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

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
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
2.
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
3.
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
4.
Nat Med ; 3(10): 1117-23, 1997 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-9334723

RESUMEN

The HIV-1 accessory gene product Vpr can influence viral pathogenesis by affecting viral replication as well as host cell transcription and proliferation. We have investigated the effects of Vpr on host cell activation and confirm that it influences cellular proliferation. However, we have also found that Vpr modulates T-cell receptor (TCR)-triggered apoptosis in a manner similar to that of glucocorticoids. In the absence of TCR-mediated activation, Vpr induces apoptosis whereas in its presence, Vpr interrupts the expected induction of apoptosis. This regulation of apoptosis is linked to Vpr suppression of NF-kappa B activity via the induction of I kappa B, an inhibitor of NF-kappa B. Further, Vpr suppresses expression of IL-2, IL-10, IL-12, TNF alpha and IL-4, all of which are NF-kappa B-dependent. The effects of Vpr could be reversed by RU486. Our finding that Vpr can regulate NF-kappa B supports the hypothesis that some aspects of viral pathogenesis are the consequence of cell dysregulation by Vpr.


Asunto(s)
Apoptosis/efectos de los fármacos , Citocinas/biosíntesis , Productos del Gen vpr/farmacología , VIH-1/inmunología , Activación de Linfocitos/efectos de los fármacos , Linfocitos/inmunología , FN-kappa B/metabolismo , Animales , Línea Celular , Células Cultivadas , Dexametasona/farmacología , Productos del Gen vpr/biosíntesis , Humanos , Hidrocortisona/farmacología , Interleucinas/biosíntesis , Linfocitos/efectos de los fármacos , Linfocitos/virología , Fitohemaglutininas , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/farmacología , Spodoptera , Transfección , Factor de Necrosis Tumoral alfa/biosíntesis , Productos del Gen vpr del Virus de la Inmunodeficiencia Humana
5.
Nat Med ; 3(5): 526-32, 1997 May.
Artículo en Inglés | MEDLINE | ID: mdl-9142121

RESUMEN

Novel approaches for the generation of more effective vaccines for HIV-1 are of significant importance. In this report we analyze the immunogenicity and efficacy of an HIV-1 DNA vaccine encoding env, rev and gag/pol in a chimpanzee model system. The immunized animals developed specific cellular and humoral immune responses. Animals were challenged with a heterologous chimpanzee titered stock of HIV-1 SF2 virus and followed for 48 weeks after challenge. Polymerase chain reaction coupled with reverse transcription (RT-PCR) results indicated infection in the control animal, whereas those animals vaccinated with the DNA constructs were protected from the establishment of infection. These studies serve as an important benchmark for the use of DNA vaccine technology for the production of protective immune responses.


Asunto(s)
Vacunas contra el SIDA/uso terapéutico , Infecciones por VIH/prevención & control , VIH-1/inmunología , Vacunas de ADN/uso terapéutico , Animales , Antígenos CD28/sangre , ADN Viral/análisis , Femenino , Anticuerpos Anti-VIH/sangre , Infecciones por VIH/inmunología , Leucocitos Mononucleares/inmunología , Ganglios Linfáticos/virología , Masculino , Pruebas de Neutralización , Pan troglodytes , Linfocitos T Citotóxicos/inmunología , Vacunación , Carga Viral
6.
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
7.
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
8.
J Clin Invest ; 90(2): 326-33, 1992 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-1386608

RESUMEN

RA is characterized by massive proliferation of synovial tissue, accompanying infiltration of the tissue with CD4+ T lymphocytes, and a genetic linkage to the MHC antigen HLA-DR4. Since T cells are restricted by class II MHC molecules such as DR4, this suggests a direct role for these CD4+ cells in pathogenesis. To investigate T cell receptor (TCR) usage in RA, we used oligonucleotide primers specific for each of the major alpha and beta TCR subfamilies to amplify cDNA derived from whole synovium or synovial tissue T cell lines in a family-specific manner. Detection of amplified DNA was facilitated by utilizing oligonucleotide probes derived from the constant regions of the TCRs. The TCR repertoire present in the synovial T cell lines was quite heterogeneous, with an average of 15 alpha chains and 15.8 beta chains detected. When synovial tissue was analyzed, the predominant TCR subfamilies detected tended to be more restricted, with an average of 4.6 alpha chains and 8.6 beta chains detected. This compared with an average of six alpha chains and 12 beta chains in nonrheumatoid synovial samples. The average percentage of synovia positive per TCR beta family was significantly lower for RA versus non-RA specimens (46.1 vs 65.6%, P = 0.034). These findings indicate that while a polyclonal population of T cells is present in RA synovium, the predominant patterns of TCR transcript expression may be somewhat more restricted, suggesting that TCR-based therapy of RA is possible.


Asunto(s)
Artritis Reumatoide/genética , Receptores de Antígenos de Linfocitos T alfa-beta/genética , Linfocitos T/fisiología , Secuencia de Bases , Expresión Génica , Humanos , Datos de Secuencia Molecular , Oligodesoxirribonucleótidos/química , Reacción en Cadena de la Polimerasa , ARN Mensajero/genética , Membrana Sinovial/fisiología
9.
J Clin Invest ; 103(6): 869-77, 1999 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-10079108

RESUMEN

The potential roles of adhesion molecules in the expansion of T cell-mediated immune responses in the periphery were examined using DNA immunogen constructs as model antigens. We coimmunized cDNA expression cassettes encoding the adhesion molecules intracellular adhesion molecule-1 (ICAM-1), lymphocyte function associated-3 (LFA-3), and vascular cell adhesion molecule-1 (VCAM-1) along with DNA immunogens, and we analyzed the resulting antigen-specific immune responses. We observed that antigen-specific T-cell responses can be enhanced by the coexpression of DNA immunogen and adhesion molecules ICAM-1 and LFA-3. Coexpression of ICAM-1 or LFA-3 molecules along with DNA immunogens resulted in a significant enhancement of T-helper cell proliferative responses. In addition, coimmunization with pCICAM-1 (and more moderately with pCLFA-3) resulted in a dramatic enhancement of CD8-restricted cytotoxic T-lymphocyte responses. Although VCAM-1 and ICAM-1 are similar in size, VCAM-1 coimmunization did not have any measurable effect on cell-mediated responses. These results suggest that ICAM-1 and LFA-3 provide direct T-cell costimulation. These observations are further supported by the finding that coinjection with ICAM-1 dramatically enhanced the level of interferon-gamma (IFN-gamma) and beta-chemokines macrophage inflammatory protein-1alpha (MIP-1alpha), MIP-1beta, and regulated on activation normal T-cell expression and secreted (RANTES) produced by stimulated T cells. Through comparative studies, we observed that ICAM-1/LFA-1 T-cell costimulatory pathways are independent of CD86/CD28 pathways and that they may synergistically expand T-cell responses in vivo.


Asunto(s)
Molécula 1 de Adhesión Intercelular/inmunología , Activación de Linfocitos , Proteínas Inflamatorias de Macrófagos/biosíntesis , Linfocitos T/inmunología , Vacunas contra el SIDA/inmunología , Adyuvantes Inmunológicos , Animales , Antígenos CD/inmunología , Antígeno B7-1/inmunología , Antígeno B7-2 , Antígenos CD58/genética , Antígenos CD58/inmunología , Quimiocina CCL3 , Quimiocina CCL4 , Citotoxicidad Inmunológica , Femenino , Proteínas de Fusión gag-pol/inmunología , VIH-1/inmunología , Humanos , Molécula 1 de Adhesión Intercelular/genética , Interferón gamma/biosíntesis , Glicoproteínas de Membrana/inmunología , Ratones , Ratones Endogámicos BALB C , Proteínas Recombinantes/inmunología , Linfocitos T Colaboradores-Inductores/inmunología , Vacunas de ADN/inmunología , Molécula 1 de Adhesión Celular Vascular/genética , Molécula 1 de Adhesión Celular Vascular/inmunología
10.
J Clin Invest ; 102(6): 1112-24, 1998 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-9739045

RESUMEN

The potential roles of CD8(+) T-cell-induced chemokines in the expansion of immune responses were examined using DNA immunogen constructs as model antigens. We coimmunized cDNA expression cassettes encoding the alpha-chemokines IL-8 and SDF-1alpha and the beta-chemokines MIP-1alpha, RANTES, and MCP-1 along with DNA immunogens and analyzed the resulting antigen-specific immune responses. In a manner more similar to the traditional immune modulatory role of CD4(+) T cells via the expression of Th1 or Th2 cytokines, CD8(+) T cells appeared to play an important role in immune expansion and effector function by producing chemokines. For instance, IL-8 was a strong inducer of CD4(+) T cells, indicated by strong T helper proliferative responses as well as an enhancement of antibody responses. MIP-1alpha had a dramatic effect on antibody responses and modulated the shift of immune responses to a Th2-type response. RANTES coimmunization enhanced the levels of antigen-specific Th1 and cytotoxic T lymphocyte (CTL) responses. Among the chemokines examined, MCP-1 was the most potent activator of CD8(+) CTL activity. The enhanced CTL results are supported by the increased expression of Th1 cytokines IFN-gamma and TNF-alpha and the reduction of IgG1/IgG2a ratio. Our results support that CD8(+) T cells may expand both humoral and cellular responses in vivo through the elaboration of specific chemokines at the peripheral site of infection during the effector stage of the immune response.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Quimiocinas/biosíntesis , Activación de Linfocitos , Vacunas contra el SIDA/inmunología , Animales , Quimiocina CCL3 , Quimiocina CCL4 , Quimiocina CCL5/biosíntesis , Quimiocina CXCL12 , Quimiocinas CXC/biosíntesis , Femenino , Proteínas de Fusión gag-pol/genética , Proteínas de Fusión gag-pol/inmunología , VIH-1/inmunología , Interleucina-8/biosíntesis , Proteínas Inflamatorias de Macrófagos/biosíntesis , Ratones , Ratones Endogámicos BALB C , Modelos Inmunológicos , Linfocitos T Citotóxicos , Células TH1/inmunología , Vacunas de ADN/inmunología
11.
Nat Biotechnol ; 15(6): 547-51, 1997 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-9181577

RESUMEN

Cancer-related, mucin-type carbohydrate epitopes, principally mannose and sialo-syl residues, are expressed on the envelope protein gp 160 of the human immunodeficiency virus (HIV). Anticarbohydrate antibodies directed toward these and other carbohydrate epitopes are known to neutralize HIV-1 infection by cell-free virus. Carbohydrates, however, being T cell-independent antigens, typically elicit diminished immune responses. To overcome this potential draw back, we have examined the ability of peptides that mimic such epitopes to elicit immune responses that cross-react with carbohydrate structures. We report that mouse polyclonal antisera generated against peptides that mimic mucin-related carbohydrate epitopes have anti-HIV-1 activity. Generation of antibodies was not lr-gene restricted, as at least two different strains of mice. Balb/c (H-2d) and C57Bl/6 (H-2b), responded equally to the peptides. The antipeptide sera displayed neutralizing activity against HIV-I/MN and HIV-I/3B viral strains. This neutralization was as good as human anti-HIV sera. These results indicate that peptide mimics of carbohydrates provide a novel strategy for the further development of reagents that elicit immune responses to carbohydrate epitopes associated with many infectious organisms and tumor cells.


Asunto(s)
Epítopos/inmunología , Proteínas gp160 de Envoltorio del VIH/inmunología , Infecciones por VIH/terapia , VIH-1/inmunología , Oligopéptidos/inmunología , Síndrome de Inmunodeficiencia Adquirida/terapia , Secuencia de Aminoácidos , Animales , Formación de Anticuerpos , Carbohidratos/inmunología , Línea Celular , Antígenos H-2 , Anticuerpos Anti-VIH/biosíntesis , Anticuerpos Anti-VIH/sangre , Proteínas gp160 de Envoltorio del VIH/química , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Pruebas de Neutralización , Oligopéptidos/química , Relación Estructura-Actividad
12.
Nat Biotechnol ; 18(9): 974-9, 2000 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-10973219

RESUMEN

Immunity to tumors as well as to viral and bacterial pathogens is often mediated by cytotoxic T lymphocytes (CTLs). Thus, the ability to induce a strong cell-mediated immune response is an important requirement of novel immunotherapies. Antigen-presenting cells (APCs), including dendritic cells (DCs), are specialized in initiating T-cell immunity. Harnessing this innate ability of these cells to acquire and present antigens, we sought to improve antigen presentation by targeting antigens directly to DCs in vivo through apoptosis. We engineered Fas-mediated apoptotic death of antigen-bearing cells in vivo by co-expressing the immunogen and Fas in the same cell. We then observed that the death of antigen-bearing cells results in increased antigen acquisition by APCs including DCs. This in vivo strategy led to enhanced antigen-specific CTLs, and the elaboration of T helper-1 (Th1) type cytokines and chemokines. This adjuvant approach has important implications for viral and nonviral delivery strategies for vaccines or gene therapies.


Asunto(s)
Células Presentadoras de Antígenos/inmunología , Células Presentadoras de Antígenos/metabolismo , Antígenos/metabolismo , Apoptosis , Células Dendríticas/metabolismo , Receptor fas/metabolismo , Animales , Anexina A5/metabolismo , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Separación Celular , ADN Complementario/metabolismo , Células Dendríticas/inmunología , Células Dendríticas/patología , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Técnicas de Transferencia de Gen , Terapia Genética/métodos , Vectores Genéticos , Inmunohistoquímica , Inmunoterapia/métodos , Interferón gamma/metabolismo , Interleucina-12/metabolismo , Ganglios Linfáticos/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Músculos/metabolismo , Plásmidos/metabolismo , Bazo/citología , Linfocitos T Citotóxicos/inmunología , Linfocitos T Citotóxicos/patología , Células TH1/inmunología , Células TH1/patología , Factores de Tiempo , Transfección
13.
Nat Biotechnol ; 15(7): 641-6, 1997 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-9219266

RESUMEN

Nucleic acid immunization is a novel vaccination technique to induce antigen-specific immune responses. We have developed expression cassettes for cell surface markers CD80 and CD86, two functionally related costimulatory molecules that play an important role in the induction of T cell-mediated immune responses. Coimmunization of these expression plasmids, along with plasmid DNA encoding for HIV-1 antigens, did not result in any significant change in the humoral response; however, we observed a dramatic increase in cytotoxic T-lymphocyte (CTL) induction as well as T-helper cell proliferation after the coadministration of CD86 genes. In contrast, coimmunization with a CD80 expression cassette resulted in a minor, but positive increase in T-helper cell or CTL responses. This strategy may be of value for the generation of rationally designed vaccines and immune therapeutics.


Asunto(s)
Vacunas de ADN/genética , Vacunas de ADN/farmacología , Animales , Formación de Anticuerpos , Antígenos CD/genética , Antígeno B7-1/genética , Antígeno B7-2 , Secuencia de Bases , Biotecnología , Cartilla de ADN/genética , Humanos , Inmunización , Técnicas In Vitro , Activación de Linfocitos , Glicoproteínas de Membrana/genética , Ratones , Ratones Endogámicos BALB C , Ingeniería de Proteínas , Linfocitos T Citotóxicos/inmunología , Linfocitos T Colaboradores-Inductores/inmunología
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.
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
16.
Cancer Res ; 50(2): 421-5, 1990 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-1967224

RESUMEN

The neu protooncogene is a recently described transforming gene originally isolated from ethylnitrosourea-induced rat neuroblastomas. We have examined the expression of the neu gene in human non-small cell lung carcinomas using immunoprecipitation and immunohistochemistry. The neu protein product (p185neu) was present in eight of 22 non-small cell carcinoma cell lines derived from human lung tumors. Expression of p185neu was found in all histological subtypes of non-small cell carcinomas including large cell carcinomas, squamous cell carcinomas, and adenocarcinomas. Extension of these data to biopsy specimens of human lung tumors demonstrated that normal ciliated bronchial epithelium of the peripheral airways expressed p185neu at low levels. Neoplastic cells in four of 12 adenocarcinomas and three of five squamous cell carcinomas also expressed p185neu at levels higher than the normal ciliated bronchial epithelium. Together these studies indicate that p185neu expression is a common feature of human lung tumors.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/análisis , Neoplasias Pulmonares/análisis , Proteínas Proto-Oncogénicas/análisis , Proto-Oncogenes , Biopsia , Amplificación de Genes , Humanos , Inmunohistoquímica , Pulmón/análisis , Proteínas Proto-Oncogénicas/inmunología , Receptor ErbB-2 , Células Tumorales Cultivadas
17.
Cancer Res ; 50(16): 5184-7, 1990 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-1974168

RESUMEN

p185neu is the protein product of the HER2/neu protooncogene. This protein has characteristics of a tyrosine kinase growth factor receptor and is postulated to be important in human carcinogenesis. To define the significance of the expression of this protein in human non-small cell lung cancer, 55 tumors from patients with squamous cell carcinoma (16), adenocarcinoma (29), or large cell carcinoma (10) of the lung were examined for p185neu using immunohistological methods. Five of 16 squamous cell carcinomas and 10 of 29 adenocarcinomas were found to overexpress p185neu relative to levels of expression seen in uninvolved bronchiolar epithelium. For the adenocarcinomas, p185neu expression was associated with older age (66.6 +/- 10.1 versus 57.5 +/- 10.8 years) (P = 0.04) and shortened survival (83.7 +/- 94.1 versus 188.5 +/- 120 weeks) (P = 0.01). In this group, using Cox's multivariate survival analysis, p185neu expression was found to be a significant determinant of survival (P = 0.04) even after accounting for the effect of tumor stage. For the squamous cell carcinomas, p185neu expression was not correlated with any of our clinicopathological parameters. Our findings indicate that non-small cell lung cancers which express p185neu do so at levels higher than that found in normal bronchiolar epithelium, and expression in adenocarcinomas of the lung is independently associated with diminished survival intervals.


Asunto(s)
Adenocarcinoma/genética , Biomarcadores de Tumor/análisis , Neoplasias Pulmonares/genética , Proteínas Proto-Oncogénicas/genética , Proto-Oncogenes , Adenocarcinoma/patología , Línea Celular , Femenino , Humanos , Sueros Inmunes , Neoplasias Pulmonares/patología , Masculino , Ploidias , Pronóstico , Proteínas Proto-Oncogénicas/análisis , Receptor ErbB-2 , Fumar , Células Tumorales Cultivadas/citología
18.
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
19.
Oncogene ; 4(10): 1175-83, 1989 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-2571965

RESUMEN

The neu oncogene encodes a 185 kd glycoprotein (p185neu) with intrinsic tyrosine kinase activity. Sequencing data has demonstrated that oncogenic p185neu differs from c-neu by a single point mutation within the transmembrane region of the glycoprotein. This mutation results in the substitution of a glutamic acid residue for a nonpolar valine residue at amino acid position 664 of the rat neu gene product. Recent studies have demonstrated that this mutation results in specific aggregation of the p185neu oncoprotein mimicking ligand induced dimerization events. The cellular consequences of the aggregated phenotype may include the enzymatic activation of p185neu. We demonstrate that the oncoprotein p185neu possesses higher intrinsic tyrosine kinase activity and that this increase in enzymatic activity is apparent within the plasma membrane, manifesting itself through the increased tyrosine phosphorylation of substrates. Furthermore, the neu oncoprotein itself is also phosphorylated on tyrosine to a higher extent than the proto-oncoprotein. These results strongly link enzymatic activation of p185neu to cellular transformation events. To test directly the effect of p185neu tyrosine kinase activity on cellular transformation we constructed mutant p185neu devoid of ATP binding ability. This mutant protein is expressed at high levels, but is unable to induce the transforming phenotype. The point mutation within the transmembrane region of p185neu mimics aspects of ligand induced activation events including increases in the specific tyrosine kinase activity of the molecule leading to cellular transformation.


Asunto(s)
Transformación Celular Neoplásica , Proteínas Tirosina Quinasas/análisis , Proteínas Proto-Oncogénicas/análisis , Adenosina Trifosfato/metabolismo , Aminoácidos/análisis , Línea Celular , Fosforilación , Pruebas de Precipitina , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Receptor ErbB-2
20.
Oncogene ; 4(1): 81-8, 1989 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-2563577

RESUMEN

The neu gene (also called NGL, erbB-2, and HER-2) encodes a 185-190 kDa transmembrane glycoprotein, p185neu, which has tyrosine-specific kinase activity and is homologous to but distinct from the epidermal growth factor receptor. The normal expression of neu mRNA and protein has been demonstrated in epithelial tissues of adult animals. Also, activation of the neu oncogene has been implicated in a variety of human adenocarcinomas. In the present study, we examined the expression of the p185neu protein in normal and transformed digestive tract tissues and in a panel of digestive tract-derived cell lines. By immunohistochemistry, strong reactivity was observed in the mucosal epithelium of the stomach, small intestine, and colon of both rodents and humans. In the small intestine, there was prominent p185neu expression by mucosal epithelium of the villus, with little or no staining in the crypts. Prominent expression was observed in the liver parenchyma, the endocrine and exocrine portions of the pancreas, and in the salivary gland. Immunoreactive p185neu was also demonstrated in fetal human intestinal epithelium. Tissue sections of selected benign and malignant colonic neoplasms were also examined. Immunoreactivity was consistently greater in adenomatous polyps than in adjacent normal colonic epithelium or areas showing malignant degeneration. By radioimmunoprecipitation, there was decreased expression in cell lines derived from more anaplastic colonic tumors. The p185neu protein is expressed widely in normal and transformed epithelial tissues of the digestive tract of the adult rat and human. This finding suggests that p185neu, a putative growth factor receptor, may play a role in the regulation of normal growth and function or in the malignant transformation of these cells.


Asunto(s)
Neoplasias del Sistema Digestivo/metabolismo , Proteínas Proto-Oncogénicas/biosíntesis , Proto-Oncogenes , Animales , Biomarcadores de Tumor/biosíntesis , Biomarcadores de Tumor/genética , Línea Celular , Neoplasias del Colon/genética , Neoplasias del Colon/metabolismo , Sistema Digestivo/embriología , Sistema Digestivo/metabolismo , Neoplasias del Sistema Digestivo/genética , Humanos , Inmunohistoquímica , Mucosa Intestinal/metabolismo , Intestino Delgado/metabolismo , Glicoproteínas de Membrana/biosíntesis , Glicoproteínas de Membrana/genética , Pruebas de Precipitina , Proteínas Proto-Oncogénicas/genética , Receptor ErbB-2 , Receptores de Superficie Celular/biosíntesis , Receptores de Superficie Celular/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA