Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 188
Filtrar
1.
J Control Release ; 370: 379-391, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38697317

RESUMEN

Although various types of mRNA-based vaccines have been explored, the optimal conditions for induction of both humoral and cellular immunity remain rather unknown. In this study, mRNA vaccines of nucleoside-modified mRNA in lipoplexes (LPXs) or lipid nanoparticles (LNPs) were evaluated after administration in mice through different routes, assessing mRNA delivery, tolerability and immunogenicity. In addition, we investigated whether mRNA vaccines could benefit from the inclusion of the adjuvant alpha-galactosylceramide (αGC), an invariant Natural Killer T (iNKT) cell ligand. Intramuscular (IM) vaccination with ovalbumin (OVA)-encoding mRNA encapsulated in LNPs adjuvanted with αGC showed the highest antibody- and CD8+ T cell responses. Furthermore, we observed that addition of signal peptides and endocytic sorting signals of either LAMP1 or HLA-B7 in the OVA-encoding mRNA sequence further enhanced CD8+ T cell activation although reducing the induction of IgG antibody responses. Moreover, mRNA LNPs with the ionizable lipidoid C12-200 exhibited higher pro-inflammatory- and reactogenic activity compared to mRNA LNPs with SM-102, correlating with increased T cell activation and antitumor potential. We also observed that αGC could further enhance the cellular immunity of clinically relevant mRNA LNP vaccines, thereby promoting therapeutic antitumor potential. Finally, a Listeria monocytogenes mRNA LNP vaccine supplemented with αGC showed synergistic protective effects against listeriosis, highlighting a key advantage of co-activating iNKT cells in antibacterial mRNA vaccines. Taken together, our study offers multiple insights for optimizing the design of mRNA vaccines for disease applications, such as cancer and intracellular bacterial infections.


Asunto(s)
Vacunas contra el Cáncer , Galactosilceramidas , Ratones Endogámicos C57BL , Nanopartículas , Ovalbúmina , Animales , Galactosilceramidas/administración & dosificación , Galactosilceramidas/química , Vacunas contra el Cáncer/administración & dosificación , Vacunas contra el Cáncer/inmunología , Femenino , Nanopartículas/química , Nanopartículas/administración & dosificación , Ovalbúmina/inmunología , Ovalbúmina/administración & dosificación , Vacunas de ARNm , Adyuvantes Inmunológicos/administración & dosificación , Linfocitos T CD8-positivos/inmunología , ARN Mensajero/administración & dosificación , Ratones , Vacunas Bacterianas/administración & dosificación , Vacunas Bacterianas/inmunología , Neoplasias/inmunología , Neoplasias/terapia , Lípidos/química , Liposomas
2.
Dev Comp Immunol ; 114: 103843, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-32871161

RESUMEN

Influenza A viruses (IAV) are a major cause of respiratory diseases in pigs. Invariant natural killer T (iNKT) cells are an innate-like T cell subset that contribute significantly to IAV resistance in mice. In the current work, we explored whether expanding and activating iNKT cells with the iNKT cell superagonist α-galactosylceramide (α-GalCer) would change the course of an IAV infection in pigs. In one study, α-GalCer was administered to pigs intramuscularly (i.m.) 9 days before infection, which systemically expanded iNKT cells. In another study, α-GalCer was administered intranasally (i.n.) 2 days before virus infection to activate mucosal iNKT cells. Despite a synergistic increase in iNKT cells when α-GalCer i.m. treated pigs were infected with IAV, neither approach reduced disease signs, lung pathology, or virus replication. Our results indicate that prophylactic use of iNKT cell agonists to prevent IAV infection is ineffective in pigs. This is significant because this type of approach has been considered for humans whose iNKT cell levels and IAV infections are more similar to those of pigs than mice.


Asunto(s)
Galactosilceramidas/administración & dosificación , Virus de la Influenza A/fisiología , Gripe Humana/inmunología , Pulmón/patología , Mucosa Nasal/inmunología , Células T Asesinas Naturales/inmunología , Infecciones por Orthomyxoviridae/inmunología , Porcinos/inmunología , Animales , Humanos , Inyecciones Intramusculares , Activación de Linfocitos , Ratones , Eficacia de las Vacunas , Replicación Viral
3.
Front Immunol ; 11: 581301, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33193386

RESUMEN

Murine and human invariant natural killer T (iNKT) lymphocytes are activated by α-galactosylceramide (α-GalCer) presented on CD1d. α-GalCer was first described as a lipid that had strong anti-metastatic effects in a mouse melanoma model, and it has subsequently been shown to induce efficient iNKT cell dependent tumor immunity in several tumor models. We have shown that α-GalCer treatment leads to a weak reduction of polyp burden in the autochthonous ApcMin/+ mouse model for human colon cancer, however this treatment resulted in upregulation of the inhibitory receptor PD-1 on iNKT cells. While anti-PD-1 treatment can prevent immune-suppression in other cancer types, human colon cancer is generally resistant to this treatment. Here we have used the ApcMin/+ model to investigate whether a combined treatment with α-GalCer and PD-1 blockade results in improved effects on polyp development. We find that PD-1 expression was high on T cells in polyps and lamina propria (LP) of ApcMin/+ mice compared to polyp free Apc+/+ littermates. Anti-PD-1 treatment alone promoted Tbet expression in iNKT cells and CD4 T cells, but did not significantly reduce polyp numbers. However, the combined treatment with anti-PD-1 and α-GalCer had synergistic effects, resulting in highly significant reduction of polyp numbers in the small and large intestine. Addition of PD-1 blockade to α-GalCer treatment prevented loss of iNKT cells that were skewed towards a TH1-like iNKT1 phenotype specifically in polyps. It also resulted in TH1 skewing and increased granzyme B expression of CD4 T cells. Taken together this demonstrates that a combination of immune stimulation targeting iNKT cells and checkpoint blockade may be a promising approach to develop for improved tumor immunotherapy.


Asunto(s)
Neoplasias del Colon/inmunología , Neoplasias del Colon/prevención & control , Galactosilceramidas/administración & dosificación , Células T Asesinas Naturales/inmunología , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Proteína de la Poliposis Adenomatosa del Colon/deficiencia , Proteína de la Poliposis Adenomatosa del Colon/genética , Animales , Anticuerpos Bloqueadores/administración & dosificación , Femenino , Humanos , Mucosa Intestinal/inmunología , Pólipos Intestinales/inmunología , Pólipos Intestinales/prevención & control , Macrófagos/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Neoplasias Experimentales/inmunología , Neoplasias Experimentales/prevención & control , Receptor de Muerte Celular Programada 1/inmunología , Linfocitos T Reguladores/inmunología , Células TH1/inmunología
4.
Front Immunol ; 11: 2043, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32973811

RESUMEN

Active co-delivery of tumor antigens (Ag) and α-galactosylceramide (α-GalCer), a potent agonist for invariant Natural Killer T (iNKT) cells, to cross-priming CD8α+ dendritic cells (DCs) was previously shown to promote strong anti-tumor responses in mice. Here, we designed a nanoparticle-based vaccine able to target human CD141+ (BDCA3+) DCs - the equivalent of murine CD8α+ DCs - and deliver both tumor Ag (Melan A) and α-GalCer. This nanovaccine was inoculated into humanized mice that mimic the human immune system (HIS) and possess functional iNKT cells and CD8+ T cells, called HIS-CD8/NKT mice. We found that multiple immunizations of HIS-CD8/NKT mice with the nanovaccine resulted in the activation and/or expansion of human CD141+ DCs and iNKT cells and ultimately elicited a potent Melan-A-specific CD8+ T cell response, as determined by tetramer staining and ELISpot assay. Single-cell proteomics further detailed the highly polyfunctional CD8+ T cells induced by the nanovaccine and revealed their predictive potential for vaccine potency. This finding demonstrates for the first time the unique ability of human iNKT cells to license cross-priming DCs in vivo and adds a new dimension to the current strategy of cancer vaccine development.


Asunto(s)
Antígenos de Neoplasias/inmunología , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Epítopos de Linfocito T/inmunología , Galactosilceramidas/administración & dosificación , Trombomodulina/metabolismo , Animales , Antígenos de Neoplasias/administración & dosificación , Biomarcadores , Vacunas contra el Cáncer/inmunología , Humanos , Inmunofenotipificación , Lectinas Tipo C/antagonistas & inhibidores , Lectinas Tipo C/inmunología , Ratones , Ratones Endogámicos NOD , Ratones Transgénicos , Proteómica/métodos , Receptores Mitogénicos/antagonistas & inhibidores , Receptores Mitogénicos/inmunología , Análisis de la Célula Individual , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo
5.
Clin Immunol ; 215: 108457, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32387537

RESUMEN

We conducted a phase I study of the trans-bronchial injection of α-galactosylceramide (αGalCer)-pulsed antigen presenting cells (APCs) to evaluate their safety, immune responses, and anti-tumor activities. Patients with advanced or recurrent non-small cell lung cancer (NSCLC) refractory to standard treatments were eligible. αGalCer-pulsed APCs were administered intratumorally or intranodally by bronchoscopy. Twenty-one patients were enrolled in this study. No severe adverse events related to the cell therapy were observed during this study in any patient. After αGalCer-pulsed APCs were administrated, increased iNKT cell numbers were observed in PBMCs from eight cases, and IFN-γ producing cells were increased in the peripheral blood of 10 cases. Regarding clinical responses, one case exhibited a partial response and eight were classified as stable disease. In the tumor microenvironment, IFN-γ expression was upregulated after treatment in partial response or stable disease cases and TGF-ß was upregulated in progressive disease cases.


Asunto(s)
Células Presentadoras de Antígenos/inmunología , Bronquios/inmunología , Galactosilceramidas/administración & dosificación , Galactosilceramidas/inmunología , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/terapia , Adulto , Anciano , Carcinoma de Pulmón de Células no Pequeñas/inmunología , Carcinoma de Pulmón de Células no Pequeñas/terapia , Femenino , Humanos , Inmunoterapia/métodos , Interferón gamma/inmunología , Masculino , Persona de Mediana Edad , Células T Asesinas Naturales/inmunología , Recurrencia Local de Neoplasia/inmunología , Recurrencia Local de Neoplasia/terapia , Microambiente Tumoral/inmunología
6.
Iran J Allergy Asthma Immunol ; 19(1): 35-44, 2020 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-32245319

RESUMEN

Whether different injection modes of α-galactosylceramide (α-GalCer) affect the activation of different subsets of invariant natural killer T (iNKT) cells in different tissues and organs of mice is unclear. This study included healthy control, subcutaneous injection, and intraperitoneal injection groups (n=10 in each group). The subcutaneous and intraperitoneal injection groups were injected with α-Galcer (0.1 mg/kg weight), and then the changes in thymus, spleen, and liver iNKT cell frequencies and subsets were observed. The intraperitoneal injection of α-GalCer could increase the frequency of splenic iNKT cells, but the subcutaneous injection did not affect the frequency. Neither injection had any effect on the frequency of iNKT cells in the thymus and liver. The subcutaneous injection of α-GalCer increased the rate of iNKT2 subsets in the thymus but did not affect the rate of iNKT1 subsets. However, the intraperitoneal injection of α-GalCer did not affect thymus iNKT1 and iNKT2 subsets. Interestingly, the subcutaneous injection of α-GalCer significantly increased the proportion of iNKT1 in the spleen and liver but did not significantly change the proportion of iNKT2. The intraperitoneal injection of α-GalCer significantly increased the rate of iNKT2 in spleen and liver but decreased the rate of iNKT1. Subsets of iNKT1 or iNKT2 cells in the spleen and liver were selectively activated by the subcutaneous or intraperitoneal injection of α-GalCer. It provides a valuable means for treating tumors and certain autoimmune diseases. Further exploration of the activation mechanism may provide new ideas about the development of related vaccines.


Asunto(s)
Galactosilceramidas/administración & dosificación , Activación de Linfocitos/efectos de los fármacos , Células T Asesinas Naturales/efectos de los fármacos , Subgrupos de Linfocitos T/efectos de los fármacos , Animales , Galactosilceramidas/inmunología , Inyecciones Intraperitoneales , Inyecciones Subcutáneas , Hígado/efectos de los fármacos , Hígado/inmunología , Activación de Linfocitos/inmunología , Ratones Endogámicos C57BL , Células T Asesinas Naturales/inmunología , Bazo/efectos de los fármacos , Bazo/inmunología , Subgrupos de Linfocitos T/inmunología , Timo/efectos de los fármacos , Timo/inmunología
7.
Mol Immunol ; 121: 167-185, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32229377

RESUMEN

Some studies have shown that maturation of dendritic cells (DCs) is modulated directly by pathogen components via pattern recognition receptors such as Toll-like receptors, but also by signal like CD40 ligand (CD40 L or CD154) mediated by activated T cells. Several reports indicate that invariant natural killer T (iNKT) cells up-regulate CD40 L upon stimulation and thereby induce activation and maturation of DCs through crosslink with CD40. Our previous findings indicated that iNKT cells promote Th2 cell responses through the induction of immunogenic maturation of lung DCs (LDCs) in the asthmatic murine, but its mechanism remains unclear. Therefore, we investigated the immunomodulatory effects of blockade of CD40 L using anti-CD40 L treatment on Th2 cell responses and immunogenic maturation of LDCs, and further analyzed whether these influences of blockade of CD40 L were related to lung iNKT cells using iNKT cell-deficient mice and the combination treatment of specific iNKT cell activation with anti-CD40 L treatment in murine models of asthma. Our findings showed that blockade of CD40 L using anti-CD40 L treatment attenuated Th2 cell responses in wild-type (WT) mice, but not in CD1d-deficient mice sensitized and challenged with ovalbumin (OVA) or house dust mite (HDM). Meanwhile, blockade of CD40 L down-regulated immunogenic maturation of LDCs in WT mice, but not in CD1d-deficient mice sensitized and challenged with OVA. Additionally, agonistic anti-CD40 treatment reversed the inhibitory effects of anti-CD40 L treatment on Th2 cell responses and LDC activation in an OVA-induced mouse model of asthma. Furthermore, LDCs from asthmatic mice treated with anti-CD40 L could significantly reduce the influence on Th2 cell responses in vivo and in vitro. Finally, α-Galactosylceramide plus anti-CD40 L treatment stimulated lung iNKT cells, but suppressed Th2 cell responses in the asthmatic mice. Taken together, our data raise an evidence that blockade of CD40 L attenuates Th2 cell responses through the inhibition of immunogenic maturation of LDCs, which may be at least partially related to lung iNKT cells in murine models of asthma.


Asunto(s)
Asma/tratamiento farmacológico , Ligando de CD40/antagonistas & inhibidores , Células Dendríticas/inmunología , Células T Asesinas Naturales/efectos de los fármacos , Células Th2/inmunología , Animales , Antígenos CD1d/genética , Asma/inmunología , Asma/patología , Ligando de CD40/inmunología , Ligando de CD40/metabolismo , Comunicación Celular/inmunología , Modelos Animales de Enfermedad , Femenino , Galactosilceramidas/administración & dosificación , Humanos , Pulmón/citología , Pulmón/inmunología , Pulmón/patología , Ratones , Ratones Noqueados , Células T Asesinas Naturales/inmunología , Células T Asesinas Naturales/metabolismo , Ovalbúmina/inmunología , Pyroglyphidae/inmunología
8.
Mol Pharm ; 17(2): 417-425, 2020 02 03.
Artículo en Inglés | MEDLINE | ID: mdl-31841011

RESUMEN

Constructing an effective therapeutic cancer vaccine is very attractive and promising for cancer immunotherapy. However, the poor immunogenicity of tumor antigens and suppression of the immune system in the tumor microenvironment are two major obstacles for developing effective cancer vaccines. Invariant NKT cells (iNKT cells), which are essential bridges between the innate and adaptive immune systems, can be rapidly activated by their agonists and, consequently, evoke whole immune systems. Herein, we conjugated a potent agonist of the iNKT cell, α-galactosylceramide (α-GalCer), with the tumor-associated MUC1 glycopeptide antigens as novel self-adjuvanting cancer vaccines through click chemistry. Immunological studies revealed that the mouse immune system was potently evoked and that high levels of tumor-specific IgG antibodies were elicited by vaccine conjugates without an external adjuvant. The produced antibodies could specifically recognize and bind to antigen-expressing cancer cells and, subsequently, induce cytotoxicity through complement-dependent cytotoxicity. Thus, the insertion of α-GalCer significantly improved the immunogenicity of the MUC1 glycopeptide and induced strong antigen-specific antitumor responses, indicating that α-GalCer is an effective built-in adjuvant for constructing potent chemical synthetic antitumor vaccines.


Asunto(s)
Adyuvantes Inmunológicos/administración & dosificación , Vacunas contra el Cáncer/inmunología , Galactosilceramidas/administración & dosificación , Inmunización/métodos , Inmunogenicidad Vacunal , Células T Asesinas Naturales/inmunología , Vacunas Sintéticas/inmunología , Adyuvantes Inmunológicos/química , Animales , Células Presentadoras de Antígenos/inmunología , Antígenos de Neoplasias/química , Antígenos de Neoplasias/genética , Vacunas contra el Cáncer/administración & dosificación , Química Clic/métodos , Células Dendríticas/inmunología , Femenino , Galactosilceramidas/química , Humanos , Inmunoglobulina G/sangre , Inmunoglobulina G/inmunología , Células MCF-7 , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Mucina-1/química , Mucina-1/genética , Transfección , Vacunas Sintéticas/administración & dosificación
9.
J Diabetes Res ; 2019: 9430473, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31781669

RESUMEN

Type 1 diabetes (T1D) is an autoimmune disease caused by the destruction of pancreatic ß cells by autoantigen-reactive diabetogenic cells. Antigen-specific therapies using islet autoantigens for restoring immune tolerance have emerged as promising approaches for the treatment of T1D but have been unsuccessful in humans. Herein, we report that RGI-3100-iB, a novel liposomal formulation carrying both α-galactosylceramide (α-GalCer), which is a representative ligand for invariant natural killer T (iNKT) cells, and insulin B chain 9-23 peptide, which is an epitope for CD4+ T cells, could induce the accumulation of regulatory T cells (Tregs) in islets in a peptide-dependent manner, followed by the remarkable prevention of diabetes onset in nonobese diabetic (NOD) mice. While multiple administrations of a monotherapy using either α-GalCer or insulin B peptide in a liposomal formulation was confirmed to delay/prevent T1D in NOD mice, RGI-3100-iB synergistically enhanced the prevention effect of each monotherapy and alleviated insulitis in NOD mice. Immunopathological analysis showed that Foxp3+ Tregs accumulated in the islets in RGI-3100-iB-treated mice. Cotransfer of diabetogenic T cells and splenocytes of NOD mice treated with RGI-3100-iB, but not liposomal α-GalCer encapsulating an unrelated peptide, to NOD-SCID mice resulted in the prevention of diabetes and elevation of Foxp3 mRNA expression in the islets. These data indicate that the migration of insulin B-peptide-specific Tregs to islet of NOD mice that are involved in the suppression of pathogenic T cells related to diabetes onset and progression could be enhanced by the administration of liposomes containing α-GalCer and insulin B peptide.


Asunto(s)
Diabetes Mellitus Tipo 1/prevención & control , Galactosilceramidas/administración & dosificación , Hipoglucemiantes/administración & dosificación , Insulina/administración & dosificación , Islotes Pancreáticos/efectos de los fármacos , Células T Asesinas Naturales/efectos de los fármacos , Fragmentos de Péptidos/administración & dosificación , Linfocitos T Reguladores/efectos de los fármacos , Traslado Adoptivo , Animales , Diabetes Mellitus Tipo 1/inmunología , Diabetes Mellitus Tipo 1/metabolismo , Modelos Animales de Enfermedad , Composición de Medicamentos , Femenino , Factores de Transcripción Forkhead/metabolismo , Islotes Pancreáticos/inmunología , Islotes Pancreáticos/metabolismo , Liposomas , Ratones Endogámicos NOD , Ratones SCID , Células T Asesinas Naturales/inmunología , Células T Asesinas Naturales/metabolismo , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Linfocitos T Reguladores/trasplante
10.
Nat Commun ; 10(1): 3258, 2019 07 22.
Artículo en Inglés | MEDLINE | ID: mdl-31332204

RESUMEN

CD160 and BTLA both bind to herpes virus entry mediator. Although a negative regulatory function of BTLA in natural killer T (NKT) cell activation has been reported, whether CD160 is also involved is unclear. By analyzing CD160-/- mice and mixed bone marrow chimeras, we show that CD160 is not essential for NKT cell development. However, CD160-/- mice exhibit severe liver injury after in vivo challenge with α-galactosylceramide (α-GalCer). Moreover, CD160-/- mice are more susceptible to Concanavalin A challenge, and display elevated serum AST and ALT levels, hyperactivation of NKT cells, and enhanced IFN-γ, TNF, and IL-4 production. Lastly, inhibition of BTLA by anti-BTLA mAb aggravates α-GalCer-induced hepatic injury in CD160-/- mice, suggesting that both CD160 and BTLA serve as non-overlapping negative regulators of NKT cells. Our data thus implicate CD160 as a co-inhibitory receptor that delivers antigen-dependent signals in NKT cells to dampen cytokine production during early innate immune activation.


Asunto(s)
Antígenos CD/metabolismo , Enfermedad Hepática Inducida por Sustancias y Drogas/metabolismo , Hígado/metabolismo , Células T Asesinas Naturales/metabolismo , Receptores Inmunológicos/metabolismo , Animales , Antígenos CD/genética , Enfermedad Hepática Inducida por Sustancias y Drogas/etiología , Enfermedad Hepática Inducida por Sustancias y Drogas/genética , Concanavalina A/administración & dosificación , Concanavalina A/toxicidad , Citocinas/metabolismo , Proteínas Ligadas a GPI/genética , Proteínas Ligadas a GPI/metabolismo , Galactosilceramidas/administración & dosificación , Galactosilceramidas/toxicidad , Hígado/efectos de los fármacos , Hígado/inmunología , Activación de Linfocitos/efectos de los fármacos , Activación de Linfocitos/inmunología , Ratones Endogámicos C57BL , Ratones Noqueados , Células T Asesinas Naturales/inmunología , Receptores Inmunológicos/genética , Miembro 14 de Receptores del Factor de Necrosis Tumoral/metabolismo , Análisis de Supervivencia
11.
EMBO J ; 38(14): e101260, 2019 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-31304630

RESUMEN

Tissue-resident iNKT cells maintain tissue homeostasis and peripheral surveillance against pathogens; however, studying these cells is challenging due to their low abundance and poor recovery from tissues. We here show that iNKT transnuclear mice, generated by somatic cell nuclear transfer, have increased tissue resident iNKT cells. We examined expression of PLZF, T-bet, and RORγt, as well as cytokine/chemokine profiles, and found that both monoclonal and polyclonal iNKT cells differentiated into functional subsets that faithfully replicated those seen in wild-type mice. We detected iNKT cells from tissues in which they are rare, including adipose, lung, skin-draining lymph nodes, and a previously undescribed population in Peyer's patches (PP). PP-NKT cells produce the majority of the IL-4 in Peyer's patches and provide indirect help for B-cell class switching to IgG1 in both transnuclear and wild-type mice. Oral vaccination with α-galactosylceramide shows enhanced fecal IgG1 titers in iNKT cell-sufficient mice. Transcriptional profiling reveals a unique signature of PP-NKT cells, characterized by tissue residency. We thus define PP-NKT as potentially important for surveillance for mucosal pathogens.


Asunto(s)
Perfilación de la Expresión Génica/métodos , Cambio de Clase de Inmunoglobulina , Inmunoglobulina G/genética , Células T Asesinas Naturales/metabolismo , Ganglios Linfáticos Agregados/inmunología , Animales , Diferenciación Celular , Células Cultivadas , Femenino , Galactosilceramidas/administración & dosificación , Galactosilceramidas/inmunología , Interleucina-4/genética , Ratones , Células T Asesinas Naturales/citología , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/genética , Técnicas de Transferencia Nuclear , Proteína de la Leucemia Promielocítica con Dedos de Zinc/genética , Proteínas de Dominio T Box/genética , Vacunación
12.
Viral Immunol ; 32(6): 269-275, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31199716

RESUMEN

CD8+ T cell immune response plays a critical role in the clearance of human papillomavirus (HPV)-infected cells. During the natural history of HPV infection, the E1 protein, an early-expressed helicase highly conserved among papillomaviruses, is involved in the replication of HPV genomes. We have previously shown, in a murine model, that immunization with HPV18 E1 protein combined with α-galactosylceramide elicits a specific CD8+ T cell response. We further proved those findings by analyzing whether CD8+ T cells from mice immunized with α-galactosylceramide plus HPV18 E1 protein could have a cytotoxic effect on cells expressing the carboxyl-terminal domain from the E1 proteins of other HPV types. Interestingly, CD8+ T cells raised against HPV18 E1 antigen presented cross-reactivity against the E1 protein from HPV53, 33, 16, and 31. Poor cross-reactivity was observed for HPV11, and none for HPV6. This outcome may be relevant for the design of broad-spectrum immune-protective agents against HPV infections.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Reacciones Cruzadas , Galactosilceramidas/inmunología , Proteínas Oncogénicas Virales/inmunología , Animales , Pruebas Inmunológicas de Citotoxicidad , Femenino , Galactosilceramidas/administración & dosificación , Antígenos de Histocompatibilidad Clase I/inmunología , Humanos , Inmunización , Ratones , Ratones Endogámicos C57BL , Proteínas Oncogénicas Virales/administración & dosificación , Papillomaviridae/clasificación , Infecciones por Papillomavirus/inmunología , Infecciones por Papillomavirus/prevención & control , Bazo/citología , Bazo/inmunología
13.
Front Immunol ; 10: 1126, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31244823

RESUMEN

NKT cells are CD1d-restricted innate-like T cells expressing both T cell receptor and NK cell markers. The major group of NKT cells in both human and mice is the invariant NKT (iNKT) cells and the best-known function of iNKT cells is their potent anti-tumor function in mice. Since its discovery 25 years ago, the prototype ligand of iNKT cells, α-galactosylceramide (α-GalCer) has been used in over 30 anti-tumor clinical trials with mostly suboptimal outcomes. To realize its therapeutic potential, numerous preclinical models have been developed to optimize the scheme and strategies for α-GalCer-based cancer immunotherapies. Nevertheless, since there is no standard protocol for α-GalCer delivery, we reviewed the preclinical studies with a focus on B16 melanoma model in the goal of identifying the best treatment schemes for α-GalCer treatment. We then reviewed the current progress in developing more clinically relevant mouse models for these preclinical studies, most notably the generation of new mouse models with a humanized CD1d/iNKT cell system. With ever-emerging novel iNKT cell ligands, invention of novel α-GalCer delivery strategies and significantly improved preclinical models for optimizing these new strategies, one can be hopeful that the full potential of anti-tumor potential for α-GalCer will be realized in the not too distant future.


Asunto(s)
Galactosilceramidas/administración & dosificación , Inmunoterapia , Células T Asesinas Naturales/inmunología , Células T Asesinas Naturales/metabolismo , Neoplasias/inmunología , Neoplasias/terapia , Animales , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Humanos , Inmunomodulación/efectos de los fármacos , Inmunoterapia/métodos , Ratones , Neoplasias/patología , Resultado del Tratamiento
14.
Int J Pharm ; 565: 123-132, 2019 Jun 30.
Artículo en Inglés | MEDLINE | ID: mdl-31075434

RESUMEN

The recognition of α-galactosylceramide (αGC), a high-affinity CD1d antigen, by the invariant Natural Killer T (iNKT) lymphocytes results in potent immunostimulatory responses that have been exploited in advanced cancer patients. Therefore, to improve αGC biological activity, several studies vectorized this agonist in PLGA and/or PEG-based nanoparticles. Despite promising findings, these approaches require several steps, from organic solvent decontamination through extrusion in membrane systems. Using a nano spray dryer, we vectorized αGC into a cationic copolymer (dimethylaminoethyl methacrylate, butyl methacrylate and methyl methacrylate - DBM) in a single step process, free of organic solvent. This methodology allowed the production of stable αGC-vectorized nanoparticles (DBM + αGC) with a more potent biological activity than the free agonist. DBM nanoparticles improved in vivo αGC loading into the CD1d molecule and induced a higher frequency of IFN-γ-expressing iNKT cells. Consequently, mice treated with DBM + αGC presented higher levels of serum IFN-γ than those treated with free agonist. Also, vectorized nanoparticles improved αGC ability to control the growth of murine lung metastatic carcinoma. Thus, this is the first study showing that nano spray dryer technology is a simple and alternative approach to enhance iNKT responses.


Asunto(s)
Portadores de Fármacos/administración & dosificación , Galactosilceramidas/administración & dosificación , Nanotecnología/métodos , Células T Asesinas Naturales/efectos de los fármacos , Animales , Línea Celular , Citocinas/inmunología , Desecación , Células Asesinas Naturales/efectos de los fármacos , Células Asesinas Naturales/inmunología , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/secundario , Masculino , Melanoma Experimental/tratamiento farmacológico , Melanoma Experimental/patología , Metacrilatos/administración & dosificación , Ratones Endogámicos C57BL , Nanopartículas/administración & dosificación , Células T Asesinas Naturales/inmunología
15.
Mucosal Immunol ; 12(4): 1055-1064, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30953000

RESUMEN

Cholera is a severe diarrheal disease caused by the bacterium Vibrio cholerae (V. cholerae) that results in 3-4 million cases globally with 100,000-150,000 deaths reported annually. Mostly confined to developing nations, current strategies to control the spread of cholera include the provision of safe drinking water and improved sanitation and hygiene, ideally in conjunction with oral vaccination. However, difficulties associated with the costs and logistics of these strategies have hampered their widespread implementation. Specific challenges pertaining to oral cholera vaccines (OCVs) include a lack of safe and effective adjuvants to further enhance gut immune responses, the complex and costly multicomponent vaccine manufacturing, limitations of conventional liquid formulation and the lack of an integrated delivery platform. Herein we describe the use of the orally active adjuvant α-Galactosylceramide (α-GalCer) to strongly enhance intestinal bacterium- and toxin-specific IgA responses to the OCV, Dukoral® in C57BL/6 and BALB/c mice. We further demonstrate the mucosal immunogenicity of a novel multi-antigen, single-component whole-cell killed V. cholerae strain and the enhancement of its immunogenicity by adding α-GalCer. Finally, we report that combining these components and recombinant cholera toxin B subunit in the SmPill® minisphere delivery system induced strong intestinal and systemic antigen-specific antibody responses.


Asunto(s)
Vacunas contra el Cólera/inmunología , Galactosilceramidas/farmacología , Inmunidad Mucosa/efectos de los fármacos , Inmunomodulación/efectos de los fármacos , Adyuvantes Inmunológicos/administración & dosificación , Administración Oral , Animales , Anticuerpos Antibacterianos/inmunología , Cólera/inmunología , Cólera/prevención & control , Vacunas contra el Cólera/administración & dosificación , Modelos Animales de Enfermedad , Femenino , Galactosilceramidas/administración & dosificación , Inmunización , Masculino , Ratones , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Células TH1/inmunología , Células TH1/metabolismo , Vibrio cholerae/inmunología
16.
Vaccine ; 37(9): 1219-1228, 2019 02 21.
Artículo en Inglés | MEDLINE | ID: mdl-30704821

RESUMEN

CD8+ T cell-mediated immune response plays a major role in the clearance of virus-infected cells, including human papillomavirus (HPV). The effective treatment of women with normal cytology but persistent high risk-HPV infection or with low-grade intraepithelial lesions could take advantage of novel strategies based on vaccination with viral immunological targets with a wide spectrum of cross-protection. The helicase E1, expressed early during viral replication in HPV infection, is among the most conserved papillomavirus proteins, which makes it a good vaccine candidate. In the present study, we examined E1-specific CD8+ T cell and NK immune responses in a mouse model with α-galactosylceramide (α-GalCer) as an adjuvant. We found that mice immunized with E1 combined with α-GalCer elicited an E1-specific CD8+ T and NK cell cytotoxic responses, which correlated with growth inhibition of grafted melanoma B16-F0 cells expressing E1, both in prophylactic and therapeutic protocols.


Asunto(s)
Vacunas contra el Cáncer/inmunología , Citotoxicidad Inmunológica , Galactosilceramidas/administración & dosificación , Proteínas Oncogénicas Virales/inmunología , Linfocitos T Citotóxicos/inmunología , Adyuvantes Inmunológicos/administración & dosificación , Animales , Femenino , Galactosilceramidas/inmunología , Papillomavirus Humano 18 , Humanos , Células Asesinas Naturales/inmunología , Melanoma Experimental/prevención & control , Melanoma Experimental/terapia , Melanoma Experimental/virología , Ratones , Ratones Endogámicos C57BL , Proteínas Oncogénicas Virales/administración & dosificación , Infecciones por Papillomavirus/inmunología , Infecciones por Papillomavirus/prevención & control , Infecciones por Papillomavirus/terapia , Trasplantes , Células Tumorales Cultivadas/inmunología , Vacunación
17.
Cell Mol Gastroenterol Hepatol ; 7(3): 623-639, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30630119

RESUMEN

BACKGROUND & AIMS: Chemokine-mediated immune cell recruitment plays pivotal roles in liver inflammation. C-C motif chemokine ligand 5 (CCL5) has been shown to be responsible for the recruitment of monocytes/macrophages and has been implicated in various liver diseases, including nonalcoholic fatty liver disease, fibrosis, and hepatocellular carcinoma. Previous studies have also shown that inhibition of CCL5 appears to be a promising therapeutic approach for several chronic liver diseases. However, whether blocking CCL5 could benefit immune cell-mediated hepatitis remains largely elusive. METHODS: By adopting a specific agonist, alpha-galactosylceramide (α-Galcer), of invariant natural killer T cells (iNKTs), we investigated the function and mechanism of CCL5 in the iNKT induced murine hepatitis model. RESULTS: We found significantly increased CCL5 expression in α-Galcer-induced hepatitis murine model. Such an increase in CCL5 is mainly enriched in non-parenchymal cells such as macrophages and iNKTs but not in hepatocytes. Surprisingly, CCL5 blockage by genetic deletion of Ccl5 does not affect the α-Galcer-induced iNKT activation but greatly worsens α-Galcer-induced liver injury accompanied by an increased hepatic neutrophil infiltration. Mechanistically, we demonstrated that greater neutrophil accumulation in the liver is responsible for the enhanced liver injury in Ccl5-/- mice. Such an increased hepatic neutrophil infiltration is mainly caused by an enhanced CXCL1-CXCR2 signal in Ccl5-/- mice. Therapeutically, either antibody-mediated neutrophil depletion or a CXCR2 antagonist, SB225002, mediated CXCR2 signaling blockage significantly ameliorated α-Galcer-induced liver injury in Ccl5-/- mice. CONCLUSIONS: Our present study demonstrates that (1) α-Galcer-induced murine hepatitis could greatly induce CCL5 production in macrophages and iNKT cells; (2) loss of CCL5 could enhance CXCL1 expression in hepatocytes and activate CXCL1-CXCR2 axis in neutrophils to augment their hepatic infiltration; and (3) neutrophil depletion or blockage of CXCL1-CXCR2 axis greatly improves α-Galcer-induced liver injury in Ccl5-/- mice. This study suggests that clinical utilization of CCL5 blockage may compensatorily induce the activation of other chemokine pathways to enhance neutrophil recruitment and liver injury in hepatitis.


Asunto(s)
Quimiocina CCL5/deficiencia , Eliminación de Gen , Hepatitis/inmunología , Células T Asesinas Naturales/inmunología , Receptores de Interleucina-8B/genética , Regulación hacia Arriba , Adulto , Anciano , Animales , Quimiocina CCL5/metabolismo , Quimiocina CXCL1/genética , Quimiocina CXCL1/metabolismo , Citocinas/biosíntesis , Modelos Animales de Enfermedad , Galactosilceramidas/administración & dosificación , Hepatitis/sangre , Hepatitis/patología , Hepatocitos/metabolismo , Hepatocitos/patología , Humanos , Hígado/lesiones , Hígado/metabolismo , Hígado/patología , Activación de Linfocitos/inmunología , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Infiltración Neutrófila , Neutrófilos/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores de Interleucina-8B/metabolismo , Bazo/metabolismo , Adulto Joven
18.
AAPS J ; 21(2): 18, 2019 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-30635795

RESUMEN

Chemoimmunotherapy with chemotherapeutics and immunoadjuvant inhibits tumor growth by activating cytotoxic T cells. However, this process also upregulates the expression of PD-1/PD-L1 and consequently leads to immune suppression. To maximize the anti-tumor immune responses and alleviate immunosuppression, PD-L1 antibody was combined with paclitaxel (PTX) and the immunoadjuvant α-galactosylceramide (αGC), which were coencapsulated into pH-sensitive TH peptide-modified liposomes (PTX/αGC/TH-Lip) to treat melanoma and lung metastasis. Compared to treatment with PD-L1 antibody or PTX/αGC/TH-Lip alone, the combination of PD-L1 antibody and PTX/αGC/TH-Lip further elevated the tumor-specific cytotoxic T cell responses and promoted apoptosis in tumor cells, leading to enhanced anti-tumor and anti-metastatic effects. In adoptive therapy, PD-L1 antibody further alleviated immunosuppression and enhanced the anti-tumor effect of CD8+ T cells. The combination of PD-L1 antibody and chemoimmunotherapy PTX/αGC/TH-Lip provides a promising strategy for enhancing treatment for melanoma and lung metastasis.


Asunto(s)
Antineoplásicos Inmunológicos/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Neoplasias Pulmonares/tratamiento farmacológico , Melanoma Experimental/tratamiento farmacológico , Neoplasias Cutáneas/tratamiento farmacológico , Adyuvantes Inmunológicos/administración & dosificación , Animales , Antineoplásicos Inmunológicos/uso terapéutico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Antígeno B7-H1/antagonistas & inhibidores , Antígeno B7-H1/inmunología , Línea Celular Tumoral/trasplante , Femenino , Galactosilceramidas/administración & dosificación , Tolerancia Inmunológica/efectos de los fármacos , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/secundario , Melanoma Experimental/inmunología , Melanoma Experimental/secundario , Ratones , Paclitaxel/administración & dosificación , Paclitaxel/inmunología , Neoplasias Cutáneas/inmunología , Neoplasias Cutáneas/patología , Linfocitos T Citotóxicos/efectos de los fármacos
19.
Sci Rep ; 8(1): 10081, 2018 07 04.
Artículo en Inglés | MEDLINE | ID: mdl-29973666

RESUMEN

Graphene oxide (GO) modulates the functions of antigen-presenting cells including dendritic cells (DCs). Although carbon nanotubes affect expression of the MHC class I-like CD1d molecule, whether GO can influence immune responses of CD1d-dependent invariant natural killer T (iNKT) cells remains unclear. Here, we investigated the impact of GO on inflammatory responses mediated by α-galactosylceramide (α-GalCer), an iNKT cell agonist. We found that in vivo GO treatment substantially inhibited the capacity of α-GalCer to induce the iNKT cell-mediated trans-activation of and cytokine production by innate and innate-like cells, including DCs, macrophages, NK cells, and γδ T cells. Such effects of GO on α-GalCer-induced inflammatory responses closely correlated with iNKT cell polarization towards TGFß production, which also explains the capacity of GO to expand regulatory T cells. Interestingly, the absence of TLR4, a receptor for GO, failed to downregulate, and instead partially enhanced the anti-inflammatory activity of GO against α-GalCer-elicited responses, implying negative effects of TLR4 signaling on the anti-inflammatory properties of GO. By employing an α-GalCer-induced sepsis model, we further demonstrated that GO treatment significantly protected mice from α-GalCer-induced lethality. Taken together, we provide strong evidence that GO holds promise as an adjuvant to modulate iNKT cell responses for immunotherapy.


Asunto(s)
Grafito/administración & dosificación , Inflamación/tratamiento farmacológico , Sepsis/tratamiento farmacológico , Receptor Toll-Like 4/genética , Factor de Crecimiento Transformador beta/genética , Animales , Antígenos CD1d/genética , Antígenos CD1d/inmunología , Polaridad Celular/efectos de los fármacos , Células Dendríticas/efectos de los fármacos , Células Dendríticas/inmunología , Modelos Animales de Enfermedad , Galactosilceramidas/administración & dosificación , Humanos , Inflamación/inmunología , Inflamación/patología , Linfocitos Intraepiteliales/efectos de los fármacos , Linfocitos Intraepiteliales/inmunología , Activación de Linfocitos/efectos de los fármacos , Ratones , Nanotubos de Carbono/química , Células T Asesinas Naturales/efectos de los fármacos , Células T Asesinas Naturales/inmunología , Sepsis/inmunología , Sepsis/patología
20.
Front Immunol ; 9: 833, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29720984

RESUMEN

Sepsis is the third leading cause of death in the neonatal population, due to susceptibility to infection conferred by immaturity of both the innate and adaptive components of the immune system. Invariant natural killer T (iNKT) cells are specialized adaptive immune cells that possess important innate-like characteristics and have not yet been well-studied in septic neonates. We hypothesized that iNKT cells would play an important role in mediating the neonatal immune response to sepsis. To study this, we subjected 5- to 7-day-old neonatal C57BL/6 mice to sepsis by intraperitoneal (i.p.) cecal slurry (CS) injection. Thirty hours prior to or immediately following sepsis induction, pups received i.p. injection of the iNKT stimulator KRN7000 (KRN, 0.2 µg/g) or vehicle. Ten hours after CS injection, blood and tissues were collected for various analyses. Thirty-hour pretreatment with KRN resulted in better outcomes in inflammation, lung injury, and survival, while immediate treatment with KRN resulted in worse outcomes compared to vehicle treatment. We further analyzed the activation status of neonatal iNKT cells for 30 h after KRN administration, and showed a peak in frequency of CD69 expression on iNKT cells and serum IFN-γ levels at 5 and 10 h, respectively. We then used CD1d knockout neonatal mice to demonstrate that KRN acts through the major histocompatibility complex-like molecule CD1d to improve outcomes in neonatal sepsis. Finally, we identified that KRN pretreatment exerts its protective effect by increasing systemic levels of TGF-ß1. These findings support the importance of iNKT cells for prophylactic immunomodulation in neonates susceptible to sepsis.


Asunto(s)
Inmunomodulación , Inflamación/inmunología , Células T Asesinas Naturales/inmunología , Sepsis Neonatal/inmunología , Adyuvantes Inmunológicos/administración & dosificación , Animales , Animales Recién Nacidos , Antígenos CD/genética , Antígenos CD/inmunología , Antígenos CD1d/genética , Antígenos CD1d/inmunología , Antígenos de Diferenciación de Linfocitos T/genética , Antígenos de Diferenciación de Linfocitos T/inmunología , Femenino , Galactosilceramidas/administración & dosificación , Galactosilceramidas/uso terapéutico , Interferón gamma/sangre , Interferón gamma/inmunología , Lectinas Tipo C/genética , Lectinas Tipo C/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Factor de Crecimiento Transformador beta1/sangre
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...