Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
J Infect Dis ; 220(5): 743-751, 2019 07 31.
Artículo en Inglés | MEDLINE | ID: mdl-31045222

RESUMEN

BACKGROUND: Human immunodeficiency virus (HIV)-infected persons are at a higher risk of severe influenza. Although we have shown that a standard-dose intradermal influenza vaccine versus a standard-dose intramuscular influenza vaccine does not result in differences in hemagglutination-inhibition titers in this population, a comprehensive examination of cell-mediated immune responses remains lacking. METHODS: Serological, antigen-specific B-cell, and interleukin 2-, interferon γ-, and tumor necrosis factor α-secreting T-cell responses were assessed in 79 HIV-infected men and 79 HIV-uninfected men. RESULTS: The route of vaccination did not affect the immunoglobulin A and immunoglobulin G (IgG) plasmablast or memory B-cell response, although these were severely impaired in the group with a CD4+ T-cell count of <200 cells/µL. The frequencies of IgG memory B cells measured on day 28 after vaccination were highest in the HIV-uninfected group, followed by the group with a CD4+ T-cell count of ≥200 cells/µL and the group with a CD4+ T-cell count of <200 cells/µL. The route of vaccination did not affect the CD4+ or CD8+ T-cell responses measured at various times after vaccination. CONCLUSIONS: The route of vaccination had no effect on antibody responses, antibody avidity, T-cell responses, or B-cell responses in HIV-infected or HIV-uninfected subjects. With the serological and cellular immune responses to influenza vaccination being impaired in HIV-infected individuals with a CD4+ T-cell count of <200 cells/µL, passive immunization strategies need to be explored to protect this population. CLINICAL TRIALS REGISTRATION: NCT01538940.


Asunto(s)
Infecciones por VIH/inmunología , Inmunidad Celular/inmunología , Vacunas contra la Influenza/administración & dosificación , Vacunas contra la Influenza/inmunología , Vacunas contra la Influenza/normas , Gripe Humana/prevención & control , Adulto , Anticuerpos Antivirales/inmunología , Formación de Anticuerpos , Linfocitos B/inmunología , Recuento de Linfocito CD4 , Linfocitos T CD4-Positivos , Linfocitos T CD8-positivos , Infecciones por VIH/complicaciones , Pruebas de Inhibición de Hemaglutinación , Glicoproteínas Hemaglutininas del Virus de la Influenza/inmunología , Humanos , Inmunoglobulina A , Inmunoglobulina G , Subtipo H1N1 del Virus de la Influenza A/inmunología , Interferón gamma/metabolismo , Interleucina-2/metabolismo , Masculino , Persona de Mediana Edad , Tailandia , Factor de Necrosis Tumoral alfa/metabolismo , Vacunación
2.
J Infect Dis ; 214(7): 1030-8, 2016 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-27534687

RESUMEN

BACKGROUND: Emergence of antigenically drifted influenza A(H3N2) viruses resulted in reduced vaccine effectiveness in all age groups during the 2014-2015 influenza season. In children, inactivated influenza vaccine (IIV) elicited neutralizing antibodies (Abs) against drifted strains at significantly lower levels than against the vaccine strain. Little is known about the cross-reactivity of cell-mediated immunity against drifted strains in children. METHODS: Children aged 3-17 years (n = 48) received IIV during the 2014-2015 influenza season. Peripheral blood mononuclear cells, collected before (on day 0) and after (on days 7 and 21) vaccination were evaluated for induction of cross-reactive plasmablasts, memory B cells, and cytokine-secreting CD4(+) and CD8(+) T cells against the vaccine and drifted A(H3N2) viruses by an enzyme-linked immunospot assay and flow cytometry. RESULTS: IIV increased frequencies of plasmablasts and memory B cells. The overall induction of the T-cell response was not significant. Both B-cell and T-cell responses showed significant cross-reactivity against A(H3N2) viruses. Age and preexisting immunity affected virus-specific plasmablast responses and fold-change of T-cell responses, respectively. The proportion of T-helper type 1-prone (ie, interferon γ- or tumor necrosis factor α-secreting) CD4(+) T cell responses also increased with age. CONCLUSIONS: In children aged 3-17 years, B- and T-cell responses following IIV receipt showed significant cross-reactivity against A(H3N2) viruses during a vaccine mismatch season.


Asunto(s)
Linfocitos B/inmunología , Flujo Genético , Inmunidad Heteróloga , Subtipo H3N2 del Virus de la Influenza A/inmunología , Gripe Humana/inmunología , Gripe Humana/virología , Linfocitos T/inmunología , Adolescente , Niño , Preescolar , Reacciones Cruzadas , Femenino , Humanos , Subtipo H3N2 del Virus de la Influenza A/genética , Masculino
3.
J Infect Dis ; 214(10): 1477-1486, 2016 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-27571905

RESUMEN

BACKGROUND: Influenza viruses gradually accumulate point mutations, reducing the effectiveness of prior immune protection. METHODS: Children aged 9-14 years received 2010-2011 trivalent inactivated influenza vaccine (TIV). Vaccination history, hemagglutination-inhibition (HI) titers, and cell-mediated immune responses were assessed to investigate the cross-reactivity with past and future influenza virus strains. RESULTS: 2010-2011 TIV induced significant T-cell responses and HI titers of ≥160, with a fold-rise of ≥4 and titers of ≥100 maintained for >7 months in the majority of children. Pre-existing memory B cells in these children differentiated quickly to antibody-secreting cells to the new vaccine antigens. Children vaccinated in the previous year maintained high HI titers well into 2010, demonstrating elevated HI titers against A/Perth/16/2009, the future (in 2010-2011) H3N2 component. Prior vaccination enhanced CD8+ T-cell responses to A/Perth/16/2009. Children vaccinated with the prior 2009-2010 seasonal vaccine also demonstrated higher preexisting levels of interferon γ-secreting CD4+CD69+ T cells to 2009 pandemic influenza A(H1N1). Children previously vaccinated with 2009-2010 seasonal influenza vaccine also showed greater expansion of tumor necrosis factor α-secreting CD8+CD69+ T cells to 2009 pandemic influenza A(H1N1) upon vaccination in the 2010-2011 season than those who were not previously vaccinated. CONCLUSIONS: Seasonal influenza viruses continuously drift, which allows them to circumvent protective immunity, but conserved epitopes provide immunological cross-reactivity in children through either vaccination directly or through prime/boost in the prior influenza season.


Asunto(s)
Inmunidad Celular , Inmunidad Humoral , Vacunas contra la Influenza/inmunología , Gripe Humana/prevención & control , Orthomyxoviridae/inmunología , Adolescente , Anticuerpos Antivirales/sangre , Niño , Reacciones Cruzadas , Femenino , Pruebas de Inhibición de Hemaglutinación , Humanos , Vacunas contra la Influenza/administración & dosificación , Gripe Humana/inmunología , Masculino , Linfocitos T/inmunología , Factores de Tiempo , Vacunas de Productos Inactivados/administración & dosificación , Vacunas de Productos Inactivados/inmunología
4.
Cell Immunol ; 304-305: 55-8, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27106062

RESUMEN

Avian H7N9 influenza virus infection with fatal outcomes continues to pose a pandemic threat and highly immunogenic vaccines are urgently needed. In this report we show that baculovirus-derived recombinant H7 hemagglutinin protein, when delivered with RIG-I ligand, induced enhanced antibody and T cell responses and conferred protection against lethal challenge with a homologous H7N9 virus. These findings indicate the potential utility of RIG-I ligands as vaccine adjuvants to increase the immunogenicity of recombinant H7 hemagglutinin.


Asunto(s)
Proteína 58 DEAD Box/inmunología , Glicoproteínas Hemaglutininas del Virus de la Influenza/inmunología , Subtipo H7N9 del Virus de la Influenza A/inmunología , Vacunas contra la Influenza/inmunología , Gripe Humana/prevención & control , Infecciones por Orthomyxoviridae/prevención & control , Linfocitos T/inmunología , Adyuvantes Inmunológicos , Animales , Células Cultivadas , Femenino , Humanos , Inmunidad Humoral , Subtipo H7N9 del Virus de la Influenza A/metabolismo , Gripe Humana/inmunología , Activación de Linfocitos , Ratones , Ratones Endogámicos BALB C , Infecciones por Orthomyxoviridae/inmunología , Receptores Inmunológicos , Receptores de Reconocimiento de Patrones/metabolismo , Linfocitos T/virología , Vacunas Sintéticas
5.
Nanomedicine ; 12(7): 1909-1917, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27112307

RESUMEN

To enhance the immunogenicity of the Influenza H5N1 vaccine, we developed an oil-in-water nanoemulsion (NE) adjuvant. NE displayed good temperature stability and maintained particle size. More importantly, it significantly enhanced IL-6 and MCP-1 production to recruit innate cells, including neutrophils, monocytes/macrophages and dendritic cells to the local environment. Furthermore, NE enhanced dendritic cell function to induce robust antigen-specific T and B cell immune responses. NE-adjuvanted H5N1 vaccine not only elicited significantly higher and long-lasting antibody responses, but also conferred enhanced protection against homologous clade 1 as well as heterologous clade 2 H5N1 virus challenge in young as well as in aged mice. The pre-existing immunity to seasonal influenza did not affect the immunogenicity of NE-adjuvanted H5N1 vaccine.


Asunto(s)
Subtipo H5N1 del Virus de la Influenza A , Vacunas contra la Influenza/administración & dosificación , Nanopartículas/química , Adyuvantes Inmunológicos , Animales , Anticuerpos Antivirales , Emulsiones , Humanos , Gripe Humana/prevención & control , Ratones
6.
J Infect Dis ; 212(12): 1923-9, 2015 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-26068782

RESUMEN

We compared the innate immune response to a newly emerged swine-origin influenza A(H3N2) variant containing the M gene from 2009 pandemic influenza A(H1N1), termed "A(H3N2)vpM," to the immune responses to the 2010 swine-origin influenza A(H3N2) variant and seasonal influenza A(H3N2). Our results demonstrated that A(H3N2)vpM-induced myeloid dendritic cells secreted significantly lower levels of type I interferon (IFN) but produced significantly higher levels of proinflammatory cytokines and induced potent inflammasome activation. The reduction in antiviral immunity with increased inflammatory responses upon A(H3N2)vpM infection suggest that these viruses have the potential for increased disease severity in susceptible hosts.


Asunto(s)
Inflamasomas/metabolismo , Subtipo H1N1 del Virus de la Influenza A/inmunología , Subtipo H3N2 del Virus de la Influenza A/inmunología , Leucocitos Mononucleares/inmunología , Animales , Línea Celular , Citocinas/metabolismo , Células Dendríticas/inmunología , Humanos , Subtipo H1N1 del Virus de la Influenza A/aislamiento & purificación , Subtipo H3N2 del Virus de la Influenza A/aislamiento & purificación , Infecciones por Orthomyxoviridae/virología , Porcinos , Enfermedades de los Porcinos/virología
7.
Proc Natl Acad Sci U S A ; 109(34): 13751-6, 2012 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-22869731

RESUMEN

Original antigenic sin is a phenomenon wherein sequential exposure to closely related influenza virus variants reduces antibody (Ab) response to novel antigenic determinants in the second strain and, consequently, impairs the development of immune memory. This could pose a risk to the development of immune memory in persons previously infected with or vaccinated against influenza. Here, we explored strategies to overcome original antigenic sin responses in mice sequentially exposed to two closely related hemagglutinin 1 neuraminidase 1 (H1N1) influenza strains A/PR/8/34 and A/FM/1/47. We found that dendritic cell-activating adjuvants [Bordetella pertussis toxin (PT) or CpG ODN or a squalene-based oil-in-water nanoemulsion (NE)], upon administration during the second viral exposure, completely protected mice from a lethal challenge and enhanced neutralizing-Ab titers against the second virus. Interestingly, PT and NE adjuvants when administered during the first immunization even prevented original antigenic sin in subsequent immunization without any adjuvants. As an alternative to using adjuvants, we also found that repeated immunization with the second viral strain relieved the effects of original antigenic sin. Taken together, our studies provide at least three ways of overcoming original antigenic sin.


Asunto(s)
Formación de Anticuerpos , Inmunización/métodos , Memoria Inmunológica , Orthomyxoviridae/genética , Animales , Presentación de Antígeno , Bordetella/metabolismo , Línea Celular , Islas de CpG , Perros , Glicoproteínas Hemaglutininas del Virus de la Influenza/inmunología , Humanos , Sistema Inmunológico , Subtipo H1N1 del Virus de la Influenza A/inmunología , Ratones , Ratones Endogámicos BALB C , Oligonucleótidos , Toxina del Pertussis/metabolismo
8.
J Immunol ; 189(6): 2758-67, 2012 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-22908331

RESUMEN

The interaction of CD28, which is constitutively expressed on T cells, with B7.1/B7.2 expressed on APCs is critical for T cell activation. CD28 is also expressed on murine and human plasma cells but its function on these cells remains unclear. There are two types of plasma cells: short-lived ones that appear in the secondary lymphoid tissue shortly after Ag exposure, and long-lived plasma cells that mainly reside in the bone marrow. We demonstrate that CD28-deficient murine short- and long-lived plasma cells produce significantly higher levels of Abs than do their wild-type counterparts. This was owing to both increased frequencies of plasma cells as well as increased Ab production per plasma cell. Plasma cells also express the ligand for CD28, B7.1, and B7.2. Surprisingly, deficiency of B7.1 and B7.2 in B cells also led to higher Ab levels, analogous to Cd28(-/-) plasma cells. Collectively, our results suggest that the CD28-B7 interaction operates as a key modulator of plasma cell function.


Asunto(s)
Antígenos B7/fisiología , Antígenos CD28/fisiología , Supervivencia Celular/inmunología , Senescencia Celular/inmunología , Células Plasmáticas/citología , Células Plasmáticas/inmunología , Animales , Antígenos B7/deficiencia , Células de la Médula Ósea/citología , Células de la Médula Ósea/inmunología , Células de la Médula Ósea/metabolismo , Antígenos CD28/biosíntesis , Antígenos CD28/deficiencia , Línea Celular , Femenino , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Células Plasmáticas/metabolismo , Bazo/citología , Bazo/inmunología , Bazo/metabolismo , Factores de Tiempo
9.
J Immunol ; 189(5): 2257-65, 2012 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-22855715

RESUMEN

Myeloid dendritic cells (mDCs) have long been thought to function as classical APCs for T cell responses. However, we demonstrate that influenza viruses induce rapid differentiation of human monocytes into mDCs. Unlike the classic mDCs, the virus-induced mDCs failed to upregulate DC maturation markers and were unable to induce allogeneic lymphoproliferation. Virus-induced mDCs secreted little, if any, proinflammatory cytokines; however, they secreted a substantial amount of chemoattractants for monocytes (MCP-1 and IP-10). Interestingly, the differentiated mDCs secreted type I IFN and upregulated the expression of IFN-stimulated genes (tetherin, IFITM3, and viperin), as well as cytosolic viral RNA sensors (RIG-I and MDA5). Additionally, culture supernatants from virus-induced mDCs suppressed the replication of virus in vitro. Furthermore, depletion of monocytes in a mouse model of influenza infection caused significant reduction of lung mDC numbers, as well as type I IFN production in the lung. Consequently, increased lung virus titer and higher mortality were observed. Taken together, our results demonstrate that the host responds to influenza virus infection by initiating rapid differentiation of circulating monocytes into IFN-producing mDCs, which contribute to innate antiviral immune responses.


Asunto(s)
Diferenciación Celular/inmunología , Células Dendríticas/inmunología , Interferón Tipo I/biosíntesis , Monocitos/inmunología , Células Mieloides/inmunología , Infecciones por Orthomyxoviridae/prevención & control , Animales , Células Cultivadas , Células Dendríticas/patología , Células Dendríticas/virología , Femenino , Humanos , Subtipo H1N1 del Virus de la Influenza A/inmunología , Subtipo H3N2 del Virus de la Influenza A/inmunología , Gripe Humana/inmunología , Gripe Humana/patología , Gripe Humana/prevención & control , Interferón Tipo I/fisiología , Ratones , Ratones Endogámicos C57BL , Monocitos/metabolismo , Monocitos/patología , Células Mieloides/patología , Células Mieloides/virología , Infecciones por Orthomyxoviridae/inmunología , Infecciones por Orthomyxoviridae/patología , Factores de Tiempo
10.
J Virol ; 86(20): 10988-98, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22837197

RESUMEN

Recognition of pathogen-associated molecular patterns by pattern recognition receptors of the innate immune system is crucial for the initiation of innate and adaptive responses and for immunological memory. We investigated the role of TLR7 in the induction of adaptive immunity and long-term memory following influenza virus infection and vaccination in C57BL/6 mice. During infection with influenza A/PR8/34 virus, the absence of either TLR7 or MyD88 leads to reduced virus-specific antibodies in the serum and antibody-secreting cells in their secondary lymphoid organs, particularly in bone marrow. In spite of this, the absence of TLR7/MyD88 signaling did not impair the production of protective antibodies. Following immunization with the 2009 pandemic inactivated split vaccine, TLR7(-/-) mice had significantly lower levels of germinal center formation, antibody-secreting cells, and circulating influenza virus-specific antibodies than control animals. Consequently, TLR7(-/-) mice failed to develop protective immunological memory upon challenge. Furthermore, the immunogenicity of the split vaccine was likely due to TLR7 recognition of virion RNA, as its removal from the split vaccine significantly reduced the levels of influenza virus-specific antibodies and compromised the vaccine protective efficacy in mice. Taken together, our data demonstrate that TLR7 plays an important role in vaccine-induced humoral immune responses to influenza virus through the interaction with viral RNA present in the split vaccine.


Asunto(s)
Anticuerpos Antivirales/biosíntesis , Glicoproteínas Hemaglutininas del Virus de la Influenza/inmunología , Subtipo H1N1 del Virus de la Influenza A/inmunología , Vacunas contra la Influenza/inmunología , Glicoproteínas de Membrana/inmunología , Infecciones por Orthomyxoviridae/inmunología , Receptor Toll-Like 7/inmunología , Animales , Anticuerpos Antivirales/sangre , Anticuerpos Antivirales/inmunología , Células Productoras de Anticuerpos/inmunología , Centro Germinal/virología , Memoria Inmunológica , Glicoproteínas de Membrana/deficiencia , Glicoproteínas de Membrana/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Factor 88 de Diferenciación Mieloide/deficiencia , Factor 88 de Diferenciación Mieloide/genética , Factor 88 de Diferenciación Mieloide/metabolismo , Infecciones por Orthomyxoviridae/prevención & control , Infecciones por Orthomyxoviridae/virología , ARN Viral/inmunología , ARN Viral/metabolismo , Receptor Toll-Like 7/deficiencia , Receptor Toll-Like 7/fisiología
11.
J Immunol ; 185(3): 1642-9, 2010 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-20585035

RESUMEN

The 2009 H1N1 influenza virus outbreak is the first pandemic of the twenty-first century. Epidemiological data reveal that of all the people afflicted with H1N1 virus, <5% are over 51 y of age. Interestingly, in the uninfected population, 33% of those >60 y old have pre-existing neutralizing Abs against the 2009 H1N1 virus. This finding suggests that influenza strains that circulated 50-60 y ago might provide cross-protection against the swine-origin 2009 H1N1 influenza virus. To test this, we determined the ability of representative H1N1 influenza viruses that circulated in the human population from 1930 to 2000, to induce cross-reactivity to and cross-protection against the pandemic swine-origin H1N1 virus, A/California/04/09. We show that exposure of mice to the 1947 virus, A/FM/1/47, or the 1934 virus, A/PR/8/34, induced robust cross-protective immune responses and these mice were protected against a lethal challenge with mouse-adapted A/California/04/09 H1N1 virus. Conversely, we observed that mice exposed to the 2009 H1N1 virus were protected against a lethal challenge with mouse-adapted 1947 or 1934 H1N1 viruses. In addition, exposure to the 2009 H1N1 virus induced broad cross-reactivity against H1N1 as well as H3N2 influenza viruses. Finally, we show that vaccination with the older H1N1 viruses, particularly A/FM/1/47, confers protective immunity against the 2009 pandemic H1N1 virus. Taken together, our data provide an explanation for the decreased susceptibility of the elderly to the 2009 H1N1 outbreak and demonstrate that vaccination with the pre-1950 influenza strains can cross-protect against the pandemic swine-origin 2009 H1N1 influenza virus.


Asunto(s)
Protección Cruzada/inmunología , Brotes de Enfermedades , Subtipo H1N1 del Virus de la Influenza A/inmunología , Infecciones por Orthomyxoviridae/inmunología , Infecciones por Orthomyxoviridae/prevención & control , Animales , Anticuerpos Antivirales/biosíntesis , Pruebas de Inhibición de Hemaglutinación , Humanos , Inmunoglobulina G/biosíntesis , Virus de la Influenza A/inmunología , Vacunas contra la Influenza/administración & dosificación , Vacunas contra la Influenza/inmunología , Ratones , Ratones Endogámicos BALB C , Infecciones por Orthomyxoviridae/mortalidad
12.
Influenza Other Respir Viruses ; 16(3): 562-567, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-34859584

RESUMEN

Individuals with type 2 diabetes mellitus experience high rates of influenza virus infection and complications. We compared the magnitude and duration of serologic response to trivalent influenza vaccine in adults aged 50-80 with and without type 2 diabetes mellitus. Serologic response to influenza vaccination was similar in both groups: greater fold-increases in antibody titer occurred among participants with lower pre-vaccination antibody titers. Waning of antibody titers was not influenced by diabetes status.


Asunto(s)
Diabetes Mellitus Tipo 2 , Vacunas contra la Influenza , Gripe Humana , Anciano , Anciano de 80 o más Años , Anticuerpos Antivirales , Pruebas de Inhibición de Hemaglutinación , Humanos , Subtipo H3N2 del Virus de la Influenza A , Gripe Humana/prevención & control , Persona de Mediana Edad , Vacunas de Productos Inactivados
13.
J Immunol ; 183(5): 3294-301, 2009 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-19648276

RESUMEN

Most immune responses follow Burnet's rule in that Ag recruits specific lymphocytes from a large repertoire and induces them to proliferate and differentiate into effector cells. However, the phenomenon of "original antigenic sin" stands out as a paradox to Burnet's rule of B cell engagement. Humans, upon infection with a novel influenza strain, produce Abs against older viral strains at the expense of responses to novel, protective antigenic determinants. This exacerbates the severity of the current infection. This blind spot of the immune system and the redirection of responses to the "original Ag" rather than to novel epitopes were described fifty years ago. Recent reports have questioned the existence of this phenomenon. Hence, we revisited this issue to determine the extent to which original antigenic sin is induced by variant influenza viruses. Using two related strains of influenza A virus, we show that original antigenic sin leads to a significant decrease in development of protective immunity and recall responses to the second virus. In addition, we show that sequential infection of mice with two live influenza virus strains leads to almost exclusive Ab responses to the first viral strain, suggesting that original antigenic sin could be a potential strategy by which variant influenza viruses subvert the immune system.


Asunto(s)
Anticuerpos Antivirales/biosíntesis , Subtipo H1N1 del Virus de la Influenza A/inmunología , Vacunas contra la Influenza/inmunología , Animales , Línea Celular , Perros , Glicoproteínas Hemaglutininas del Virus de la Influenza/administración & dosificación , Glicoproteínas Hemaglutininas del Virus de la Influenza/inmunología , Vacunas contra la Influenza/administración & dosificación , Ratones , Ratones Endogámicos BALB C , Pruebas de Neutralización , Factores de Tiempo , Vacunas Atenuadas/administración & dosificación , Vacunas Atenuadas/inmunología , Vacunas de ADN/administración & dosificación , Vacunas de ADN/inmunología , Vacunas de Productos Inactivados/administración & dosificación , Vacunas de Productos Inactivados/inmunología , Carga Viral
14.
Nat Commun ; 12(1): 1222, 2021 02 22.
Artículo en Inglés | MEDLINE | ID: mdl-33619272

RESUMEN

Programmed death-ligand 1 is a glycoprotein expressed on antigen presenting cells, hepatocytes, and tumors which upon interaction with programmed death-1, results in inhibition of antigen-specific T cell responses. Here, we report a mechanism of inhibiting programmed death-ligand 1 through small molecule-induced dimerization and internalization. This represents a mechanism of checkpoint inhibition, which differentiates from anti-programmed death-ligand 1 antibodies which function through molecular disruption of the programmed death 1 interaction. Testing of programmed death ligand 1 small molecule inhibition in a humanized mouse model of colorectal cancer results in a significant reduction in tumor size and promotes T cell proliferation. In addition, antigen-specific T and B cell responses from patients with chronic hepatitis B infection are significantly elevated upon programmed death ligand 1 small molecule inhibitor treatment. Taken together, these data identify a mechanism of small molecule-induced programmed death ligand 1 internalization with potential therapeutic implications in oncology and chronic viral infections.


Asunto(s)
Antígeno B7-H1/metabolismo , Endocitosis , Inhibidores de Puntos de Control Inmunológico/farmacología , Bibliotecas de Moléculas Pequeñas/farmacología , Animales , Antineoplásicos/farmacología , Antivirales/farmacología , Células CHO , Proliferación Celular/efectos de los fármacos , Neoplasias Colorrectales/patología , Cricetulus , Modelos Animales de Enfermedad , Femenino , Virus de la Hepatitis B/efectos de los fármacos , Humanos , Ratones Endogámicos C57BL , Receptor de Muerte Celular Programada 1/metabolismo , Multimerización de Proteína/efectos de los fármacos , Bibliotecas de Moléculas Pequeñas/química
15.
Curr Top Microbiol Immunol ; 333: 197-210, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19768407

RESUMEN

As an attractive alternative to conventional vaccines, DNA vaccines play a critical role in inducing protection against several infectious diseases. In this review, we discuss the advantages that DNA vaccines offer in comparison to conventional protein-based vaccines. We discuss strategies to improve the potency and efficacy of DNA vaccines. Specifically, we focus on the potential use of DNA-based vaccines to elicit broad-spectrum humoral and cellular immunity against influenza virus. Finally, we discuss the advances made in the use of DNA vaccines to prevent avian H5N1 influenza.


Asunto(s)
Subtipo H1N1 del Virus de la Influenza A/inmunología , Subtipo H5N1 del Virus de la Influenza A/inmunología , Vacunas contra la Influenza/inmunología , Vacunas de ADN/inmunología , Animales , Humanos
16.
Hepat Oncol ; 8(1): HEP35, 2020 Dec 29.
Artículo en Inglés | MEDLINE | ID: mdl-33680431

RESUMEN

Chronic hepatitis B virus (HBV) infection is a major public health challenge on the global scale. Affecting hundreds of millions worldwide, HBV is a leading risk factor for hepatocellular carcinoma (HCC). Clinical outcomes from chronic HBV infection are varied and appear to be influenced by a complex and dysregulated host immune response. In turn, much attention has been given to the immunologic response to HBV in an effort to identify host factors that lead to the development of HCC. However, the role of nonimmunologic host factors, such as chronic stress, in HBV-related HCC is poorly defined. Indeed, a growing appreciation for the effects of stress on chronic liver diseases raises the question of its role in chronic HBV infection. In this light, the present review will untangle the roles of key host factors in HBV-related HCC with an emphasis on chronic stress as a viable contributor. First discussed is the interplay of stress, inflammation and chronic liver disease. The host immune response's role as a driver of HBV-related HCC is then reviewed, allowing for a close exploration of the effects of stress on immune function in chronic hepatitis B and as a potential risk factor for HBV-related HCC.

17.
18.
Sci Rep ; 10(1): 1835, 2020 02 04.
Artículo en Inglés | MEDLINE | ID: mdl-32020034

RESUMEN

Chronic hepatitis B (CHB) infection functional cure is defined as sustained loss of HBsAg and several therapeutic strategies are in clinical development designed to pharmacologically reduce serum HBsAg, break immune tolerance, and increase functional cure rates. However, little is known about pre-treatment HBsAg levels as an indicator of HBV immune potential. Here, we compared the phenotypes and HBV-specific response of lymphocytes in CHB patients stratified by serum HBsAg levels <500 (HBslo) or >50,000 IU/ml (HBshi) using immunological assays (flow cytometry, ICS, ELISPOT). HBshi patients had significantly higher expression of inhibitory PD-1 on CD4+ T cells, particularly among TEMRA subset, and higher FcRL5 expression on B cells. Upon HBcAg(core) or HBsAg(env)-stimulation, 85% and 60% of HBslo patients had IFNγ+TNFα+ and IFNγ+ IL2+ CD4+ T cell responses respectively, in comparison to 33% and 13% of HBshi patients. Checkpoint blockade with αPD-1 improved HBV-specific CD4+ T cell function only in HBslo patients. HBsAg-specific antibody-secreting cells (ASCs) response was not different between these groups, yet αPD-1 treatment resulted in significantly higher fold change in ASCs among patients with HBsAg <100 IU/ml compared to patients with HBsAg >5,000 IU/ml. Thus, serum HBsAg correlates with inhibitory receptor expression, HBV-specific CD4+ T cell responses, and augmentation by checkpoint blockade.


Asunto(s)
Linfocitos B/inmunología , Antígenos de Superficie de la Hepatitis B/sangre , Virus de la Hepatitis B/inmunología , Hepatitis B Crónica/inmunología , Linfocitos T/inmunología , Biomarcadores/sangre , Citometría de Flujo , Hepatitis B Crónica/sangre , Humanos , Receptor de Muerte Celular Programada 1/metabolismo
19.
Sci Rep ; 8(1): 6112, 2018 04 17.
Artículo en Inglés | MEDLINE | ID: mdl-29666412

RESUMEN

Influenza virus causes widespread, yearly epidemics by accumulating surface protein mutations to escape neutralizing antibodies established from prior exposure. In contrast to antibody epitopes, T cell mediated immunity targets influenza epitopes that are more highly conserved and have potential for cross-protection. The extent of T cell cross-reactivity between a diverse array of contemporary and historical influenza strains was investigated in ferrets challenged with 2009 pandemic H1N1 influenza or the seasonal H3N2 strain, A/Perth/16/2009. Post-challenge cell-mediated immune responses demonstrated extensive cross-reactivity with a wide variety of contemporary and historical influenza A strains as well as influenza B. Responses in peripheral blood were undetectable by 36d post-challenge, but cross-reactivity persisted in spleen. The strongest responses targeted peptides from the NP protein and demonstrated cross-reactivity in both the CD4+ and CD8+ T cell populations. Cross-reactive CD4+ T cells also targeted HA and NA epitopes, while cross-reactive CD8+ T cells targeted internal M1, NS2, and PA. T cell epitopes demonstrated extensive cross-reactivity between diverse influenza strains in outbred animals, with NP implicated as a significant antigenic target demonstrating extensive cross-reactivity for both CD4+ and CD8+ T cells.


Asunto(s)
Epítopos de Linfocito T/inmunología , Hurones/virología , Subtipo H1N1 del Virus de la Influenza A/inmunología , Subtipo H3N2 del Virus de la Influenza A/inmunología , Infecciones por Orthomyxoviridae/inmunología , Linfocitos T/inmunología , Animales , Reacciones Cruzadas , Modelos Animales de Enfermedad , Hurones/inmunología , Inmunidad Celular , Masculino , Estaciones del Año
20.
Vaccine ; 35(25): 3318-3325, 2017 06 05.
Artículo en Inglés | MEDLINE | ID: mdl-28499553

RESUMEN

Sporadic, yet frequent human infections with avian H5N1 influenza A viruses continue to pose a potential pandemic threat. Poor immunogenicity of unadjuvanted H5N1 vaccines warrants developing novel adjuvants and formulations as well as alternate delivery systems to improve their immunogenicity and efficacy. Here, we show that Protollin, a nasal adjuvant composed of Neisseria meningitides outer membrane proteins non-covalently linked to Shigella flexneri 2a lipopolysaccharide, is a potent nasal adjuvant for an inactivated split virion H5N1 clade 1 A/Viet Nam1203/2004 (A/VN/1203/04) vaccine in a mouse model. Protollin-adjuvanted vaccines elicited enhanced serum protective hemagglutination inhibition titers, mucosal IgA responses, and H5N1-specific cell-mediated immunity that resulted in complete protection against a lethal challenge with a homologous virus as well as a heterologous clade 2 virus A/Indonesia/05/2005 (A/IN/05/05). Detailed analysis of adaptive immunity revealed that Protollin increased the frequency of lymphoid- as well as local tissue-resident antibody-secreting cells, local germinal center reaction of B cells, broad-spectrum of CD4 T cell response. Our findings suggest that nasal delivery of H5N1 vaccine with Protollin adjuvant can overcome the poor immunogenicity of H5N1 vaccines, induce both cellular and humoral immune responses, enhance protection against challenge with clade 1 and clade 2 H5N1 viruses and achieve significant antigen dose-sparing.


Asunto(s)
Adyuvantes Inmunológicos , Cisteína Endopeptidasas/inmunología , Inmunidad Mucosa , Subtipo H5N1 del Virus de la Influenza A/inmunología , Vacunas contra la Influenza/inmunología , Lipopolisacáridos/inmunología , Infecciones por Orthomyxoviridae/inmunología , Animales , Anticuerpos Antivirales/sangre , Células Productoras de Anticuerpos/inmunología , Linfocitos T CD4-Positivos/inmunología , Modelos Animales de Enfermedad , Combinación de Medicamentos , Pruebas de Inhibición de Hemaglutinación , Inmunidad Celular , Inmunidad Humoral , Inmunogenicidad Vacunal , Inmunoglobulina A/biosíntesis , Inmunoglobulina A/inmunología , Vacunas contra la Influenza/administración & dosificación , Ratones , Infecciones por Orthomyxoviridae/prevención & control
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA