Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Ann Behav Med ; 54(2): 87-93, 2020 01 24.
Artículo en Inglés | MEDLINE | ID: mdl-31282543

RESUMEN

BACKGROUND: Previous research documents an association between adverse childhood experiences (ACEs) and immune system inflammation. High chronic inflammation is believed to be one biological pathway through which childhood adversity may affect health into adulthood. The Blackfeet tribal community has high rates of childhood trauma and community members are disproportionately affected by inflammatory diseases. PURPOSE: To investigate whether belonging to the tribal community may moderate the relationship between childhood trauma and immune system inflammation in the Blackfeet tribal community. METHODS: In a sample of 90 adults residing on the Blackfeet reservation, we measured ACEs belonging to the tribal community and two markers of immune system inflammation, interleukin-6 (IL-6) and C-reactive protein (CRP). RESULTS: We found that independent of age, gender, annual income, body mass index, and depressive symptoms, belonging to the tribal community and ACEs interacted to predict levels of both IL-6 and CRP (B= -.37, t[81] = -3.82, p < .001, R2 change = .07 and B = -.29, t[81] = -2.75, p = .01, R2 change = .08, respectively). The association between ACEs and markers of immune system inflammation was statistically significant for community members who reported low levels of belonging to the community. CONCLUSIONS: The findings of this study have important implications for intervention research seeking to reduce risk for inflammatory diseases for at-risk populations. Fostering stronger connections to the larger tribal community may positively affect risk for inflammatory diseases. Future work should examine the behavioral and psychosocial pathways through which stronger connections to community may confer health benefits.


Asunto(s)
Experiencias Adversas de la Infancia/etnología , Indígenas Norteamericanos/etnología , Inflamación/etnología , Trauma Psicológico/etnología , Medio Social , Adulto , Proteína C-Reactiva/metabolismo , Enfermedad Crónica/etnología , Femenino , Humanos , Inflamación/sangre , Inflamación/inmunología , Interleucina-6/sangre , Masculino , Montana/etnología , Factores Protectores , Factores de Riesgo
2.
Am J Community Psychol ; 64(1-2): 118-125, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31290568

RESUMEN

American Indian (AI) communities have high levels of stress and trauma and are disproportionately affected by numerous preventable diseases. Here, we describe an academic-community partnership based on a collaboration between Blackfeet Community College students and faculty in Psychology and Immunology at Montana State University (MSU). The collaboration, which has spanned over 5 years, was sparked by community interest in the relationship between stress and disease on the Blackfeet reservation. Specifically, community members wanted to understand how the experience of psychological stress and trauma may affect disease risk in their community and identify factors that promote resilience. In doing so, they hoped to identify pathways through which health could be improved for individual community members. Here, we discuss all stages of the collaborative process, including development of measures and methods and themes of research projects, challenges for community members and non-indigenous collaborators, future directions for research, and the lessons learned. Finally, we note the ways in which this partnership and experience has advanced the science of community engagement in tribal communities, with the hope that our experiences will positively affect future collaborations between indigenous community members and non-indigenous scientists.


Asunto(s)
Investigación Biomédica/organización & administración , Investigación Participativa Basada en la Comunidad/métodos , Indígenas Norteamericanos , Investigación Participativa Basada en la Comunidad/organización & administración , Relaciones Comunidad-Institución , Estado de Salud , Humanos , Montana , Desarrollo de Programa , Estrés Psicológico/complicaciones , Estrés Psicológico/etnología , Universidades
3.
J Immunol ; 196(12): 5036-46, 2016 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-27194787

RESUMEN

Regulatory T cells (Tregs) induced during autoimmunity often become quiescent and unable to resolve disease, suggesting inadequate activation. Resolution of established experimental autoimmune encephalomyelitis (EAE) can be achieved with myelin oligodendrocyte glycoprotein (MOG) fused to reovirus protein σ1 (MOG-pσ1), which activates Tregs, restoring protection, but requiring other regulatory cells to revitalize them. B cells have a dichotomous role in both the pathogenesis and recovery from EAE. Although inflammatory B cells contribute to EAE's pathogenesis, treatment of EAE mice with MOG-pσ1, but not OVA-pσ1, resulted in an influx of IL-10-producing B220(+)CD5(+) B regulatory cells (Bregs) enabling Tregs to recover their inhibitory activity, and in turn, leading to the rapid amelioration of EAE. These findings implicate direct interactions between Bregs and Tregs to facilitate this recovery. Adoptive transfer of B220(+)CD5(-) B cells from MOG-pσ1-treated EAE or Bregs from PBS-treated EAE mice did not resolve disease, whereas the adoptive transfer of MOG-pσ1-induced B220(+)CD5(+) Bregs greatly ameliorated EAE. MOG-pσ1-, but not OVA-pσ1-induced IL-10-producing Bregs, expressed elevated levels of B and T lymphocyte attenuator (BTLA) relative to CD5(-) B cells, as opposed to Tregs or effector T (Teff) cells, whose BTLA expression was not affected. These induced Bregs restored EAE Treg function in a BTLA-dependent manner. BTLA(-/-) mice showed more pronounced EAE with fewer Tregs, but upon adoptive transfer of MOG-pσ1-induced BTLA(+) Bregs, BTLA(-/-) mice were protected against EAE. Hence, this evidence shows the importance of BTLA in activating Tregs to facilitate recovery from EAE.


Asunto(s)
Linfocitos B Reguladores/inmunología , Encefalomielitis Autoinmune Experimental/inmunología , Receptores Inmunológicos/inmunología , Linfocitos T Reguladores/inmunología , Traslado Adoptivo , Animales , Linfocitos B Reguladores/efectos de los fármacos , Linfocitos B Reguladores/fisiología , Antígenos CD5/genética , Antígenos CD5/inmunología , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/fisiopatología , Interleucina-10/biosíntesis , Interleucina-10/inmunología , Antígenos Comunes de Leucocito/genética , Antígenos Comunes de Leucocito/inmunología , Activación de Linfocitos , Ratones , Ratones Endogámicos C57BL , Glicoproteína Mielina-Oligodendrócito/administración & dosificación , Glicoproteína Mielina-Oligodendrócito/inmunología , Receptores Inmunológicos/deficiencia , Receptores Inmunológicos/genética , Linfocitos T Reguladores/patología , Linfocitos T Reguladores/fisiología
4.
Infect Immun ; 83(10): 3764-70, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26216421

RESUMEN

Seasonal influenza virus infection presents a major strain on the health care system. Influenza virus infection has pandemic potential, which was repeatedly observed during the last century. Severe disease may occur in the young, in the elderly, in those with preexisting lung disease, and in previously healthy individuals. A common cause of severe influenza pathogenesis is superinfection with bacterial pathogens, namely, Staphylococcus aureus and Streptococcus pneumoniae. A great deal of recent research has focused on the immune pathways involved in influenza-induced susceptibility to secondary bacterial pneumonia. Both innate and adaptive antibacterial host defenses are impaired in the context of preceding influenza virus infection. The goal of this minireview is to highlight these findings and synthesize these data into a shared central theme of pathogenesis.


Asunto(s)
Gripe Humana/complicaciones , Infecciones Estafilocócicas/inmunología , Sobreinfección/inmunología , Animales , Humanos , Virus de la Influenza A/fisiología , Gripe Humana/virología , Infecciones Estafilocócicas/microbiología , Staphylococcus aureus/fisiología , Streptococcus pneumoniae/fisiología , Sobreinfección/microbiología
5.
Eur J Immunol ; 44(2): 397-408, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24222381

RESUMEN

Recent evidence suggests that an individual's unique history and sequence of exposures to pathogens and antigens may dictate downstream immune responses to disparate antigens. We show that the i.n. delivery of nonreplicative virus-like particles (VLPs), which bear structural but no antigenic similarities to respiratory pathogens, acts to prime the lungs of both C56BL/6 and BALB/c mice, facilitating heightened and accelerated primary immune responses to high-dose influenza challenge, thus providing a nonpathogenic model of innate imprinting. These responses correspond closely to those observed following natural infection with the opportunistic fungus, Pneumocystis murina, and are characterized by accelerated antigen processing by DCs and alveolar macrophages, an enhanced influx of cells to the local tracheobronchial lymph node, and early upregulation of T-cell co-stimulatory/adhesion molecules. CD11c⁺ cells, which have been directly exposed to VLPs or Pneumocystis are necessary in facilitating enhanced clearance of influenza virus, and the repopulation of the lung by Ly-6C⁺ precursors relies on CCR2 expression. Thus, immune imprinting 72 h after VLP-priming, or 2 weeks after Pneumocystis-priming is CCR2-mediated and results from the enhanced antigen processing, maturation, and trafficking abilities of DCs and alveolar macrophages, which cause accelerated influenza-specific primary immune responses and result in superior viral clearance.


Asunto(s)
Antígenos/inmunología , Antígeno CD11c/inmunología , Inmunidad Innata/inmunología , Infecciones por Orthomyxoviridae/inmunología , Orthomyxoviridae/inmunología , Animales , Presentación de Antígeno/inmunología , Antígenos Ly/inmunología , Células Dendríticas/inmunología , Células Dendríticas/microbiología , Células Dendríticas/virología , Pulmón/inmunología , Pulmón/microbiología , Pulmón/virología , Ganglios Linfáticos/inmunología , Ganglios Linfáticos/microbiología , Ganglios Linfáticos/virología , Macrófagos Alveolares/inmunología , Macrófagos Alveolares/microbiología , Macrófagos Alveolares/virología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Infecciones por Orthomyxoviridae/prevención & control , Infecciones por Orthomyxoviridae/virología , Pneumocystis/inmunología , Infecciones por Pneumocystis/inmunología , Receptores CCR2/inmunología , Linfocitos T/inmunología , Linfocitos T/microbiología , Linfocitos T/virología , Regulación hacia Arriba/inmunología , Vacunas de Partículas Similares a Virus/inmunología
6.
Eur J Immunol ; 44(11): 3263-72, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25091976

RESUMEN

Superinfection in mice at day 7 postinfluenza infection exacerbates bacterial pneumonia at least in part via downstream effects of increased IFN-γ signaling. Here we show that up to 3 days postinfluenza infection, mice have reduced susceptibility to superinfection with methicillin-resistant Staphylococcus aureus (MRSA), but that superinfection during that time exacerbated influenza disease. This was due to IL-13 signaling that was advantageous for resolving MRSA infection via inhibition of IFN-γ, but was detrimental to the clearance of influenza virus. However, if superinfection did not occur until the near resolution of influenza infection (day 7), IL-13 signaling was inhibited, at least in part by upregulation of IL-13 decoy receptor (IL-13Rα2), which in turn caused increases in IFN-γ signaling and exacerbation of bacterial infection. Understanding these cytokine sequelae is critical to development of immunotherapies for influenza-MRSA coinfection since perturbations of these sequelae at the wrong time could increase susceptibility to MRSA and/or influenza.


Asunto(s)
Interferón gamma/genética , Subunidad alfa2 del Receptor de Interleucina-13/inmunología , Interleucina-13/inmunología , Staphylococcus aureus Resistente a Meticilina/inmunología , Infecciones por Orthomyxoviridae/inmunología , Infecciones Estafilocócicas/inmunología , Sobreinfección/inmunología , Animales , Carga Bacteriana , Coinfección , Susceptibilidad a Enfermedades , Interleucina-13/genética , Interleucina-13/farmacología , Subunidad alfa2 del Receptor de Interleucina-13/antagonistas & inhibidores , Subunidad alfa2 del Receptor de Interleucina-13/biosíntesis , Pulmón/microbiología , Pulmón/virología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Neumonía Bacteriana/inmunología , Proteínas Recombinantes/farmacología , Sobreinfección/microbiología , Carga Viral
7.
J Immunol ; 188(12): 6389-98, 2012 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-22586039

RESUMEN

The infiltration of suppressive myeloid cells into the tumor microenvironment restrains anti-tumor immunity. However, cytokines may alter the function of myeloid lineage cells to support tumor rejection, regulating the balance between pro- and anti-tumor immunity. In this study, it is shown that effector cytokines secreted by adoptively transferred T cells expressing a chimeric Ag receptor (CAR) shape the function of myeloid cells to promote endogenous immunity and tumor destruction. Mice bearing the ovarian ID8 tumor were treated with T cells transduced with a chimeric NKG2D receptor. GM-CSF secreted by the adoptively transferred T cells recruited peripheral F4/80(lo)Ly-6C(+) myeloid cells to the tumor microenvironment in a CCR2-dependent fashion. T cell IFN-γ and GM-CSF activated local, tumor-associated macrophages, decreased expression of regulatory factors, increased IL-12p40 production, and augmented Ag processing and presentation by host macrophages to Ag-specific T cells. In addition, T cell-derived IFN-γ, but not GM-CSF, induced the production of NO by F4/80(hi) macrophages and enhanced their lysis of tumor cells. The ability of CAR T cell therapy to eliminate tumor was moderately impaired when inducible NO synthase was inhibited and greatly impaired in the absence of peritoneal macrophages after depletion with clodronate encapsulated liposomes. This study demonstrates that the activation of host macrophages by CAR T cell-derived cytokines transformed the tumor microenvironment from immunosuppressive to immunostimulatory and contributed to inhibition of ovarian tumor growth.


Asunto(s)
Factor Estimulante de Colonias de Granulocitos y Macrófagos/inmunología , Interferón gamma/inmunología , Macrófagos/inmunología , Neoplasias Experimentales/inmunología , Receptores de Antígenos de Linfocitos T/inmunología , Microambiente Tumoral/inmunología , Traslado Adoptivo , Animales , Presentación de Antígeno/inmunología , Quimera , Citometría de Flujo , Linfocitos Infiltrantes de Tumor/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Células Mieloides/inmunología , Subfamilia K de Receptores Similares a Lectina de Células NK/inmunología , Reacción en Cadena en Tiempo Real de la Polimerasa , Linfocitos T/inmunología , Linfocitos T/trasplante
8.
Viruses ; 16(6)2024 May 29.
Artículo en Inglés | MEDLINE | ID: mdl-38932288

RESUMEN

The Viruses Editorial Office retracts the article, "Contribution of Host Immune Responses Against Influenza D Virus Infection Toward Secondary Bacterial Infection in a Mouse Model" [...].

9.
Immunol Cell Biol ; 91(6): 435-40, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23628805

RESUMEN

Tumor heterogeneity presents a substantial barrier to increasing clinical responses mediated by targeted therapies. Broadening the immune response elicited by treatments that target a single antigen is necessary for the elimination of tumor variants that fail to express the targeted antigen. In this study, it is shown that adoptive transfer of T cells bearing a chimeric antigen receptor (CAR) inhibited the growth of target-expressing and -deficient tumor cells within ovarian and lymphoma tumors. Mice bearing the ID8 ovarian or RMA lymphoma tumors were treated with T cells transduced with a NKG2D-based CAR (chNKG2D). NKG2D CAR T-cell therapy protected mice from heterogeneous RMA tumors. Moreover, adoptive transfer of chNKG2D T cells mediated tumor protection against highly heterogeneous ovarian tumors in which 50, 20 or only 7% of tumor cells expressed significant amounts of NKG2D ligands. CAR T cells did not mediate an in vivo response against tumor cells that did not express sufficient amounts of NKG2D ligands, and the number of ligand-expressing tumor cells correlated with therapeutic efficacy. In addition, tumor-free surviving mice were protected against a tumor re-challenge with NKG2D ligand-negative ovarian tumor cells. These data indicate that NKG2D CAR T-cell treatment can be an effective therapy against heterogeneous tumors and induce tumor-specific immunity against ligand-deficient tumor cells.


Asunto(s)
Inmunoterapia Adoptiva/métodos , Linfoma/terapia , Subfamilia K de Receptores Similares a Lectina de Células NK/metabolismo , Células T Asesinas Naturales/inmunología , Neoplasias Ováricas/terapia , Proteínas Recombinantes de Fusión/metabolismo , Animales , Procesos de Crecimiento Celular/inmunología , Línea Celular Tumoral , Citotoxicidad Inmunológica , Femenino , Humanos , Interferón gamma/metabolismo , Linfoma/inmunología , Ratones , Ratones Endogámicos C57BL , Subfamilia K de Receptores Similares a Lectina de Células NK/genética , Células T Asesinas Naturales/trasplante , Neoplasias Ováricas/inmunología , Proteínas Recombinantes de Fusión/genética
10.
Am J Pathol ; 181(1): 196-210, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22642909

RESUMEN

The importance of the priming of the lung environment by past infections is being increasingly recognized. Exposure to any given antigen can either improve or worsen the outcome of subsequent lung infections, depending on the immunological history of the host. Thus, an ability to impart transient alterations in the lung environment in anticipation of future insult could provide an important novel therapy for emerging infectious diseases. In this study, we show that nasal administration of virus-like particles (VLPs) before, or immediately after, lethal challenge with methicillin-resistant Staphylococcus aureus (MRSA) of mice i) ensures complete recovery from lung infection and near absolute clearance of bacteria within 12 hours of challenge, ii) reduces host response-induced lung tissue damage, iii) promotes recruitment and efficient bacterial clearance by neutrophils and CD11c(+) cells, and iv) protects macrophages from MRSA-induced necrosis. VLP-mediated protection against MRSA relied on innate immunity. Complete recovery occurred in VLP-dosed mice with severe combined immunodeficiency, but not in wild-type mice depleted of either Ly6G(+) or CD11c(+) cells. Early IL-13 production associated with VLP-induced CD11c(+) cells was essential for VLP-induced protection. These results indicate that VLP-induced alteration of the lung environment protects the host from lethal MRSA pneumonia by enhancing phagocyte recruitment and killing and by reducing inflammation-induced tissue damage via IL-13-dependent mechanisms.


Asunto(s)
Staphylococcus aureus Resistente a Meticilina/aislamiento & purificación , Fagocitos/inmunología , Neumonía Estafilocócica/prevención & control , Vacunas Estafilocócicas/inmunología , Vacunas de Partículas Similares a Virus/inmunología , Inmunidad Adaptativa , Administración Intranasal , Animales , Carga Bacteriana , Antígeno CD11c/análisis , Células Dendríticas/inmunología , Femenino , Inmunofenotipificación , Interleucina-13/inmunología , Pulmón/microbiología , Macrófagos Alveolares/inmunología , Masculino , Ratones , Ratones Endogámicos , Ratones Noqueados , Monocitos/inmunología , Neutrófilos/inmunología , Fagocitosis/inmunología , Neumonía Estafilocócica/inmunología , Neumonía Estafilocócica/patología , Vacunas Estafilocócicas/administración & dosificación , Factores de Tiempo , Vacunas de Partículas Similares a Virus/administración & dosificación
11.
bioRxiv ; 2023 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-38077027

RESUMEN

Land-use change may drive viral spillover from bats into humans, partly through dietary shifts caused by decreased availability of native foods and increased availability of cultivated foods. We manipulated diets of Jamaican fruit bats to investigate whether diet influences shedding of a virus they naturally host. To reflect dietary changes experienced by wild bats during periods of nutritional stress, bats were fed either standard or putative suboptimal diets which were deprived of protein (suboptimal-sugar) and/or supplemented with fat (suboptimal-fat). Upon H18N11 influenza A-virus infection, bats fed the suboptimal-sugar diet shed the most viral RNA for the longest period, but bats fed the suboptimal-fat diet shed the least viral RNA for the shortest period. Unlike mice and humans, bats fed the suboptimal-fat diet displayed higher pre-infection levels of metabolic markers associated with gut health. Diet-driven heterogeneity in viral shedding may influence population-level viral dynamics in wild bats and alter risk of shedding and spillover to humans.

12.
Eur J Immunol ; 41(2): 313-23, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21268002

RESUMEN

Stimulation of Ag-specific inducible Treg can enhance resolution of autoimmune disease. Conventional methods to induce Treg often require induction of autoimmune disease or subjection to infection. Reovirus adhesin, protein σ1 (pσ1), can successfully facilitate tolerance when fused to a tolerogen. We tested whether myelin oligodendrocyte glycoprotein (MOG) fused to pσ1 (MOG-pσ1) can stimulate Ag-specific Treg. We show that C57BL/6 mice treated nasally with MOG-pσ1 fail to induce MOG-specific Abs and delayed-type hypersensitivity (DTH) responses and resist EAE. Such resistance was attributed to stimulation of Foxp3(+) Treg, as well as Th2 cells. MOG-pσ1's protective capacity was abrogated in IL-10(-/-) mice, but restored when adoptively transferred with MOG-pσ1-induced Treg. As a therapeutic, MOG-pσ1 diminished EAE within 24 h of nasal application, unlike recombinant MOG (rMOG), pσ1, or pσ1+rMOG, implicating the importance of Ag specificity by pσ1-based therapeutics. MOG-pσ1-treated mice showed elevated IL-4, IL-10, and IL-28 production by CD4(+) T cells, unlike rMOG treated or control mice that produced elevated IFN-γ or IL-17, respectively. These data show the feasibility of using pσ1 as a tolerogen platform for Ag-specific tolerance induction and highlight its potential use as an immunotherapeutic for autoimmunity.


Asunto(s)
Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/terapia , Interleucina-10/inmunología , Linfocitos T Reguladores/inmunología , Vacunación/métodos , Traslado Adoptivo , Animales , Proteínas de la Cápside/administración & dosificación , Proteínas de la Cápside/genética , Proteínas de la Cápside/inmunología , Recuento de Células , Citocinas/metabolismo , Encefalomielitis Autoinmune Experimental/diagnóstico , Femenino , Glicoproteínas/inmunología , Hipersensibilidad Tardía/inmunología , Tolerancia Inmunológica/inmunología , Inmunoglobulina G/sangre , Inmunoglobulina G/inmunología , Interleucina-10/genética , Subunidad alfa del Receptor de Interleucina-2/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas de la Mielina , Glicoproteína Asociada a Mielina/administración & dosificación , Glicoproteína Asociada a Mielina/genética , Glicoproteína Asociada a Mielina/inmunología , Glicoproteína Mielina-Oligodendrócito , Fragmentos de Péptidos/inmunología , Proteínas Recombinantes de Fusión/administración & dosificación , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/inmunología , Médula Espinal/citología , Médula Espinal/inmunología , Bazo/citología , Bazo/inmunología , Linfocitos T Reguladores/citología , Linfocitos T Reguladores/metabolismo , Linfocitos T Reguladores/trasplante , Células Th2/inmunología , Células Th2/metabolismo , Vacunas Sintéticas/administración & dosificación , Vacunas Sintéticas/genética , Vacunas Sintéticas/inmunología
13.
Viruses ; 14(7)2022 06 28.
Artículo en Inglés | MEDLINE | ID: mdl-35891403

RESUMEN

Polymicrobial pneumonias occur frequently in cattle, swine, and sheep, resulting in major economic losses. Individual pathogens comprising these complex infections may be mild on their own but can instead exhibit synergism or increase host susceptibility. Two examples of such pathogens, Mycoplasma ovipneumoniae (M. ovipneumoniae) and influenza D viruses (IDVs), naturally infect domestic sheep. In sheep, the role of M. ovipneumoniae in chronic nonprogressive pneumonia is well-established, but the pathogenesis of IDV infection has not previously been studied. We utilized a specific-pathogen-free sheep flock to study the clinical response to IDV infection in naïve vs. M. ovipneumoniae-exposed lambs. Lambs were inoculated intranasally with M. ovipneumoniae or mock infection, followed after four weeks by infection with IDV. Pathogen shedding was tracked, and immunological responses were evaluated by measuring acute phase response and IDV-neutralizing antibody titers. While lamb health statuses remained subclinical, M. ovipneumoniae-exposed lambs had significantly elevated body temperatures during IDV infection compared to M. ovipneumoniae-naïve, IDV-infected lambs. Moreover, we found a positive correlation between prior M. ovipneumoniae burden, early-infection IDV shedding, and IDV-neutralizing antibody response. Our findings suggest that IDV infection may not induce clinical symptoms in domestic sheep, but previous M. ovipneumoniae exposure may promote mild IDV-associated inflammation.


Asunto(s)
Enfermedades Transmisibles , Mycoplasma ovipneumoniae , Infecciones por Orthomyxoviridae , Orthomyxoviridae , Neumonía , Enfermedades de las Ovejas , Thogotovirus , Animales , Anticuerpos Neutralizantes , Bovinos , Infecciones por Orthomyxoviridae/veterinaria , Ovinos , Porcinos
14.
Front Immunol ; 13: 918881, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35757773

RESUMEN

Angiotensin Converting Enzyme 2 (ACE2) is the primary cell entry receptor for SARS-CoV and SARS-CoV-2 viruses. A disintegrin and metalloproteinase 17 (ADAM17) is a protease that cleaves ectodomains of transmembrane proteins, including that of ACE2 and the proinflammatory cytokine TNF-α, from cell surfaces upon cellular activation. We hypothesized that blockade of ADAM17 activity would alter COVID-19 pathogenesis. To assess this pathway, we blocked the function of ADAM17 using the monoclonal antibody MEDI3622 in the K18-hACE2 transgenic mouse model of COVID-19. Antibody-treated mice were healthier, less moribund, and had significantly lower lung pathology than saline-treated mice. However, the viral burden in the lungs of MEDI3622-treated mice was significantly increased. Thus, ADAM17 appears to have a critical anti-viral role, but also may promote inflammatory damage. Since the inflammatory cascade is ultimately the reason for adverse outcomes in COVID-19 patients, there may be a therapeutic application for the MEDI3622 antibody.


Asunto(s)
Proteína ADAM17 , Anticuerpos Neutralizantes , COVID-19 , SARS-CoV-2 , Proteína ADAM17/antagonistas & inhibidores , Proteína ADAM17/inmunología , Enzima Convertidora de Angiotensina 2/metabolismo , Animales , Anticuerpos Neutralizantes/inmunología , Anticuerpos Neutralizantes/farmacología , COVID-19/inmunología , COVID-19/terapia , COVID-19/virología , Modelos Animales de Enfermedad , Humanos , Ratones , Ratones Transgénicos , Peptidil-Dipeptidasa A/metabolismo , SARS-CoV-2/inmunología , Carga Viral
15.
Vet Microbiol ; 265: 109334, 2022 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-35033769

RESUMEN

Mycoplasma ovipneumoniae (M. ovipneumoniae) is a respiratory pathogen associated with mild to moderate respiratory disease in domestic lambs and severe pneumonia outbreaks in wild ruminants such as bighorn sheep. However, whether M. ovipneumoniae by itself causes clinical respiratory disease in domestic sheep in the absence of secondary bacterial pathogens is still unclear. The goal of our study was to better understand the role of M. ovipneumoniae as a respiratory pathogen in domestic sheep and to explore potential antibiotic treatment approaches. Therefore, we inoculated four 4-month-old, specific-pathogen-free lambs with fresh nasal wash fluids from M. ovipneumoniae-infected sheep. The lambs were monitored for M. ovipneumoniae colonization, M. ovipneumoniae-specific antibodies, clinical signs, and cellular and molecular correlates of lung inflammation for eight weeks. All lambs then were treated with gamithromycin and observed for an additional four weeks. M. ovipneumoniae inoculation resulted in stable colonization of the upper respiratory tract in all M. ovipneumoniae-inoculated, but in none of the four mock-infected control lambs. All M. ovipneumoniae-infected lambs developed a robust antibody response to M. ovipneumoniae within 2 weeks. However, we did not observe significant signs of respiratory disease, evidence of lung damage or inflammation in any of the infected lambs. Interestingly, treatment with gamithromycin, which blocked growth of the M. ovipneumoniae in vitro, failed to reduce M. ovipneumoniae colonization. These observations indicate that, in the absence of co-infections, M. ovipneumoniae caused asymptomatic colonization of the upper respiratory tract that was resistant to clearance by the host immune response and by gamithromycin treatment.


Asunto(s)
Mycoplasma ovipneumoniae , Enfermedades de las Ovejas , Borrego Cimarrón , Animales , Antibacterianos/farmacología , Antibacterianos/uso terapéutico , Infecciones Asintomáticas , Ovinos , Enfermedades de las Ovejas/epidemiología
16.
J Autoimmun ; 37(4): 328-41, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22018711

RESUMEN

Natural killer (NK) cells and dendritic cells (DCs) have been shown to link the innate and adaptive immune systems. Likewise, a new innate cell subset, interferon-producing killer DCs (IKDCs), shares phenotypic and functional characteristics with both DCs and NK cells. Here, we show IKDCs play an essential role in the resolution of experimental autoimmune encephalomyelitis (EAE) upon treatment with the tolerizing agent, myelin oligodendrocyte glycoprotein (MOG), genetically fused to reovirus protein σ1 (termed MOG-pσ1). Activated IKDCs were recruited subsequent MOG-pσ1 treatment of EAE, and disease resolution was abated upon NK1.1 cell depletion. These IKDCs were able to kill activated CD4(+) T cells and mature dendritic DCs, thus, contributing to EAE remission. In addition, IKDCs were responsible for MOG-pσ1-mediated MOG-specific regulatory T cell recruitment to the CNS. The IKDCs induced by MOG-pσ1 expressed elevated levels of HVEM for interactions with cognate ligand-positive cells: LIGHT(+) NK and T(eff) cells and BTLA(+) B cells. Further characterization revealed these activated IKDCs being MHC class II(high), and upon their adoptive transfer (CD11c(+)NK1.1(+)MHC class II(high)), IKDCs, but not CD11c(+)NK1.1(+)MHC class II(intermediate/low) (unactivated) cells, conferred protection against EAE. These activated IKDCs showed enhanced CD107a, PD-L1, and granzyme B expression and could present OVA, unlike unactivated IKDCs. Thus, these results demonstrate the interventional potency induced HVEM(+) IKDCs to resolve autoimmune disease.


Asunto(s)
Células Dendríticas/metabolismo , Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/terapia , Proteínas de la Mielina/metabolismo , Proteínas Virales/metabolismo , Traslado Adoptivo , Animales , Antígenos Ly/metabolismo , Citotoxicidad Inmunológica , Células Dendríticas/inmunología , Células Dendríticas/patología , Progresión de la Enfermedad , Encefalomielitis Autoinmune Experimental/patología , Encefalomielitis Autoinmune Experimental/fisiopatología , Tolerancia Inmunológica , Interferones/metabolismo , Activación de Linfocitos , Ratones , Proteínas de la Mielina/genética , Proteínas de la Mielina/inmunología , Glicoproteína Mielina-Oligodendrócito , Subfamilia B de Receptores Similares a Lectina de Células NK/metabolismo , Miembro 14 de Receptores del Factor de Necrosis Tumoral/metabolismo , Proteínas Recombinantes de Fusión/administración & dosificación , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/inmunología , Proteínas Recombinantes de Fusión/metabolismo , Recurrencia , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Subgrupos de Linfocitos T/patología , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Linfocitos T Reguladores/patología , Proteínas Virales/genética , Proteínas Virales/inmunología
17.
Nat Commun ; 12(1): 1717, 2021 03 19.
Artículo en Inglés | MEDLINE | ID: mdl-33741967

RESUMEN

Myeloid-derived suppressor cells (MDSC) are pathologically activated neutrophils and monocytes with potent immune suppressive activity. These cells play an important role in accelerating tumor progression and undermining the efficacy of anti-cancer therapies. The natural mechanisms limiting MDSC activity are not well understood. Here, we present evidence that type I interferons (IFN1) receptor signaling serves as a universal mechanism that restricts acquisition of suppressive activity by these cells. Downregulation of the IFNAR1 chain of this receptor is found in MDSC from cancer patients and mouse tumor models. The decrease in IFNAR1 depends on the activation of the p38 protein kinase and is required for activation of the immune suppressive phenotype. Whereas deletion of IFNAR1 is not sufficient to convert neutrophils and monocytes to MDSC, genetic stabilization of IFNAR1 in tumor bearing mice undermines suppressive activity of MDSC and has potent antitumor effect. Stabilizing IFNAR1 using inhibitor of p38 combined with the interferon induction therapy elicits a robust anti-tumor effect. Thus, negative regulatory mechanisms of MDSC function can be exploited therapeutically.


Asunto(s)
Interferón Tipo I/metabolismo , Células Supresoras de Origen Mieloide/inmunología , Neoplasias/metabolismo , Receptor de Interferón alfa y beta/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Animales , Antineoplásicos/farmacología , Médula Ósea , Línea Celular Tumoral , Modelos Animales de Enfermedad , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Persona de Mediana Edad , Monocitos/inmunología , Neutrófilos/inmunología , Receptor de Interferón alfa y beta/genética , Proteínas Quinasas p38 Activadas por Mitógenos/efectos de los fármacos , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
18.
ACS Appl Mater Interfaces ; 12(16): 18211-18224, 2020 Apr 22.
Artículo en Inglés | MEDLINE | ID: mdl-32233444

RESUMEN

The continuous evolution of influenza A virus (IAV) requires the influenza vaccine formulations to be updated annually to provide adequate protection. Recombinant protein-based vaccines provide safer, faster, and a more scalable alternative to the conventional embryonated egg approach for developing vaccines. However, these vaccines are typically poorer in immunogenicity than the vaccines containing inactivated or attenuated influenza viruses and require administration of a large antigen dosage together with potent adjuvants. The presentation of protein antigens on the surface of virus-like particles (VLP) provides an attractive strategy to rapidly induce stronger antigen-specific immune responses. Here we have examined the immunogenic potential and protective efficacy of P22 VLPs conjugated with multiple copies of the globular head domain of the hemagglutinin (HA) protein from the PR8 strain of IAV in a murine model of influenza pathogenesis. Using a covalent attachment strategy (SpyTag/SpyCatcher), we conjugated the HA globular head, which was recombinantly expressed in a genetically modified E. coli strain and found to refold as a monomer, to preassembled P22 VLPs. Immunization of mice with this P22-HAhead conjugate provided full protection from morbidity and mortality following infection with a homologous IAV strain. Moreover, the P22-HAhead conjugate also elicited an accelerated and enhanced HA head specific IgG response, which was significantly higher than the soluble HA head, or the admixture of P22 and HA head without the need for adjuvants. Thus, our results show that the HA head can be easily prepared by in vitro refolding in a modified E. coli strain, maintaining its intact structure and enabling the induction of a strong immune response when conjugated to P22 VLPs, even when presented as a monomer. These results also demonstrate that the P22 VLPs can be rapidly modified in a modular fashion, resulting in an effective vaccine construct that can generate protective immunity without the need for additional adjuvants.


Asunto(s)
Antígenos Virales , Virus de la Influenza A , Vacunas de Partículas Similares a Virus , Virión , Animales , Antígenos Virales/genética , Antígenos Virales/inmunología , Modelos Animales de Enfermedad , Escherichia coli , Femenino , Glicoproteínas Hemaglutininas del Virus de la Influenza/genética , Glicoproteínas Hemaglutininas del Virus de la Influenza/inmunología , Virus de la Influenza A/genética , Virus de la Influenza A/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Vacunas de Partículas Similares a Virus/genética , Vacunas de Partículas Similares a Virus/inmunología , Virión/genética , Virión/inmunología
19.
Physiol Biochem Zool ; 92(1): 37-48, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30481115

RESUMEN

Variation in immune defense influences infectious disease dynamics within and among species. Understanding how variation in immunity drives pathogen transmission among species is especially important for animals that are reservoir hosts for zoonotic pathogens. Bats, in particular, have a propensity to host serious viral zoonoses without developing clinical disease themselves. The immunological adaptations that allow bats to host viruses without disease may be related to their adaptations for flight (e.g., in metabolism and mediation of oxidative stress). A number of analyses report greater richness of zoonotic pathogens in bats than in other taxa, such as birds (i.e., mostly volant vertebrates) and rodents (i.e., nonvolant small mammals), but immunological comparisons between bats and these other taxa are rare. To examine interspecific differences in bacterial killing ability (BKA), a functional measure of overall constitutive innate immunity, we use a phylogenetic meta-analysis to compare how BKA responds to the acute stress of capture and to storage time of frozen samples across the orders Aves and Chiroptera. After adjusting for host phylogeny, sample size, and total microbe colony-forming units, we find preliminary evidence that the constitutive innate immune defense of bats may be more resilient to handling stress and storage time than that of birds. This pattern was also similar when we analyzed the proportion of nonnegative and positive effect sizes per species, using phylogenetic comparative methods. We discuss potential physiological and evolutionary mechanisms by which complement proteins may differ between species orders and suggest future avenues for comparative field studies of immunity between sympatric bats, birds, and rodents in particular.


Asunto(s)
Aves/inmunología , Quirópteros/inmunología , Animales , Actividad Bactericida de la Sangre/inmunología , Recolección de Muestras de Sangre/métodos , Escherichia coli , Inmunidad Innata , Filogenia , Estrés Fisiológico/inmunología
20.
Viruses ; 11(11)2019 10 29.
Artículo en Inglés | MEDLINE | ID: mdl-31671825

RESUMEN

Influenza D viruses (IDV) are known to co-circulate with viral and bacterial pathogens in cattle and other ruminants. Currently, there is limited knowledge regarding host responses to IDV infection and whether IDV infection affects host susceptibility to secondary bacterial infections. To begin to address this gap in knowledge, the current study utilized a combination of in vivo and in vitro approaches to evaluate host cellular responses against primary IDV infection and secondary bacterial infection with Staphylococcus aureus (S. aureus). Primary IDV infection in mice did not result in clinical signs of disease and it did not enhance the susceptibility to secondary S. aureus infection. Rather, IDV infection appeared to protect mice from the usual clinical features of secondary bacterial infection, as demonstrated by improved weight loss, survival, and recovery when compared to S. aureus infection alone. We found a notable increase in IFN-ß expression following IDV infection while utilizing human alveolar epithelial A549 cells to analyze early anti-viral responses to IDV infection. These results demonstrate for the first time that IDV infection does not increase the susceptibility to secondary bacterial infection with S. aureus, with evidence that anti-viral immune responses during IDV infection might protect the host against these potentially deadly outcomes.


Asunto(s)
Coinfección/inmunología , Infecciones por Orthomyxoviridae/inmunología , Infecciones Estafilocócicas/inmunología , Células A549 , Animales , Modelos Animales de Enfermedad , Femenino , Humanos , Interferón beta/metabolismo , Ratones , Ratones Endogámicos C57BL , Infecciones por Orthomyxoviridae/virología , Infecciones Estafilocócicas/prevención & control , Staphylococcus aureus/inmunología , Análisis de Supervivencia , Thogotovirus/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA