Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
J Clin Immunol ; 43(2): 371-390, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36282455

RESUMEN

PURPOSE: About 15% of patients with common variable immunodeficiency (CVID) develop a small intestinal enteropathy, which resembles celiac disease with regard to histopathology but evolves from a distinct, poorly defined pathogenesis that has been linked in some cases to chronic norovirus (NV) infection. Interferon-driven inflammation is a prominent feature of CVID enteropathy, but it remains unknown how NV infection may contribute. METHODS: Duodenal biopsies of CVID patients, stratified according to the presence of villous atrophy (VA), IgA plasma cells (PCs), and chronic NV infection, were investigated by flow cytometry, multi-epitope-ligand cartography, bulk RNA-sequencing, and RT-qPCR of genes of interest. RESULTS: VA development was connected to the lack of intestinal (IgA+) PC, a T helper 1/T helper 17 cell imbalance, and increased recruitment of granzyme+CD8+ T cells and pro-inflammatory macrophages to the affected site. A mixed interferon type I/III and II signature occurred already in the absence of histopathological changes and increased with the severity of the disease and in the absence of (IgA+) PCs. Chronic NV infection exacerbated this signature when compared to stage-matched NV-negative samples. CONCLUSIONS: Our study suggests that increased IFN signaling and T-cell cytotoxicity are present already in mild and are aggravated in severe stages (VA) of CVID enteropathy. NV infection preempts local high IFN-driven inflammation, usually only seen in VA, at milder disease stages. Thus, revealing the impact of different drivers of the pathological mixed IFN type I/III and II signature may allow for more targeted treatment strategies in CVID enteropathy and supports the goal of viral elimination.


Asunto(s)
Infecciones por Caliciviridae , Inmunodeficiencia Variable Común , Norovirus , Humanos , Atrofia/complicaciones , Atrofia/patología , Infecciones por Caliciviridae/inmunología , Linfocitos T CD8-positivos , Inmunodeficiencia Variable Común/complicaciones , Inmunodeficiencia Variable Común/inmunología , Inmunoglobulina A , Inflamación/complicaciones , Interferones , Norovirus/fisiología
2.
Virol J ; 17(1): 146, 2020 10 07.
Artículo en Inglés | MEDLINE | ID: mdl-33028368

RESUMEN

BACKGROUND: An effective vaccine that prevents disease caused by hantaviruses is a global public health priority, but up to now, no vaccine has been approved for worldwide use. Therefore, novel vaccines with high prophylaxis efficacy are urgently needed. METHODS: Herein, we designed and synthesized Hantaan virus (HTNV) linear multi-epitope peptide consisting of HLA-A*02-restricted HTNV cytotoxic T cell (CTL) epitope and pan HLA-DR-binding epitope (PADRE), and evaluated the immunogenicity, as well as effectiveness, of multi-epitope peptides in HLA-A2.1/Kb transgenic mice with interferon (IFN)-γ enzyme-linked immunospot assay, cytotoxic mediator detection, proliferation assay and HTNV-challenge test. RESULTS: The results showed that a much higher frequency of specific IFN-γ-secreting CTLs, high levels of granzyme B production, and a strong proliferation capacity of specific CTLs were observed in splenocytes of mice immunized with multi-epitope peptide than in those of a single CTL epitope. Moreover, pre-immunization of multi-epitope peptide could reduce the levels of HTNV RNA loads in the liver, spleen and kidneys of mice, indicating that specific CTL responses induced by multi-epitope peptide could reduce HTNV RNA loads in vivo. CONCLUSIONS: This study may provide an important foundation for the development of novel peptide vaccines for HTNV prophylaxis.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Epítopos de Linfocito T/inmunología , Antígeno HLA-A2/genética , Antígeno HLA-A2/inmunología , Virus Hantaan/inmunología , Fiebre Hemorrágica con Síndrome Renal/prevención & control , Vacunas contra la Malaria/inmunología , Animales , Antígenos Virales/genética , Antígenos Virales/inmunología , Epítopos de Linfocito T/genética , Virus Hantaan/genética , Inmunización , Vacunas contra la Malaria/administración & dosificación , Vacunas contra la Malaria/genética , Masculino , Ratones , Ratones Transgénicos , Linfocitos T Citotóxicos/inmunología , Vacunas de Subunidad/administración & dosificación , Vacunas de Subunidad/inmunología
3.
Artif Cells Nanomed Biotechnol ; 47(1): 665-673, 2019 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-30829072

RESUMEN

The present study was aimed to develop an effective nanoliposomal vaccine delivery system with P435 HER2/neu-derived peptide conjugated to Maleimide-PEG2000-DSPE. The nanoliposome formulation composed of DSPC/DSPG/Chol/DOPE and monophosphoryl lipid A was used as an adjuvant. Liposomal formulations were prepared and their physical properties were characterized. Anti-tumoral efficacy of formulations was evaluated by immunization of tumor-bearing BALB/c mice and the generated immune response was studied by using ELISpot and flow cytometry analysis. The results of the study demonstrated Lip + DOPE + P535 formulation caused the lowest tumor size and the longest survival time in TUBO mice model and could make it a promising candidate in developing effective vaccines against HER2-positive breast cancers.


Asunto(s)
Vacunas contra el Cáncer , Neoplasias Mamarias Experimentales , Nanopartículas , Péptidos , Receptor ErbB-2 , Animales , Vacunas contra el Cáncer/química , Vacunas contra el Cáncer/inmunología , Vacunas contra el Cáncer/farmacología , Línea Celular Tumoral , Femenino , Liposomas , Neoplasias Mamarias Experimentales/inmunología , Neoplasias Mamarias Experimentales/prevención & control , Ratones Endogámicos BALB C , Nanopartículas/química , Nanopartículas/uso terapéutico , Péptidos/química , Péptidos/inmunología , Péptidos/farmacología , Receptor ErbB-2/química , Receptor ErbB-2/inmunología , Receptor ErbB-2/uso terapéutico
4.
Front Immunol ; 10: 2, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30723467

RESUMEN

Robust anti-tumor immunity requires innate as well as adaptive immune responses. We have shown that plasmacytoid dendritic cells develop killer cell-like activity in melanoma cell cocultures after exposure to the infectious but replication-deficient herpes simplex virus 1 (HSV-1) d106S. To combine this innate effect with an enhanced adaptive immune response, the gene encoding human MelanA/MART-1 was inserted into HSV-1 d106S via homologous recombination to increase direct expression of this tumor antigen. Infection of Vero cells using this recombinant virus confirmed MelanA expression by Western blotting, flow cytometry, and immunofluorescence. HSV-1 d106S-MelanA induced expression of the transgene in fibroblast and melanoma cell lines not naturally expressing MelanA. Infection of a melanoma cell line with CRISPR-Cas9-mediated knockout of MelanA confirmed de novo expression of the transgene in the viral context. Dependent on MelanA expression, infected fibroblast and melanoma cell lines induced degranulation of HLA-matched MelanA-specific CD8+ T cells, followed by killing of infected cells. To study infection of immune cells, we exposed peripheral blood mononuclear cells and in vitro-differentiated macrophages to the parental HSV-1 d106S, resulting in expression of the transgene GFP in CD11c+ cells and macrophages. These data provide evidence that the application of MelanA-encoding HSV-1 d106S could enhance adaptive immune responses and re-direct MelanA-specific CD8+ T cells to tumor lesions, which have escaped adaptive immune responses via downregulation of their tumor antigen. Hence, HSV-1 d106S-MelanA harbors the potential to induce innate immune responses in conjunction with adaptive anti-tumor responses by CD8+ T cells, which should be evaluated in further studies.


Asunto(s)
Expresión Génica , Vectores Genéticos/genética , Herpesvirus Humano 1 , Antígeno MART-1/genética , Virus Oncolíticos , Transgenes , Células Presentadoras de Antígenos/inmunología , Células Presentadoras de Antígenos/metabolismo , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Línea Celular Tumoral , Fibroblastos/metabolismo , Marcación de Gen , Ingeniería Genética , Herpesvirus Humano 1/genética , Humanos , Leucocitos Mononucleares/inmunología , Leucocitos Mononucleares/metabolismo , Melanoma , Virus Oncolíticos/genética , Linfocitos T/inmunología , Linfocitos T/metabolismo
5.
J Control Release ; 266: 27-35, 2017 Nov 28.
Artículo en Inglés | MEDLINE | ID: mdl-28917531

RESUMEN

The skin is an attractive organ for immunization due to the presence of a large number of epidermal and dermal antigen-presenting cells. Hollow microneedles allow for precise and non-invasive intradermal delivery of vaccines. In this study, ovalbumin (OVA)-loaded poly(lactic-co-glycolic acid) (PLGA) nanoparticles with and without TLR3 agonist poly(I:C) were prepared and administered intradermally by hollow microneedles. The capacity of the PLGA nanoparticles to induce a cytotoxic T cell response, contributing to protection against intracellular pathogens, was examined. We show that a single injection of OVA-loaded PLGA nanoparticles, compared to soluble OVA, primed both adoptively transferred antigen-specific naïve transgenic CD8+ and CD4+ T cells with markedly high efficiency. Applying a triple immunization protocol, PLGA nanoparticles primed also endogenous OVA-specific CD8+ T cells. Immune response, following immunization with in particular anionic PLGA nanoparticles co-encapsulated with OVA and poly(I:C), provided protection against a recombinant strain of the intracellular bacterium Listeria monocytogenes, secreting OVA. Taken together, we show that PLGA nanoparticle formulation is an excellent delivery system for protein antigen into the skin and that protective cellular immune responses can be induced using hollow microneedles for intradermal immunizations.


Asunto(s)
Antígenos/administración & dosificación , Ácido Láctico/administración & dosificación , Nanopartículas/administración & dosificación , Agujas , Ovalbúmina/administración & dosificación , Poli I-C/administración & dosificación , Ácido Poliglicólico/administración & dosificación , Vacunación/instrumentación , Vacunas/administración & dosificación , Animales , Inyecciones Intradérmicas , Listeria monocytogenes/inmunología , Masculino , Ratones Transgénicos , Microinyecciones , Copolímero de Ácido Poliláctico-Ácido Poliglicólico , Linfocitos T/inmunología , Linfocitos T/trasplante , Receptor Toll-Like 3/agonistas , Vacunación/métodos
6.
Immunology ; 152(4): 660-676, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-28767147

RESUMEN

Growing evidence points to a deregulated response to Epstein-Barr virus (EBV) in the central nervous system of patients with multiple sclerosis (MS) as a possible cause of disease. We have investigated the response of a subpopulation of effector CD8+ T cells to EBV in 36 healthy donors and in 35 patients with MS in active and inactive disease. We have measured the expression of markers of degranulation, the release of cytokines, cytotoxicity and the regulation of effector functions by inhibitory receptors, such as programmed death 1 (PD-1) and human inhibitor receptor immunoglobulin-like transcript 2 (ILT2). We demonstrate that polyfunctional cytotoxic CD8+ CD57+ T cells are able to kill EBV-infected cells in healthy donors. In contrast, an anergic exhaustion-like phenotype of CD8+ CD57+ T cells with high expression of PD-1 was observed in inactive patients with MS compared with active patients with MS or healthy donors. Detection of CD8+ CD57+ T cells in meningeal inflammatory infiltrates from post-mortem MS tissue confirmed the association of this cell phenotype with the disease pathological process. The overall results suggest that ineffective immune control of EBV in patietns with MS during remission may be one factor preceding and enabling the reactivation of the virus in the central nervous system and may cause exacerbation of the disease.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Regulación de la Expresión Génica/inmunología , Herpesvirus Humano 4/inmunología , Esclerosis Múltiple/inmunología , Receptor de Muerte Celular Programada 1/inmunología , Adulto , Antígenos CD/inmunología , Antígenos CD57/inmunología , Linfocitos T CD8-positivos/patología , Femenino , Humanos , Receptor Leucocitario Tipo Inmunoglobulina B1/inmunología , Masculino , Persona de Mediana Edad , Esclerosis Múltiple/patología , Esclerosis Múltiple/virología
7.
Front Immunol ; 8: 1797, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29312318

RESUMEN

Hantaan virus (HTNV) infections can cause severe hemorrhagic fever with renal syndrome (HFRS) in humans, which is associated with high fatality rates. Cytotoxic T cell (CTL) responses contribute to virus elimination; however, to date, HLA class I allele-restricted HTNV glycoprotein (GP) epitopes recognized by CTLs have not been reported, limiting our understanding of CTL responses against HTNV infection in humans. In this study, 34 HTNV GP nine-mer epitopes that may bind to HLA-A*0201 molecules were predicted using the BIMAS and SYFPEITHI database. Seven of the epitopes were demonstrated to bind to HLA-A*0201 molecules with high affinity via the T2 cell binding assay and were successfully used to synthesize peptide/HLA-A*0201 tetramers. The results of tetramer staining showed that the frequencies of each epitope-specific CTL were higher in patients with milder HFRS, which indicated that the epitopes may induce protective CTL responses after HTNV infection. IFN-γ-enzyme-linked immunospot analysis further confirmed the immunoreactivity of epitopes by eliciting epitope-specific IFN-γ-producing CTL responses. In an HTNV challenge trial, significant inhibition of HTNV replication characterized by lower levels of antigens and RNA loads was observed in major target organs (liver, spleen, and kidneys) of HLA-A2.1/Kb transgenic mice pre-vaccinated with nonapeptides VV9 (aa8-aa16, VMASLVWPV), SL9 (aa996-aa1004, SLTECPTFL) and LL9 (aa358-aa366, LIWTGMIDL). Importantly, LL9 exhibited the best ability to induce protective CTL responses and showed a prominent effect on the kidneys, potentially preventing kidney injury after HTNV infection. Taken together, our results highlight that HTNV GP-derived HLA-A*0201-restricted epitopes could elicit protective CTL responses against the virus, and that epitope LL9 functions as an immunodominant protective epitope that may advance the design of safe and effective CTL-based HTNV peptide vaccines for humans.

8.
Cancer Cell Int ; 16: 45, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27330408

RESUMEN

BACKGROUND: Since cytotoxic T cell (CTL) response is the major cellular type in attacking tumor cells, most immunotherapy targets to manipulate the CTL response. Immunotherapies targeting melanoma-specific antigens (MAGEs), a group of tumor-specific shared antigen, have shown to be promising. Our previous study has shown that MAGE1/TBHSP70 and MAGE3/TBHSP70 could induce a robust immune response against B-16 melanoma cells in C57BL/6 mice. In this study, we used an animal model to further demonstrate MAGEs as a potential immunotherapy target for tumorigenesis in vivo. METHODS: In the current study, we developed a MAGE1/MAGE3/TBHSP70 recombinant protein vaccine and evaluated its protective efficacy against tumor development by challenge vaccine-immunized mice with MAGE-expressing human tumor cell lines in a Hu-PBL-SCID mouse model. The cellular immune reactions were monitored by ELISPOT and cytotoxicity assays. RESULTS: Splenocytes isolated from vaccine-immunized mice presented potent cytokine secretion capacity and CTL-specific cytotoxic. Vaccine-immunized mice had a significant tumor regression and prolonged survival compared with controls (both p < 0.05). In vitro, rMAGE1-MAGE3-TBHSP70 showed a potent tumor-antigen-specific immune response in both hepatocellular carcinoma and pulmonary carcinoma cell lines. CONCLUSION: This newly-developed recombinant protein vaccine may serve as a new immunotherapy for cancer.

9.
Cell Immunol ; 308: 35-43, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27318760

RESUMEN

Imiquimod, a toll-like receptor 7 (TLR7) agonist, is an active pharmaceutical ingredient (API) established for the topical treatment of several dermal cancerous and precancerous skin lesions. Within this work, the immunostimulatory effect of imiquimod is further exploited in a transcutaneous immunization (TCI) approach based on a solid nanoemulsion (SN) formulation. SN contains a combination of imiquimod with the model peptide antigen SIINFEKL as a novel approach to omit needle and syringe and optimize dermal antigen administration. Excipients including sucrose fatty acid esters and the pharmaceutically acceptable oils MCT (middle chain triglycerides), avocado oil, jojoba wax and squalene are high pressure homogenized together with the antigen SIINFEKL. Freeze drying was performed to eliminate water and to achieve spreadable properties of the formulation for dermal administration. The influence of the different oil components was assessed regarding in vitro drug permeation in a Franz diffusion cell model using a murine skin setup. In vivo performance in terms of cytotoxic T-cell response was assessed in a C57BL/6 mouse model. Whereas Aldara® cream contains imiquimod in a dissolved state, the SN formulations carry the active in a suspended state. This resulted in a reduction of imiquimod permeation across murine skin from the SN when compared to Aldara® cream. In spite of this permeation rate reduction, each SN induced an in vivo immune response by specific T-cell lysis. A stabilized solid nanosuspension containing squalene/tocopherol exhibited a significantly higher performance (p⩽0.05) in comparison with Aldara® cream. MCT based SN exerted an in vivo effect comparable to Aldara®. In conclusion, anhydrous highly dispersed vehicles containing imiquimod in a submicron particle size distribution can represent promising formulations for TCI. The choice of the oil component has a strong influence on SN performance, independent of in vitro drug permeation.


Asunto(s)
Adyuvantes Inmunológicos/administración & dosificación , Aminoquinolinas/uso terapéutico , Antineoplásicos/uso terapéutico , Sistemas de Liberación de Medicamentos , Células de Langerhans/inmunología , Nanoestructuras/administración & dosificación , Lesiones Precancerosas/tratamiento farmacológico , Neoplasias Cutáneas/tratamiento farmacológico , Adyuvantes Inmunológicos/química , Administración Cutánea , Animales , Química Farmacéutica , Humanos , Imiquimod , Inmunización , Ratones , Ratones Endogámicos C57BL , Nanoestructuras/química , Aceites/química , Vacunación
10.
Oncotarget ; 6(33): 34537-48, 2015 Oct 27.
Artículo en Inglés | MEDLINE | ID: mdl-26417929

RESUMEN

Induction of cytotoxic T lymphocytes (CTL) is critical to cancer vaccine based immunotherapy. Efforts to elicit CTLs against tumor MUC1 with peptide based vaccine have not been successful in clinical application. We have design a MUC1 vaccine by replacing B cell epitope of CTB with MUC1 VNTR peptide. Immunization with hybrid CTB-MUC1 plus aluminum hydroxide and CpG adujuvant (CTB-MUC1-Alum-CpG) induce MUC1-specific CTLs in mice. Moreover, this vaccination can prevent tumor growth and reduce tumor burden in MUC1+B16 mice model. Meanwhile, CTB-MUC1-Alum-CpG vaccination can promote Th1 cells and CD8+ T cells inflate to tumor tissue. Our approach might be applicable to other cancer vaccine design.


Asunto(s)
Vacunas contra el Cáncer/inmunología , Toxina del Cólera/inmunología , Mucina-1/inmunología , Linfocitos T Citotóxicos/inmunología , Adyuvantes Inmunológicos/farmacología , Animales , Presentación de Antígeno/inmunología , Western Blotting , Modelos Animales de Enfermedad , Epítopos de Linfocito B/inmunología , Femenino , Técnica del Anticuerpo Fluorescente , Activación de Linfocitos/inmunología , Ratones , Ratones Endogámicos C57BL , Repeticiones de Minisatélite/inmunología , Reacción en Cadena de la Polimerasa , Vacunas de Subunidad/inmunología
11.
World J Gastroenterol ; 21(12): 3480-91, 2015 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-25834312

RESUMEN

Hepatitis C virus (HCV)-specific cytotoxic T cell (CTL) response plays a major role in viral control during spontaneous infection resolution. These cells develop an exhausted and pro-apoptotic status during chronic onset, being unable to get rid of HCV. The role of this response in contributing to sustained viral response (SVR) after anti-HCV is controversial. Recent studies show that after successful interferon-based anti-HCV treatment, HCV traces are still detectable and this correlates with a peak of HCV-specific CTL response activation, probably responsible for maintaining SVR by subsequent complete HCV clearing. Moreover, SVR patients' serum is still able to induce HCV infection in naïve chimpanzees, suggesting that the infection could be under the control of the immune system after a successful treatment, being transmissible in absence of this adaptive response. At least theoretically, treatment-induced viral load decrease could allow an effective HCV-specific CTL response reestablishment. This effect has been recently described with anti-HCV interferon-free regimes, based on direct-acting antivirals. Nevertheless, this is to some extent controversial with interferon-based therapies, due to the detrimental immunoregulatory α-interferon effect on T cells. Moreover, HCV-specific CTL response features during anti-HCV treatment could be a predictive factor of SVR that could have clinical implications in patient management. In this review, the recent knowledge about the role of HCV-specific CTL response in the development of SVR after anti-HCV treatment is discussed.


Asunto(s)
Antivirales/uso terapéutico , Hepacivirus/efectos de los fármacos , Hepatitis C Crónica/tratamiento farmacológico , Interferones/uso terapéutico , Activación de Linfocitos/efectos de los fármacos , Linfocitos T Citotóxicos/efectos de los fármacos , Animales , Hepacivirus/inmunología , Hepatitis C Crónica/inmunología , Hepatitis C Crónica/virología , Humanos , Linfocitos T Citotóxicos/inmunología , Linfocitos T Citotóxicos/virología , Factores de Tiempo , Resultado del Tratamiento , Carga Viral
12.
Front Immunol ; 4: 487, 2014 Jan 07.
Artículo en Inglés | MEDLINE | ID: mdl-24432018

RESUMEN

Native type I heat-labile toxins (LTs) produced by enterotoxigenic Escherichia coli (ETEC) strains exert strong adjuvant effects on both antibody and T cell responses to soluble and particulate antigens following co-administration via mucosal routes. However, inherent enterotoxicity and neurotoxicity (following intra-nasal delivery) had reduced the interest in the use of these toxins as mucosal adjuvants. LTs can also behave as powerful and safe adjuvants following delivery via parenteral routes, particularly for activation of cytotoxic lymphocytes. In the present study, we evaluated the adjuvant effects of a new natural LT polymorphic form (LT2), after delivery via intradermal (i.d.) and subcutaneous (s.c.) routes, with regard to both antibody and T cell responses. A recombinant HIV-1 p24 protein was employed as a model antigen for determination of antigen-specific immune responses while the reference LT (LT1), produced by the ETEC H10407 strain, and a non-toxigenic LT form (LTK63) were employed as previously characterized LT types. LT-treated mice submitted to a four dose-base immunization regimen elicited similar p24-specific serum IgG responses and CD4(+) T cell activation. Nonetheless, mice immunized with LT1 or LT2 induced higher numbers of antigen-specific CD8(+) T cells and in vivo cytotoxic responses compared to mice immunized with the non-toxic LT derivative. These effects were correlated with stronger activation of local dendritic cell populations. In addition, mice immunized with LT1 and LT2, but not with LTK63, via s.c. or i.d. routes developed local inflammatory reactions. Altogether, the present results confirmed that the two most prevalent natural polymorphic LT variants (LT1 or LT2) display similar and strong adjuvant effects for subunit vaccines administered via i.d. or s.c. routes.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA