Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
Int J Mol Sci ; 22(10)2021 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-34067854

RESUMEN

Numerous viruses hijack cellular protein trafficking pathways to mediate cell entry or to rearrange membrane structures thereby promoting viral replication and antagonizing the immune response. Adaptor protein complexes (AP), which mediate protein sorting in endocytic and secretory transport pathways, are one of the conserved viral targets with many viruses possessing AP-interacting motifs. We present here different mechanisms of viral interference with AP complexes and the functional consequences that allow for efficient viral propagation and evasion of host immune defense. The ubiquity of this phenomenon is evidenced by the fact that there are representatives for AP interference in all major viral families, covered in this review. The best described examples are interactions of human immunodeficiency virus and human herpesviruses with AP complexes. Several other viruses, like Ebola, Nipah, and SARS-CoV-2, are pointed out as high priority disease-causative agents supporting the need for deeper understanding of virus-AP interplay which can be exploited in the design of novel antiviral therapies.


Asunto(s)
Proteínas Adaptadoras del Transporte Vesicular/metabolismo , VIH-1/metabolismo , Herpesviridae/metabolismo , SARS-CoV-2/metabolismo , Ebolavirus/metabolismo , Endocitosis , Humanos , Virus Nipah/metabolismo , Transporte de Proteínas , Liberación del Virus , Replicación Viral
2.
Eur J Immunol ; 43(8): 2151-61, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23640773

RESUMEN

The activity of NK cells is controlled by inhibitory and activating receptors. The inhibitory receptors interact mostly with MHC class I proteins, however, inhibitory receptors such as CD300a, which bind to non-MHC class I ligands, also exist. Recently, it was discovered that phosphatidylserine (PS) is a ligand for CD300a and that the interaction between PS expressed on apoptotic cells and CD300a inhibits the uptake of apoptotic cells by phagocytic cells. Whether PS can inhibit NK-cell activity through CD300a is unknown. Here, we have generated specific antibodies directed against CD300a and we used these mAbs to demonstrate that various NK-cell clones express different levels of CD300a. We further demonstrated that both CD300a and its highly homologous molecule CD300c bind to the tumor cells equally well and that they recognize PS and additional unknown ligand(s) expressed by tumor cells. Finally, we showed that blocking the PS-CD300a interaction resulted in increased NK-cell killing of tumor cells. Collectively, we demonstrate a new tumor immune evasion mechanism that is mediated through the interaction between PS and CD300a and we suggest that CD300c, similarly to CD300a, also interacts with PS.


Asunto(s)
Antígenos CD/metabolismo , Antígenos de Superficie/metabolismo , Células Asesinas Naturales/inmunología , Glicoproteínas de Membrana/metabolismo , Fosfatidilserinas/metabolismo , Receptores Inmunológicos/metabolismo , Apoptosis/inmunología , Células Cultivadas , Citotoxicidad Inmunológica , Antígenos de Histocompatibilidad Clase I/inmunología , Antígenos de Histocompatibilidad Clase I/metabolismo , Humanos , Hibridomas/metabolismo , Activación de Linfocitos , Fagocitosis , Escape del Tumor
3.
Eur J Immunol ; 43(8): 2138-50, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23677581

RESUMEN

The activity of natural killer (NK) cells is controlled by a balance of signals derived from inhibitory and activating receptors. TIGIT is a novel inhibitory receptor, recently shown in humans to interact with two ligands: PVR and Nectin2 and to inhibit human NK-cell cytotoxicity. Whether mouse TIGIT (mTIGIT) inhibits mouse NK-cell cytotoxicity is unknown. Here we show that mTIGIT is expressed by mouse NK cells and interacts with mouse PVR. Using mouse and human Ig fusion proteins we show that while the human TIGIT (hTIGIT) cross-reacts with mouse PVR (mPVR), the binding of mTIGIT is restricted to mPVR. We further demonstrate using surface plasmon resonance (SPR) and staining with Ig fusion proteins that mTIGIT binds to mPVR with higher affinity than the co-stimulatory PVR-binding receptor mouse DNAM1 (mDNAM1). Functionally, we show that triggering of mTIGIT leads to the inhibition of NK-cell cytotoxicity, that IFN-γ secretion is enhanced when mTIGIT is blocked and that the TIGIT-mediated inhibition is dominant over the signals delivered by the PVR-binding co-stimulatory receptors. Additionally, we identify the inhibitory motif responsible for mTIGIT inhibition. In conclusion, we show that TIGIT is a powerful inhibitory receptor for mouse NK cells.


Asunto(s)
Moléculas de Adhesión Celular/metabolismo , Citotoxicidad Inmunológica , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Receptores Inmunológicos/metabolismo , Receptores Virales/metabolismo , Secuencia de Aminoácidos , Animales , Antígenos CD/metabolismo , Antígenos de Diferenciación de Linfocitos T/metabolismo , Antígenos de Histocompatibilidad Clase I/inmunología , Interferón gamma/biosíntesis , Ratones , Ratones Endogámicos C57BL , Nectinas , Receptores Inmunológicos/química , Proteínas Recombinantes de Fusión/metabolismo , Alineación de Secuencia , Transducción de Señal , Resonancia por Plasmón de Superficie
4.
Front Immunol ; 14: 1231782, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37753084

RESUMEN

In recent years, reoviruses have been of major interest in immunotherapy because of their oncolytic properties. Preclinical and clinical trials, in which reovirus was used for the treatment of melanoma and glioblastoma, have paved the way for future clinical use of reovirus. However, little is known about how reovirus infection affects the tumor microenvironment and immune response towards infected tumor cells. Studies have shown that reovirus can directly stimulate natural killer (NK) cells, but how reovirus affects cellular ligands on tumor cells, which are ultimately key to tumor recognition and elimination by NK cells, has not been investigated. We tested how reovirus infection affects the binding of the NK Group-2 member D (NKG2D) receptor, which is a dominant mediator of NK cell anti-tumor activity. Using models of human-derived melanoma and glioblastoma tumors, we demonstrated that NKG2D ligands are downregulated in tumor cells post-reovirus-infection due to the impaired translation of these ligands in reovirus-infected cells. Moreover, we showed that downregulation of NKG2D ligands significantly impaired the binding of NKG2D to infected tumor cells. We further demonstrated that reduced recognition of NKG2D ligands significantly alters NK cell anti-tumor cytotoxicity in human primary NK cells and in the NK cell line NK-92. Thus, this study provides novel insights into reovirus-host interactions and could lead to the development of novel reovirus-based therapeutics that enhance the anti-tumor immune response.


Asunto(s)
Glioblastoma , Melanoma , Orthoreovirus , Infecciones por Reoviridae , Reoviridae , Humanos , Anticuerpos Antivirales , Glioblastoma/terapia , Ligandos , Melanoma/terapia , Subfamilia K de Receptores Similares a Lectina de Células NK , Microambiente Tumoral
5.
Nat Commun ; 14(1): 5016, 2023 08 18.
Artículo en Inglés | MEDLINE | ID: mdl-37596248

RESUMEN

TIGIT is an inhibitory receptor expressed on lymphocytes and can inhibit T cells by preventing CD226 co-stimulation through interactions in cis or through competition of shared ligands. Whether TIGIT directly delivers cell-intrinsic inhibitory signals in T cells remains unclear. Here we show, by analysing lymphocytes from matched human tumour and peripheral blood samples, that TIGIT and CD226 co-expression is rare on tumour-infiltrating lymphocytes. Using super-resolution microscopy and other techniques, we demonstrate that ligation with CD155 causes TIGIT to reorganise into dense nanoclusters, which coalesce with T cell receptor (TCR)-rich clusters at immune synapses. Functionally, this reduces cytokine secretion in a manner dependent on TIGIT's intracellular ITT-like signalling motif. Thus, we provide evidence that TIGIT directly inhibits lymphocyte activation, acting independently of CD226, requiring intracellular signalling that is proximal to the TCR. Within the subset of tumours where TIGIT-expressing cells do not commonly co-express CD226, this will likely be the dominant mechanism of action.


Asunto(s)
Activación de Linfocitos , Linfocitos Infiltrantes de Tumor , Humanos , Microscopía , Receptores Inmunológicos/genética , Transducción de Señal
6.
Cancer Gene Ther ; 29(11): 1676-1685, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-35681020

RESUMEN

Inhibitory receptors (IRs), such as the carcinoembryonic antigen-related cell adhesion molecule 1 (CEACAM1), are cell surface molecules expressed on both normal epithelial, endothelial, and hematopoietic cells and on neoplastic cells. IRs are usually used by cancer cells to inhibit immune cell functions. Thus, CEACAM1 positive tumor cells can interact homophilically with CEACAM1 expressed on T and NK cells to inhibit their antibody-dependent cell-mediated cytotoxicity (ADCC). In this study, we investigated the effect of agonistic/activating anti-CEACAM1 monoclonal antibody (mAb) on melanoma cell lines in vitro and in vivo, following our hypothesis that activation of CEACAM1 on melanoma cells by distinct mAbs may induce inhibition of cancer cell proliferation and/or their death. To address this, we established an activating anti-CEACAM1 mAb (CCM5.01) and characterized its binding to the CEACAM1 receptor. Using this mAb, we assessed the expression of CEACAM1 on four different human melanoma cell lines by western blot and flow cytometry and determined its effect on cell viability in vitro by MTT assay. Furthermore, we evaluated the mAb mechanism of action and found that binding of CEACAM1 with CCM5.01 induced SHP1 phosphorylation and p53 activation resulting in melanoma cell apoptosis. For in vivo studies, a xenograft model of melanoma was performed by injection of Mel-14 cells subcutaneously (s.c.) in SCID/Beige mice followed by intraperitoneal (i.p.) injection of CCM5.01 or of IgG1 isotype control every other day. CCM5.01 treated mice showed a slight but not significant decrease in tumor weight in comparison to the control group. Based on the obtained data, we suggest that activating CEACAM1 on melanoma cells might be a promising novel approach to fight cancers expressing this IR.


Asunto(s)
Anticuerpos Monoclonales , Melanoma , Humanos , Ratones , Animales , Anticuerpos Monoclonales/farmacología , Molécula 1 de Adhesión Celular , Ratones SCID , Línea Celular Tumoral , Melanoma/metabolismo , Antígeno Carcinoembrionario/metabolismo
7.
Front Mol Biosci ; 8: 745360, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34722634

RESUMEN

Despite the considerable progress toward the eradication of meningococcal disease with the introduction of glycoconjugate vaccines, previously unremarkable serogroup X has emerged in recent years, recording several outbreaks throughout the African continent. Different serogroup X polysaccharide-based vaccines have been tested in preclinical trials, establishing the principles for further improvement. To elucidate the antigenic determinants of the MenX capsular polysaccharide, we generated a monoclonal antibody, and its bactericidal nature was confirmed using the rabbit serum bactericidal assay. The antibody was tested by the inhibition enzyme-linked immunosorbent assay and surface plasmon resonance against a set of oligosaccharide fragments of different lengths. The epitope was shown to be contained within five to six α-(1-4) phosphodiester mannosamine repeating units. The molecular interactions between the protective monoclonal antibody and the MenX capsular polysaccharide fragment were further detailed at the atomic level by saturation transfer difference nuclear magnetic resonance (NMR) spectroscopy. The NMR results were used for validation of the in silico docking analysis between the X-ray crystal structure of the antibody (Fab fragment) and the modeled hexamer oligosaccharide. The antibody recognizes the MenX fragment by binding all six repeating units of the oligosaccharide via hydrogen bonding, salt bridges, and hydrophobic interactions. In vivo studies demonstrated that conjugates containing five to six repeating units can produce high functional antibody levels. These results provide an insight into the molecular basis of MenX vaccine-induced protection and highlight the requirements for the epitope-based vaccine design.

8.
J Exp Med ; 216(8): 1809-1827, 2019 08 05.
Artículo en Inglés | MEDLINE | ID: mdl-31142589

RESUMEN

CMVs efficiently target MHC I molecules to avoid recognition by cytotoxic T cells. However, the lack of MHC I on the cell surface renders the infected cell susceptible to NK cell killing upon missing self recognition. To counter this, mouse CMV (MCMV) rescues some MHC I molecules to engage inhibitory Ly49 receptors. Here we identify a new viral protein, MATp1, that is essential for MHC I surface rescue. Rescued altered-self MHC I molecules show increased affinity to inhibitory Ly49 receptors, resulting in inhibition of NK cells despite substantially reduced MHC I surface levels. This enables the virus to evade recognition by licensed NK cells. During evolution, this novel viral immune evasion mechanism could have prompted the development of activating NK cell receptors that are specific for MATp1-modified altered-self MHC I molecules. Our study solves a long-standing conundrum of how MCMV avoids recognition by NK cells, unravels a fundamental new viral immune evasion mechanism, and demonstrates how this forced the evolution of virus-specific activating MHC I-restricted Ly49 receptors.


Asunto(s)
Infecciones por Herpesviridae/inmunología , Antígenos de Histocompatibilidad Clase I/metabolismo , Evasión Inmune/inmunología , Células Asesinas Naturales/inmunología , Muromegalovirus/metabolismo , Subfamilia A de Receptores Similares a Lectina de Células NK/metabolismo , Proteínas Virales/metabolismo , Animales , Antígenos Ly/genética , Citotoxicidad Inmunológica , Modelos Animales de Enfermedad , Femenino , Fibroblastos/metabolismo , Infecciones por Herpesviridae/virología , Antígenos de Histocompatibilidad Clase I/inmunología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Transgénicos , Receptor 1 Gatillante de la Citotoxidad Natural/genética
10.
MAbs ; 8(3): 491-500, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26926266

RESUMEN

Pregnancy-specific glycoproteins (PSGs) are immunoglobulin superfamily members related to the carcinoembryonic antigen-related cell adhesion molecule (CEACAM) family and are encoded by 10 genes in the human. They are secreted at high levels by placental syncytiotrophoblast into maternal blood during pregnancy, and are implicated in immunoregulation, thromboregulation, and angiogenesis. To determine whether PSGs are expressed in tumors, we characterized 16 novel monoclonal antibodies to human PSG1 and used 2 that do not cross-react with CEACAMs to study PSG expression in tumors and in the gastrointestinal (GI) tract using tissue arrays and immunohistochemistry. Staining was frequently observed in primary squamous cell carcinomas and colonic adenocarcinomas and was correlated with the degree of tumor differentiation, being largely absent from metastatic samples. Staining was also observed in normal oesophageal and colonic epithelium. PSG expression in the human and mouse GI tract was confirmed using quantitative RT-PCR. However, mRNA expression was several orders of magnitude lower in the GI tract compared to placenta. Our results identify a non-placental site of PSG expression in the gut and associated tumors, with implications for determining whether PSGs have a role in tumor progression, and utility as tumor biomarkers.


Asunto(s)
Anticuerpos Monoclonales de Origen Murino , Anticuerpos Antineoplásicos , Biomarcadores de Tumor/inmunología , Neoplasias Gastrointestinales/inmunología , Regulación Neoplásica de la Expresión Génica/inmunología , Proteínas de Neoplasias/inmunología , Glicoproteínas beta 1 Específicas del Embarazo/inmunología , Adulto , Animales , Anticuerpos Monoclonales de Origen Murino/química , Anticuerpos Monoclonales de Origen Murino/inmunología , Anticuerpos Antineoplásicos/química , Anticuerpos Antineoplásicos/inmunología , Femenino , Neoplasias Gastrointestinales/patología , Células HeLa , Humanos , Inmunohistoquímica , Ratones , Embarazo
11.
Viruses ; 6(10): 3719-37, 2014 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-25275428

RESUMEN

Prion diseases or Transmissible Spongiform Encephalopathies (TSEs) are lethal neurodegenerative disorders involving the misfolding of the host encoded cellular prion protein, PrPC. This physiological form of the protein is expressed throughout the body, and it reaches the highest levels in the central nervous system where the pathology occurs. The conversion into the pathogenic isoform denoted as prion or PrPSc is the key event in prion disorders. Prominent candidates for the treatment of prion diseases are antibodies and their derivatives. Anti-PrPC antibodies are able to clear PrPSc from cell culture of infected cells. Furthermore, application of anti-PrPC antibodies suppresses prion replication in experimental animal models. Major drawbacks of immunotherapy are immune tolerance, the risks of neurotoxic side effects, limited ability of compounds to cross the blood-brain barrier and their unfavorable pharmacokinetic. The focus of this review is to recapitulate the current understanding of the molecular mechanisms for antibody mediated anti-prion activity. Although relevant for designing immunotherapeutic tools, the characterization of key antibody parameters shaping the molecular mechanism of the PrPC to PrPSc conversion remains elusive. Moreover, this review illustrates the various attempts towards the development of anti-PrP antibody compounds and discusses therapeutic candidates that modulate PrP expression.


Asunto(s)
Anticuerpos/inmunología , Proteínas PrPC/inmunología , Proteínas PrPSc/inmunología , Enfermedades por Prión/terapia , Animales , Expresión Génica , Humanos , Inmunoterapia , Proteínas PrPC/metabolismo , Proteínas PrPSc/metabolismo , Transporte de Proteínas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA