Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 50
Filtrar
1.
J Immunol ; 206(9): 2122-2134, 2021 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-33827894

RESUMEN

Respiratory syncytial virus (RSV) is a leading cause of lower respiratory tract infection in both young children and in older adults. Despite the morbidity, mortality, and high economic burden caused by RSV worldwide, no licensed vaccine is currently available. We have developed a novel RSV vaccine composed of a prefusion-stabilized variant of the fusion (F) protein (DS-Cav1) and a CpG oligodeoxynucleotide adjuvant encapsulated within polyanhydride nanoparticles, termed RSVNanoVax. A prime-boost intranasal administration of RSVNanoVax in BALB/c mice significantly alleviated weight loss and pulmonary dysfunction in response to an RSV challenge, with protection maintained up to at least 6 mo postvaccination. In addition, vaccinated mice exhibited rapid viral clearance in the lungs as early as 2 d after RSV infection in both inbred and outbred populations. Vaccination induced tissue-resident memory CD4 and CD8 T cells in the lungs, as well as RSV F-directed neutralizing Abs. Based on the robust immune response elicited and the high level of durable protection observed, our prefusion RSV F nanovaccine is a promising new RSV vaccine candidate.


Asunto(s)
Inmunidad Celular/inmunología , Polianhídridos/química , Vacunas contra Virus Sincitial Respiratorio/inmunología , Virus Sincitial Respiratorio Humano/inmunología , Animales , Femenino , Ratones , Ratones Endogámicos BALB C
2.
J Immunol ; 203(12): 3313-3324, 2019 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-31712384

RESUMEN

Chronic alcohol consumption is associated with an increased incidence of disease severity during pulmonary infections. Our previous work in a mouse model of chronic alcohol consumption has detailed that the primary influenza A virus (IAV)-specific CD8 T cell response in mice that consumed ethanol (EtOH) had a reduced proliferative capacity as well as the ability to kill IAV target cells. Interestingly, recent studies have highlighted that human alcoholics have an increased susceptibility to IAV infections, even though they likely possess pre-existing immunity to IAV. However, the effects of chronic alcohol consumption on pre-existing immune responses (i.e., memory) to IAV have not been explored. Our results presented in this study show that IAV-immune mice that then chronically consumed alcohol (X31→EtOH) exhibited increased morbidity and mortality following IAV re-exposure compared with IAV-immune mice that had consumed water (X31→H2O). This increased susceptibility in X31→EtOH mice was associated with reduced IAV-specific killing of target cells and a reduction in the number of IAV-specific CD8 T cells within the lungs. Furthermore, upon IAV challenge, recruitment of the remaining memory IAV-specific CD8 T cells into the lungs is reduced in X31→EtOH mice. This altered recruitment is associated with a reduced pulmonary expression of CXCL10 and CXCL11, which are chemokines that are important for T cell recruitment to the lungs. Overall, these results demonstrate that chronic alcohol consumption negatively affects the resting memory CD8 T cell response and reduces the ability of memory T cells to be recruited to the site of infection upon subsequent exposures, therein contributing to an enhanced susceptibility to IAV infections.


Asunto(s)
Bebidas Alcohólicas/efectos adversos , Linfocitos T CD8-positivos/inmunología , Etanol/farmacología , Memoria Inmunológica/efectos de los fármacos , Virus de la Influenza A , Infecciones por Orthomyxoviridae/inmunología , Animales , Quimiocina CXCL10/metabolismo , Quimiocina CXCL11/metabolismo , Susceptibilidad a Enfermedades/inducido químicamente , Etanol/administración & dosificación , Pulmón/efectos de los fármacos , Pulmón/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Infecciones por Orthomyxoviridae/virología , Receptores CXCR3/metabolismo , Transducción de Señal/efectos de los fármacos
3.
J Immunol ; 200(3): 1188-1197, 2018 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-29282312

RESUMEN

Exaggerated inflammatory responses during influenza A virus (IAV) infection are typically associated with severe disease. Neutrophils are among the immune cells that can drive this excessive and detrimental inflammation. In moderation, however, neutrophils are necessary for optimal viral control. In this study, we explore the role of the nucleotide-binding domain leucine-rich repeat containing receptor family member Nlrp12 in modulating neutrophilic responses during lethal IAV infection. Nlrp12-/- mice are protected from lethality during IAV infection and show decreased vascular permeability, fewer pulmonary neutrophils, and a reduction in levels of neutrophil chemoattractant CXCL1 in their lungs compared with wild-type mice. Nlrp12-/- neutrophils and dendritic cells within the IAV-infected lungs produce less CXCL1 than their wild-type counterparts. Decreased CXCL1 production by Nlrp12-/- dendritic cells was not due to a difference in CXCL1 protein stability, but instead to a decrease in Cxcl1 mRNA stability. Together, these data demonstrate a previously unappreciated role for Nlrp12 in exacerbating the pathogenesis of IAV infection through the regulation of CXCL1-mediated neutrophilic responses.


Asunto(s)
Quimiocina CXCL1/metabolismo , Virus de la Influenza A/inmunología , Péptidos y Proteínas de Señalización Intracelular/genética , Infiltración Neutrófila/inmunología , Neutrófilos/inmunología , Infecciones por Orthomyxoviridae/inmunología , Animales , Permeabilidad Capilar/genética , Quimiocina CXCL1/genética , Células Dendríticas/inmunología , Femenino , Pulmón/inmunología , Pulmón/patología , Pulmón/virología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Infecciones por Orthomyxoviridae/mortalidad , Infecciones por Orthomyxoviridae/virología , Estabilidad del ARN/genética , ARN Mensajero/genética
4.
J Immunol ; 197(11): 4301-4311, 2016 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-27798171

RESUMEN

Patients surviving acute stages of sepsis often display impaired adaptive-immune responses. Using the cecal ligation and puncture model, we demonstrated that sepsis leads to substantial and long-lasting changes in the naive CD8 T cell repertoire, affecting the capacity of the host to respond to new infections. However, the identity of CD8 T cell-extrinsic factor(s) and mechanism(s) that contribute to impaired CD8 T cell responses after sepsis is unknown. Priming of naive CD8 T cells is critically dependent on the ability of dendritic cells (DCs) to provide Ag, costimulation, and inflammatory signal 3 cytokines; therefore, the sepsis-induced changes in the DC compartment might represent a contributing factor leading to diminished CD8 T cell immunity in septic hosts. In a direct test of this hypothesis, we show that, in addition to numerical decline, sepsis leads to functional impairments in DCs, diminishing their capacity to produce cytokines upon TLR stimulation in vitro or postinfection in vivo. Importantly, we demonstrated a direct link between DC dysfunction and impairments in CD8 T cell immunity after sepsis by directly targeting Ag to DCs. Finally, postsepsis Flt3 ligand treatment increased the number of DCs and improved DC function, including the ability to sense inflammation and produce IL-12, leading to improved primary CD8 T cell responses to newly encountered Ags. Thus, sepsis-induced numerical and functional loss of DCs contributes to the observed defects in CD8 T cell immunity, and therapeutic approaches designed to improve the status of the DC compartment after sepsis might facilitate the recovery of CD8 T cell immunity.


Asunto(s)
Presentación de Antígeno , Linfocitos T CD8-positivos/inmunología , Células Dendríticas/inmunología , Sepsis/inmunología , Animales , Antígenos/genética , Antígenos/inmunología , Linfocitos T CD8-positivos/patología , Citocinas/genética , Citocinas/inmunología , Células Dendríticas/patología , Ratones , Ratones Transgénicos , Sepsis/genética , Sepsis/patología
5.
J Virol ; 90(6): 2830-7, 2015 Dec 30.
Artículo en Inglés | MEDLINE | ID: mdl-26719269

RESUMEN

UNLABELLED: Following influenza A virus (IAV) infection, development of a robust IAV-specific CD8 T cell response is required for clearance of primary infection and enhances memory protection. Following IAV infection, plasmacytoid dendritic cells (pDC) or CD8α(+) DC regulate pulmonary effector CD8 T cell responses within the lung. Without this DC-T cell interaction, insufficient effector CD8 T cells are maintained in the lungs, leading to enhanced morbidity and mortality. Previous studies have demonstrated that pDC are capable of classical presentation or cross-presentation of IAV antigens and could potentially regulate IAV-specific CD8 T cell responses through either mechanism. Our results demonstrate that pDC from the lungs of donor mice infected with an IAV that is not able to replicate in hematopoietic cells (142t-IAV), unlike donor pDC isolated from the lungs of control infected mice, are not able to rescue the host IAV-specific CD8 T cell response from apoptosis. This indicates that pDC must utilize the direct presentation pathway for this rescue. This inability of pDC from 142t-IAV donors to rescue the IAV-specific CD8 T cell response is not due to differences in the overall ability of 142t-IAV to replicate within the lungs or generate defective viral genomes or to differences in levels of costimulatory molecules required for this interaction. We further demonstrate that bypassing the antigen presentation pathway by coating the 142t-IAV pDC with IAV peptide epitopes restores their ability to rescue the IAV-specific CD8 T cell response. IMPORTANCE: IAV continues to be a global health burden, infecting 5 to 20% of the global population annually. Continued investigation into the mechanisms that mediate protective immune responses against IAV is important to improving current vaccination and antiviral strategies antagonistic toward IAV. Our findings presented herein demonstrate a key requirement for pDC promotion of effector CD8 T cell survival: that rather than utilizing cross-presentation, pDC must be infected and utilize the endogenous pathway for presentation of antigens to CD8 T cells during in vivo IAV infections. This suggests that targeting presentation via the endogenous pathway in pDC could be important for the development of unique antiviral cellular therapies.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Células Dendríticas/inmunología , Células Dendríticas/virología , Virus de la Influenza A/inmunología , Pulmón/inmunología , Animales , Ratones Endogámicos BALB C
6.
J Immunol ; 192(12): 5906-12, 2014 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-24829418

RESUMEN

Although lymphopenia is a hallmark of severe infection with highly pathogenic H5N1 and the newly emerged H7N9 influenza viruses in humans, the mechanism(s) by which lethal H5N1 viruses cause lymphopenia in mammalian hosts remains poorly understood. Because influenza-specific T cell responses are initiated in the lung draining lymph nodes (LNs), and lymphocytes subsequently traffic to the lungs or peripheral circulation, we compared the immune responses in the lung draining LNs postinfection with a lethal A/HK/483/97 or nonlethal A/HK/486/97 (H5N1) virus in a mouse model. We found that lethal H5N1, but not nonlethal H5N1, virus infection in mice enhances Fas ligand (FasL) expression on plasmacytoid dendritic cells (pDCs), resulting in apoptosis of influenza-specific CD8(+) T cells via a Fas-FasL-mediated pathway. We also found that pDCs, but not other DC subsets, preferentially accumulate in the lung draining LNs of lethal H5N1 virus-infected mice, and that the induction of FasL expression on pDCs correlates with high levels of IL-12p40 monomer/homodimer in the lung draining LNs. Our data suggest that one of the mechanisms of lymphopenia associated with lethal H5N1 virus infection involves a deleterious role for pDCs.


Asunto(s)
Apoptosis/inmunología , Células Dendríticas/inmunología , Subtipo H5N1 del Virus de la Influenza A/inmunología , Linfopenia/inmunología , Infecciones por Orthomyxoviridae/inmunología , Células Plasmáticas/inmunología , Linfocitos T/inmunología , Animales , Células Dendríticas/patología , Células Dendríticas/virología , Proteína Ligando Fas/inmunología , Subtipo H5N1 del Virus de la Influenza A/patogenicidad , Interleucina-12/inmunología , Pulmón/inmunología , Pulmón/patología , Pulmón/virología , Linfopenia/etiología , Linfopenia/patología , Ratones , Ratones Endogámicos BALB C , Infecciones por Orthomyxoviridae/complicaciones , Infecciones por Orthomyxoviridae/patología , Células Plasmáticas/patología , Células Plasmáticas/virología , Linfocitos T/patología , Linfocitos T/virología , Receptor fas/inmunología
7.
J Immunol ; 193(12): 5873-82, 2014 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-25378592

RESUMEN

The extent to which obesity compromises the differentiation and maintenance of protective memory CD8 T cell responses and renders obese individuals susceptible to infection remains unknown. In this study, we show that diet-induced obesity did not impact the maintenance of pre-existing memory CD8 T cells, including acquisition of a long-term memory phenotype (i.e., CD27(hi), CD62L(hi), KLRG1(lo)) and function (i.e., cytokine production, secondary expansion, and memory CD8 T cell-mediated protection). Additionally, obesity did not influence the differentiation and maintenance of newly evoked memory CD8 T cell responses in inbred and outbred hosts generated in response to different types of systemic (LCMV, L. monocytogenes) and/or localized (influenza virus) infections. Interestingly, the rate of naive-to-memory CD8 T cell differentiation after a peptide-coated dendritic cell immunization was similar in lean and obese hosts, suggesting that obesity-associated inflammation, unlike pathogen- or adjuvant-induced inflammation, did not influence the development of endogenous memory CD8 T cell responses. Therefore, our studies reveal that the obese environment does not influence the development or maintenance of memory CD8 T cell responses that are either primed before or after obesity is established, a surprising notion with important implications for future studies aiming to elucidate the role obesity plays in host susceptibility to infections.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Dieta/efectos adversos , Memoria Inmunológica/inmunología , Obesidad/etiología , Animales , Antígenos/inmunología , Infecciones Bacterianas/inmunología , Linfocitos T CD8-positivos/metabolismo , Células Dendríticas/inmunología , Dieta Alta en Grasa/efectos adversos , Modelos Animales de Enfermedad , Epítopos de Linfocito T/inmunología , Femenino , Inmunofenotipificación , Inflamación/inmunología , Activación de Linfocitos/efectos de los fármacos , Masculino , Ratones , Obesidad/inmunología , Péptidos/inmunología , Fenotipo , Virosis/inmunología
8.
J Immunol ; 191(5): 2486-94, 2013 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-23885108

RESUMEN

The development of influenza A virus (IAV) vaccines capable of inducing cytotoxic CD8 T cell responses could potentially provide superior, long-term protection against multiple, heterologous strains of IAV. Although prior studies demonstrated the effectiveness of baculovirus-derived virus-like particle (VLP) vaccination in generating Ab-mediated protection, the role that CD8 T cell immunity plays in overall VLP-mediated protection is less-well understood. In this article, we demonstrate that intranasal vaccination of mice with a VLP containing the hemagglutinin and matrix 1 proteins of IAV/PR/8/34 leads to a significant increase in hemagglutinin 533-specific CD8 T cells in the lungs and protection following subsequent homologous challenge with IAV. VLP-mediated protection was significantly reduced by CD8 T cell depletion, indicating a critical role for CD8 T cells in protective immunity. Importantly, our results show that VLP vaccine-induced CD8 T cell-mediated protection is not limited to homologous IAV strains. VLP vaccination leads to an increase in protection following heterosubtypic challenge with a strain of IAV that avoids vaccine-induced neutralizing Abs but contains conserved, immunodominant CD8 T cell epitopes. Overall, our results demonstrate the ability of influenza protein-containing VLPs to prime IAV-specific CD8 T cell responses that contribute to protection from homo- and heterosubtypic IAV infections. These results further suggest that vaccination strategies focused on the development of cross-protective CD8 T cell responses may contribute to the development of "universal" IAV vaccines.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Virus de la Influenza A/inmunología , Vacunas contra la Influenza/inmunología , Infecciones por Orthomyxoviridae/inmunología , Vacunas de Partículas Similares a Virus/inmunología , Animales , Ratones , Ratones Endogámicos BALB C , Infecciones por Orthomyxoviridae/prevención & control , Proteínas de la Matriz Viral/inmunología
9.
Circ Res ; 111(9): 1190-7, 2012 Oct 12.
Artículo en Inglés | MEDLINE | ID: mdl-22904093

RESUMEN

RATIONALE: Inflammation and autonomic dysfunction contribute to the pathophysiology of hypertension. Cholinergic stimulation suppresses innate immune responses. Angiotensin II (Ang II) induces hypertension and is associated with proinflammatory immune responses. OBJECTIVE: Our goal was to define the innate immune response in a model of genetic hypertension and the influences of cholinergic stimulation and Ang II. METHODS AND RESULTS: Studies were conducted on 4- to 5-week-old prehypertensive spontaneously hypertensive rats (SHRs) and age-matched normotensive control, Wistar Kyoto (WKY) rats. Isolated splenocytes were preexposed to nicotine or Ang II before Toll-like receptor (TLR) activation. Culture supernatants were tested for cytokines (tumor necrosis factor-α, interleukin [IL]-10, and IL-6). TLR-mediated cytokine responses were most pronounced with TLR7/8 and TLR9 activation and similar between WKY rats and SHRs. Nicotine and Ang II enhanced this TLR-mediated IL-6 response in prehypertensive SHR splenocytes. In contrast, nicotine suppressed the TLR-mediated IL-6 response in WKY rats, whereas Ang II had no effect. In vivo, nicotine enhanced plasma levels of TLR7/8-mediated IL-6 and IL-1ß responses in prehypertensive SHRs but suppressed these responses in WKY rats. Flow cytometry revealed an increase in a CD161+ innate immune cell population, which was enhanced by nicotine in the prehypertensive SHR spleen but not in WKY. CONCLUSIONS: There is a pronounced anti-inflammatory nicotinic/cholinergic modulation of the innate immune system in WKY rats, which is reversed in prehypertensive SHRs. The results support the novel concept that neurohormonal regulation of the innate immune system plays a role in the pathogenesis of genetic hypertension and provide putative molecular targets for treatment of hypertension.


Asunto(s)
Hipertensión/fisiopatología , Inmunidad Innata/fisiología , Inflamación/fisiopatología , Neurotransmisores/fisiología , Prehipertensión/fisiopatología , Angiotensina II/farmacología , Animales , Células Cultivadas , Citocinas/metabolismo , Modelos Animales de Enfermedad , Macrófagos/efectos de los fármacos , Macrófagos/patología , Masculino , Nicotina/farmacología , Ratas , Ratas Endogámicas SHR , Ratas Endogámicas WKY , Bazo/efectos de los fármacos , Bazo/patología
10.
Alcohol Clin Exp Res ; 38(9): 2403-13, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25160044

RESUMEN

BACKGROUND: It is well established that chronic ethanol (EtOH) consumption is associated with increased incidence and disease severity of respiratory infections. Our recent work demonstrates this increase in disease severity to influenza A virus (IAV) infections is due, in part, to a failure to mount a robust IAV-specific CD8 T cell response along with a specific impairment in the ability of these T cells to produce interferon γ (IFNγ). However, the full extent of the lesion in the effector CD8 T cell compartment during chronic EtOH consumption remains unknown. METHODS: Utilizing the Meadows-Cook murine model of chronic alcohol consumption, mice received EtOH in their drinking water for 8 or 12 weeks. Mice were challenged intranasally with IAV, and the activation and effector functions of IAV-specific CD8 T cells were determined in both the lung-draining lymph nodes (dLN) and lungs. RESULTS: Our results confirm the defect in IFNγ production; however, the ability of IAV-specific T cells to produce tumor necrosis factor α (TNFα) and interleukin-2 (IL-2) in EtOH-consuming mice remains unaltered. In contrast, EtOH consumption significantly reduces the ability of CD8 T cells to degranulate and kill IAV-specific targets. Finally, our findings suggest the lesion begins during the initial activation of CD8 T cells, as we observe early defects in proliferation in the dLN of IAV-infected, EtOH-consuming mice. CONCLUSIONS: These findings highlight the previously unrecognized depth of the lesion in the IAV-specific CD8 T cell response during chronic EtOH consumption. Given the important role CD8 T cell immunity plays in control of IAV, these findings may aid in the development of vaccination and/or therapeutic strategies to reverse these defects in the CD8 T cell response and reduce serious disease outcomes associated with IAV infections in alcoholics.


Asunto(s)
Consumo de Bebidas Alcohólicas/metabolismo , Linfocitos T CD8-positivos/efectos de los fármacos , Linfocitos T CD8-positivos/metabolismo , Etanol/administración & dosificación , Virus de la Influenza A/efectos de los fármacos , Infecciones por Orthomyxoviridae/metabolismo , Consumo de Bebidas Alcohólicas/efectos adversos , Animales , Etanol/toxicidad , Mediadores de Inflamación/metabolismo , Virus de la Influenza A/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Transgénicos
11.
NPJ Vaccines ; 9(1): 96, 2024 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-38822003

RESUMEN

Influenza A virus (IAV) causes significant morbidity and mortality worldwide due to seasonal epidemics and periodic pandemics. The antigenic drift/shift of IAV continually gives rise to new strains and subtypes, aiding IAV in circumventing previously established immunity. As a result, there has been substantial interest in developing a broadly protective IAV vaccine that induces, durable immunity against multiple IAVs. Previously, a polyanhydride nanoparticle-based vaccine or nanovaccine (IAV-nanovax) encapsulating H1N1 IAV antigens was reported, which induced pulmonary B and T cell immunity and resulted in cross-strain protection against IAV. A key feature of IAV-nanovax is its ability to easily incorporate diverse proteins/payloads, potentially increasing its ability to provide broad protection against IAV and/or other pathogens. Due to human susceptibility to both H1N1 and H3N2 IAV, several H3N2 nanovaccines were formulated herein with multiple IAV antigens to examine the "plug-and-play" nature of the polyanhydride nanovaccine platform and determine their ability to induce humoral and cellular immunity and broad-based protection similar to IAV-nanovax. The H3N2-based IAV nanovaccine formulations induced systemic and mucosal B cell responses which were associated with antigen-specific antibodies. Additionally, systemic and lung-tissue resident CD4 and CD8 T cell responses were enhanced post-vaccination. These immune responses corresponded with protection against both homologous and heterosubtypic IAV infection. Overall, these results demonstrate the plug-and-play nature of the polyanhydride nanovaccine platform and its ability to generate immunity and protection against IAV utilizing diverse antigenic payloads.

12.
J Immunol ; 187(9): 4581-8, 2011 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-21940678

RESUMEN

An immune response of appropriate magnitude should be robust enough to control pathogen spread but not simultaneously lead to immunopathology. Primary infection with influenza A virus (IAV) results in a localized pulmonary infection and inflammation and elicits an IAV-specific CD8 T cell immune response necessary for viral clearance. Clearance of IAV-infected cells, and recovery from infection, is mediated by perforin/granzyme B- and Fas/FasL-mediated mechanisms. We recently reported that TRAIL is another means by which IAV-specific CD8 T cells can kill IAV-infected cells. The current study examined the role of TRAIL in the pulmonary CD8 T cell response to a clinically significant IAV [A/PR/8/34 (PR8; H1N1)] infection (i.e., leads to observable, but limited, morbidity and mortality in wild-type [WT] mice). Compared with WT mice, IAV-infected Trail(-/-) mice experienced increased morbidity and mortality despite similar rates of viral clearance from the lungs. The increased morbidity and mortality in Trail(-/-) mice correlated with increased pulmonary pathology and inflammatory chemokine production. Analysis of lung-infiltrating lymphocytes revealed increased numbers of IAV-specific CD8 T cells in infected Trail(-/-) mice, which correlated with increased pulmonary cytotoxic activity and increased pulmonary expression of MIG and MIP-1α. In addition, there was decreased apoptosis and increased proliferation of IAV-specific CD8 T cells in the lungs of Trail(-/-) mice compared with WT mice. Together, these data suggest that TRAIL regulates the magnitude of the IAV-specific CD8 T cell response during a clinically significant IAV infection to decrease the chance for infection-induced immunopathology.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/virología , Subtipo H1N1 del Virus de la Influenza A/inmunología , Subtipo H3N2 del Virus de la Influenza A/inmunología , Ligando Inductor de Apoptosis Relacionado con TNF/fisiología , Animales , Linfocitos T CD8-positivos/patología , Relación Dosis-Respuesta Inmunológica , Epítopos de Linfocito T/inmunología , Pulmón/inmunología , Pulmón/patología , Pulmón/virología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Infecciones por Orthomyxoviridae/inmunología , Infecciones por Orthomyxoviridae/mortalidad , Infecciones por Orthomyxoviridae/virología , Análisis de Supervivencia , Ligando Inductor de Apoptosis Relacionado con TNF/deficiencia , Ligando Inductor de Apoptosis Relacionado con TNF/genética
13.
Proc Natl Acad Sci U S A ; 107(27): 12198-203, 2010 Jul 06.
Artículo en Inglés | MEDLINE | ID: mdl-20616089

RESUMEN

The number of memory CD8 T cells generated by infection or vaccination correlates strongly with the degree of protection observed in infection and tumor models. Therefore, rapid induction of protective numbers of effector and memory CD8 T cells may be crucial in the case of malignancy, pandemic infection, or bioterrorism. Many studies have shown that amplifying T-cell numbers by prime-boost vaccination is most effective with a substantial time interval between immunizations. In contrast, immunization with peptide-coated mature dendritic cells (DCs) results in a CD8 T-cell response exhibiting accelerated acquisition of memory characteristics, including the ability to respond to booster immunization within days of initial priming. However, personalized DC immunization is too costly, labor intensive, and time-consuming for large-scale vaccination. Here, we demonstrate that in vivo cross-priming with cell-associated antigens or antigen-coated, biodegradable microspheres in the absence of adjuvant quickly generates CD8 T cells that display the phenotype and function of long-term memory populations. Importantly, cross-primed CD8 T cells can respond to booster immunization within days of the initial immunization to generate rapidly large numbers of effector and memory T cells that can protect against bacterial, viral, and parasitic infections, including lethal influenza and malaria-causing Plasmodium infection. Thus, accelerated CD8 T-cell memory after in vivo cross-priming in the absence of adjuvant is generalizable and can be exploited to generate protective immunity rapidly.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Reactividad Cruzada/inmunología , Inmunidad/inmunología , Memoria Inmunológica/inmunología , Animales , Linfocitos T CD8-positivos/citología , Linfocitos T CD8-positivos/metabolismo , Células Dendríticas/inmunología , Inmunización/métodos , Inmunización Secundaria , Leucocitos Mononucleares/inmunología , Listeria monocytogenes/inmunología , Listeriosis/inmunología , Listeriosis/microbiología , Malaria/inmunología , Malaria/parasitología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Transgénicos , Modelos Inmunológicos , Ovalbúmina/inmunología , Plasmodium berghei/inmunología , Factores de Tiempo
14.
ACS Nano ; 17(15): 14586-14603, 2023 08 08.
Artículo en Inglés | MEDLINE | ID: mdl-37463491

RESUMEN

It has been shown that inhalation exposure to copper oxide nanoparticles (CuO NPs) results in pulmonary inflammation. However, immunomodulatory consequences after CuO NP inhalation exposure have been less explored. We tested the effect of CuO NP aerosols on immune responses in healthy, house dust mite (HDM) asthmatic, or allergen immunotherapy (AIT)-treated asthmatic mice (BALB/c, females). The AIT consisted of a vaccine comprising HDM allergens and CpG-loaded nanoparticles (CpG NPs). AIT treatment involved mice being immunized (via subcutaneous (sc) injection; 2 doses) while concomitantly being exposed to CuO NP aerosols (over a 2 week period), starting on the day of the first vaccination. Mice were then sensitized twice by sc injection and subsequently challenged with HDM extract 10 times by intranasal instillation. The asthmatic model followed the same timeline except that no immunizations were administered. All mice were necropsied 24 h after the end of the HDM challenge. CuO NP-exposed healthy mice showed a significant decrease in TH1 and TH2 cells, and an elevation in T-bet+ Treg cells, even 40 days after the last exposure to CuO NPs. Similarly, the CuO NP-exposed HDM asthma model demonstrated decreased TH2 responses and increased T-bet+ Treg cells. Conversely, CuO NP inhalation exposure to AIT-treated asthmatic mice resulted in an increase in TH2 cells. In conclusion, immunomodulatory effects of inhalation exposure to CuO NPs are dependent on immune conditions prior to exposure.


Asunto(s)
Asma , Nanopartículas , Femenino , Ratones , Animales , Cobre , Exposición por Inhalación , Asma/inducido químicamente , Asma/terapia , Pyroglyphidae , Inmunidad , Óxidos
15.
Viruses ; 15(6)2023 06 12.
Artículo en Inglés | MEDLINE | ID: mdl-37376652

RESUMEN

Macrophages are critical in the pathogenesis of a diverse group of viral pathogens, both as targets of infection and for eliciting primary defense mechanisms. Our prior in vitro work identified that CD40 signaling in murine peritoneal macrophages protects against several RNA viruses by eliciting IL-12, which stimulates the production of interferon gamma (IFN-γ). Here, we examine the role of CD40 signaling in vivo. We show that CD40 signaling is a critical, but currently poorly appreciated, component of the innate immune response using two distinct infectious agents: mouse-adapted influenza A virus (IAV, PR8) and recombinant VSV encoding the Ebola virus glycoprotein (rVSV-EBOV GP). We find that stimulation of CD40 signaling decreases early IAV titers, whereas loss of CD40 elevated early titers and compromised lung function by day 3 of infection. Protection conferred by CD40 signaling against IAV is dependent on IFN-γ production, consistent with our in vitro studies. Using rVSV-EBOV GP that serves as a low-biocontainment model of filovirus infection, we demonstrate that macrophages are a CD40-expressing population critical for protection within the peritoneum and T-cells are the key source of CD40L (CD154). These experiments reveal the in vivo mechanisms by which CD40 signaling in macrophages regulates the early host responses to RNA virus infection and highlight how CD40 agonists currently under investigation for clinical use may function as a novel class of broad antiviral treatments.


Asunto(s)
Antígenos CD40 , Infecciones por Virus ARN , Virus ARN , Animales , Ratones , Antígenos CD40/metabolismo , Interferón gamma , Macrófagos , Infecciones por Virus ARN/inmunología
16.
J Immunol ; 184(8): 4440-6, 2010 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-20220091

RESUMEN

Previous studies have shown that the reduction in CD8 T cell immunity observed during high-dose influenza A virus (IAV) infection is mediated via lymph node (LN) dendritic cells (DCs) that express Fas ligand (FasL) and drive FasL-Fas (DC-T)-induced apoptosis. However, the specific DC subset(s) within the LN and the additional factors required for DC-mediated elimination of IAV-specific CD8 T cells remain unknown. In this paper, we demonstrate that plasmacytoid DCs (pDCs), which downregulate FasL during sublethal, but not lethal, IAV infection, accumulate to greater numbers within the LNs of lethal dose-infected mice. Further our findings show that pDCs from lethal, but not sublethal, dose IAV infections drive elimination of Fas(+) CD8 T cells and that this elimination occurs only in the absence of TCR recognition of IAV peptide-MHC class I complexes. Together, these results suggest that pDCs play a heretofore unknown deleterious role during lethal dose IAV infections by limiting the CD8 T cell response.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/patología , Células Dendríticas/inmunología , Depleción Linfocítica , Infecciones por Orthomyxoviridae/inmunología , Infecciones por Orthomyxoviridae/mortalidad , Traslado Adoptivo , Animales , Apoptosis/inmunología , Antígenos CD8/biosíntesis , Linfocitos T CD8-positivos/metabolismo , Movimiento Celular/inmunología , Células Dendríticas/metabolismo , Células Dendríticas/trasplante , Regulación hacia Abajo/inmunología , Proteína Ligando Fas/antagonistas & inhibidores , Proteína Ligando Fas/biosíntesis , Proteína Ligando Fas/fisiología , Inmunofenotipificación , Subtipo H1N1 del Virus de la Influenza A/inmunología , Subtipo H2N2 del Virus de la Influenza A/inmunología , Ganglios Linfáticos/inmunología , Ganglios Linfáticos/patología , Ganglios Linfáticos/virología , Activación de Linfocitos/inmunología , Recuento de Linfocitos , Depleción Linfocítica/métodos , Ratones , Ratones Endogámicos BALB C , Ratones Transgénicos , Infecciones por Orthomyxoviridae/patología , Receptor fas/biosíntesis , Receptor fas/fisiología
17.
Immunology ; 133(4): 452-68, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21635248

RESUMEN

Germinal centre (GC) reactions are central features of T-cell-driven B-cell responses, and the site where antibody-producing cells and memory B cells are generated. Within GCs, a range of complex cellular and molecular events occur which are critical for the generation of high affinity antibodies. These processes require exquisite regulation not only to ensure the production of desired antibodies, but to minimize unwanted autoreactive or low affinity antibodies. To assess whether T regulatory (Treg) cells participate in the control of GC responses, immunized mice were treated with an anti-glucocorticoid-induced tumour necrosis factor receptor-related protein (GITR) monoclonal antibody (mAb) to disrupt Treg-cell activity. In anti-GITR-treated mice, the GC B-cell pool was significantly larger compared with control-treated animals, with switched GC B cells composing an abnormally high proportion of the response. Dysregulated GCs were also observed regardless of strain, T helper type 1 or 2 polarizing antigens, and were also seen after anti-CD25 mAb treatment. Within the spleens of immunized mice, CXCR5(+) and CCR7(-) Treg cells were documented by flow cytometry and Foxp3(+) cells were found within GCs using immunohistology. Final studies demonstrated administration of either anti-transforming growth factor-ß or anti-interleukin-10 receptor blocking mAb to likewise result in dysregulated GCs, suggesting that generation of inducible Treg cells is important in controlling the GC response. Taken together, these findings indicate that Treg cells contribute to the overall size and quality of the humoral response by controlling homeostasis within GCs.


Asunto(s)
Centro Germinal/inmunología , Linfocitos T Reguladores/inmunología , Animales , Centro Germinal/citología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Transgénicos , Linfocitos T Reguladores/citología
18.
Alcohol Clin Exp Res ; 35(1): 47-59, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21039629

RESUMEN

BACKGROUND: As initiators of immune responses, dendritic cells (DCs) are required for antigen (Ag)-specific activation of naïve T cells in the defense against infectious agents. The increased susceptibility to and severity of infection seen in chronic alcoholics could be because of impaired DCs initiation of naïve T-cell responses. Specifically, these DCs may not provide adequate Signals 1 (Ag presentation), 2 (costimulation), or 3 (cytokine production) to these T cells. METHODS: Using the Meadows-Cook murine model of chronic alcohol abuse, the ability of ethanol (EtOH)-exposed DCs to stimulate T-cell proliferation, acquire and process Ag, express costimulatory molecules, and produce inflammatory cytokines was assessed. RESULTS: Normal naïve T cells primed by EtOH-exposed DCs showed decreased proliferation in vitro and in vivo, compared to water-fed control mice. These EtOH-exposed DCs, after activation by CpG or tumor necrosis factor alpha (TNFα), were less able to upregulate costimulatory molecules CD40, CD80, or CD86, and produced less IL-12 p40, TNFα, and IFNα than DCs from water-fed mice. TLR9 and TNF receptor expression were also reduced in/on EtOH-exposed DCs. No evidence of defective Ag acquisition or processing as a result of EtOH feeding was identified. CONCLUSIONS: Inadequate proliferation of normal T cells following stimulation by EtOH-exposed DCs is likely a result of diminished Signal 2 and Signal 3. Lack of adequate inflammatory stimulation of EtOH-exposed DCs because of diminished receptors for inflammatory mediators appears to be at least partially responsible for their dysfunction. These findings provide a mechanism to explain increased morbidity and mortality from infectious diseases in alcoholics and suggest targets for therapeutic intervention.


Asunto(s)
Alcoholismo/inmunología , Células Dendríticas/inmunología , Etanol/administración & dosificación , Activación de Linfocitos , Linfocitos T/inmunología , Animales , Presentación de Antígeno , Antígenos CD/inmunología , Diferenciación Celular , Cricetinae , Citocinas/inmunología , Citocinas/metabolismo , Modelos Animales de Enfermedad , Femenino , Ratones , Ratones Endogámicos C57BL , Ratas , Receptores del Factor de Necrosis Tumoral/metabolismo , Bazo/citología , Bazo/inmunología , Linfocitos T/metabolismo , Factores de Tiempo , Receptor Toll-Like 9/metabolismo
19.
J Immunol ; 183(7): 4177-81, 2009 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-19767567

RESUMEN

Following influenza virus infection, CD8 T cells encounter mature, Ag-bearing dendritic cells within the draining lymph nodes and undergo activation, programmed proliferation, and differentiation to effector cells before migrating to the lungs to mediate viral clearance. However, it remains unclear whether CD8 T cells continue their proliferation after arriving in the lungs. To address this question, we developed a novel, in vivo, dual-label system using intranasal CFSE and BrdU administration to identify virus-specific CD8 T cells that are actively undergoing cell division while in the lungs. With this technique we demonstrate that a high frequency of virus-specific CD8 T cells incorporate BrdU while in the lungs and that this lung-resident proliferation contributes significantly to the magnitude of the Ag-specific CD8 T cell response following influenza virus infection.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/virología , Proliferación Celular , Epítopos de Linfocito T/inmunología , Subtipo H1N1 del Virus de la Influenza A/inmunología , Pulmón/inmunología , Pulmón/virología , Infecciones por Orthomyxoviridae/inmunología , Animales , Linfocitos T CD8-positivos/metabolismo , Movimiento Celular/inmunología , Femenino , Clorhidrato de Fingolimod , Pulmón/patología , Ratones , Ratones Endogámicos BALB C , Infecciones por Orthomyxoviridae/patología , Infecciones por Orthomyxoviridae/virología , Glicoles de Propileno/farmacología , Esfingosina/análogos & derivados , Esfingosina/farmacología
20.
J Immunol ; 182(12): 7803-8, 2009 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-19494304

RESUMEN

Alcohol use by pregnant women is a significant public health issue despite well-described risks to the fetus including physical and intellectual growth retardation and malformations. Although clinical studies are limited, they suggest that in utero alcohol exposure also results in significant immune deficiencies in naive neonates. However, little is known about fetal alcohol exposure (FAE) effects on adult infections. Therefore, to determine the long-term effects of FAE on disease susceptibility and the adult immune system, we infected FAE adult mice with influenza virus. In this study, we demonstrate that mice exposed to ethanol during gestation and nursing exhibit enhanced disease severity as well as increased and sustained pulmonary viral titers following influenza virus infection. Secondary exposure to alcohol as an adult further exacerbates these effects. Moreover, we demonstrate that FAE mice have impaired adaptive immune responses, including decreased numbers of virus-specific pulmonary CD8 T cells, a decreased size and frequency of pulmonary B cell foci, and reduced production of influenza-specific Ab following influenza infection. Together, our results suggest that FAE induces significant and long-term defects in immunity and susceptibility to influenza virus infection and that FAE individuals could be at increased risk for severe and fatal respiratory infections.


Asunto(s)
Etanol/farmacología , Trastornos del Espectro Alcohólico Fetal/inmunología , Subtipo H1N1 del Virus de la Influenza A/inmunología , Subtipo H2N2 del Virus de la Influenza A/inmunología , Infecciones por Orthomyxoviridae/inmunología , Infecciones por Orthomyxoviridae/patología , Efectos Tardíos de la Exposición Prenatal , Animales , Linfocitos B/citología , Linfocitos B/inmunología , Modelos Animales de Enfermedad , Femenino , Recuento de Linfocitos , Ratones , Infecciones por Orthomyxoviridae/complicaciones , Infecciones por Orthomyxoviridae/virología , Embarazo , Tasa de Supervivencia , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA