDLINE | ID: mdl-31023715

RESUMEN

A link between chronic inflammation and development of tumors is well established. Moreover, it has become evident that tumorigenesis is not a cell autonomous disease, and an inflammatory microenvironment is a prerequisite of basically all tumors, including those that emerge in the absence of overt inflammation. This knowledge has led to the development of anti-inflammatory concepts to treat and prevent cancer. In contrast, immunotherapies, in particular checkpoint inhibitors, representing the most significant progress in the therapy of several malignancies depend on the presence of a pro-inflammatory "hot" environment. Here, we discuss pro- and anti-inflammatory concepts for the treatment of cancer.


Asunto(s)
Inflamación/patología , Neoplasias/patología , Neoplasias/terapia , Animales , Humanos , Inmunoterapia , Neoplasias/inmunología , Microambiente Tumoral
14.
Methods Mol Biol ; 1182: 195-201, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25055912

RESUMEN

The majority of regulatory RNA sequences exert their function through interaction with proteins. Therefore, the identification of RNA-binding proteins is the key step in understanding the role of many RNA motifs. Here, we describe a straightforward method to identify RNA-binding proteins. In this approach, RNAs are immobilized on beads and incubated with protein lysates. After removing unbound fraction of proteins, the bound proteins are eluted by successive increasing of salt concentration. The lower the salt concentration, the lower is the binding affinity of a protein to the RNA. According to this principle, each elution fraction can contain a number of proteins which bind with similar affinity to the RNA. After gel electrophoretic separation and staining, each single protein band can be identified, isolated, and analyzed by mass spectrometry.


Asunto(s)
Proteínas de Unión al ARN/aislamiento & purificación , Cromatografía de Afinidad , Espectrometría de Masas , ARN/metabolismo , Proteínas de Unión al ARN/química , Proteínas de Unión al ARN/metabolismo
15.
J Mol Biol ; 407(4): 492-504, 2011 Apr 08.
Artículo en Inglés | MEDLINE | ID: mdl-21300069

RESUMEN

Heterogeneous nuclear ribonucleoprotein D-like protein (JKTBP) 1 was implicated in cap-independent translation by binding to the internal ribosome entry site in the 5' untranslated region (UTR) of NF-κB-repressing factor (NRF). Two different NRF mRNAs have been identified so far, both sharing the common 5' internal ribosome entry site but having different length of 3' UTRs. Here, we used a series of DNA and RNA luciferase reporter constructs comprising 5', 3' or both NRF UTRs to study the effect of JKTBP1 on translation of NRF mRNA variants. The results indicate that JKTBP1 regulates the level of NRF protein expression by binding to both NRF 5' and 3' UTRs. Using successive deletion and point mutations as well as RNA binding studies, we define two distinct JKTBP1 binding elements in NRF 5' and 3' UTRs. Furthermore, JKTBP1 requires two distinct RNA binding domains to interact with NRF UTRs and a short C-terminal region for its effect on NRF expression. Together, our study shows that JKTBP1 contributes to NRF protein expression via two disparate mechanisms: mRNA stabilization and cap-independent translation. By binding to 5' UTR, JKTBP1 increases the internal translation initiation in both NRF mRNA variants, whereas its binding to 3' UTR elevated primarily the stability of the major NRF mRNA. Thus, JKTBP1 is a key regulatory factor linking two pivotal control mechanisms of NRF gene expression: the cap-independent translation initiation and mRNA stabilization.


Asunto(s)
Regiones no Traducidas 3' , Regiones no Traducidas 5' , Biosíntesis de Proteínas , ARN Mensajero/metabolismo , Proteínas Represoras/biosíntesis , Ribonucleoproteínas/metabolismo , Fusión Artificial Génica , Genes Reporteros , Luciferasas/genética , Luciferasas/metabolismo , Mutación Puntual , Unión Proteica , Estabilidad del ARN , ARN Mensajero/genética , Proteínas de Unión al ARN/metabolismo , Eliminación de Secuencia
16.
RNA ; 13(8): 1328-40, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17592041

RESUMEN

The mRNA of human NF-kappaB repressing factor (NRF) contains a long 5'-untranslated region (UTR) that directs ribosomes to the downstream start codon by a cap-independent mechanism. Comparison of the nucleotide (nt) sequences of human and mouse NRF mRNAs reveals a high degree of identity throughout a fragment of 150 nt proximal to the start codon. Here, we show that this region constitutes a minimal internal ribosome entry segment (IRES) module. Enzymatic RNA structure analysis reveals a secondary structure model of the NRF IRES module. Point mutation analysis of the module determines a short, 14-nt RNA element (nt 640-653) as a mediator of IRES function. Purification of IRES binding cellular proteins and subsequent ESI/MS/MS sequence analysis led to identification of the RNA-binding protein, JKTBP1. EMSA experiments show that JKTBP1 binds upstream to the 14-nt RNA element in the NRF IRES module (nt 579-639). Over-expression of JKTBP1 significantly enhances activity of the NRF IRES module in dicistronic constructs. Moreover, siRNA experiments demonstrate that down-regulation of endogenous JKTBP1 decreases NRF IRES activity and the level of endogenous NRF protein. The data of this study show that JKTBP1 and the 14-nt element act independently to mediate NRF IRES activity.


Asunto(s)
Regiones no Traducidas 5' , Proteínas de Unión al ADN/genética , Proteínas Represoras/genética , Ribonucleoproteínas/metabolismo , Ribosomas , Animales , Secuencia de Bases , Genes Reporteros , Humanos , Ratones , Datos de Secuencia Molecular , Conformación de Ácido Nucleico , Isoformas de Proteínas/metabolismo
17.
Biol Chem ; 386(12): 1287-93, 2005 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16336123

RESUMEN

Post-transcriptional mechanisms contribute to the changes in gene expression induced by cell stress. The effect of UV-B light on mRNA degradation in HeLa cells was investigated using a transcriptional chase system to determine the decay kinetics of tet-off vector-derived mRNAs containing or lacking a destabilizing AU-rich element. Degradation of both mRNAs was strongly inhibited in cells exposed to UV-B light. Removal of the poly(A)-tail, considered a crucial step in mRNA degradation, was strikingly impaired. UV light also inhibited deadenylation and degradation of endogenous mRNA of the chemoattractant cytokine interleukin (IL)-8. Both effects occurred rapidly and independently of newly induced genes. Importantly, stabilization of IL-8 mRNA was accompanied by a strong increase in the duration of IL-8 protein formation. Furthermore, general inhibition of protein synthesis, a hallmark of the response to cell stress, required far higher doses of UV-B than inhibition of mRNA deadenylation and degradation. The difference in sensitivity of cells to these effects of UV-B light establishes a dose range in which mRNA stabilization can lead to dramatically enhanced expression of proteins derived from normally unstable mRNAs, such as those of inflammatory cytokines, growth factors and proto-oncogenes, and thereby have a major impact on the response to UV light.


Asunto(s)
Adenina/metabolismo , Estabilidad del ARN/efectos de la radiación , ARN Mensajero/efectos de la radiación , Rayos Ultravioleta , Adenina/antagonistas & inhibidores , Citocinas/genética , Citocinas/metabolismo , Electroforesis en Gel de Poliacrilamida , Células HeLa/metabolismo , Células HeLa/efectos de la radiación , Humanos , Interleucina-8/genética , Interleucina-8/metabolismo , Cinética , Poli A/genética , Poli A/metabolismo , Estabilidad del ARN/genética , Estabilidad del ARN/fisiología , ARN Mensajero/antagonistas & inhibidores , ARN Mensajero/metabolismo
18.
Eur J Biochem ; 269(23): 5830-9, 2002 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-12444971

RESUMEN

mRNA stabilization plays an important role in the changes in protein expression initiated by inducers of inflammation or direct cell stress such as UV light. This study provides evidence that stabilization in response to UV light differs from that induced by proinflammatory stimuli such as bacterial lipopolysaccharide or interleukin (IL)-1. Firstly, UV-induced stabilization is independent of the p38 MAP kinase pathway, which has previously been shown to mediate stabilization induced by IL-1 or lipopolysaccharide. UV-induced mRNA stabilization was insensitive to the dominant negative forms of p38 MAP kinase and its substrate MAP kinase-activated protein kinase 2 (MK2), or to the p38 MAP kinase inhibitor SB 203580, demonstrating that it occurs through a different signaling mechanism. Secondly, UV-induced stabilization exhibits a different transcript selectivity. Activation of the p38 MAP kinase pathway, by expressing active MAP kinase kinase 6, induced stabilization only of transcripts containing AU-rich elements. UV light also induced stabilization of transcripts lacking AU-rich elements. This effect could not be mimicked by expressing MEKK1, an upstream activator of the p38, JNK, ERK and NF-kappaB pathways. UV light also stabilized endogenous histone mRNA, which lacks AU-rich elements and a poly(A) tail. This effect was not mimicked by active MAP kinase kinase 6 and not sensitive to a p38 MAP kinase inhibitor. This suggests that UV light induces stabilization through a mechanism that is independent of p38 MAP kinase and affects a broad spectrum of mRNAs.


Asunto(s)
Antígenos de Superficie , ARN Mensajero/efectos de la radiación , Rayos Ultravioleta , Proteínas ELAV , Proteína 1 Similar a ELAV , Ensayo de Cambio de Movilidad Electroforética , Activación Enzimática , Células HeLa , Histonas/genética , Humanos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , ARN Mensajero/química , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas de Unión al ARN/metabolismo , Transcripción Genética , Proteínas Quinasas p38 Activadas por Mitógenos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA