Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 66
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
BMC Cancer ; 24(1): 339, 2024 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-38486205

RESUMEN

BACKGROUND: Beta-(1,3)(1,6)-D-glucan is a complex polysaccharide, which is found in the cell wall of various fungi, yeasts, bacteria, algae, barley, and oats and has immunomodulatory, anticancer and antiviral effects. In the present study, we investigated the effect of beta-(1,3)(1,6)-D-glucan derived from yeast on the proliferation of primary NK cells and breast cancer cell lines in 2D and 3D models, and on the cytotoxicity of primary NK cells against breast cancer cell lines in 2D and 3D models. METHODS: In this study, we investigated the effects of different concentrations of yeast-derived beta-(1→3)(1→6)-D-glucan on the proliferation and cytotoxicity of human NK cells and breast cancer cell lines in 2D and 3D models using the XTT cell proliferation assay and the CellTiter-Glo® 2.0 assay to determine the cytotoxicity of human NK cells on breast cancer cell lines in 2D and 3D models. RESULTS: We found that the co-incubation of NK cells with beta-glucan in the absence of IL2 at 48 h significantly increased the proliferation of NK cells, whereas the co-incubation of NK cells with beta-glucan in the presence of IL2 (70 U/ml) increased the proliferation of NK cells but not significantly. Moreover, beta-glucan significantly inhibited the proliferation of breast cancer cell lines in 2D model and induced a weak, non-significant growth inhibitory effect on breast cancer multicellular tumor spheroids (3D). In addition, the cytotoxicity of NK cells against breast cancer cell lines was examined in 2D and 3D models, and beta-glucan significantly increased the cytotoxicity of NK cells against MCF-7 (in 2D). CONCLUSIONS: Yeast derived beta-(1,3)(1,6)-D-glucan could contribute to the treatment of cancer by enhancing NK cell immune response as well as contributing to inhibition of breast cancer cell growth.


Asunto(s)
Neoplasias de la Mama , beta-Glucanos , Humanos , Femenino , Células MCF-7 , Glucanos/farmacología , Neoplasias de la Mama/patología , Saccharomyces cerevisiae , Interleucina-2 , Células Asesinas Naturales , beta-Glucanos/farmacología
2.
Int J Mol Sci ; 24(10)2023 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-37240385

RESUMEN

There is a medical need to develop new and effective therapies against triple-negative breast cancer (TNBC). Chimeric antigen receptor (CAR) natural killer (NK) cells are a promising alternative to CAR-T cell therapy for cancer. A search for a suitable target in TNBC identified CD44v6, an adhesion molecule expressed in lymphomas, leukemias and solid tumors that is implicated in tumorigenesis and metastases. We have developed a next-generation CAR targeting CD44v6 that incorporates IL-15 superagonist and checkpoint inhibitor molecules. We could show that CD44v6 CAR-NK cells demonstrated effective cytotoxicity against TNBC in 3D spheroid models. The IL-15 superagonist was specifically released upon recognition of CD44v6 on TNBC and contributed to the cytotoxic attack. PD1 ligands are upregulated in TNBC and contribute to the immunosuppressive tumor microenvironment (TME). Competitive inhibition of PD1 neutralized inhibition by PD1 ligands expressed on TNBC. In total, CD44v6 CAR-NK cells are resistant to TME immunosuppression and offer a new therapeutic option for the treatment of BC, including TNBC.


Asunto(s)
Receptores Quiméricos de Antígenos , Neoplasias de la Mama Triple Negativas , Humanos , Neoplasias de la Mama Triple Negativas/patología , Interleucina-15/metabolismo , Ligandos , Línea Celular Tumoral , Células Asesinas Naturales , Inmunoterapia Adoptiva , Microambiente Tumoral
3.
Virus Genes ; 53(6): 762-773, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28451945

RESUMEN

Herpesviruses have acquired numerous genes from their hosts. Although these homologs are not essential for viral replication, they often have important immunomodulatory functions that ensure viral persistence in the host. Some of these viral molecules are called virokines as they mimic cellular cytokines of their host such as interleukin-10 (cIL-10). In recent years, many viral homologs of IL-10 (vIL-10s) have been discovered in the genome of members of the order Herpesvirales. For some, gene and protein structure as well as biological activity and potential use in the clinical context have been explored. Besides virokines, herpesviruses have also captured genes encoding membrane-bound host immunomodulatory proteins such as major histocompatibility complex (MHC) molecules. These viral MHC mimics also retain many of the functions of the cellular genes, in particular directly or indirectly modulating the activity of natural killer cells. The mechanisms underlying capture of cellular genes by large DNA viruses are still enigmatic. In this review, we provide an update of the advances in the field of herpesviral gene piracy and discuss possible scenarios that could explain how the gene transfer from host to viral genome was achieved.


Asunto(s)
Herpesviridae/genética , Interacciones Huésped-Patógeno/genética , Factores Inmunológicos/genética , Inmunomodulación/genética , Animales , Citocinas/genética , Humanos , Proteínas Virales/genética
4.
Proc Natl Acad Sci U S A ; 111(10): 3817-22, 2014 Mar 11.
Artículo en Inglés | MEDLINE | ID: mdl-24569863

RESUMEN

Insulin resistance and associated metabolic sequelae are common in chronic kidney disease (CKD) and are positively and independently associated with increased cardiovascular mortality. However, the pathogenesis has yet to be fully elucidated. 11ß-Hydroxysteroid dehydrogenase type 1 (11ßHSD1) catalyzes intracellular regeneration of active glucocorticoids, promoting insulin resistance in liver and other metabolic tissues. Using two experimental rat models of CKD (subtotal nephrectomy and adenine diet) which show early insulin resistance, we found that 11ßHSD1 mRNA and protein increase in hepatic and adipose tissue, together with increased hepatic 11ßHSD1 activity. This was associated with intrahepatic but not circulating glucocorticoid excess, and increased hepatic gluconeogenesis and lipogenesis. Oral administration of the 11ßHSD inhibitor carbenoxolone to uremic rats for 2 wk improved glucose tolerance and insulin sensitivity, improved insulin signaling, and reduced hepatic expression of gluconeogenic and lipogenic genes. Furthermore, 11ßHSD1(-/-) mice and rats treated with a specific 11ßHSD1 inhibitor (UE2316) were protected from metabolic disturbances despite similar renal dysfunction following adenine experimental uremia. Therefore, we demonstrate that elevated hepatic 11ßHSD1 is an important contributor to early insulin resistance and dyslipidemia in uremia. Specific 11ßHSD1 inhibitors potentially represent a novel therapeutic approach for management of insulin resistance in patients with CKD.


Asunto(s)
11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 1/metabolismo , Resistencia a la Insulina/fisiología , ARN Mensajero/metabolismo , Insuficiencia Renal Crónica/complicaciones , Uremia/enzimología , 11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 1/antagonistas & inhibidores , 11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 1/genética , Análisis de Varianza , Animales , Glucemia , Carbenoxolona/administración & dosificación , Carbenoxolona/farmacología , Corticosterona/sangre , Citocinas/sangre , Ensayo de Inmunoadsorción Enzimática , Glucocorticoides/metabolismo , Immunoblotting , Insulina/sangre , Hígado/metabolismo , Ratones , Ratones Noqueados , Ratas , Ratas Wistar , Reacción en Cadena en Tiempo Real de la Polimerasa , Uremia/etiología
5.
Proc Natl Acad Sci U S A ; 111(25): 9229-34, 2014 Jun 24.
Artículo en Inglés | MEDLINE | ID: mdl-24927527

RESUMEN

In the bone marrow, a population of memory T cells has been described that promotes efficient secondary immune responses and has been considered to be preactivated, owing to its expression of CD69 and CD25. Here we show that human bone marrow professional memory T cells are not activated but are resting in terms of proliferation, transcription, and mobility. They are in the G0 phase of the cell cycle, and their transcriptome is that of resting T cells. The repertoire of CD4(+) bone marrow memory T cells compared with CD4(+) memory T cells from the blood is significantly enriched for T cells specific for cytomegalovirus-pp65 (immunodominant protein), tetanus toxoid, measles, mumps, and rubella. It is not enriched for vaccinia virus and Candida albicans-MP65 (immunodominant protein), typical pathogens of skin and/or mucosa. CD4(+) memory T cells specific for measles are maintained nearly exclusively in the bone marrow. Thus, CD4(+) memory T cells from the bone marrow provide long-term memory for systemic pathogens.


Asunto(s)
Antígenos CD/inmunología , Antígenos de Diferenciación de Linfocitos T/inmunología , Células de la Médula Ósea/inmunología , Linfocitos T CD4-Positivos/inmunología , Memoria Inmunológica/fisiología , Subunidad alfa del Receptor de Interleucina-2/inmunología , Lectinas Tipo C/inmunología , Fase de Descanso del Ciclo Celular/inmunología , Adulto , Células de la Médula Ósea/citología , Linfocitos T CD4-Positivos/citología , Femenino , Humanos , Masculino , Persona de Mediana Edad
6.
J Immunol ; 192(9): 4294-302, 2014 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-24683192

RESUMEN

Invariant NKT cells (iNKT cells) are innate lymphocytes that recognize lipid-derived Ags presented by the MHC class I-related protein CD1d. In this study, we analyzed the role of iNKT cells in the generation of Abs against HSV type 1 (HSV-1). In sera from healthy hman donors, we found a correlation between HSV-1-specific IgG titers and proportions of CD4(+) iNKT cells. In HSV-1-infected iNKT cell-deficient mice, the amount of specific IgM and IgG Abs were significantly reduced compared with wild-type mice. Moreover, iNKT cell-deficient mice were unable to upregulate CD1d on B cells and failed to establish an IFN-γ-driven subtype profile of HSV-1-specific IgG Abs. In spleens of HSV-1-infected wild-type mice, the percentage of iNKT cells expressing CCR6, a marker for inflammatory iNKT cells secreting IFN-γ, was significantly decreased at 6 mo postinfection, suggesting that these cells were released from the spleen to other tissues. Finally, in vitro experiments showed that in the absence of CD1d-restricted cells, HSV-1 induced markedly lower IFN-γ production in splenocytes from naive mice. Taken together, our results indicate that iNKT cells shape the Ab response to HSV-1 infection and provide a basis for rational development of antiviral vaccines.


Asunto(s)
Anticuerpos Antivirales/inmunología , Herpes Simple/inmunología , Células T Asesinas Naturales/inmunología , Animales , Anticuerpos Antivirales/sangre , Western Blotting , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Humanos , Inmunoglobulina G/sangre , Inmunoglobulina G/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
7.
J Gen Virol ; 96(Pt 6): 1258-1263, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25678530

RESUMEN

Hantaviruses are emerging zoonotic pathogens that can cause severe disease in humans. Clinical observations suggest that human immune components contribute to hantavirus-induced pathology. To address this issue we generated mice with a humanized immune system. Hantavirus infection of these animals resulted in systemic infection associated with weight loss, decreased activity, ruffled fur and inflammatory infiltrates of lung tissue. Intriguingly, after infection, humanized mice harbouring human leukocyte antigen (HLA) class I-restricted human CD8+ T cells started to lose weight earlier (day 10) than HLA class I-negative humanized mice (day 15). Moreover, in these mice the number of human platelets dropped by 77 % whereas the number of murine platelets did not change, illustrating how differences between rodent and human haemato-lymphoid systems may contribute to disease development. To our knowledge this is the first description of a humanized mouse model of hantavirus infection, and our results indicate a role for human immune cells in hantaviral pathogenesis.


Asunto(s)
Modelos Animales de Enfermedad , Infecciones por Hantavirus/patología , Infecciones por Hantavirus/virología , Interacciones Huésped-Patógeno , Orthohantavirus/fisiología , Animales , Peso Corporal , Orthohantavirus/crecimiento & desarrollo , Orthohantavirus/patogenicidad , Humanos , Ratones SCID , Recuento de Plaquetas
8.
Eur J Immunol ; 43(10): 2566-76, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23824566

RESUMEN

Hantaviruses are emerging human pathogens. They induce an unusually strong antiviral response of human HLA class I (HLA-I) restricted CD8⁺ T cells that may contribute to tissue damage and hantavirus-associated disease. In this study, we analyzed possible hantaviral mechanisms that enhance the HLA-I antigen presentation machinery. Upon hantavirus infection of various human and primate cell lines, we observed transactivation of promoters controlling classical HLA molecules. Hantavirus-induced HLA-I upregulation required proteasomal activity and was associated with increased TAP expression. Intriguingly, human DCs acquired the capacity to cross-present antigen upon hantavirus infection. Furthermore, knockdown of TIR domain containing adaptor inducing IFN-ß or retinoic acid inducible gene I abolished hantavirus-driven HLA-I induction. In contrast, MyD88-dependent viral sensors were not involved in HLA-I induction. Our results show that hantaviruses strongly boost the HLA-I antigen presentation machinery by mechanisms that are dependent on both retinoic acid inducible gene I and TIR domain containing adaptor inducing IFN-ß.


Asunto(s)
Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Presentación de Antígeno , Células Dendríticas/inmunología , Infecciones por Hantavirus/inmunología , Orthohantavirus/inmunología , Receptores de Ácido Retinoico/metabolismo , Transportadoras de Casetes de Unión a ATP/metabolismo , Proteínas Adaptadoras del Transporte Vesicular/genética , Animales , Chlorocebus aethiops , Reactividad Cruzada/genética , Células Dendríticas/virología , Antígenos HLA/genética , Antígenos HLA/metabolismo , Humanos , Complejo de la Endopetidasa Proteasomal/metabolismo , ARN Interferente Pequeño/genética , Receptores de Ácido Retinoico/genética , Regulación hacia Arriba , Células Vero
9.
Cancers (Basel) ; 16(2)2024 Jan 16.
Artículo en Inglés | MEDLINE | ID: mdl-38254876

RESUMEN

Colorectal carcinoma (CRC) presents a formidable medical challenge, demanding innovative therapeutic strategies. Chimeric antigen receptor (CAR) natural killer (NK) cell therapy has emerged as a promising alternative to CAR T-cell therapy for cancer. A suitable tumor antigen target on CRC is carcinoembryonic antigen (CEA), given its widespread expression and role in tumorigenesis and metastasis. CEA is known to be prolifically shed from tumor cells in a soluble form, thus hindering CAR recognition of tumors and migration through the TME. We have developed a next-generation CAR construct exclusively targeting cell-associated CEA, incorporating a PD1-checkpoint inhibitor and a CCR4 chemokine receptor to enhance homing and infiltration of the CAR-NK-92 cell line through the TME, and which does not induce fratricidal killing of CAR-NK-92-cells. To evaluate this therapeutic approach, we harnessed intricate 3D multicellular tumor spheroid models (MCTS), which emulate key elements of the TME. Our results demonstrate the effective cytotoxicity of CEA-CAR-NK-92 cells against CRC in colorectal cell lines and MCTS models. Importantly, minimal off-target activity against non-cancerous cell lines underscores the precision of this therapy. Furthermore, the integration of the CCR4 migration receptor augments homing by recognizing target ligands, CCL17 and CCL22. Notably, our CAR design results in no significant trogocytosis-induced fratricide. In summary, the proposed CEA-targeting CAR-NK cell therapy could offer a promising solution for CRC treatment, combining precision and efficacy in a tailored approach.

10.
Front Immunol ; 14: 1166765, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37520530

RESUMEN

Introduction: The coronavirus disease 2019 (COVID-19) pandemic caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has highlighted the danger posed by human coronaviruses. Rapid emergence of immunoevasive variants and waning antiviral immunity decrease the effect of the currently available vaccines, which aim at induction of neutralizing antibodies. In contrast, T cells are marginally affected by antigen evolution although they represent the major mediators of virus control and vaccine protection against virus-induced disease. Materials and methods: We generated a multi-epitope vaccine (PanCoVac) that encodes the conserved T cell epitopes from all structural proteins of coronaviruses. PanCoVac contains elements that facilitate efficient processing and presentation of PanCoVac-encoded T cell epitopes and can be uploaded to any available vaccine platform. For proof of principle, we cloned PanCoVac into a non-integrating lentivirus vector (NILV-PanCoVac). We chose Roborovski dwarf hamsters for a first step in evaluating PanCoVac in vivo. Unlike mice, they are naturally susceptible to SARS-CoV-2 infection. Moreover, Roborovski dwarf hamsters develop COVID-19-like disease after infection with SARS-CoV-2 enabling us to look at pathology and clinical symptoms. Results: Using HLA-A*0201-restricted reporter T cells and U251 cells expressing a tagged version of PanCoVac, we confirmed in vitro that PanCoVac is processed and presented by HLA-A*0201. As mucosal immunity in the respiratory tract is crucial for protection against respiratory viruses such as SARS-CoV-2, we tested the protective effect of single-low dose of NILV-PanCoVac administered via the intranasal (i.n.) route in the Roborovski dwarf hamster model of COVID-19. After infection with ancestral SARS-CoV-2, animals immunized with a single-low dose of NILV-PanCoVac i.n. did not show symptoms and had significantly decreased viral loads in the lung tissue. This protective effect was observed in the early phase (2 days post infection) after challenge and was not dependent on neutralizing antibodies. Conclusion: PanCoVac, a multi-epitope vaccine covering conserved T cell epitopes from all structural proteins of coronaviruses, might protect from severe disease caused by SARS-CoV-2 variants and future pathogenic coronaviruses. The use of (HLA-) humanized animal models will allow for further efficacy studies of PanCoVac-based vaccines in vivo.


Asunto(s)
COVID-19 , Vacunas Virales , Cricetinae , Humanos , Animales , Ratones , COVID-19/prevención & control , SARS-CoV-2 , Vacunas contra la COVID-19 , Epítopos de Linfocito T , Administración Intranasal , Anticuerpos Neutralizantes , Antígenos HLA-A
11.
J Immunol ; 185(1): 488-97, 2010 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-20525895

RESUMEN

Virulent varicella-zoster virus (VZV) can spread in immunocompetent humans, resulting in symptoms mostly of the skin. In contrast, vaccine Oka (V-Oka), the attenuated VZV vaccine strain, only rarely causes clinical reactions. The mechanisms underlying these pathogenetic differences are unclear. In this study, we comparatively analyzed the ability of virulent VZV and V-Oka to modulate instruction of dendritic cells (DCs) by innate signals. DCs isolated from normal human skin were susceptible to infection with VZV and V-Oka. Moreover, inflammatory DCs, which play a crucial role in the stimulation of Th1 immune responses, accumulated in herpes zoster lesions. Infection of inflammatory DCs generated in vitro with virulent VZV or V-Oka resulted in upregulation of CD1c. Upon coculture with CD1c-restricted innate cells, DCs developed a mature phenotype whether infected with virulent VZV or V-Oka. Intriguingly, a striking difference was detected on the functional level. The release of IFN-gamma and IL-12, the signature cytokines of Th1 responses, was enhanced by V-Oka but blocked by virulent VZV. V-Oka and virulent VZV efficiently synergized with CD40L, eliminating the possibility that CD40 signaling was a target of VZV-associated immune evasion. Instead, virulent VZV selectively interfered with signaling through TLR2, which is known to sense VZV. Thus, virulent VZV subverts Th1-promoting instruction of human DCs by blocking TLR2-mediated innate signals that prime IL-12 production by DCs. Taken together, our results demonstrate a novel immune-evasion mechanism of virulent VZV that has been lost during the attenuation process leading to the VZV vaccine strain.


Asunto(s)
Vacuna contra la Varicela/inmunología , Células Dendríticas/inmunología , Células Dendríticas/virología , Herpesvirus Humano 3/inmunología , Herpesvirus Humano 3/patogenicidad , Vacunas contra Herpesvirus/inmunología , Transducción de Señal/inmunología , Células Cultivadas , Técnicas de Cocultivo , Células Dendríticas/citología , Herpes Zóster/inmunología , Herpes Zóster/virología , Herpesvirus Humano 3/aislamiento & purificación , Humanos , Evasión Inmune/inmunología , Interleucina-12/biosíntesis , Persona de Mediana Edad , Monocitos/citología , Monocitos/inmunología , Monocitos/virología , Células TH1/inmunología , Células TH1/virología , Vacunas Atenuadas/inmunología , Virulencia
12.
Immunol Rev ; 225: 163-89, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18837782

RESUMEN

SUMMARY: Hantaviruses are predominantly rodent-borne pathogens, although recently novel shrew-associated hantaviruses were found. Within natural reservoir hosts, hantairuses do not cause obvious pathogenetic effects; transmission to humans, however, can lead to hemorrhagic fever with renal syndrome or hantavirus cardiopulmonary syndrome, depending on the virus species involved. This review is focussed on the recent knowledge on hantavirus-induced immune responses in rodent reservoirs and humans and their impact on susceptibility, transmission, and outcome of hantavirus infections. In addition, this review incorporates a discussion on the potential role of direct cell-virus interactions in the pathogenesis of hantavirus infections in humans. Finally, questions for further research efforts on the immune responses in potential hantavirus reservoir hosts and humans are summarized.


Asunto(s)
Células Dendríticas/inmunología , Reservorios de Enfermedades/virología , Infecciones por Hantavirus/inmunología , Orthohantavirus/fisiología , Subgrupos de Linfocitos T/inmunología , Animales , Anticuerpos Antivirales/inmunología , Anticuerpos Antivirales/metabolismo , Apoptosis/inmunología , Células Dendríticas/metabolismo , Orthohantavirus/genética , Orthohantavirus/inmunología , Infecciones por Hantavirus/patología , Infecciones por Hantavirus/transmisión , Infecciones por Hantavirus/virología , Humanos , Inmunidad Activa , Inmunidad Innata , Integrinas/inmunología , Integrinas/metabolismo , Roedores/inmunología , Roedores/virología , Subgrupos de Linfocitos T/metabolismo , Factor A de Crecimiento Endotelial Vascular/inmunología , Factor A de Crecimiento Endotelial Vascular/metabolismo
13.
Cancers (Basel) ; 14(13)2022 Jun 29.
Artículo en Inglés | MEDLINE | ID: mdl-35804950

RESUMEN

Breast cancer is poorly immunogenic due to immunosuppressive mechanisms produced in part by the tumor microenvironment (TME). The TME is a peritumoral area containing significant quantities of (1) cancer-associated fibroblasts (CAF), (2) tumor-infiltrating lymphocytes (TIL) and (3) tumor-associated macrophages (TAM). This combination protects the tumor from effective immune responses. How these protective cell types are generated and how the changes in the developing tumor relate to these subsets is only partially understood. Immunotherapies targeting solid tumors have proven ineffective largely due to this protective TME barrier. Therefore, a better understanding of the interplay between the tumor, the tumor microenvironment and immune cells would both advance immunotherapeutic research and lead to more effective immunotherapies. This review will summarize the current understanding of the microenvironment of breast cancer giving implications for future immunotherapeutic strategies.

14.
Front Immunol ; 13: 1028972, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36275700

RESUMEN

Multiple Sclerosis (MS) is an autoimmune disease that is characterized by inflammation and demyelination of nerve cells. There is strong evidence that Epstein-Barr virus (EBV), a human herpesvirus infecting B cells, greatly increases the risk of subsequent MS. Intriguingly, EBV not only induces human interleukin-10 but also encodes a homologue of this molecule, which is a key anti-inflammatory cytokine of the immune system. Although EBV-encoded IL-10 (ebvIL-10) has a high amino acid identity with its cellular counterpart (cIL-10), it shows more restricted and partially weaker functionality. We propose that both EBV-induced cIL-10 and ebvIL-10 act in a temporally and functionally coordinated manner helping the pathogen to establish latency in B cells and, at the same time, to balance the function of antiviral T cells. As a result, the EBV load persisting in the immune system is kept at a constant but individually different level (set point). During this immunological tug of war between virus and host, however, MS can be induced as collateral damage if the set point is too high. Here, we discuss a possible role of ebvIL-10 and EBV-induced cIL-10 in EBV-driven pathogenesis of MS.


Asunto(s)
Infecciones por Virus de Epstein-Barr , Esclerosis Múltiple , Humanos , Aminoácidos/metabolismo , Antivirales/metabolismo , Herpesvirus Humano 4 , Interleucina-10/metabolismo , Esclerosis Múltiple/etiología
15.
J Gen Virol ; 92(Pt 9): 2191-2200, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21632559

RESUMEN

Hantaan virus (HTNV) causes severe human disease. The HTNV genome consists of three ssRNA segments of negative polarity that are complexed with viral nucleocapsid (N) protein. How the human innate immune system detects HTNV is unclear. RNA helicase retinoic acid-inducible gene I (RIG-I) does not sense genomic HTNV RNA. So far it has not been analysed whether pathogen-associated molecular patterns generated during the HTNV replication trigger RIG-I-mediated innate responses. Indeed, we found that knock-down of RIG-I in A549 cells, an alveolar epithelial cell line, increases HTNV replication and prevents induction of 2',5'-oligoadenylate synthetase, an interferon-stimulated gene. Moreover, overexpression of wild-type or constitutive active RIG-I in Huh7.5 cells lacking a functional RIG-I diminished HTNV virion production. Intriguingly, reporter assays revealed that in vitro-transcribed HTNV N RNA and expression of the HTNV N ORF triggers RIG-I signalling. This effect was completely blocked by the RNA-binding domain of vaccinia virus E3 protein, suggesting that dsRNA-like secondary structures of HTNV N RNA stimulate RIG-I. Finally, transfection of HTNV N RNA into A549 cells resulted in a 2 log-reduction of viral titres upon challenge with virus. Our study is the first demonstration that RIG-I mediates antiviral innate responses induced by HTNV N RNA during HTNV replication and interferes with HTNV growth.


Asunto(s)
ARN Helicasas DEAD-box/metabolismo , Virus Hantaan/fisiología , Interacciones Huésped-Patógeno , Replicación Viral , Línea Celular , Proteína 58 DEAD Box , ARN Helicasas DEAD-box/inmunología , Técnicas de Silenciamiento del Gen , Virus Hantaan/inmunología , Humanos , Receptores Inmunológicos
16.
J Virol ; 84(2): 1034-46, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19906927

RESUMEN

Herpes simplex virus type 1 (HSV-1) is one of the most frequent and successful human pathogens. It targets immature dendritic cells (iDCs) to interfere with the antiviral immune response. The mechanisms underlying apoptosis of HSV-1-infected iDCs are not fully understood. Previously, we have shown that HSV-1-induced apoptosis of iDCs is associated with downregulation of the cellular FLICE-inhibitory protein (c-FLIP), a potent inhibitor of caspase-8-mediated apoptosis. In this study, we prove that HSV-1 induces degradation of c-FLIP in a proteasome-independent manner. In addition, by using c-FLIP-specific small interfering RNA (siRNA) we show for the first time that downregulation of c-FLIP expression is sufficient to drive uninfected iDCs into apoptosis, underlining the importance of this molecule for iDC survival. Surprisingly, we also observed virus-induced c-FLIP downregulation in epithelial cells and many other cell types that do not undergo apoptosis after HSV-1 infection. Microarray analyses revealed that HSV-1-encoded latency-associated transcript (LAT) sequences, which can substitute for c-FLIP as an inhibitor of caspase-8-mediated apoptosis, are much less abundant in iDCs as compared to epithelial cells. Finally, iDCs infected with an HSV-1 LAT knockout mutant showed increased apoptosis when compared to iDCs infected with the corresponding wild-type HSV-1. Taken together, our results demonstrate that apoptosis of HSV-1-infected iDCs requires both c-FLIP downregulation and diminished expression of viral LAT.


Asunto(s)
Apoptosis/fisiología , Proteína Reguladora de Apoptosis Similar a CASP8 y FADD/metabolismo , Células Dendríticas , Regulación hacia Abajo , Herpesvirus Humano 1/patogenicidad , MicroARNs/metabolismo , Proteína Reguladora de Apoptosis Similar a CASP8 y FADD/genética , Línea Celular , Células Dendríticas/citología , Células Dendríticas/fisiología , Células Dendríticas/virología , Herpesvirus Humano 1/genética , Herpesvirus Humano 1/fisiología , Humanos , Latencia del Virus
17.
Viruses ; 13(1)2021 Jan 09.
Artículo en Inglés | MEDLINE | ID: mdl-33435377

RESUMEN

Human cytomegalovirus (HCMV) has been implicated in the development of human malignancies, for instance in colon cancer. Proteasome inhibitors were developed for cancer therapy and have also been shown to influence HCMV infection. The aim of this study was to investigate if proteasome inhibitors have therapeutic potential for colon carcinoma and how this is influenced by HCMV infection. We show by immunofluorescence and flow cytometry that the colon carcinoma cell line Caco-2 is susceptible to HCMV infection. Growth curve analysis as well as protein expression kinetics and quantitative genome analysis further confirm these results. HCMV has an anti-apoptotic effect on Caco-2 cells by inhibiting very early events of the apoptosis cascade. Further investigations showed that HCMV stabilizes the membrane potential of the mitochondria, which is typically lost very early during apoptosis. This stabilization is resistant to proteasome inhibitor Bortezomib treatment, allowing HCMV-infected cells to survive apoptotic signals. Our findings indicate a possible role of proteasome inhibitors in colon carcinoma therapy.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Bortezomib/farmacología , Infecciones por Citomegalovirus/metabolismo , Infecciones por Citomegalovirus/virología , Citomegalovirus/fisiología , Células CACO-2 , Muerte Celular , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Técnica del Anticuerpo Fluorescente , Genoma Humano , Interacciones Huésped-Patógeno/efectos de los fármacos , Humanos , Potencial de la Membrana Mitocondrial , Inhibidores de Proteasoma/farmacología
18.
J Am Soc Nephrol ; 20(9): 2075-84, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19608703

RESUMEN

Bicarbonate supplementation preserves renal function in experimental chronic kidney disease (CKD), but whether the same benefit occurs in humans is unknown. Here, we randomly assigned 134 adult patients with CKD (creatinine clearance [CrCl] 15 to 30 ml/min per 1.73 m(2)) and serum bicarbonate 16 to 20 mmol/L to either supplementation with oral sodium bicarbonate or standard care for 2 yr. The primary end points were rate of CrCl decline, the proportion of patients with rapid decline of CrCl (>3 ml/min per 1.73 m(2)/yr), and ESRD (CrCl <10 ml/min). Secondary end points were dietary protein intake, normalized protein nitrogen appearance, serum albumin, and mid-arm muscle circumference. Compared with the control group, decline in CrCl was slower with bicarbonate supplementation (5.93 versus 1.88 ml/min 1.73 m(2); P < 0.0001). Patients supplemented with bicarbonate were significantly less likely to experience rapid progression (9 versus 45%; relative risk 0.15; 95% confidence interval 0.06 to 0.40; P < 0.0001). Similarly, fewer patients supplemented with bicarbonate developed ESRD (6.5 versus 33%; relative risk 0.13; 95% confidence interval 0.04 to 0.40; P < 0.001). Nutritional parameters improved significantly with bicarbonate supplementation, which was well tolerated. This study demonstrates that bicarbonate supplementation slows the rate of progression of renal failure to ESRD and improves nutritional status among patients with CKD.


Asunto(s)
Acidosis/tratamiento farmacológico , Fallo Renal Crónico/tratamiento farmacológico , Estado Nutricional , Bicarbonato de Sodio/administración & dosificación , Acidosis/complicaciones , Acidosis/metabolismo , Administración Oral , Proteínas Sanguíneas/metabolismo , Creatinina/sangre , Proteínas en la Dieta/administración & dosificación , Progresión de la Enfermedad , Femenino , Humanos , Estimación de Kaplan-Meier , Fallo Renal Crónico/complicaciones , Fallo Renal Crónico/metabolismo , Masculino , Persona de Mediana Edad , Albúmina Sérica/metabolismo , Bicarbonato de Sodio/efectos adversos , Bicarbonato de Sodio/sangre , Resultado del Tratamiento
19.
J Am Soc Nephrol ; 20(11): 2412-25, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19797168

RESUMEN

In the setting of renal ischemia-reperfusion injury (IRI), the effect and mechanism of action of glucocorticoids are not well understood. In rat renal IRI, a single dose of dexamethasone administered before ischemia, or at the onset of reperfusion, ameliorated biochemical and histologic acute kidney injury after 24 h. Dexamethasone upregulated Bcl-xL, downregulated ischemia-induced Bax, inhibited caspase-9 and caspase-3 activation, and reduced apoptosis and necrosis of proximal tubular cells. In addition, dexamethasone decreased the number of infiltrating neutrophils and ICAM-1. We observed the protective effect of dexamethasone in neutrophil-depleted mice, suggesting a neutrophil-independent mechanism. In vitro, dexamethasone protected human kidney proximal tubular (HK-2) cells during serum starvation and IRI-induced apoptosis, but inhibition of MEK 1/2 abolished its anti-apoptotic effects in these conditions. Dexamethasone stimulated rapid and transient phosphorylation of ERK 1/2, which required the presence of the glucocorticoid receptor and was independent of transcriptional activity. In summary, in the setting of renal ischemia-reperfusion injury, dexamethasone directly protects against kidney injury by a receptor-dependent, nongenomic mechanism.


Asunto(s)
Dexametasona/uso terapéutico , Glucocorticoides/uso terapéutico , Riñón/irrigación sanguínea , Daño por Reperfusión/tratamiento farmacológico , Animales , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Neutrófilos/efectos de los fármacos , Neutrófilos/fisiología , Ratas , Ratas Wistar
20.
Adv Biol Regul ; 77: 100741, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32773102

RESUMEN

Pandemic coronavirus disease 2019 (COVID-19) is caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and poses an unprecedented challenge to healthcare systems due to the lack of a vaccine and specific treatment options. Accordingly, there is an urgent need to understand precisely the pathogenic mechanisms underlying this multifaceted disease. There is increasing evidence that the immune system reacts insufficiently to SARS-CoV-2 and thus contributes to organ damage and to lethality. In this review, we suggest that the overwhelming production of reactive oxygen species (ROS) resulting in oxidative stress is a major cause of local or systemic tissue damage that leads to severe COVID-19. It increases the formation of neutrophil extracellular traps (NETs) and suppresses the adaptive arm of the immune system, i.e. T cells that are necessary to kill virus-infected cells. This creates a vicious cycle that prevents a specific immune response against SARS-CoV-2. The key role of oxidative stress in the pathogenesis of severe COVID-19 implies that therapeutic counterbalancing of ROS by antioxidants such as vitamin C or NAC and/or by antagonizing ROS production by cells of the mononuclear phagocyte system (MPS) and neutrophil granulocytes and/or by blocking of TNF-α can prevent COVID-19 from becoming severe. Controlled clinical trials and preclinical models of COVID-19 are needed to evaluate this hypothesis.


Asunto(s)
Antioxidantes/uso terapéutico , Infecciones por Coronavirus/epidemiología , Trampas Extracelulares/inmunología , Linfopenia/epidemiología , Neutrófilos/inmunología , Pandemias , Neumonía Viral/epidemiología , Acetilcisteína/uso terapéutico , Ácido Ascórbico/uso terapéutico , Betacoronavirus/inmunología , Betacoronavirus/patogenicidad , COVID-19 , Infecciones por Coronavirus/tratamiento farmacológico , Infecciones por Coronavirus/inmunología , Infecciones por Coronavirus/virología , Citocinas/genética , Citocinas/inmunología , Trampas Extracelulares/efectos de los fármacos , Trampas Extracelulares/metabolismo , Interacciones Huésped-Patógeno/efectos de los fármacos , Interacciones Huésped-Patógeno/genética , Interacciones Huésped-Patógeno/inmunología , Humanos , Inmunidad Innata/efectos de los fármacos , Linfopenia/tratamiento farmacológico , Linfopenia/inmunología , Linfopenia/virología , FN-kappa B/genética , FN-kappa B/inmunología , Neutrófilos/efectos de los fármacos , Neutrófilos/virología , Estrés Oxidativo/efectos de los fármacos , Neumonía Viral/tratamiento farmacológico , Neumonía Viral/inmunología , Neumonía Viral/virología , Especies Reactivas de Oxígeno/antagonistas & inhibidores , Especies Reactivas de Oxígeno/inmunología , Especies Reactivas de Oxígeno/metabolismo , SARS-CoV-2 , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología , Linfocitos T/virología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA