Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
1.
Proc Natl Acad Sci U S A ; 119(26): e2116738119, 2022 06 28.
Artículo en Inglés | MEDLINE | ID: mdl-35749366

RESUMEN

Tumor infiltration by T cells profoundly affects cancer progression and responses to immunotherapy. However, the tumor immunosuppressive microenvironment can impair the induction, trafficking, and local activity of antitumor T cells. Here, we investigated whether intratumoral injection of virus-derived peptide epitopes could activate preexisting antiviral T cell responses locally and promote antitumor responses or antigen spreading. We focused on a mouse model of cytomegalovirus (CMV), a highly prevalent human infection that induces vigorous and durable T cell responses. Mice persistently infected with murine CMV (MCMV) were challenged with lung (TC-1), colon (MC-38), or melanoma (B16-F10) tumor cells. Intratumoral injection of MCMV-derived T cell epitopes triggered in situ and systemic expansion of their cognate, MCMV-specific CD4+ or CD8+ T cells. The MCMV CD8+ T cell epitopes injected alone provoked arrest of tumor growth and some durable remissions. Intratumoral injection of MCMV CD4+ T cell epitopes with polyinosinic acid:polycytidylic acid (pI:C) preferentially elicited tumor antigen-specific CD8+ T cells, promoted tumor clearance, and conferred long-term protection against tumor rechallenge. Notably, secondary proliferation of MCMV-specific CD8+ T cells correlated with better tumor control. Importantly, intratumoral injection of MCMV-derived CD8+ T cell-peptide epitopes alone or CD4+ T cell-peptide epitopes with pI:C induced potent adaptive and innate immune activation of the tumor microenvironment. Thus, CMV-derived peptide epitopes, delivered intratumorally, act as cytotoxic and immunotherapeutic agents to promote immediate tumor control and long-term antitumor immunity that could be used as a stand-alone therapy. The tumor antigen-agnostic nature of this approach makes it applicable across a broad range of solid tumors regardless of their origin.


Asunto(s)
Linfocitos T CD8-positivos , Infecciones por Citomegalovirus , Citomegalovirus , Epítopos de Linfocito T , Neoplasias , Animales , Antígenos de Neoplasias/inmunología , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/virología , Citomegalovirus/inmunología , Infecciones por Citomegalovirus/inmunología , Epítopos de Linfocito T/administración & dosificación , Epítopos de Linfocito T/inmunología , Inmunoterapia , Ratones , Neoplasias/inmunología , Neoplasias/terapia , Poli I-C/administración & dosificación , Poli I-C/inmunología , Microambiente Tumoral
2.
J Immunol ; 202(4): 1250-1264, 2019 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-30635393

RESUMEN

Recent insight into the mechanisms of induction of tissue-resident memory (TRM) CD8+ T cells (CD8+ TRM) enables the development of novel vaccine strategies against sexually transmitted infections. To maximize both systemic and genital intraepithelial CD8+ T cells against vaccine Ags, we assessed combinations of i.m. and intravaginal routes in heterologous prime-boost immunization regimens with unrelated viral vectors. Only i.m. prime followed by intravaginal boost induced concomitant strong systemic and intraepithelial genital-resident CD8+ T cell responses. Intravaginal boost with vectors expressing vaccine Ags was far superior to intravaginal instillation of CXCR3 chemokine receptor ligands or TLR 3, 7, and 9 agonists to recruit and increase the pool of cervicovaginal CD8+ TRM Transient Ag presentation increased trafficking of cognate and bystander circulating activated, but not naive, CD8+ T cells into the genital tract and induced in situ proliferation and differentiation of cognate CD8+ TRM Secondary genital CD8+ TRM were induced in the absence of CD4+ T cell help and shared a similar TCR repertoire with systemic CD8+ T cells. This prime-pull-amplify approach elicited systemic and genital CD8+ T cell responses against high-risk human papillomavirus type 16 E7 oncoprotein and conferred CD8-mediated protection to a vaccinia virus genital challenge. These results underscore the importance of the delivery route of nonreplicating vectors in prime-boost immunization to shape the tissue distribution of CD8+ T cell responses. In this context, the importance of local Ag presentation to elicit genital CD8+ TRM provides a rationale to develop novel vaccines against sexually transmitted infections and to treat human papillomavirus neoplasia.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Papillomavirus Humano 16/inmunología , Vacunas contra Papillomavirus/inmunología , Animales , Células HEK293 , Humanos , Ratones , Ratones Congénicos , Ratones Endogámicos C57BL , Vacunas contra Papillomavirus/genética , Vacunación
3.
Proc Natl Acad Sci U S A ; 115(4): E733-E742, 2018 01 23.
Artículo en Inglés | MEDLINE | ID: mdl-29311317

RESUMEN

Protein-based drugs are very active in treating cancer, but their efficacy can be limited by the formation of neutralizing antidrug antibodies (ADAs). Recombinant immunotoxins are proteins that are very effective in patients with leukemia, where immunity is suppressed, but induce ADAs, which compromise their activity, in patients with intact immunity. Here we induced a specific, durable, and transferable immune tolerance to recombinant immunotoxins by combining them with nanoparticles containing rapamycin (SVP-R). SVP-R mitigated the formation of inhibitory ADAs in naïve and sensitized mice, resulting in restoration of antitumor activity. The immune tolerance is mediated by colocalization of the SVP-R and immunotoxin to dendritic cells and macrophages in the spleen and is abrogated by depletion of regulatory T cells. Tolerance induced by SVPs was not blocked by checkpoint inhibitors or costimulatory agonist monoclonal antibodies that by themselves enhance ADA formation.


Asunto(s)
Inmunomodulación , Inmunosupresores/administración & dosificación , Inmunotoxinas/administración & dosificación , Leucemia/terapia , Sirolimus/administración & dosificación , Animales , Anticuerpos Neutralizantes , Proteínas Ligadas a GPI/inmunología , Humanos , Inmunotoxinas/inmunología , Mesotelina , Nanopartículas , Factores de Tiempo
4.
J Immunol ; 200(6): 2038-2045, 2018 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-29431691

RESUMEN

Recombinant immunotoxins (RITs) are chimeric proteins being developed for cancer treatment. They are composed of an Ab fragment that targets a cancer Ag and a cytotoxic portion of Pseudomonas exotoxin A. They are effective for patients with hematologic malignancies with defective immunity, but their efficacy against solid tumors is limited by anti-drug Ab (ADA) responses in immune-competent patients. Pre-existing Abs or immune memory owing to previous toxin exposure represent additional hurdles because they induce rapid and strong ADA responses. Here, we evaluated the efficacy of methotrexate (MTX) to prevent ADA formation against the mesothelin-targeting RIT LMB-100 in naive mice and in mice with pre-existing Abs. We found that low-dose MTX combined with LMB-100 completely suppressed the formation of ADAs in a dose- and frequency-dependent manner. Suppression of the immune response restored blood levels of LMB-100 and prevented its neutralization. Furthermore, combination of MTX with LMB-100 did not compromise the immune response against a second Ag given after stopping MTX, indicating specific immune tolerance. Adoptive transfer of splenocytes suppressed Ab responses to LMB-100 in recipient mice, indicating a durable immune tolerance. We conclude that combination of MTX and LMB-100 is effective at preventing immune responses in a durable, Ag-specific manner. We propose combining low-dose MTX in immune-competent cancer patients receiving RIT therapy to prevent immunogenicity. This approach could be applied to other immunogenic therapeutic agents and to proteins for which there is pre-existing immunity.


Asunto(s)
Tolerancia Inmunológica/efectos de los fármacos , Inmunidad Humoral/efectos de los fármacos , Inmunotoxinas/inmunología , Metotrexato/farmacología , Proteínas Recombinantes/inmunología , ADP Ribosa Transferasas/inmunología , Traslado Adoptivo/métodos , Animales , Anticuerpos Monoclonales/inmunología , Formación de Anticuerpos/efectos de los fármacos , Toxinas Bacterianas/inmunología , Células Cultivadas , Exotoxinas/inmunología , Femenino , Proteínas Ligadas a GPI/farmacología , Tolerancia Inmunológica/inmunología , Inmunidad Humoral/inmunología , Inmunoterapia/métodos , Mesotelina , Ratones , Ratones Endogámicos BALB C , Neoplasias/inmunología , Factores de Virulencia/inmunología , Exotoxina A de Pseudomonas aeruginosa
5.
Int J Cancer ; 142(7): 1467-1479, 2018 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-29159802

RESUMEN

Recent advances in immunotherapy against cancer underscore the importance of T lymphocytes and tumor microenvironment, but few vaccines targeting cancer have been approved likely due in part to the dearth of common tumor antigens, insufficient immunogenicity and the evolution of immune evasion mechanisms during the progression to malignancy. Human papillomaviruses (HPVs) are the primary etiologic agents of cervical cancer and progression from persistent HPV-infection to cervical intraepithelial lesions and eventually cancer requires persistent expression of the oncoproteins E6 and E7. This offers the opportunity to specifically target these virus-specific antigens for vaccine-induced clearance of infected cells before cancers develop. Here we have evaluated the immunogenicity of Adenovirus Types 26 and 35 derived vectors expressing a fusion of HPV16 E6 and E7 oncoproteins after intramuscular (IM) and/or intravaginal (Ivag) immunization in mice. The adenovirus vectors were shown to transduce an intact cervicovaginal epithelium. IM prime followed by Ivag boost maximized the induction and trafficking of HPV-specific CD8+ T cells producing IFN-γ and TNF-α to the cervicovaginal tract. Importantly, the cervicovaginal CD8+ T cells expressed CD69 and CD103; hallmarks of intraepithelial tissue-resident memory CD8+ T cells. This prime-boost strategy targeting heterologous locations also induced circulating HPV-specific CD8+ T cell responses. Our study prompts further evaluation of Ivag immunization with adenoviral vectors expressing modified E6 and E7 antigens for therapeutic vaccination against persistent HPV infection and cervical intraepithelial neoplasia.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Vacunas contra el Cáncer/inmunología , Inmunoterapia/métodos , Infecciones por Papillomavirus/inmunología , Neoplasias del Cuello Uterino/virología , Adenoviridae , Animales , Femenino , Vectores Genéticos , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Proteínas Oncogénicas Virales/inmunología , Proteínas E7 de Papillomavirus/inmunología , Infecciones por Papillomavirus/complicaciones , Vacunas contra Papillomavirus/inmunología , Proteínas Represoras/inmunología , Transducción Genética , Vacunación
6.
Eur J Immunol ; 45(8): 2389-95, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25973715

RESUMEN

Sos-1 and Sos-2 are ubiquitously expressed Ras-guanine exchange factors involved in Erk-MAP kinase pathway activation. Using mice lacking genes encoding Sos-1 and Sos-2, we evaluated the role of these proteins in peripheral T-cell signaling and function. Our results confirmed that TCR-mediated Erk activation in peripheral CD4(+) T cells does not depend on Sos-1 and Sos-2, although IL-2-mediated Erk activation does. Unexpectedly, however, we show an increase in AKT phosphorylation in Sos-1/2dKO CD4(+) T cells upon TCR and IL-2 stimulation. Activation of AKT was likely a consequence of increased recruitment of PI3K to Grb2 upon TCR and/or IL-2 stimulation in Sos-1/2dKO CD4(+) T cells. The increased activity of the PI3K/AKT pathway led to downregulation of the surface receptor CD62L in Sos-1/2dKO T cells and a subsequent impairment in T-cell migration.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Movimiento Celular/inmunología , Fosfatidilinositol 3-Quinasas/inmunología , Proteína SOS1/inmunología , Transducción de Señal/inmunología , Proteínas Son Of Sevenless/inmunología , Animales , Movimiento Celular/genética , Activación Enzimática/genética , Activación Enzimática/inmunología , Proteína Adaptadora GRB2/genética , Proteína Adaptadora GRB2/inmunología , Interleucina-2/genética , Interleucina-2/inmunología , Selectina L/genética , Selectina L/inmunología , Ratones , Ratones Noqueados , Fosfatidilinositol 3-Quinasas/genética , Fosforilación/genética , Fosforilación/inmunología , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/inmunología , Receptores de Antígenos de Linfocitos T/genética , Receptores de Antígenos de Linfocitos T/inmunología , Proteína SOS1/genética , Transducción de Señal/genética , Proteínas Son Of Sevenless/genética
7.
J Virol ; 89(1): 83-96, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25320297

RESUMEN

UNLABELLED: No herpes simplex virus 2 (HSV-2) vaccine has been licensed for use in humans. HSV-2 glycoproteins B (gB) and D (gD) are targets of neutralizing antibodies and T cells, but clinical trials involving intramuscular (i.m.) injection of HSV-2 gB and gD in adjuvants have not been effective. Here we evaluated intravaginal (ivag) genetic immunization of C57BL/6 mice with a replication-defective human papillomavirus pseudovirus (HPV PsV) expressing HSV-2 gB (HPV-gB) or gD (HPV-gD) constructs to target different subcellular compartments. HPV PsV expressing a secreted ectodomain of gB (gBsec) or gD (gDsec), but not PsV expressing a cytoplasmic or membrane-bound form, induced circulating and intravaginal-tissue-resident memory CD8(+) T cells that were able to secrete gamma interferon (IFN-γ) and tumor necrosis factor alpha (TNF-α) as well as moderate levels of serum HSV neutralizing antibodies. Combined immunization with HPV-gBsec and HPV-gDsec (HPV-gBsec/gDsec) vaccines conferred longer survival after vaginal challenge with HSV-2 than immunization with HPV-gBsec or HPV-gDsec alone. HPV-gBsec/gDsec ivag vaccination was associated with a reduced severity of genital lesions and lower levels of viral shedding in the genital tract after HSV-2 challenge. In contrast, intramuscular vaccination with a soluble truncated gD protein (gD2t) in alum and monophosphoryl lipid A (MPL) elicited high neutralizing antibody titers and improved survival but did not reduce genital lesions and viral shedding. Vaccination combining ivag HPV-gBsec/gDsec and i.m. gD2t-alum-MPL improved survival and reduced genital lesions and viral shedding. Finally, high levels of circulating HSV-2-specific CD8(+) T cells, but not serum antibodies, correlated with reduced viral shedding. Taken together, our data underscore the potential of HPV PsV as a platform for a topical mucosal vaccine to control local manifestations of primary HSV-2 infection. IMPORTANCE: Genital herpes is a highly prevalent chronic disease caused by HSV infection. To date, there is no licensed vaccine against HSV infection. This study describes intravaginal vaccination with a nonreplicating HPV-based vector expressing HSV glycoprotein antigens. The data presented in this study underscore the potential of HPV-based vectors as a platform for the induction of genital-tissue-resident memory T cell responses and the control of local manifestations of primary HSV infection.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Herpes Genital/prevención & control , Vacunas contra Herpesvirus/inmunología , Papillomaviridae/genética , Proteínas del Envoltorio Viral/inmunología , Esparcimiento de Virus , Administración Intravaginal , Animales , Anticuerpos Neutralizantes/sangre , Anticuerpos Antivirales/sangre , Modelos Animales de Enfermedad , Femenino , Vectores Genéticos , Herpes Genital/inmunología , Herpes Genital/patología , Herpesvirus Humano 2/genética , Herpesvirus Humano 2/inmunología , Vacunas contra Herpesvirus/administración & dosificación , Vacunas contra Herpesvirus/genética , Memoria Inmunológica , Inyecciones Intramusculares , Interferón gamma/metabolismo , Ratones Endogámicos C57BL , Proteínas Recombinantes/genética , Proteínas Recombinantes/inmunología , Análisis de Supervivencia , Factor de Necrosis Tumoral alfa/metabolismo , Vacunación/métodos , Vacunas Sintéticas/administración & dosificación , Vacunas Sintéticas/genética , Vacunas Sintéticas/inmunología , Proteínas del Envoltorio Viral/genética
8.
J Immunol ; 193(12): 6172-83, 2014 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-25398324

RESUMEN

The human papillomavirus pseudovirions (HPV-PsVs) approach is an effective gene-delivery system that can prime or boost an immune response in the vaginal tract of nonhuman primates and mice. Intravaginal vaccination with HPV-PsVs expressing SIV genes, combined with an i.m. gp120 protein injection, induced humoral and cellular SIV-specific responses in macaques. Priming systemic immune responses with i.m. immunization with ALVAC-SIV vaccines, followed by intravaginal HPV-PsV-SIV/gp120 boosting, expanded and/or recruited T cells in the female genital tract. Using a stringent repeated low-dose intravaginal challenge with the highly pathogenic SIVmac251, we show that although these regimens did not demonstrate significant protection from virus acquisition, they provided control of viremia in a number of animals. High-avidity Ab responses to the envelope gp120 V1/V2 region correlated with delayed SIVmac251 acquisition, whereas virus levels in mucosal tissues were inversely correlated with antienvelope CD4(+) T cell responses. CD8(+) T cell depletion in animals with controlled viremia caused an increase in tissue virus load in some animals, suggesting a role for CD8(+) T cells in virus control. This study highlights the importance of CD8(+) cells and antienvelope CD4(+) T cells in curtailing virus replication and antienvelope V1/V2 Abs in preventing SIVmac251 acquisition.


Asunto(s)
Anticuerpos Antivirales/inmunología , Fragmentos de Péptidos/inmunología , Virus de la Inmunodeficiencia de los Simios/inmunología , Subgrupos de Linfocitos T/inmunología , Vagina/inmunología , Proteínas del Envoltorio Viral/inmunología , Viremia/inmunología , Alphapapillomavirus/genética , Animales , Especificidad de Anticuerpos/inmunología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Modelos Animales de Enfermedad , Femenino , Vectores Genéticos/genética , Depleción Linfocítica , Macaca mulatta , Datos de Secuencia Molecular , Membrana Mucosa/inmunología , Membrana Mucosa/virología , Vacunas contra el SIDAS/genética , Vacunas contra el SIDAS/inmunología , Síndrome de Inmunodeficiencia Adquirida del Simio/inmunología , Síndrome de Inmunodeficiencia Adquirida del Simio/prevención & control , Síndrome de Inmunodeficiencia Adquirida del Simio/virología , Virus de la Inmunodeficiencia de los Simios/genética , Subgrupos de Linfocitos T/virología , Vacunación , Vacunas de ADN/genética , Vacunas de ADN/inmunología , Vagina/virología , Proteínas del Envoltorio Viral/química , Proteínas del Envoltorio Viral/genética , Viremia/prevención & control , Viremia/virología
9.
J Virol ; 87(18): 10105-13, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23843634

RESUMEN

BK polyomavirus (BKV) causes significant urinary tract pathogenesis in immunosuppressed individuals, including kidney and bone marrow transplant recipients. It is currently unclear whether BKV-neutralizing antibodies can moderate or prevent BKV disease. We developed reporter pseudoviruses based on seven divergent BKV isolates and performed neutralization assays on sera from healthy human subjects. The results demonstrate that BKV genotypes I, II, III, and IV are fully distinct serotypes. While nearly all healthy subjects had BKV genotype I-neutralizing antibodies, a majority of subjects did not detectably neutralize genotype III or IV. Surprisingly, BKV subgenotypes Ib1 and Ib2 can behave as fully distinct serotypes. This difference is governed by as few as two residues adjacent to the cellular glycan receptor-binding site on the virion surface. Serological analysis of mice given virus-like particle (VLP)-based BKV vaccines confirmed these findings. Mice administered a multivalent VLP vaccine showed high-titer serum antibody responses that potently cross-neutralized all tested BKV genotypes. Interestingly, each of the neutralization serotypes bound a distinct spectrum of cell surface receptors, suggesting a possible connection between escape from recognition by neutralizing antibodies and cellular attachment mechanisms. The finding implies that different BKV genotypes have different cellular tropisms and pathogenic potentials in vivo. Individuals who are infected with one BKV serotype may remain humorally vulnerable to other BKV serotypes after implementation of T cell immunosuppression. Thus, prevaccinating organ transplant recipients with a multivalent BKV VLP vaccine might reduce the risk of developing posttransplant BKV disease.


Asunto(s)
Virus BK/genética , Virus BK/fisiología , Tropismo Viral , Internalización del Virus , Animales , Anticuerpos Neutralizantes/sangre , Anticuerpos Antivirales/sangre , Virus BK/clasificación , ADN Viral/química , ADN Viral/genética , Femenino , Genotipo , Humanos , Ratones , Ratones Endogámicos BALB C , Datos de Secuencia Molecular , Análisis de Secuencia de ADN , Serotipificación , Vacunas Virales/administración & dosificación , Vacunas Virales/inmunología
10.
PLoS Pathog ; 8(4): e1002650, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22511874

RESUMEN

BK polyomavirus (BKV or BKPyV) associated nephropathy affects up to 10% of kidney transplant recipients (KTRs). BKV isolates are categorized into four genotypes. It is currently unclear whether the four genotypes are also serotypes. To address this issue, we developed high-throughput serological assays based on antibody-mediated neutralization of BKV genotype I and IV reporter vectors (pseudoviruses). Neutralization-based testing of sera from mice immunized with BKV-I or BKV-IV virus-like particles (VLPs) or sera from naturally infected human subjects revealed that BKV-I specific serum antibodies are poorly neutralizing against BKV-IV and vice versa. The fact that BKV-I and BKV-IV are distinct serotypes was less evident in traditional VLP-based ELISAs. BKV-I and BKV-IV neutralization assays were used to examine BKV type-specific neutralizing antibody responses in KTRs at various time points after transplantation. At study entry, sera from 5% and 49% of KTRs showed no detectable neutralizing activity for BKV-I or BKV-IV neutralization, respectively. By one year after transplantation, all KTRs were neutralization seropositive for BKV-I, and 43% of the initially BKV-IV seronegative subjects showed evidence of acute seroconversion for BKV-IV neutralization. The results suggest a model in which BKV-IV-specific seroconversion reflects a de novo BKV-IV infection in KTRs who initially lack protective antibody responses capable of neutralizing genotype IV BKVs. If this model is correct, it suggests that pre-vaccinating prospective KTRs with a multivalent VLP-based vaccine against all BKV serotypes, or administration of BKV-neutralizing antibodies, might offer protection against graft loss or dysfunction due to BKV associated nephropathy.


Asunto(s)
Anticuerpos Neutralizantes/inmunología , Virus BK/inmunología , Enfermedades Renales/prevención & control , Trasplante de Riñón , Infecciones por Polyomavirus/prevención & control , Animales , Anticuerpos Neutralizantes/sangre , Anticuerpos Neutralizantes/farmacología , Anticuerpos Antivirales , Virus BK/metabolismo , Modelos Animales de Enfermedad , Femenino , Humanos , Enfermedades Renales/inmunología , Masculino , Ratones , Ratones Endogámicos BALB C , Infecciones por Polyomavirus/sangre , Infecciones por Polyomavirus/inmunología , Factores de Tiempo , Vacunación/métodos , Vacunas Virales/inmunología , Vacunas Virales/farmacología
11.
J Immunol ; 188(2): 714-23, 2012 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-22174446

RESUMEN

The majority of HIV infections occur via mucosal transmission. Vaccines that induce memory T and B cells in the female genital tract may prevent the establishment and systemic dissemination of HIV. We tested the immunogenicity of a vaccine that uses human papillomavirus (HPV)-based gene transfer vectors, also called pseudovirions (PsVs), to deliver SIV genes to the vaginal epithelium. Our findings demonstrate that this vaccine platform induces gene expression in the genital tract in both cynomolgus and rhesus macaques. Intravaginal vaccination with HPV16, HPV45, and HPV58 PsVs delivering SIV Gag DNA induced Gag-specific Abs in serum and the vaginal tract, and T cell responses in blood, vaginal mucosa, and draining lymph nodes that rapidly expanded following intravaginal exposure to SIV(mac251.) HPV PsV-based vehicles are immunogenic, which warrant further testing as vaccine candidates for HIV and may provide a useful model to evaluate the benefits and risks of inducing high levels of SIV-specific immune responses at mucosal sites prior to SIV infection.


Asunto(s)
ADN Viral/administración & dosificación , Productos del Gen gag/genética , Técnicas de Transferencia de Gen , Vacunas contra Papillomavirus/administración & dosificación , Vacunas contra Papillomavirus/genética , Virus de la Inmunodeficiencia de los Simios/genética , Vagina/inmunología , Virión/genética , Alphapapillomavirus/genética , Alphapapillomavirus/inmunología , Animales , ADN Viral/inmunología , Femenino , Productos del Gen gag/administración & dosificación , Productos del Gen gag/inmunología , Células HEK293 , Humanos , Inmunidad Mucosa/genética , Proteínas Luminiscentes/administración & dosificación , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/inmunología , Macaca fascicularis , Macaca mulatta , Infecciones por Papillomavirus/genética , Infecciones por Papillomavirus/inmunología , Infecciones por Papillomavirus/virología , Vacunas contra Papillomavirus/inmunología , Síndrome de Inmunodeficiencia Adquirida del Simio/genética , Síndrome de Inmunodeficiencia Adquirida del Simio/inmunología , Síndrome de Inmunodeficiencia Adquirida del Simio/virología , Virus de la Inmunodeficiencia de los Simios/inmunología , Vagina/metabolismo , Vagina/virología , Virión/inmunología , Proteína Fluorescente Roja
12.
PLoS One ; 19(1): e0297262, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38277395

RESUMEN

The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) evades the innate immune machinery through multiple viral proteins, including nonstructural protein 1 (NSP1). While NSP1 is known to suppress translation of host mRNAs, the mechanisms underlying its immune evasion properties remain elusive. By integrating RNA-seq, ribosome footprinting, and ChIP-seq in A549 cells we found that NSP1 predominantly represses transcription of immune-related genes by favoring Histone 3 Lysine 9 dimethylation (H3K9me2). G9a/GLP H3K9 methyltransferase inhibitor UNC0638 restored expression of antiviral genes and restricted SARS-CoV-2 replication. Our multi-omics study unravels an epigenetic mechanism underlying host immune evasion by SARS-CoV-2 NSP1. Elucidating the factors involved in this phenomenon, may have implications for understanding and treating viral infections and other immunomodulatory diseases.


Asunto(s)
COVID-19 , SARS-CoV-2 , Humanos , COVID-19/genética , COVID-19/inmunología , COVID-19/virología , Represión Epigenética , SARS-CoV-2/genética , SARS-CoV-2/inmunología , SARS-CoV-2/metabolismo , Proteínas no Estructurales Virales/metabolismo
13.
J Immunol ; 187(6): 3044-52, 2011 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-21832166

RESUMEN

Previous studies demonstrated cross talk between mucosal and reproductive organs during secretory IgA Ab induction. In this study, we aimed to clarify the underlying mechanisms of this cross talk. We found significantly higher titers of Ag-specific secretory IgA Ab in the vaginal wash after mucosal vaccination by both the intranasal (i.n.) and the intravaginal routes but not by the s.c. route. Interestingly, Ag-specific IgA Ab-secreting cells (ASCs) were found mainly in the uterus but not in the cervix and vaginal canal after i.n. vaccination. The fact that most Ag-specific IgA ASCs isolated from the uteri of vaccinated mice migrated toward mucosa-associated epithelial chemokine (MEC)/CCL28 suggests dominant expression of CCR10 on the IgA ASCs. Further, IgA ASCs in the uteri of vaccinated mice were reduced drastically in mice treated with neutralizing anti-MEC/CCL28 Ab. Most intriguingly, the female sex hormone estrogen directly regulated MEC/CCL28 expression and was augmented by i.n. vaccination with cholera toxin or stimulators for innate immunity. Further, blockage of estrogen function in the uterus by oral administration of the estrogen antagonist raloxifene significantly inhibited migration of Ag-specific IgA ASCs after i.n. vaccination with OVA plus cholera toxin. Taken together, these data strongly suggest that CCR10(+) IgA ASCs induced by mucosal vaccination via the i.n. route migrate into the uterus in a MEC/CCL28-dependent manner and that estrogen might have a crucial role in the protection against genital infection by regulating MEC/CCL28 expression in the uterus.


Asunto(s)
Quimiocinas CC/biosíntesis , Estrógenos/inmunología , Inmunidad Mucosa/inmunología , Células Plasmáticas/inmunología , Receptores CCR10/metabolismo , Útero/inmunología , Administración Intranasal , Animales , Células 3T3 BALB , Western Blotting , Quimiocinas CC/inmunología , Quimiotaxis de Leucocito/inmunología , Toxina del Cólera/administración & dosificación , Toxina del Cólera/inmunología , Ensayo de Inmunoadsorción Enzimática , Estrógenos/metabolismo , Femenino , Inmunoglobulina A Secretora/inmunología , Inmunoglobulina A Secretora/metabolismo , Ratones , Ratones Endogámicos C57BL , Ovalbúmina/administración & dosificación , Ovalbúmina/inmunología , Células Plasmáticas/metabolismo , Receptores CCR10/inmunología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Útero/metabolismo , Vacunación
15.
NPJ Vaccines ; 7(1): 40, 2022 Mar 29.
Artículo en Inglés | MEDLINE | ID: mdl-35351898

RESUMEN

The HPV vaccine has shown sustained efficacy and consistent stabilization of antibody levels, even after a single dose. We defined the HPV16-VLP antibody avidity patterns over 11 years among women who received one- or three doses of the bivalent HPV vaccine in the Costa Rica HPV Vaccine Trial. Absolute HPV16 avidity was lower in women who received one compared to three doses, although the patterns were similar (increased in years 2 and 3 and remained stable over the remaining 8 years). HPV16 avidity among women who were HPV16-seropositive women at HPV vaccination, a marker of natural immune response to HPV16 infection, was significantly lower than those of HPV16-seronegative women, a difference that was more pronounced among one-dose recipients. No differences in HPV16 avidity were observed by HPV18 serostatus at vaccination, confirming the specificity of the findings. Importantly, point estimates for vaccine efficacy against incident, six-month persistent HPV16 infections was similar between women who were HPV16 seronegative and seropositive at the time of initial HPV vaccination for both one-dose and three-dose participants. It is therefore likely that this lower avidity level is still sufficient to enable antibody-mediated protection. It is encouraging for long-term HPV-vaccine protection that HPV16 antibody avidity was maintained for over a decade, even after a single dose.

16.
J Immunol ; 182(11): 6851-60, 2009 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-19454681

RESUMEN

Our previous studies demonstrated the potential of the sublingual (s.l.) route for delivering vaccines capable of inducing mucosal as well as systemic immune responses. Those findings prompted us to attempt to identify possible inductive mechanism of s.l. vaccination for immune responses. Within 2 h after s.l. administration with cholera toxin (CT), significantly higher numbers of MHC class II(+) cells accumulated in the s.l. mucosa. Of note, there were brisk expression levels of both CCL19 and CCL21 in cervical lymph nodes (CLN) 24 h after s.l. vaccination with CT. In reconstitution experiments using OVA-specific CD4(+) or CD8(+) T cells, s.l. vaccination elicited strong Ag-specific T cell proliferation mainly in CLN. Interestingly, Ag-specific T cell proliferation completely disappeared in CD11c-depleted and CCR7(-/-) mice but not in Langerin-depleted, macrophage-depleted, and CCR6(-/-) mice. Similar to CD4(+) T cell responses, induction of Ag-specific IgG (systemic) and IgA (mucosal) Ab responses were significantly reduced in CD11c-depleted and CCR7(-/-) mice after s.l. vaccination with OVA plus CT. Although CD8alpha(-) dendritic cells ferried Ag from the s.l. mucosa, both migratory CD8alpha(-) and resident CD8alpha(+) dendritic cells were essential to prime CD4(+) T cells in the CLN. On the basis of these findings, we believe that CCR7 expressed CD8alpha(-)CD11c(+) cells ferry Ag in the s.l. mucosa, migrate into the CLN, and share the Ag with resident CD8alpha(+)CD11c(+) cells for the initiation of Ag-specific T and B cell responses following s.l. challenge. We propose that the s.l. mucosa is one of the effective mucosal inductive sites regulated by the CCR7-CCL19/CCL21 pathway.


Asunto(s)
Quimiocina CCL19/metabolismo , Quimiocina CCL21/metabolismo , Células Dendríticas/inmunología , Receptores CCR7/metabolismo , Vacunas/administración & dosificación , Administración Sublingual , Animales , Linfocitos B , Toxina del Cólera/administración & dosificación , Toxina del Cólera/farmacología , Ganglios Linfáticos , Ratones , Ratones Noqueados , Mucosa Bucal/inmunología , Ovalbúmina/administración & dosificación , Ovalbúmina/inmunología , Farmacocinética , Linfocitos T , Vacunas/inmunología , Vacunas/farmacocinética
17.
J Immunol ; 183(12): 7851-9, 2009 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-19933861

RESUMEN

We have recently reported that the sublingual (s.l.) mucosa is an efficient site for inducing systemic and mucosal immune responses. In this study, the potential of s.l. immunization to induce remote Ab responses and CD8(+) cytotoxic responses in the female genital tract was examined in mice by using a nonreplicating Ag, OVA, and cholera toxin (CT) as an adjuvant. Sublingual administration of OVA and CT induced Ag-specific IgA and IgG Abs in blood and in cervicovaginal secretions. These responses were associated with large numbers of IgA Ab-secreting cells (ASCs) in the genital mucosa. Genital ASC responses were similar in magnitude and isotype distribution after s.l., intranasal, or vaginal immunization and were superior to those seen after intragastric immunization. Genital, but not blood or spleen, IgA ASC responses were inhibited by treatment with anti-CCL28 Abs, suggesting that the chemokine CCL28 plays a major role in the migration of IgA ASC progenitors to the reproductive tract mucosa. Furthermore, s.l. immunization with OVA induced OVA-specific effector CD8(+) cytolytic T cells in the genital mucosa, and these responses required coadministration of the CT adjuvant. Furthermore, s.l. administration of human papillomavirus virus-like particles with or without the CT adjuvant conferred protection against genital challenge with human papillomavirus pseudovirions. Taken together, these findings underscore the potential of s.l. immunization as an efficient vaccination strategy for inducing genital immune responses and should impact on the development of vaccines against sexually transmitted diseases.


Asunto(s)
Anticuerpos Antibacterianos/biosíntesis , Células Productoras de Anticuerpos/inmunología , Toxina del Cólera/inmunología , Papillomavirus Humano 16/inmunología , Ovalbúmina/inmunología , Infecciones por Papillomavirus/prevención & control , Linfocitos T Citotóxicos/inmunología , Neoplasias del Cuello Uterino/prevención & control , Adyuvantes Inmunológicos/administración & dosificación , Administración Sublingual , Animales , Anticuerpos Antivirales/biosíntesis , Células Productoras de Anticuerpos/citología , Células Productoras de Anticuerpos/virología , Diferenciación Celular/inmunología , Células Cultivadas , Toxina del Cólera/administración & dosificación , Modelos Animales de Enfermedad , Femenino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Membrana Mucosa/inmunología , Membrana Mucosa/virología , Ovalbúmina/administración & dosificación , Infecciones por Papillomavirus/inmunología , Infecciones por Papillomavirus/virología , Linfocitos T Citotóxicos/virología , Neoplasias del Cuello Uterino/inmunología , Neoplasias del Cuello Uterino/virología , Virión/inmunología
18.
Hum Vaccin ; 7(1): 110-4, 2011 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-21263223

RESUMEN

The sublingual route has been used for many years to deliver drugs and small molecules to the bloodstream. Surprisingly, the potential of this route for delivering vaccines has received very little if any attention until recently. During the past few years, a number of laboratories have documented the efficacy of sublingual immunization for inducing a broad range of immune responses in different experimental animal systems using a variety of antigens, including soluble proteins, inert particulate antigens (killed viruses, virus-like particles, bacterial extracts) as well as live-attenuated viruses. In most cases, systemic and mucosal immune responses, including humoral and cytotoxic T-cell responses were induced in both mucosal and extra-mucosal tissues. Overall, sublingual immunization was comparable to nasal immunization regarding the magnitude, breadth, and anatomic dissemination of the induced immune responses. Importantly, and contrary to nasal administration, sublingual administration did not redirect antigens and/or adjuvants to the brain. Here we review the results of pre-clinical studies using animal models of respiratory, intestinal and genital infections. These promising results provide a foundation for testing the approach in humans.


Asunto(s)
Vacunación/métodos , Vacunas/administración & dosificación , Vacunas/inmunología , Administración Sublingual , Animales , Modelos Animales de Enfermedad , Femenino , Enfermedades Gastrointestinales/prevención & control , Enfermedades de los Genitales Femeninos/prevención & control , Infecciones del Sistema Respiratorio/prevención & control
19.
Proc Natl Acad Sci U S A ; 105(5): 1644-9, 2008 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-18227512

RESUMEN

We assessed whether the sublingual (s.l.) route would be an effective means of delivering vaccines against influenza virus in mice by using either formalin-inactivated or live influenza A/PR/8 virus (H1N1). Sublingual administration of inactivated influenza virus given on two occasions induced both systemic and mucosal antibody responses and conferred protection against a lethal intranasal (i.n.) challenge with influenza virus. Coadministration of a mucosal adjuvant (mCTA-LTB) enhanced these responses and resulted in complete protection against respiratory viral challenge. In addition, s.l. administration of formalin-inactivated A/PR/8 plus mCTA-LTB induced systemic expansion of IFN-gamma-secreting T cells and virus-specific cytotoxic T lymphocyte responses. Importantly, a single s.l. administration of live A/PR/8 virus was not pathogenic and induced protection mediated by both acquired and innate immunity. Moreover, s.l. administration of live A/PR/8 virus conferred heterosubtypic protection against respiratory challenge with H3N2 virus. Unlike the i.n. route, the A/PR/8 virus, whether live or inactivated, did not migrate to or replicate in the CNS after s.l. administration. Based on these promising findings, we propose that the s.l. mucosal route offers an attractive alternative to mucosal routes for administering influenza vaccines.


Asunto(s)
Subtipo H1N1 del Virus de la Influenza A/inmunología , Vacunas contra la Influenza/administración & dosificación , Infecciones por Orthomyxoviridae/prevención & control , Vacunas de Productos Inactivados/administración & dosificación , Administración Sublingual , Animales , Anticuerpos Antivirales/sangre , Antígenos Virales/análisis , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Sistema Nervioso Central/inmunología , Inmunidad Innata , Inmunidad Mucosa , Interferón gamma/metabolismo , Ratones , Infecciones por Orthomyxoviridae/inmunología , Vacunación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA