Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
J Biol Chem ; 297(5): 101266, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34600888

RESUMO

Functional delivery of mRNA has high clinical potential. Previous studies established that mRNAs can be delivered to cells in vitro and in vivo via RNA-loaded lipid nanoparticles (LNPs). Here we describe an alternative approach using exosomes, the only biologically normal nanovesicle. In contrast to LNPs, which elicited pronounced cellular toxicity, exosomes had no adverse effects in vitro or in vivo at any dose tested. Moreover, mRNA-loaded exosomes were characterized by efficient mRNA encapsulation (∼90%), high mRNA content, consistent size, and a polydispersity index under 0.2. Using an mRNA encoding the red light-emitting luciferase Antares2, we observed that mRNA-loaded exosomes were superior to mRNA-loaded LNPs at delivering functional mRNA into human cells in vitro. Injection of Antares2 mRNA-loaded exosomes also led to strong light emission following injection into the vitreous fluid of the eye or into the tissue of skeletal muscle in mice. Furthermore, we show that repeated injection of Antares2 mRNA-loaded exosomes drove sustained luciferase expression across six injections spanning at least 10 weeks, without evidence of signal attenuation or adverse injection site responses. Consistent with these findings, we observed that exosomes loaded with mRNAs encoding immunogenic forms of the SARS-CoV-2 Spike and Nucleocapsid proteins induced long-lasting cellular and humoral responses to both. Taken together, these results demonstrate that exosomes can be used to deliver functional mRNA to and into cells in vivo.


Assuntos
Exossomos/imunologia , RNA Mensageiro/genética , SARS-CoV-2/imunologia , Células Cultivadas , Técnicas de Transferência de Genes , Células HEK293 , Humanos , Lipídeos/química , Nanopartículas/química , RNA Mensageiro/imunologia , SARS-CoV-2/genética
2.
J Virol ; 89(8): 4158-69, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25631080

RESUMO

UNLABELLED: Broadly neutralizing antibodies (bNAbs) specific for conserved epitopes on the HIV-1 envelope (Env) are believed to be essential for protection against multiple HIV-1 clades. However, vaccines capable of stimulating the production of bNAbs remain a major challenge. Given that polyreactivity and autoreactivity are considered important characteristics of anti-HIV bNAbs, we designed an HIV vaccine incorporating the molecular adjuvants BAFF (B cell activating factor) and APRIL (a proliferation-inducing ligand) with the potential to facilitate the maturation of polyreactive and autoreactive B cells as well as to enhance the affinity and/or avidity of Env-specific antibodies. We designed recombinant DNA plasmids encoding soluble multitrimers of BAFF and APRIL using surfactant protein D as a scaffold, and we vaccinated mice with these molecular adjuvants using DNA and DNA-protein vaccination strategies. We found that immunization of mice with a DNA vaccine encoding BAFF or APRIL multitrimers, together with interleukin 12 (IL-12) and membrane-bound HIV-1 Env gp140, induced neutralizing antibodies against tier 1 and tier 2 (vaccine strain) viruses. The APRIL-containing vaccine was particularly effective at generating tier 2 neutralizing antibodies following a protein boost. These BAFF and APRIL effects coincided with an enhanced germinal center (GC) reaction, increased anti-gp120 antibody-secreting cells, and increased anti-gp120 functional avidity. Notably, BAFF and APRIL did not cause indiscriminate B cell expansion or an increase in total IgG. We propose that BAFF and APRIL multitrimers are promising molecular adjuvants for vaccines designed to induce bNAbs against HIV-1. IMPORTANCE: Recent identification of antibodies that neutralize most HIV-1 strains has revived hopes and efforts to create novel vaccines that can effectively stimulate HIV-1 neutralizing antibodies. However, the multiple immune evasion properties of HIV have hampered these efforts. These include the instability of the gp120 trimer, the inaccessibility of the conserved sequences, highly variable protein sequences, and the loss of HIV-1-specific antibody-producing cells during development. We have shown previously that tumor necrosis factor (TNF) superfamily ligands, including BAFF and APRIL, can be multitrimerized using the lung protein SP-D (surfactant protein D), enhancing immune responses. Here we show that DNA or DNA-protein vaccines encoding BAFF or APRIL multitrimers, IL-12p70, and membrane-bound HIV-1 Env gp140 induced tier 1 and tier 2 neutralizing antibodies in a mouse model. BAFF and APRIL enhanced the immune reaction, improved antibody binding, and increased the numbers of anti-HIV-1 antibody-secreting cells. Adaptation of this vaccine design may prove useful in designing preventive HIV-1 vaccines for humans.


Assuntos
Vacinas contra a AIDS/imunologia , Adjuvantes Imunológicos/farmacologia , Anticorpos Neutralizantes/efeitos dos fármacos , Proteína gp120 do Envelope de HIV/imunologia , Infecções por HIV/prevenção & controle , HIV-1/imunologia , Vacinas de DNA/imunologia , Análise de Variância , Animais , Fator Ativador de Células B/imunologia , Ensaio de Imunoadsorção Enzimática , ELISPOT , Feminino , Citometria de Fluxo , Proteína gp120 do Envelope de HIV/efeitos dos fármacos , Interleucina-12/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Testes de Neutralização , Plasmídeos/genética , Membro 13 da Superfamília de Ligantes de Fatores de Necrose Tumoral/imunologia
3.
Infect Immun ; 83(11): 4404-15, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26351279

RESUMO

Induction of adaptive immunity leads to the establishment of immunological memory; however, how innate immunity regulates memory T cell function remains obscure. Here we show a previously undefined mechanism in which innate and adaptive immunity are linked by TIR domain-containing adapter-inducing beta interferon (TRIF) during establishment and reactivation of memory T cells against Gram-negative enteropathogens. Absence of TRIF in macrophages (Mϕs) but not dendritic cells led to a predominant generation of CD4(+) central memory T cells that express IL-17 during enteric bacterial infection in mice. TRIF-dependent type I interferon (IFN) signaling in T cells was essential to Th1 lineage differentiation and reactivation of memory T cells. TRIF activated memory T cells to facilitate local neutrophil influx and enhance bacterial elimination. These results highlight the importance of TRIF as a mediator of the innate and adaptive immune interactions in achieving the protective properties of memory immunity against Gram-negative bacteria and suggest TRIF as a potential therapeutic target.


Assuntos
Proteínas Adaptadoras de Transporte Vesicular/imunologia , Memória Imunológica , Yersiniose/imunologia , Yersinia enterocolitica/imunologia , Proteínas Adaptadoras de Transporte Vesicular/genética , Animais , Células Dendríticas/imunologia , Humanos , Interferon gama/imunologia , Interleucina-17/genética , Interleucina-17/imunologia , Macrófagos/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Linfócitos T/imunologia , Yersiniose/genética , Yersiniose/microbiologia , Yersinia enterocolitica/genética
4.
Retrovirology ; 8: 39, 2011 May 18.
Artigo em Inglês | MEDLINE | ID: mdl-21592361

RESUMO

BACKGROUND: Molecular adjuvants are a promising method to enhance virus-specific immune responses and protect against HIV-1 infection. Immune activation by ligands for receptors such as CD40 can induce dendritic cell activation and maturation. Here we explore the incorporation of two CD40 mimics, Epstein Barr Virus gene LMP1 or an LMP1-CD40 chimera, into a strain of SIV that was engineered to be limited to a single cycle of infection. RESULTS: Full length LMP1 or the chimeric protein LMP1-CD40 was cloned into the nef-locus of single-cycle SIV. Human and Macaque monocyte derived macrophages and DC were infected with these viruses. Infected cells were analyzed for activation surface markers by flow cytometry. Cells were also analyzed for secretion of pro-inflammatory cytokines IL-1ß, IL-6, IL-8, IL-12p70 and TNF by cytometric bead array. CONCLUSIONS: Overall, single-cycle SIV expressing LMP1 and LMP1-CD40 produced a broad and potent T(H)1-biased immune response in human as well as rhesus macaque macrophages and DC when compared with control virus. Single-cycle SIV-LMP1 also enhanced antigen presentation by lentiviral vector vaccines, suggesting that LMP1-mediated immune activation may enhance lentiviral vector vaccines against HIV-1.


Assuntos
Adjuvantes Imunológicos/farmacologia , Portadores de Fármacos , Vetores Genéticos , Lentivirus/genética , Vacinas contra a SAIDS/imunologia , Proteínas da Matriz Viral/farmacologia , Adjuvantes Imunológicos/genética , Animais , Apresentação de Antígeno , Citocinas/metabolismo , Humanos , Macaca mulatta , Monócitos/virologia , Vacinas Sintéticas/imunologia , Proteínas da Matriz Viral/genética
5.
PLoS One ; 12(9): e0184915, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28910387

RESUMO

Dendritic cells (DC) are a promising cell type for cancer vaccines due to their high immunostimulatory capacity. However, improper maturation of DC prior to treatment may account for the limited efficacy of DC vaccine clinical trials. Latent Membrane Protein-1 (LMP1) of Epstein-Barr virus was examined for its ability to mature and activate DC as a gene-based molecular adjuvant for DC vaccines. DC were transduced with an adenovirus 5 vector (Ad5) expressing LMP1 under the control of a Tet-inducible promoter. Ad5-LMP1 was found to mature and activate both human and mouse DC. LMP1 enhanced in vitro migration of DC toward CCL19, as well as in vivo migration of DC to the inguinal lymph nodes of mice following intradermal injection. LMP1-transduced DC increased T cell proliferation in a Pmel-1 adoptive transfer model and enhanced survival in B16-F10 melanoma models. LMP1-DC also enhanced protection in a vaccinia-Gag viral challenge assay. LMP1 induced high levels of IL-12p70 secretion in mouse DC when compared to standard maturation protocols. Importantly, LMP1-transduced human DC retained the capacity to secrete IL-12p70 and TNF in response to DC restimulation. In contrast, DC matured with Monocyte Conditioned Media-Mimic cocktail (Mimic) were impaired in IL-12p70 secretion following restimulation. Overall, LMP1 matured and activated DC, induced migration to the lymph node, and generated high levels of IL-12p70 in a murine model. We propose LMP1 as a promising molecular adjuvant for DC vaccines.


Assuntos
Células Dendríticas/transplante , Herpesvirus Humano 4/metabolismo , Interleucina-12/metabolismo , Linfonodos/imunologia , Melanoma Experimental/terapia , Proteínas da Matriz Viral/genética , Animais , Vacinas Anticâncer/imunologia , Movimento Celular , Quimiocina CCL19/metabolismo , Células Dendríticas/citologia , Células Dendríticas/imunologia , Dependovirus/genética , Dependovirus/fisiologia , Feminino , Células HEK293 , Humanos , Injeções Intradérmicas , Melanoma Experimental/imunologia , Camundongos , Proteínas da Matriz Viral/imunologia
6.
J Clin Cell Immunol ; 7(5)2016 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-27853628

RESUMO

OBJECTIVE: Establishing an effective humoral immunity is an important host defense mechanism in intestinal mucosa. T follicular helper (Tfh) cells are a spectrum of CXCR5 expressing T helper cells that induce antigen-specific B cell differentiation. Because the differentiation of T helper cells is largely regulated by innate immunity, we addressed whether TRIF signaling regulates Tfh cell differentiation and its ability to trigger humoral immune responses in the intestine. METHOD: CD4+CXCR5+ T cells, B cells, and plasma cells in the Peyer's patches (PPs) of WT and TRIF-deficient (TrifLPS2) mice were analyzed by flow cytometry at the baseline, 9 days post primary infection, and 7 days post-secondary infection with Y. enterocolitica. Y. enterocolitica-specific CD4+CXCR5+ T cells were generated in vitro by co-culturing peritoneal macrophages with splenic naïve T cells in the presence of Y. enterocolitica lysate. WT and TrifLPS2 mice received CD4+CXCR5+ T cells isolated either from Y. enterocolitica-primed WT mice or generated in vitro. These mice were infected with Y. enterocolitica and followed up to 4 weeks. Y. enterocolitica-specific IgA and IgG were measured in stool and serum samples, respectively. RESULTS: At baseline, CD4+CXCR5+ T cell proportion was higher but the proportion of B cells and plasma cells was lower in the PPs of TrifLPS2 mice compared to WT mice. After infection, the proportion of plasma cells also became higher in the PPs of TrifLPS2 mice compared to WT mice. Corresponding increase of Y. enterocolitica-specific stool IgA but not serum IgG was found in TrifLPS2 mice compared to WT mice. Both in vivo isolated and in vitro generated CD4+CXCR5+ T cells induced protective immunity against Y. enterocolitica infection. CONCLUSION: Our results reveal a novel role of TRIF in the regulation of humoral immunity in the intestine that can be utilized as a basis for a unique vaccine strategy.

7.
Artigo em Inglês | MEDLINE | ID: mdl-26793623

RESUMO

Recognition of Gram-negative bacteria by toll-like receptor (TLR)4 induces MyD88 and TRIF mediated responses. We have shown that TRIF-dependent responses play an important role in intestinal defense against Gram-negative enteropathogens. In the current study, we examined underlying mechanisms of how systemic TRIF activation enhances intestinal immune defense against Gram-negative bacteria. First we confirmed that the protective effect of poly I:C against enteric infection of mice with Yersinia enterocolitica was dependent on TLR3-mediated TRIF signaling by using TLR3-deficient mice. This protection was unique in TRIF-dependent TLR signaling because systemic stimulation of mice with agonists for TLR2 (Pam3CSK4) or TLR5 (flagellin) did not reduce mortality on Y. enterocolitica infection. Systemic administration of poly I:C mobilized CD11c+, F4/80+, and Gr-1(hi) cells from lamina propria and activated NK cells in the mesenteric lymph nodes (MLN) within 24 h. This innate immune cell rearrangement was type I IFN dependent and mediated through upregulation of TLR4 followed by CCR7 expression in these innate immune cells found in the intestinal mucosa. Poly I:C induced IFN-γ expression by NK cells in the MLN, which was mediated through type I IFNs and IL-12p40 from antigen presenting cells and consequent activation of STAT1 and STAT4 in NK cells. This formation of innate immunity significantly contributed to the elimination of bacteria in the MLN. Our results demonstrated an innate immune network in the intestine that can be established by systemic stimulation of TRIF, which provides a strong host defense against Gram-negative pathogens. The mechanism underlying TRIF-mediated protective immunity may be useful to develop novel therapies for enteric bacterial infection.


Assuntos
Proteínas Adaptadoras de Transporte Vesicular/metabolismo , Intestinos/imunologia , Transdução de Sinais , Receptor 3 Toll-Like/metabolismo , Yersiniose/imunologia , Yersinia enterocolitica/imunologia , Animais , Redes Reguladoras de Genes , Fatores Imunológicos/metabolismo , Camundongos , Poli I-C/metabolismo , Análise de Sobrevida
8.
J Innate Immun ; 7(1): 87-101, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25171731

RESUMO

In the intestine, interaction between epithelial cells and macrophages (MΦs) create a unique immunoregulatory microenvironment necessary to maintain local immune and tissue homeostasis. Human intestinal epithelial cells (IECs) have been shown to express interleukin (IL)-10, which keeps epithelial integrity. We have demonstrated that bacterial signaling through Toll-like receptor (TLR) 4 induces 15-deoxy-Δ-12,14-prostaglandin J2 (15d-PGJ2) synthesis in intestinal MΦs by cyclooxygenase (Cox)-2 expression. Here, we show that TLR4 signaling generates crosstalk between IECs and MΦs that enhances IL-10 expression in IECs. Direct stimulation of TLR4 leads to the expression of IL-10 in IECs, while the presence of MΦs in a Transwell system induces another peak in IL-10 expression in IECs at a later time point. The second peak of the IL-10 expression is two times greater than the first peak. This late induction of IL-10 depends on the nuclear receptor peroxisome proliferator-activated receptor (PPAR) γ that is accumulated in IECs by TLR4-mediated inhibition of the ubiquitin-proteasomal pathway. TLR4 signaling in MΦs in turn synthesizes 15d-PGJ2 through p38 and ERK activation and Cox-2 induction, which activates PPARγ in IECs. These results suggest that TLR4 signaling maintains IL-10 production in IECs by generating epithelial-MΦs crosstalk, which is an important mechanism in the maintenance of intestinal homeostasis mediated through host-bacterial interactions.


Assuntos
Comunicação Celular/imunologia , Células Epiteliais/imunologia , Interleucina-10/imunologia , Mucosa Intestinal/imunologia , Sistema de Sinalização das MAP Quinases/imunologia , Macrófagos/imunologia , Receptor 4 Toll-Like/imunologia , Células Cultivadas , Ciclo-Oxigenase 2/imunologia , Regulação Enzimológica da Expressão Gênica/imunologia , Humanos , Prostaglandina D2/análogos & derivados , Prostaglandina D2/imunologia
9.
PLoS One ; 9(2): e90100, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24587225

RESUMO

Adenoviral vectored vaccines have shown considerable promise but could be improved by molecular adjuvants. Ligands in the TNF superfamily (TNFSF) are potential adjuvants for adenoviral vector (Ad5) vaccines based on their central role in adaptive immunity. Many TNFSF ligands require aggregation beyond the trimeric state (multi-trimerization) for optimal biological function. Here we describe Ad5 vaccines for HIV-1 Gag antigen (Ad5-Gag) adjuvanted with the TNFSF ligands 4-1BBL, BAFF, GITRL and CD27L constructed as soluble multi-trimeric proteins via fusion to Surfactant Protein D (SP-D) as a multimerization scaffold. Mice were vaccinated with Ad5-Gag combined with Ad5 expressing one of the SP-D-TNFSF constructs or single-chain IL-12p70 as adjuvant. To evaluate vaccine-induced protection, mice were challenged with vaccinia virus expressing Gag (vaccinia-Gag) which is known to target the female genital tract, a major route of sexually acquired HIV-1 infection. In this system, SP-D-4-1BBL or SP-D-BAFF led to significantly reduced vaccinia-Gag replication when compared to Ad5-Gag alone. In contrast, IL-12p70, SP-D-CD27L and SP-D-GITRL were not protective. Histological examination following vaccinia-Gag challenge showed a dramatic lymphocytic infiltration into the uterus and ovaries of SP-D-4-1BBL and SP-D-BAFF-treated animals. By day 5 post challenge, proinflammatory cytokines in the tissue were reduced, consistent with the enhanced control over viral replication. Splenocytes had no specific immune markers that correlated with protection induced by SP-D-4-1BBL and SP-D-BAFF versus other groups. IL-12p70, despite lack of anti-viral efficacy, increased the total numbers of splenic dextramer positive CD8+ T cells, effector memory T cells, and effector Gag-specific CD8+ T cells, suggesting that these markers are poor predictors of anti-viral immunity in this model. In conclusion, soluble multi-trimeric 4-1BBL and BAFF adjuvants led to strong protection from vaccinia-Gag challenge, but the protection was independent of standard immune markers. Soluble multi-trimeric SP-D-4-1BBL and SP-D-BAFF provide a novel technology to enhance adenoviral vector vaccines against HIV-1.


Assuntos
Ligante 4-1BB/imunologia , Vacinas contra a AIDS/imunologia , Adenoviridae/imunologia , Adjuvantes Imunológicos/genética , Fator Ativador de Células B/imunologia , Infecções por HIV/prevenção & controle , Vaccinia virus/imunologia , Ligante 4-1BB/administração & dosagem , Ligante 4-1BB/genética , Vacinas contra a AIDS/administração & dosagem , Vacinas contra a AIDS/genética , Adenoviridae/genética , Adjuvantes Imunológicos/biossíntese , Animais , Fator Ativador de Células B/administração & dosagem , Fator Ativador de Células B/genética , Linfócitos T CD8-Positivos/citologia , Linfócitos T CD8-Positivos/imunologia , Feminino , Expressão Gênica , Produtos do Gene gag/genética , Produtos do Gene gag/imunologia , Vetores Genéticos , Infecções por HIV/imunologia , HIV-1/imunologia , Humanos , Imunidade Ativa , Contagem de Linfócitos , Camundongos , Multimerização Proteica , Vacinação , Vacinas de Subunidades Antigênicas , Replicação Viral/efeitos dos fármacos
10.
Radiat Res ; 182(2): 149-62, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25036982

RESUMO

Radiation is a potent immune-modulator that elicits cell death upon tumor, stromal and angiogenic compartments of tumor microenvironment. Here, we test a novel approach of high-dose radiation delivery using three dimensional volume based lattice radiation therapy (LRT) to understand the impact of different volume irradiation in eliciting both local and metastatic/distant tumor control through modulation of tumor immune micro-environment. To study such effects of LRT, tumors were implanted in both hind legs of C57BL/6 mice using Lewis lung carcinoma 1 (LLC1) cells. Mice were divided into five groups: untreated; partial tumor volume groups included two 10% vertices, one 20% vertex and one 50% vertex of the total tumor volume; and 100% open-field irradiation. Tumors implanted in the left flank were irradiated with a single dose of 20 Gy while the tumors in the right flank were unirradiated. Tumor growth and regression as well as immune responses (such as Th1 and Th2; T-cell infiltration) were determined after radiation treatment. Results demonstrated that both 100% open-field irradiation and 20% volume irradiation (in two 10% volumes) resulted in significant growth delay in the irradiated tumor. Further, all types of radiation exposures, partial or 100% volume, demonstrated distal effectiveness, however, 20% volume irradiation (in two 10% volumes) and 50% tumor volume irradiation led to maximum growth delay. Mice treated with partial tumor volume radiation induced a robust IFN-γ and Th1 response when compared to whole-tumor irradiation and down-modulated Th2 functions. The presence of increased CD3+ cells and TRAIL in partially irradiated tumor volumes correlated well with tumor growth delay. Further, serum obtained from any of the LRT treated mice caused growth inhibition of endothelial cells when compared to serum obtained from either untreated or open-field irradiated groups. These results indicate that high-dose partial volume irradiation can cause an improved distant effect than the total tumor volume irradiation through activating the host immune system.


Assuntos
Imunomodulação/efeitos da radiação , Neoplasias Pulmonares/imunologia , Neoplasias Pulmonares/radioterapia , Animais , Proliferação de Células/efeitos da radiação , Fatores Quimiotáticos/metabolismo , Citocinas/metabolismo , Fracionamento da Dose de Radiação , Feminino , Regulação Neoplásica da Expressão Gênica/imunologia , Regulação Neoplásica da Expressão Gênica/efeitos da radiação , Queratinócitos/imunologia , Queratinócitos/metabolismo , Queratinócitos/efeitos da radiação , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patologia , Camundongos , Camundongos Endogâmicos C57BL , Metástase Neoplásica , Esfingomielina Fosfodiesterase/metabolismo , Baço/imunologia , Baço/efeitos da radiação , Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Microambiente Tumoral/imunologia , Microambiente Tumoral/efeitos da radiação
11.
Microbes Infect ; 15(1): 1-10, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23116944

RESUMO

Both anti-viral and anti-bacterial host defense mechanisms involve TRIF signaling. TRIF provides early clearance of pathogens and coordination of a local inflammatory ensemble through an interferon cascade, while it may trigger organ damage. The multipotentiality of TRIF-mediated immune machinery may direct the fate of our continuous battle with microbes.


Assuntos
Proteínas Adaptadoras de Transporte Vesicular/imunologia , Infecções Bacterianas/imunologia , Receptores Toll-Like/imunologia , Viroses/imunologia , Interações Hospedeiro-Patógeno , Humanos , Imunidade Inata
12.
Immunol Res ; 57(1-3): 303-10, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24198065

RESUMO

TNF superfamily ligands play a critical role in the regulation of adaptive immune responses, including the costimulation of dendritic cells, T cells, and B cells. This costimulation could potentially be exploited for the development of prophylactic vaccines and immunotherapy. Despite this, there have been only a limited number of reports on the use of this family of molecules as gene-based adjuvants to enhance DNA and/or viral vector vaccines. In addition, the molecule latent membrane protein 1 (LMP1), a viral mimic of the TNF superfamily receptor CD40, provides an alternative approach for the design of novel molecular adjuvants. Here, we discuss advances in the development of recombinant TNF superfamily ligands as adjuvants for HIV vaccines and as cancer immunotherapy, including the use of LMP1 and LMP1-CD40 chimeric fusion proteins to mimic constitutive CD40 signaling.


Assuntos
Adjuvantes Imunológicos , Mimetismo Molecular/imunologia , Fatores de Necrose Tumoral , Vacinas Sintéticas/imunologia , Vacinas/química , Vacinas/imunologia , Adjuvantes Imunológicos/química , Animais , Antígenos CD40/imunologia , Ligante de CD40/química , Ligante de CD40/metabolismo , Vacinas Anticâncer/química , Vacinas Anticâncer/imunologia , Vetores Genéticos/genética , Vetores Genéticos/imunologia , Humanos , Imunoterapia , Lentivirus/genética , Lentivirus/imunologia , Ligantes , Macaca mulatta , Neoplasias/imunologia , Neoplasias/terapia , Multimerização Proteica , Ligante Indutor de Apoptose Relacionado a TNF/química , Receptores Toll-Like/agonistas , Fatores de Necrose Tumoral/química , Vacinas Sintéticas/química , Proteínas da Matriz Viral/imunologia
13.
Gut Microbes ; 3(5): 437-41, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22713267

RESUMO

The gastrointestinal tract is the largest mucosal surface in our body. It houses diverse microorganisms that collectively form the commensal microbial community. The security of this community is kept by host-microbial interactions and is violated by foreign pathogens that induce local as well as systemic pathology. In most cases, gastrointestinal infections are caused by Gram-negative enteropathogens, which trigger host immune responses through the TLR4 signaling pathways. Although TRIF is one of the major pathways downstream of TLR4, very little is known about how the TRIF pathway contributes to intestinal defense against pathogenic infection. Recently, we reported a unique role of TRIF signaling in host response to an enterophathogen Yersinia enterocolitica, which consisted of IFN-ß induction from regional macrophages followed by activation of NK cells in the mesenteric lymph nodes. In this addendum, we show distinct roles for TRIF-dependent host response in intestinal vs. systemic infection with Gram-negative enterophathogens.


Assuntos
Proteínas Adaptadoras de Transporte Vesicular/metabolismo , Trato Gastrointestinal/imunologia , Trato Gastrointestinal/microbiologia , Transdução de Sinais , Receptor 4 Toll-Like/metabolismo , Yersinia enterocolitica/imunologia , Yersinia enterocolitica/patogenicidade , Proteínas Adaptadoras de Transporte Vesicular/genética , Animais , Fígado/microbiologia , Camundongos , Camundongos Knockout , Baço/microbiologia
14.
Vaccine ; 30(4): 691-702, 2012 Jan 17.
Artigo em Inglês | MEDLINE | ID: mdl-22146759

RESUMO

BACKGROUND: DNA vaccines remain an important component of HIV vaccination strategies, typically as part of a prime/boost vaccination strategy with viral vector or protein boost. A number of DNA prime/viral vector boost vaccines are currently being evaluated for both preclinical studies and in Phase I and Phase II clinical trials. These vaccines would benefit from molecular adjuvants that increase correlates of immunity during the DNA prime. While HIV vaccine immune correlates are still not well defined, there are a number of immune assays that have been shown to correlate with protection from viral challenge including CD8+ T cell avidity, antigen-specific proliferation, and polyfunctional cytokine secretion. METHODOLOGY AND PRINCIPAL FINDINGS: Recombinant DNA vaccine adjuvants composed of a fusion between Surfactant Protein D (SP-D) and either CD40 Ligand (CD40L) or GITR Ligand (GITRL) were previously shown to enhance HIV-1 Gag DNA vaccines. Here we show that similar fusion constructs composed of the TNF superfamily ligands (TNFSFL) 4-1BBL, OX40L, RANKL, LIGHT, CD70, and BAFF can also enhanced immune responses to a HIV-1 Gag DNA vaccine. BALB/c mice were vaccinated intramuscularly with plasmids expressing secreted Gag and SP-D-TNFSFL fusions. Initially, mice were analyzed 2 weeks or 7 weeks following vaccination to evaluate the relative efficacy of each SP-D-TNFSFL construct. All SP-D-TNFSFL constructs enhanced at least one Gag-specific immune response compared to the parent vaccine. Importantly, the constructs SP-D-4-1BBL, SP-D-OX40L, and SP-D-LIGHT enhanced CD8+ T cell avidity and CD8+/CD4+ T cell proliferation 7 weeks post vaccination. These avidity and proliferation data suggest that 4-1BBL, OX40L, and LIGHT fusion constructs may be particularly effective as vaccine adjuvants. Constructs SP-D-OX40L, SP-D-LIGHT, and SP-D-BAFF enhanced Gag-specific IL-2 secretion in memory T cells, suggesting these adjuvants can increase the number of self-renewing Gag-specific CD8+ and/or CD4+ T cells. Finally adjuvants SP-D-OX40L and SP-D-CD70 increased T(H)1 (IgG2a) but not T(H)2 (IgG1) antibody responses in the vaccinated animals. Surprisingly, the B cell-activating protein BAFF did not enhance anti-Gag antibody responses when given as an SP-D fusion adjuvant, but nonetheless enhanced CD4+ and CD8+ T cell responses. CONCLUSIONS: We present evidence that various SP-D-TNFSFL fusion constructs can enhance immune responses following DNA vaccination with HIV-1 Gag expression plasmid. These data support the continued evaluation of SP-D-TNFSFL fusion proteins as molecular adjuvants for DNA and/or viral vector vaccines. Constructs of particular interest included SP-D-OX40L, SP-D-4-1BBL, SP-D-LIGHT, and SP-D-CD70. SP-D-BAFF was surprisingly effective at enhancing T cell responses, despite its inability to enhance anti-Gag antibody secretion.


Assuntos
Vacinas contra a AIDS/imunologia , Adjuvantes Imunológicos/metabolismo , HIV-1/imunologia , Fator de Necrose Tumoral alfa/metabolismo , Vacinas de DNA/imunologia , Produtos do Gene gag do Vírus da Imunodeficiência Humana/imunologia , Vacinas contra a AIDS/administração & dosagem , Vacinas contra a AIDS/genética , Adjuvantes Imunológicos/genética , Animais , Linfócitos T CD8-Positivos/imunologia , Proliferação de Células , Feminino , HIV-1/genética , Humanos , Injeções Intramusculares , Ligantes , Camundongos , Camundongos Endogâmicos BALB C , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/imunologia , Fator de Necrose Tumoral alfa/imunologia , Vacinas de DNA/administração & dosagem , Vacinas de DNA/genética , Produtos do Gene gag do Vírus da Imunodeficiência Humana/genética
15.
J Leukoc Biol ; 90(2): 389-98, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21586676

RESUMO

HIV-1 does not significantly activate cellular immunity, which has made it difficult to use attenuated forms of HIV-1 as a vaccine. In contrast, EBV induces robust T cell responses in most infected individuals, perhaps as this virus contains LMP1, a viral mimic of CD40, which is a key activating molecule for DCs and macrophages. Consequently, studies were conducted using LMP1 and LMP1-CD40, a related construct formed by replacing the intracellular signaling domain of LMP1 with that of CD40. Upon electroporation into DCs, LMP1 and LMP1-CD40 mRNAs were sufficient to up-regulate costimulatory molecules and proinflammatory cytokines, indicating that these molecules can function in isolation as adjuvant-like molecules. As a first step toward an improved HIV vaccine, LMP1 and LMP1-CD40 were introduced into a HIV-1 construct to produce virions encoding these proteins. Transduction of DCs and macrophages with these viruses induced morphological changes and up-regulated costimulatory molecules and cytokine production by these cells. HIV-LMP1 enhanced the antigen-presenting function of DCs, as measured in an in vitro immunization assay. Taken together, these data show that LMP1 and LMP1-CD40 are portable gene cassettes with strong adjuvant properties that can be introduced into viruses such as HIV, which by themselves, are insufficient to induce protective cellular immunity.


Assuntos
Vacinas contra a AIDS/imunologia , Células Dendríticas/imunologia , Herpesvirus Humano 4/imunologia , Proteínas da Matriz Viral/imunologia , Adjuvantes Imunológicos/uso terapêutico , Apresentação de Antígeno , Antígenos CD40/uso terapêutico , HIV/genética , HIV/imunologia , Herpesvirus Humano 4/química , Herpesvirus Humano 4/genética , Humanos , Mimetismo Molecular/imunologia , Transdução Genética , Proteínas da Matriz Viral/uso terapêutico
16.
Vaccine ; 29(11): 2110-9, 2011 Mar 03.
Artigo em Inglês | MEDLINE | ID: mdl-21241732

RESUMO

BACKGROUND: Dendritic cell (DC) therapy is a promising technology for the treatment of HIV infected individuals. HIV-1 Gag- and Nef RNA-loaded DC have previously been shown to induce immune responses ex vivo following coculture with autologous lymphocytes. However, polyfunctionality and memory responses following coculture have not been evaluated. In addition, little is known regarding whether specific HIV-1 proteome components, such as highly conserved regions of the HIV-1, could enhance clinical responses following DC therapy. METHODOLOGY AND PRINCIPAL FINDINGS: To determine the breadth of the immune responses to antigen loaded DC, we analyzed polyfunctional T cell response ex vivo to Gag RNA loaded DC. Blood samples were used to generate monocyte derived DC, which were then matured and cocultured with autologous lymphocytes. We found that cytokine-matured DC loaded with Gag RNA was able to induce Gag-specific IFN-γ and IL-2 responses after a 12-day coculture. We characterized these responses by polyfunctional intracellular cytokine staining and evaluation of T cell memory phenotypes. Central memory CD8+ T cells were induced ex vivo after DC coculture from each of 3 patients, and the effector memory pool was increased by DC coculture from 2 patients. We also observed a decrease in the terminal effector and intermediate CD8+ T cell pool and an increase in the naïve/other population. There was a reduction in terminal effector and intermediate CD4+ T cells, and a corresponding increase in naïve/other CD4+ T cells. Finally, we evaluated conserved regions of Gag as a novel DC therapy immunogen and found that a conserved element (CE) p24 Gag antigen elicited IFN-γ and IL-2 responses comparable to those induced by a full-length Gag antigen. CONCLUSIONS: We showed that RNA-loaded DC therapy induced a polyfunctional T cell response ex vivo, supporting the use of such DC-therapy for HIV infection. However, the central and effector memory phenotypes of T cells did not appear to be enhanced during coculture with Gag RNA-loaded DC. Furthermore, comparable antigen-specific responses were induced in HIV infected individuals using full-length Gag or only conserved elements of the Gag p24 protein. This indicates that immune responses can be focused onto the conserved elements of Gag in the absence of other Gag components.


Assuntos
Vacinas contra a AIDS/imunologia , Células Dendríticas/imunologia , Infecções por HIV/terapia , Produtos do Gene gag do Vírus da Imunodeficiência Humana/imunologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Proteína do Núcleo p24 do HIV/imunologia , Infecções por HIV/imunologia , HIV-1/genética , HIV-1/imunologia , Humanos , Memória Imunológica , Interferon gama/imunologia , Interleucina-2/imunologia , RNA Viral/genética
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa