Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 217
Filtrar
Más filtros

Base de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Vaccine ; 38(11): 2626-2635, 2020 03 04.
Artículo en Inglés | MEDLINE | ID: mdl-32057572

RESUMEN

An effective prophylactic vaccine targeting HIV must induce a robust humoral response and must direct the bulk of this response to the mucosa-the primary site of HIV transmission. The chemokine, CCL28, is secreted by epithelial cells at mucosal surfaces and recruits' cells expressing its receptor CCR10. CCR10 is predominantly expressed by IgA + ASCs. We hypothesized that co-immunization with plasmid DNA encoding consensus envelope antigens with plasmid-encoded CCL28 would enhance anti-HIV IgA responses at mucosal surfaces. Indeed, animals receiving pCCL28 and pEnvA/C had significantly increased HIV-specific IgA in fecal extract. Surprisingly, CCL28 co-immunization induced a significant increase in anti-HIV IgG in the serum in mice compared to those receiving pEnvA/C alone. These robust antibody responses were not associated with changes in the frequency of germinal center B cells but depended upon the expression of CCR10, as these responses we abolished in CCR10-deficient animals. Finally, immunization with CCL28 led to increased frequencies in HIV-specific CCR10 + and CCR10 + IgA + B cells in the small intestine and Peyer's patches of vaccinated animals as compared to those receiving pEnvA/C alone. These data indicate that CCL28 administration can enhance antigen-specific humoral responses systemically and at mucosal surfaces.


Asunto(s)
Vacunas contra el SIDA/administración & dosificación , Adyuvantes Inmunológicos/administración & dosificación , Quimiocinas CC/administración & dosificación , Receptores CCR10/genética , Vacunas de ADN/administración & dosificación , Animales , Anticuerpos Anti-VIH/inmunología , VIH-1/inmunología , Inmunidad Mucosa , Inmunoglobulina A/inmunología , Inmunoglobulina G/sangre , Ratones , Membrana Mucosa/inmunología , Productos del Gen env del Virus de la Inmunodeficiencia Humana/inmunología
2.
Mucosal Immunol ; 9(1): 13-23, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25943275

RESUMEN

There have been encouraging results for the development of an effective HIV vaccine. However, many questions remain regarding the quality of immune responses and the role of mucosal antibodies. We addressed some of these issues by using a simian immunodeficiency virus (SIV) DNA vaccine adjuvanted with plasmid-expressed mucosal chemokines combined with an intravaginal SIV challenge in rhesus macaque (RhM) model. We previously reported on the ability of CCR9 and CCR10 ligand (L) adjuvants to enhance mucosal and systemic IgA and IgG responses in small animals. In this study, RhMs were intramuscularly immunized five times with either DNA or DNA plus chemokine adjuvant delivered by electroporation followed by challenge with SIVsmE660. Sixty-eight percent of all vaccinated animals (P<0.01) remained either uninfected or had aborted infection compared with only 14% in the vaccine naïve group. The highest protection was observed in the CCR10L chemokines group, where six of nine animals had aborted infection and two remained uninfected, leading to 89% protection (P<0.001). The induction of mucosal SIV-specific antibodies and neutralization titers correlated with trends in protection. These results indicate the need to further investigate the contribution of chemokine adjuvants to modulate immune responses and the role of mucosal antibodies in SIV/HIV protection.


Asunto(s)
Vacunas contra el SIDA/administración & dosificación , Anticuerpos Antivirales/biosíntesis , Quimiocinas/inmunología , Inmunidad Mucosa/efectos de los fármacos , Síndrome de Inmunodeficiencia Adquirida del Simio/prevención & control , Vacunas de ADN/administración & dosificación , Vacunas contra el SIDA/genética , Vacunas contra el SIDA/inmunología , Adyuvantes Inmunológicos/administración & dosificación , Animales , Quimiocinas/administración & dosificación , Quimiocinas/genética , Femenino , Inmunidad Celular/efectos de los fármacos , Ligandos , Macaca mulatta , Plásmidos/química , Plásmidos/inmunología , Receptores CCR/genética , Receptores CCR/inmunología , Receptores CCR10/genética , Receptores CCR10/inmunología , Síndrome de Inmunodeficiencia Adquirida del Simio/inmunología , Síndrome de Inmunodeficiencia Adquirida del Simio/virología , Virus de la Inmunodeficiencia de los Simios/efectos de los fármacos , Virus de la Inmunodeficiencia de los Simios/inmunología , Vacunación , Vacunas de ADN/genética , Vacunas de ADN/inmunología , Vagina/efectos de los fármacos , Vagina/inmunología , Vagina/virología
3.
Hum Vaccin Immunother ; 11(8): 1945-53, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26020621

RESUMEN

Leptospirosis is a bacterial zoonotic disease caused by an infection with a spirochete belonging to the genus Leptospira. In animals, leptospirosis displays a wide range of pathologies, including fever, abortion, icterus, and uveitis. Conversely, infection in humans is associated with multi-organ injury, resulting in an increased rate of fatalities. Pathogenic leptospires are able to translocate through cell monolayers at a rate significantly greater than that of non-pathogenic leptospires. Thus, vaccine approaches have been focused on targeting bacterial motility, lipopolysaccharides (LPSs), lipoproteins, outer-membrane proteins (OMPs) and other potential virulence factors. Previous studies have indicated that leptospiral proteins elicit long-lasting immunological memory in infected humans. In the study reported here, the efficacy of a synthetic consensus DNA vaccine developed against the Leptospira membrane lipoprotein LipL45 was tested. After in vivo electroporation (EP) mediated intramuscular immunization with a synthetic LipL45 DNA vaccine (pLipL45) immunized mice developed a significant cellular response along with the development of anti-LipL45-specific antibodies. Specifically, the pLipL45 vaccine induced a significant Th1 type immune response, indicated by the higher production of IL-12 and IFN-γ cytokines. The results presented here are the first demonstration that a LipL45 based DNA immunogen has potential as a anti-Leptospira vaccine.


Asunto(s)
Proteínas de la Membrana Bacteriana Externa/inmunología , Vacunas Bacterianas/inmunología , Leptospirosis/prevención & control , Lipoproteínas/inmunología , Vacunas de ADN/inmunología , Animales , Anticuerpos Antibacterianos/sangre , Proteínas de la Membrana Bacteriana Externa/genética , Vacunas Bacterianas/administración & dosificación , Vacunas Bacterianas/genética , Electroporación , Femenino , Inyecciones Intramusculares , Interferón gamma/metabolismo , Interleucina-12/metabolismo , Leucocitos Mononucleares/inmunología , Lipoproteínas/genética , Ratones Endogámicos BALB C , Células TH1/inmunología , Vacunas de ADN/administración & dosificación , Vacunas de ADN/genética , Vacunas Sintéticas/administración & dosificación , Vacunas Sintéticas/genética , Vacunas Sintéticas/inmunología
4.
Cancer Gene Ther ; 21(12): 507-17, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25394503

RESUMEN

Melanoma is the most deadly type of skin cancer, constituting annually ∼ 75% of all cutaneous cancer-related deaths due to metastatic spread. Currently, because of metastatic spread, there are no effective treatment options for late-stage metastatic melanoma patients. Studies over the past two decades have provided insight into several complex molecular mechanisms as to how these malignancies evade immunological control, indicating the importance of immune escape or suppression for tumor survival. Thus, it is essential to develop innovative cancer strategies and address immune obstacles with the goal of generating more effective immunotherapies. One important area of study is to further elucidate the role and significance of myeloid-derived suppressor cells (MDSCs) in the maintenance of the tumor microenvironment. These cells possess a remarkable ability to suppress immune responses and, as such, facilitate tumor growth. Thus, MDSCs represent an important new target for preventing tumor progression and escape from immune control. In this study, we investigated the role of MDSCs in immune suppression of T cells in an antigen-specific B16 melanoma murine system utilizing a novel synthetic tyrosinase (Tyr) DNA vaccine therapy in both prophylactic and therapeutic models. This Tyr vaccine induced a robust and broad immune response, including directing CD8 T-cell infiltration into tumor sites. The vaccine also reduced the number of MDSCs in the tumor microenvironment through the downregulation of monocyte chemoattractant protein 1, interleukin-10, CXCL5 and arginase II, factors important for MDSC expansion. This novel synthetic DNA vaccine significantly reduced the melanoma tumor burden and increased survival in vivo, due likely, in part, to the facilitation of a change in the tumor microenvironment through MDSC suppression.


Asunto(s)
Vacunas contra el Cáncer/inmunología , Melanoma/inmunología , Melanoma/terapia , Monofenol Monooxigenasa/inmunología , Células Mieloides/inmunología , Vacunas de ADN/inmunología , Animales , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Vacunas contra el Cáncer/administración & dosificación , Citocinas/metabolismo , Modelos Animales de Enfermedad , Femenino , Humanos , Inmunidad Celular , Inmunidad Humoral , Inmunización , Inmunomodulación , Melanoma/genética , Melanoma/mortalidad , Melanoma/patología , Melanoma/prevención & control , Melanoma Experimental , Ratones , Monofenol Monooxigenasa/antagonistas & inhibidores , Monofenol Monooxigenasa/genética , Células Mieloides/metabolismo , Especificidad del Receptor de Antígeno de Linfocitos T , Carga Tumoral/inmunología , Microambiente Tumoral , Vacunas de ADN/administración & dosificación
5.
Cancer Gene Ther ; 20(12): 652-62, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24310062

RESUMEN

There are well over a quarter of a billion chronic hepatitis B virus (HBV) carriers across the globe. Most carriers are at high risk for development of liver cirrhosis and subsequent progression to hepatocellular carcinoma. It is therefore imperative to develop new approaches for immunotherapy against this infection. Antibodies and cytotoxic T cells to different HBV antigens are believed to be important for reducing viral load and clearing HBV-infected cells from the liver. Some of the major challenges facing current vaccine candidates have been their inability to induce both humoral and cellular immunity to multiple antigenic targets and the induction of potent immune responses against the major genotypes of HBV. In this study, highly optimized synthetic DNA plasmids against the HBV consensus core (HBc) and surface (HBs) antigens genotypes A and C were developed and evaluated for their immune potential. These plasmids, which encode the most prevalent genotypes of the virus, were observed to individually induce binding antibodies to HBs antigens and drove robust cell-mediated immunity in animal models. Similar responses to both HBc and HBs antigens were observed when mice and non-human primates were inoculated with the HBc-HBs cocktails. In addition to the cytotoxic T lymphocyte activities exhibited by the immunized mice, the vaccine-induced responses were broadly distributed across multiple antigenic epitopes. These elements are believed to be important to develop an effective therapeutic vaccine. These data support further evaluation of multivalent synthetic plasmids as therapeutic HBV vaccines.


Asunto(s)
Formación de Anticuerpos/genética , Citotoxicidad Inmunológica/genética , Genotipo , Antígenos del Núcleo de la Hepatitis B/genética , Antígenos de Superficie de la Hepatitis B/genética , Virus de la Hepatitis B/genética , Vacunas de ADN/genética , Animales , Formación de Anticuerpos/inmunología , Línea Celular , Citocinas/metabolismo , Citotoxicidad Inmunológica/inmunología , Epítopos de Linfocito T/inmunología , Femenino , Orden Génico , Hepatitis B/inmunología , Hepatitis B/prevención & control , Antígenos del Núcleo de la Hepatitis B/inmunología , Antígenos de Superficie de la Hepatitis B/inmunología , Virus de la Hepatitis B/inmunología , Humanos , Inmunidad Celular , Inmunidad Humoral , Hígado/inmunología , Pruebas de Función Hepática , Macaca mulatta , Ratones , Plásmidos/genética , Bazo/inmunología , Linfocitos T Citotóxicos/inmunología , Vacunación , Vacunas de ADN/inmunología
6.
Cancer Gene Ther ; 20(10): 552-63, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23969886

RESUMEN

Hepatotropic pathogens, such as hepatitis B (HBV) and hepatitis C (HCV), often escape cellular immune clearance resulting in chronic infection. As HBV and HCV infections are the most common causes of hepatocellular carcinoma (HCC), prevention of these infections is believed to be key to the prevention of HCC. It is believed that an effective immune therapy must induce strong cytotonic T lymphocytes (CTLs) that can migrate into the liver, where they can clear infected hepatocytes. Here, we compared the induction of CD8 T cells by two different DNA immunization methods for T-cell differentiation, function, memory programming and their distribution within relevant tissues in a highly controlled fashion. We used hydrodynamic tail vein injection of plasmid to establish liver-specific LCMV-gp antigen (Ag) transient expression, and studied CD8 T cells induced using the P14 transgenic mouse model. CD8 T cells from this group exhibited unique and limited expansion, memory differentiation, polyfunctionality and cytotoxicity compared with T cells generated in intramuscularly immunized mice. This difference in liver-generated expansion resulted in lower memory CD8 T-cell frequency, leading to reduced protection against lethal viral challenge. These data show an unusual induction of naive CD8 T cells contributed to the lower frequency of Ag-specific CTLs observed after immunization in the liver, suggesting that limited priming in liver compared with peripheral tissues is responsible for this outcome.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Hepatocitos/inmunología , Neoplasias Hepáticas Experimentales/inmunología , Linfocitos T Citotóxicos/inmunología , Vacunas de ADN/administración & dosificación , Inmunidad Adaptativa/inmunología , Secuencia de Aminoácidos , Animales , Antígenos Virales/inmunología , Linfocitos T CD8-positivos/patología , Glicoproteínas/inmunología , Hepatocitos/patología , Hidrodinámica , Inmunización/métodos , Memoria Inmunológica , Neoplasias Hepáticas Experimentales/patología , Neoplasias Hepáticas Experimentales/prevención & control , Activación de Linfocitos , Virus de la Coriomeningitis Linfocítica/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Linfocitos T Citotóxicos/patología , Vacunas de ADN/inmunología
7.
Infect Immun ; 81(10): 3709-20, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23897618

RESUMEN

A vaccine candidate that elicits humoral and cellular responses to multiple sporozoite and liver-stage antigens may be able to confer protection against Plasmodium falciparum malaria; however, a technology for formulating and delivering such a vaccine has remained elusive. Here, we report the preclinical assessment of an optimized DNA vaccine approach that targets four P. falciparum antigens: circumsporozoite protein (CSP), liver stage antigen 1 (LSA1), thrombospondin-related anonymous protein (TRAP), and cell-traversal protein for ookinetes and sporozoites (CelTOS). Synthetic DNA sequences were designed for each antigen with modifications to improve expression and were delivered using in vivo electroporation (EP). Immunogenicity was evaluated in mice and nonhuman primates (NHPs) and assessed by enzyme-linked immunosorbent assay (ELISA), gamma interferon (IFN-γ) enzyme-linked immunosorbent spot (ELISpot) assay, and flow cytometry. In mice, DNA with EP delivery induced antigen-specific IFN-γ production, as measured by ELISpot assay and IgG seroconversion against all antigens. Sustained production of IFN-γ, interleukin-2, and tumor necrosis factor alpha was elicited in both the CD4(+) and CD8(+) T cell compartments. Furthermore, hepatic CD8(+) lymphocytes produced LSA1-specific IFN-γ. The immune responses conferred to mice by this approach translated to the NHP model, which showed cellular responses by ELISpot assay and intracellular cytokine staining. Notably, antigen-specific CD8(+) granzyme B(+) T cells were observed in NHPs. Collectively, the data demonstrate that delivery of gene sequences by DNA/EP encoding malaria parasite antigens is immunogenic in animal models and can harness both the humoral and cellular arms of the immune system.


Asunto(s)
Antígenos de Protozoos/inmunología , ADN Protozoario/inmunología , Hígado/parasitología , Plásmidos/genética , Plasmodium falciparum/fisiología , Esporozoítos/inmunología , Animales , Línea Celular , ADN Protozoario/genética , Femenino , Inmunidad Celular , Inmunidad Humoral , Macaca mulatta , Malaria Falciparum/inmunología , Malaria Falciparum/parasitología , Ratones , Ratones Endogámicos BALB C
8.
Cancer Gene Ther ; 19(11): 779-87, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23037809

RESUMEN

The prevalence of hepatitis B virus (HBV) infection in Asia and sub-Sahara Africa is alarming. With quarter of a billion people chronically infected worldwide and at risk of developing liver cancer, the need for a prophylactic or therapeutic vaccination approach that can effectively induce protective responses against the different genotypes of HBV is more important than ever. Such a strategy will require both the induction of a strong antigen-specific immune response and the subsequent deployment of immune response towards the liver. Here, we assessed the ability of a synthetic DNA vaccine encoding a recombinant consensus plasmid from genotype A through E of the HBV core antigen (HBcAg), to drive immunity in the liver. Intramuscular vaccination induced both strong antigen-specific T cell and high titer antibody responses systematically and in the liver. Furthermore, immunized mice showed strong cytotoxic responses that eliminate adoptively transferred HBV-coated target cells. Importantly, vaccine-induced immune responses provided protection from HBcAg plasmid-based liver transfection in a hydrodynamic liver transfection model. These data provide important insight into the generation of peripheral immune responses that are recruited to the liver-an approach that can be beneficial in the search for vaccines or immune-therapies to liver disease.


Asunto(s)
ADN Viral/inmunología , Antígenos del Núcleo de la Hepatitis B/inmunología , Vacunas contra Hepatitis B/inmunología , Hígado/inmunología , Vacunas de ADN/inmunología , Administración Intranasal , Animales , Formación de Anticuerpos , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/virología , ADN Viral/administración & dosificación , ADN Viral/genética , Ensayo de Immunospot Ligado a Enzimas , Femenino , Genes Virales , Genotipo , Hepatitis B/inmunología , Hepatitis B/terapia , Antígenos del Núcleo de la Hepatitis B/administración & dosificación , Antígenos del Núcleo de la Hepatitis B/genética , Vacunas contra Hepatitis B/administración & dosificación , Vacunas contra Hepatitis B/genética , Virus de la Hepatitis B/genética , Virus de la Hepatitis B/inmunología , Hepatocitos/inmunología , Hepatocitos/virología , Hígado/virología , Ratones , Ratones Endogámicos BALB C , Plásmidos/metabolismo , Transfección , Vacunación/métodos , Vacunas de ADN/administración & dosificación
9.
Gene Ther ; 18(11): 1070-7, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21544096

RESUMEN

DNA-based vaccines, while highly immunogenic in mice, generate significantly weaker responses in primates. Therefore, current efforts are aimed at increasing their immunogenicity, which include optimizing the plasmid/gene, the vaccine formulation and method of delivery. For example, co-immunization with molecular adjuvants encoding an immunomodulatory protein has been shown to improve the antigen (Ag)-specific immune response. Thus, the incorporation of enhancing elements, such as these, may be particularly important in the influenza model in which high titered antibody (Ab) responses are critical for protection. In this regard, we compared the ability of plasmid-encoded high-mobility group box 1 protein (HMGB1), a novel cytokine in which we have previously mutated in order to increase DNA vaccine immunogenicity, with boost Ag-specific immune responses during DNA vaccination with influenza A/PR/8/34 nucleoprotein or the hemagglutinin of A novel H1N1/09. We show that the HMGB1 adjuvant is capable of enhancing adaptive effector and memory immune responses. Although Ag-specific antibodies were detected in all vaccinated animals, a greater neutralizing Ab response was associated with the HMGB1 adjuvant. Furthermore, these responses improved CD8 T+-cell effector and memory responses and provided protection against a lethal mucosal influenza A/PR/8/34 challenge. Thus, co-immunization with HMGB1 has strong in vivo adjuvant activity during the development of immunity against plasmid-encoded Ag.


Asunto(s)
Adyuvantes Inmunológicos/administración & dosificación , Proteína HMGB1/inmunología , Subtipo H1N1 del Virus de la Influenza A/inmunología , Vacunas contra la Influenza/inmunología , Vacunas de ADN/inmunología , Animales , Anticuerpos Neutralizantes/biosíntesis , Anticuerpos Antivirales/biosíntesis , Epítopos , Femenino , Memoria Inmunológica , Ratones , Ratones Endogámicos BALB C , Infecciones por Orthomyxoviridae/prevención & control , Linfocitos T/inmunología , Vacunación/métodos
10.
Gene Ther ; 18(3): 258-65, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20962869

RESUMEN

The magnitude of the immune response to a DNA vaccine depends on three criteria--the optimized vector design, the use of a suitable adjuvant and the successful delivery and subsequent expression of the plasmid in the target tissue. In vivo electroporation (EP) has proved to be particularly effective in efficiently delivering DNA immunogens to the muscle and the skin, and indeed several devices have entered into human clinical trials. Here, we report on a novel concept of DNA delivery to the dermal tissue using a minimally invasive EP device, which is powered using low-voltage parameters. We show that this prototype device containing a novel 4 × 4-electrode array results in robust and reproducible transfection of dermal tissue and subsequent antigen expression at the injection site. Using DNA encoding for NP and M2e influenza antigens, we further show induction of potent cellular responses in a mouse model as measured by antigen-specific T-cell ELISpot assays. Importantly, 100% of the immunized animals were protected when challenged with VN/1203/04 (H5N1) strain of influenza. We have also extended our findings to a guinea-pig model and demonstrated induction of HI titers greater than 1:40 against a pandemic novel H1N1 virus showing proof of concept efficacy for DNA delivery with the prototype device in a broad spectrum of species and using multiple antigens. Finally, we were able to generate protective HI titers in macaques against the same novel H1N1 strain. Our results suggest that the minimally invasive dermal device may offer a safe, tolerable and efficient method to administer DNA vaccinations in a prophylactic setting, and thus potentially represents an important new option for improved DNA vaccine delivery in vivo.


Asunto(s)
Electroporación/instrumentación , Subtipo H1N1 del Virus de la Influenza A/inmunología , Subtipo H5N1 del Virus de la Influenza A/inmunología , Vacunas contra la Influenza/administración & dosificación , Transfección/instrumentación , Vacunas de ADN/administración & dosificación , Animales , Antígenos Virales/genética , Electrodos , Ensayo de Immunospot Ligado a Enzimas , Femenino , Cobayas , Vacunas contra la Influenza/inmunología , Ratones , Ratones Endogámicos BALB C , Vacunas de ADN/inmunología
11.
Gene Ther ; 17(1): 72-82, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19847203

RESUMEN

A hurdle facing DNA vaccine development is the ability to generate strong immune responses systemically and at local immune sites. We report a novel systemically administered DNA vaccination strategy using intramuscular codelivery of CCL27 or CCL28, which elicited elevated peripheral IFN-gamma and antigen-specific IgG while driving antigen-specific T-cell secretion of cytokine and antibody production in the gut-associated lymphoid tissue and lung. This strategy resulted in induction of long-lived antibody responses that neutralized influenza A/PR8/34 and protected mice from morbidity and mortality associated with a lethal intranasal viral challenge. This is the first example of the use of CCL27 and CCL28 chemokines as adjuvants to influence a DNA vaccine strategy, suggesting further examination of this approach for manipulation of vaccine-induced immunity impacting both quality and phenotype of responses.


Asunto(s)
Adyuvantes Inmunológicos , Quimiocina CCL27/inmunología , Quimiocinas CC/inmunología , Inmunización/métodos , Inmunoglobulina G/biosíntesis , Plásmidos , Vacunas de ADN/inmunología , Animales , Virus de la Influenza A/inmunología , Interferón gamma/biosíntesis , Ratones
12.
J Med Primatol ; 36(4-5): 276-84, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17669216

RESUMEN

BACKGROUND: The cell mediated immune profiles following immunization with a recombinant DNA vaccine was assessed in the simian-human immunodeficiency virus (SHIV) and Macaque model. Earlier work demonstrated increased numbers of antigen specific CD8 and CD4 effector cells able to secrete IFN-gamma. METHOD: The vaccine strategy included co-immunization of a DNA based vaccine alone or in combination with a macaque IL-12 expressing plasmid (pmacIL12). Antigen activated lymphocytes were studied for activation of a set of immunological molecules. RESULTS: The current study demonstrates lymphocytes isolated and activated from the group that was immunized with DNA and pmacIL12 had a higher level of IFN-gamma producing cells. We also observed a different immunological profile when comparing the cells isolated from macaques immunized with DNA as compared to those animals that also received pmacIL12. CONCLUSION: The observed immune profiles are reflective of the co-delivery of pmacIL12 and demonstrates that IL-12 can increase the magnitude and polyfunctionality of the cellular immune response.


Asunto(s)
Interleucina-12/inmunología , Macaca fascicularis , Proteínas de los Retroviridae/inmunología , Vacunas contra el SIDAS/inmunología , Síndrome de Inmunodeficiencia Adquirida del Simio/inmunología , Virus de la Inmunodeficiencia de los Simios/inmunología , Vacunas de ADN/inmunología , Animales , Citometría de Flujo , Productos del Gen env/genética , Productos del Gen env/inmunología , Productos del Gen gag/genética , Productos del Gen gag/inmunología , Productos del Gen pol/genética , Productos del Gen pol/inmunología , Humanos , Inmunidad Celular/efectos de los fármacos , Inmunidad Celular/inmunología , Interferón gamma/sangre , Interleucina-12/genética , Plásmidos/genética , Plásmidos/inmunología , ARN Viral/química , ARN Viral/genética , Distribución Aleatoria , Proteínas de los Retroviridae/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Vacunas contra el SIDAS/genética , Síndrome de Inmunodeficiencia Adquirida del Simio/prevención & control , Síndrome de Inmunodeficiencia Adquirida del Simio/orina , Vacunas de ADN/genética
13.
Cancer Gene Ther ; 13(10): 969-74, 2006 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16763607

RESUMEN

In vivo electroporation has been used to efficiently deliver drugs and 'therapeutic' genes to tumors, including melanoma lesions. This study reports on the effect of intratumoral delivery of an optimized DNA plasmid expressing interleukin-15 (pIL-15) on established murine melanoma tumors. IL-15 has been demonstrated to have a pivotal role in the function of memory CD8+ T cells and natural killer cells, which are critical for tumor immunosurveillance. In this study, C57BL/6 mice were injected with B16.F10 melanoma cells and randomized into different experimental groups: untreated (P-V-E-), treated with pIL-15 (P+) or backbone plasmid (V+), with or without electroporation (E+ or E-). Treatment was performed intratumorally with 50 microg of plasmid on days 0, 4 and 7 and tumor volume/size, tumor regression and long-term survival were measured. At day 100 after initiation of treatment, the percentage of mice surviving with complete tumor regression in the P-V+E+, P+V-E-, P+V-E+ and P-V-E- treatment groups were 0, 12.5, 37.5 and 0%, respectively. These results demonstrate the ability of pIL-15 to mediate B16 melanoma regression, with the effect being significantly enhanced by electroporative delivery. This is the first description of the ability of a naked DNA plasmid expressing IL-15 to alone mediate complete regression of B16 melanoma tumors and underscores the potential clinical use of these plasmids for the treatment of malignant tumors when delivered with in vivo electroporation.


Asunto(s)
Interleucina-15/administración & dosificación , Melanoma Experimental/terapia , Plásmidos , Animales , Electroporación , Femenino , Inyecciones Intralesiones , Melanoma Experimental/patología , Ratones , Ratones Endogámicos C57BL
14.
Cell Death Differ ; 12 Suppl 1: 962-70, 2005 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15832179

RESUMEN

The destruction of CD4(+) T cells and eventual induction of immunodeficiency is a hallmark of the human immunodeficiency virus type 1 infection (HIV-1). However, the mechanism of this destruction remains unresolved. Several auxiliary proteins have been proposed to play a role in this aspect of HIV pathogenesis including a 14 kDa protein named viral protein R (Vpr). Vpr has been implicated in the regulation of various cellular functions including apoptosis, cell cycle arrest, differentiation, and immune suppression. However, the mechanism(s) involved in Vpr-mediated apoptosis remains unresolved, and several proposed mechanisms for these effects are under investigation. In this review, we discuss the possibility that some of these proposed pathways might converge to modulate Vpr's behavior. Further, we also discuss caveats and future directions for investigation of the interesting biology of this HIV accessory gene.


Asunto(s)
Apoptosis , Linfocitos T CD4-Positivos/virología , Productos del Gen vpr/fisiología , VIH-1/fisiología , Proteínas Adaptadoras Transductoras de Señales , Animales , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Complejo del Señalosoma COP9 , Proteínas Portadoras/fisiología , Factores Eucarióticos de Iniciación/fisiología , Productos del Gen vpr/antagonistas & inhibidores , Productos del Gen vpr/farmacología , Proteínas HSP70 de Choque Térmico/farmacología , Humanos , Membranas Intracelulares/efectos de los fármacos , Potenciales de la Membrana/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Complejos Multiproteicos/fisiología , Péptido Hidrolasas/fisiología , Receptores de Glucocorticoides/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Productos del Gen vpr del Virus de la Inmunodeficiencia Humana
15.
J Med Primatol ; 31(4-5): 179-85, 2002 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-12390540

RESUMEN

We compared the immunogenicity of plasmid vaccines containing multiple human immunodeficiency virus (HIV) antigens and found that covaccination with plasmids expressing HIV-1 14 kDa vpr gene product profoundly reduces antigen-specific CD8-mediated cytotoxic T-cell activity (CTL). Interestingly, Th1 type responses against codelivered antigens (pGag-Pol, pNef, etc.) encoded by the plasmid vaccines were suppressed. This suggested that vpr might compromise CD8 T-cell immunity in vivo during infection. A pilot primate vaccine study was designed to test the hypothesis to compare the following groups: unvaccinated controls, animals vaccinated without simean immunodeficiency virus (SIV)-Nef antigen plasmid, and animals covaccinated with the identical plasmid antigen and a plasmid construct encoding SIV Vpr/Vpx. Animals were subsequently challenged intrarectally with pathogenic SIVmac251 after the final vaccination of a multiple immunization protocol. Control animals were all infected and exhibited high viral loads and rapid CD4+ T-cell loss. In contrast, the Nef plasmid-vaccinated animals were also infected but exhibited preservation of CD4+ T-cells and a multilog reduction in viral load compared with controls. Animals covaccinated multiple times with the Nef vaccine and pVpr/Vpx plasmid suffered rapid and profound loss of CD4+ T-cells. These results have important implications for the design of multicomponent and particle vaccines for HIV-1 as well as for our understanding of HIV/SIV pathogenesis in vivo.


Asunto(s)
Linfocitos T CD4-Positivos/citología , Productos del Gen nef/inmunología , Productos del Gen vpr/inmunología , Macaca mulatta/inmunología , Macaca mulatta/virología , Vacunas contra el SIDAS/genética , Vacunas contra el SIDAS/inmunología , Virus de la Inmunodeficiencia de los Simios/inmunología , Carga Viral , Animales , Recuento de Linfocito CD4 , Linfocitos T CD4-Positivos/inmunología , Modelos Animales de Enfermedad , Femenino , Productos del Gen nef/genética , Productos del Gen vpr/genética , Modelos Logísticos , Activación de Linfocitos , Masculino , Ratones , Ratones Endogámicos BALB C , Plásmidos/genética , ARN Viral/sangre , ARN Viral/genética , Síndrome de Inmunodeficiencia Adquirida del Simio/inmunología , Síndrome de Inmunodeficiencia Adquirida del Simio/prevención & control , Virus de la Inmunodeficiencia de los Simios/genética , Virus de la Inmunodeficiencia de los Simios/fisiología , Factores de Tiempo , Vacunas de ADN/genética , Vacunas de ADN/inmunología
16.
Int J Parasitol ; 32(5): 543-50, 2002 May.
Artículo en Inglés | MEDLINE | ID: mdl-11943227

RESUMEN

It is widely believed that a Th1 type CD4 response is critical for enhancement of CD8 immunity and for controlling HIV-1 infection. Th2 type responses, such as what might be seen in a chronic parasitic infection, would sacrifice cellular immunity and thus benefit the virus at the expense of the host. However, there has been little direct examination of the hypothesis in a primate model system. Accordingly, the simian immunodeficiency virus (SIV) infected rhesus macaque model was used to investigate the impact of immunisation with SIV expressing DNA constructs and co-injection with IL-4 on the SIV specific immunological responses, lymphocyte cell counts, as well as the impact on viral load. IL-4 is a Th2 type cytokine, which enhances antibody production and inhibits a CD4 Th1 phenotype. Rhesus macaques were infected with 10 AID50 of SIVmac239 and treated with 9-[2-(phosphonomethoxy)propyl]adenine (PMPA) 9 weeks post-infection. During PMPA treatment, animals were immunised with plasmids that expressed the SIV proteins, env, rev, gag and pol. In addition, they were immunised with a construct that encoded the gene for IL-4. IL-4 co-immunisation increased the neutralizing antibody titres in this group. Importantly, the viral loads in animals vaccinated with IL-4 expressing plasmid increased during the immunisation regimens despite the higher neutralizing antibody titres. In addition, neutralizing antibodies did not correlate with viral set point prior to PMPA treatment, however, there was a correlation between viral loads and antibody titres following the treatment with PMPA. Antibody titres decreased following the suppression of viral load. Importantly, vaccination in the absence of IL-4 protected CD4 levels without increasing viral load. The data support the hypothesis that inappropriate immune bias toward a Th2 pathway would ultimately enhance disease progression.


Asunto(s)
Interleucina-4/inmunología , Vacunas contra el SIDAS/inmunología , Síndrome de Inmunodeficiencia Adquirida del Simio/inmunología , Virus de la Inmunodeficiencia de los Simios/inmunología , Virus de la Inmunodeficiencia de los Simios/fisiología , Replicación Viral , Animales , Anticuerpos Antivirales/sangre , Recuento de Linfocito CD4 , Humanos , Inmunización , Interleucina-4/administración & dosificación , Interleucina-4/genética , Activación de Linfocitos , Macaca mulatta , Plásmidos/genética , Vacunas contra el SIDAS/administración & dosificación , Síndrome de Inmunodeficiencia Adquirida del Simio/prevención & control , Síndrome de Inmunodeficiencia Adquirida del Simio/virología , Carga Viral
17.
Virology ; 289(2): 334-42, 2001 Oct 25.
Artículo en Inglés | MEDLINE | ID: mdl-11689055

RESUMEN

Human immunodeficiency virus type 1 (HIV-1) Vpr, a 14-kDa virion-associated protein, plays an important role in the viral life cycle. Using a panel of truncated HIV-1 LTR-CAT constructs and Vpr expression plasmid, we have identified sequences from nucleotide -278 to -176 in LTR as Vpr-mediated transactivation domain. This region includes the glucocorticoid response element (GRE) in HIV-1 LTR. Transactivation by Vpr was noted with the HIV-1 LTR reporter constructs containing CAT or luciferase. A similar effect was also observed with a construct in which the GRE motif was linked to CAT. Studies involving Vpr mutants identified that helical domains I and III, and amino acid residues at G75 and C76, are responsible for GRE-mediated LTR transactivation. The transactivation function of Vpr is independent of its cell cycle arrest activity. Further, viral replication studies indicated that Vpr-mediated increase in viral replication is directly correlated with the ability of Vpr to transactivate HIV-1 LTR. The results presented here demonstrate that Vpr activates HIV-1 LTR through the host GR pathway and suggest that an intact GRE in the LTR is critical for Vpr activity.


Asunto(s)
Productos del Gen vpr/fisiología , Infecciones por VIH/virología , VIH-1/fisiología , Transactivadores/fisiología , Productos del Gen vpr/genética , Glucocorticoides/farmacología , Realizador del VIH , Duplicado del Terminal Largo de VIH/genética , VIH-1/química , VIH-1/patogenicidad , Células HeLa , Humanos , Luciferasas , Mutación , Elementos de Respuesta , Replicación Viral , Productos del Gen vpr del Virus de la Inmunodeficiencia Humana
18.
J Infect Dis ; 184(7): 809-16, 2001 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-11550123

RESUMEN

West Nile virus (WNV) is a vectorborne pathogen that induces brain inflammation and death. Recently, confirmed cases of infection and deaths have occurred in the United States Mid-Atlantic region. In this study, a DNA vaccine encoding the WNV capsid protein was constructed, and the in vivo immune responses generated were investigated in DNA vaccine-immunized mice. Antigen-specific humoral and cellular immune responses were observed, including a potent induction of antigen-specific Th1 and cytotoxic T lymphocyte responses. Strong induction of Th1-type immune responses included high levels of antigen-specific elaboration of the Th1-type cytokines interferon-gamma and interleukin-2 and beta-chemokines RANTES (regulated upon activation, normal T cell-expressed and secreted) and macrophage inflammatory protein-1beta. Dramatic infiltration of CD4 and CD8 T cells and macrophages also was observed at the muscle injection site. These results support the potential utility of this method as a tool for developing immunization strategies for WNV and other emerging pathogens.


Asunto(s)
Linfocitos T Citotóxicos/inmunología , Células TH1/inmunología , Vacunas de ADN/administración & dosificación , Vacunas Virales/administración & dosificación , Fiebre del Nilo Occidental/prevención & control , Virus del Nilo Occidental/inmunología , Animales , Antígenos Virales/inmunología , Cápside/inmunología , Citocinas/análisis , Modelos Animales de Enfermedad , Femenino , Ratones , Ratones Endogámicos BALB C , Músculos/inmunología , Virus del Nilo Occidental/aislamiento & purificación
19.
Oncogene ; 20(33): 4497-506, 2001 Jul 27.
Artículo en Inglés | MEDLINE | ID: mdl-11494145

RESUMEN

Prostate specific antigen (PSA) is a widely used marker for prostate cancer, which is secreted by normal prostate cells at low levels, but is produced more substantially by cancer cells. We have previously reported on the use of a DNA vaccine construct that encodes for human PSA gene to elicit host immune responses against cells producing PSA. DNA immunization strategy delivers DNA constructs encoding for a specific immunogen into the host, who becomes the in vivo protein source for the production of antigen. This antigen then is the focus of the resulting immune response. In this study, we examine the induction of immune responses and safety profiles in rhesus macaques immunized with DNA-based PSA vaccine. We observed induction of PSA-specific humoral response as well as positive PSA-specific lymphoproliferative (LPA) response in the vaccinated macaques. We also observed that the stimulated T cells from the PSA-immunized rhesus macaques produced higher levels of Th1 type cytokine IFN-gamma than the control vector immunized animals. On the other hand, DNA immunization did not result in any adverse effects in the immunized macaques, as indicated by complete blood counts, leukocyte differentials and hepatic and renal chemistries. The macaques appeared healthy, without any physical signs of toxicity throughout the observation period. In addition, we did not observe any adverse effect on the vaccination site. The apparent safety and immunogenecity of DNA immunization in this study suggest that further evaluation of this vaccination strategy is warranted.


Asunto(s)
Antígenos de Neoplasias/inmunología , Vacunas contra el Cáncer/inmunología , Antígeno Prostático Específico/inmunología , Vacunas de ADN/inmunología , Animales , Recuento de Células Sanguíneas , Vacunas contra el Cáncer/administración & dosificación , Vacunas contra el Cáncer/toxicidad , Relación Dosis-Respuesta Inmunológica , Humanos , Interferón gamma/metabolismo , Pruebas de Función Renal , Pruebas de Función Hepática , Activación de Linfocitos , Macaca mulatta , Masculino , Próstata/efectos de los fármacos , Seguridad , Células TH1/inmunología , Células TH1/metabolismo , Vacunación/efectos adversos , Vacunas de ADN/administración & dosificación , Vacunas de ADN/toxicidad , Vacunas Sintéticas/administración & dosificación , Vacunas Sintéticas/inmunología , Vacunas Sintéticas/toxicidad
20.
Virology ; 285(2): 204-17, 2001 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-11437655

RESUMEN

Multicomponent DNA vaccines were used to elicit immune responses, which can impact viral challenge in three separate rhesus macaque models. Eight rhesus macaques were immunized with DNA vaccines for HIV env/rev and SIV gag/pol and were challenged intravenously with 10 animal infective doses (AID(50)) of cell-free SHIV IIIB. Three of eight immunized rhesus macaques were protected, exhibiting no detectable virus. Animals protected from nonpathogenic SHIVIIIB challenge were rested for extended periods of time and were rechallenged first with pathogenic SIV(mac239) and subsequently with pathogenic SHIV89.6P viruses. Following the pathogenic challenges, all three vaccinated animals were negative for viral coculture and antigenemia and were negative by PCR. In contrast, the control animals exhibited antigenemia by 2 weeks postchallenge and exhibited greater than 10 logs of virus/10(6) cells in limiting dilution coculture. The control animals exhibited CD4 cell loss and developed SIV-related wasting with high viral burden and subsequently failed to thrive. Vaccinated animals remained virus-negative and were protected from the viral load, CD4 loss, disease, and death. We observed strong Th1-type cellular immune responses in the protected macaques throughout the study, suggesting their important roles in protection. These studies support the finding that multicomponent DNA vaccines can directly impact viral replication and disease in a highly pathogenic challenge system, thus potentially broadening our strategies against HIV.


Asunto(s)
Vacunas contra el SIDA/inmunología , VIH-1/inmunología , Síndrome de Inmunodeficiencia Adquirida del Simio/prevención & control , Virus de la Inmunodeficiencia de los Simios/inmunología , Vacunas de ADN/inmunología , Animales , VIH-1/genética , Humanos , Macaca mulatta
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA