Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Retrovirology ; 21(1): 8, 2024 May 02.
Artículo en Inglés | MEDLINE | ID: mdl-38693565

RESUMEN

The study of HIV infection and pathogenicity in physical reservoirs requires a biologically relevant model. The human immune system (HIS) mouse is an established model of HIV infection, but defects in immune tissue reconstitution remain a challenge for examining pathology in tissues. We utilized exogenous injection of the human recombinant FMS-like tyrosine kinase 3 ligand (rFLT-3 L) into the hematopoietic stem cell (HSC) cord blood HIS mouse model to significantly expand the total area of lymph node (LN) and the number of circulating human T cells. The results enabled visualization and quantification of HIV infectivity, CD4 T cell depletion and other measures of pathogenesis in the secondary lymphoid tissues of the spleen and LN. Treatment with the Caspase-1/4 inhibitor VX-765 limited CD4+ T cell loss in the spleen and reduced viral load in both the spleen and axillary LN. In situ hybridization further demonstrated a decrease in viral RNA in both the spleen and LN. Transcriptomic analysis revealed that in vivo inhibition of caspase-1/4 led to an upregulation in host HIV restriction factors including SAMHD1 and APOBEC3A. These findings highlight the use of rFLT-3 L to augment human immune system characteristics in HIS mice to support investigations of HIV pathogenesis and test host directed therapies, though further refinements are needed to further augment LN architecture and cellular populations. The results further provide in vivo evidence of the potential to target inflammasome pathways as an avenue of host-directed therapy to limit immune dysfunction and virus replication in tissue compartments of HIV+ persons.


Asunto(s)
Linfocitos T CD4-Positivos , Modelos Animales de Enfermedad , Infecciones por VIH , VIH-1 , Animales , Ratones , Infecciones por VIH/inmunología , Infecciones por VIH/virología , Infecciones por VIH/tratamiento farmacológico , VIH-1/fisiología , VIH-1/efectos de los fármacos , Humanos , Linfocitos T CD4-Positivos/inmunología , Tejido Linfoide/virología , Tejido Linfoide/inmunología , Carga Viral/efectos de los fármacos , Bazo/virología , Bazo/inmunología , Ganglios Linfáticos/inmunología , Ganglios Linfáticos/virología , Caspasas/metabolismo , Inhibidores de Caspasas/farmacología , Antirretrovirales/uso terapéutico
2.
J Immunol ; 207(1): 221-233, 2021 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-34183369

RESUMEN

Tuberculosis (TB) caused by infection with Mycobacterium tuberculosis is characterized by inflammatory pathology and poorly understood mechanisms of innate immunity. Pattern recognition receptors, expressed on the surface of macrophages, determine the balance of inflammatory and antimicrobial functions that influence disease outcome. Carbohydrate moieties displayed by mycobacteria can serve as pattern recognition receptor ligands for some members of the C-type lectin receptor (CLR) family, interactions that mediate a variety of incompletely understood immune outcomes. This work identifies a novel role for the CLR macrophage galactose-type lectin (MGL)-1 in a mouse model (C57BL/6 and MGL-1-/-) of experimental TB. Murine macrophages upregulated MGL-1 following in vitro exposure to M. tuberculosis, whereas MGL+ cells accumulated at sites of mycobacteria-driven inflammation in the lung. Pulmonary macrophages from MGL-1-deficient mice displayed increased production of proinflammatory cytokines (IL-1ß, IL-6, and IFN-γ) that were associated with greater lipid accumulation, following M. tuberculosis infection. Surprisingly, for a CLR, we also observed MGL-1-dependent antimycobacterial activity as evidenced by greater M. tuberculosis proliferation in bone marrow-derived macrophages, and the lung, of MGL-1-deficient mice. Differential transcriptome analysis further revealed that loss of MGL-1 perturbs the activation of various genes involved in the regulation of inflammation and lipid metabolism in the setting of M. tuberculosis infection. These results identify MGL-1 signaling as an important mechanism that regulates innate immunity against M. tuberculosis and indicates the potential for the MGL pathway as a novel therapeutic target for anti-TB immunity.


Asunto(s)
Mycobacterium tuberculosis , Tuberculosis , Animales , Galactosa , Inmunidad Innata , Lectinas Tipo C/genética , Macrófagos , Ratones , Ratones Endogámicos C57BL
3.
Retrovirology ; 18(1): 14, 2021 06 16.
Artículo en Inglés | MEDLINE | ID: mdl-34134725

RESUMEN

Humanized mice have become an important workhorse model for HIV research. Advances that enabled development of a human immune system in immune deficient mouse strains have aided new basic research in HIV pathogenesis and immune dysfunction. The small animal features facilitate development of clinical interventions that are difficult to study in clinical cohorts, and avoid the high cost and regulatory burdens of using non-human primates. The model also overcomes the host restriction of HIV for human immune cells which limits discovery and translational research related to important co-infections of people living with HIV. In this review we emphasize recent advances in modeling bacterial and viral co-infections in the setting of HIV in humanized mice, especially neurological disease, and Mycobacterium tuberculosis and HIV co-infections. Applications of current and future co-infection models to address important clinical and research questions are further discussed.


Asunto(s)
Modelos Animales de Enfermedad , Infecciones por VIH/microbiología , Infecciones por VIH/virología , VIH-1/patogenicidad , Ratones Transgénicos , Enfermedades del Sistema Nervioso/virología , Animales , Gonorrea/virología , Infecciones por VIH/inmunología , VIH-1/inmunología , Humanos , Ratones , Mycobacterium tuberculosis/patogenicidad , Neisseria gonorrhoeae/patogenicidad , Tuberculosis/virología
4.
J Virol ; 91(15)2017 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-28539439

RESUMEN

Nipah virus (NiV) is a zoonotic emerging paramyxovirus that can cause fatal respiratory illness or encephalitis in humans. Despite many efforts, the molecular mechanisms of NiV-induced acute lung injury (ALI) remain unclear. We previously showed that NiV replicates to high titers in human lung grafts in NOD-SCID/γ mice, resulting in a robust inflammatory response. Interestingly, these mice can undergo human immune system reconstitution by the bone marrow, liver, and thymus (BLT) reconstitution method, in addition to lung tissue engraftment, giving altogether a realistic model to study human respiratory viral infections. Here, we characterized NiV Bangladesh strain (NiV-B) infection of human lung grafts from human immune system-reconstituted mice in order to identify the overall effect of immune cells on NiV pathogenesis of the lung. We show that NiV-B replicated to high titers in human lung grafts and caused similar cytopathic effects irrespective of the presence of human leukocytes in mice. However, the human immune system interfered with virus spread across lung grafts, responded to infection by leukocyte migration to small airways and alveoli of the lung grafts, and accelerated oxidative stress in lung grafts. In addition, the presence of human leukocytes increased the expression of cytokines and chemokines that regulate inflammatory influx to sites of infection and tissue damage. These results advance our understanding of how the immune system limits NiV dissemination and contributes to ALI and inform efforts to identify therapeutic targets.IMPORTANCE Nipah virus (NiV) is an emerging paramyxovirus that can cause a lethal respiratory and neurological disease in humans. Only limited data are available on NiV pathogenesis in the human lung, and the relative contribution of the innate immune response and NiV to acute lung injury (ALI) is still unknown. Using human lung grafts in a human immune system-reconstituted mouse model, we showed that the NiV Bangladesh strain induced cytopathic lesions in lung grafts similar to those described in patients irrespective of the donor origin or the presence of leukocytes. However, the human immune system interfered with virus spread, responded to infection by leukocyte infiltration in the small airways and alveolar area, induced oxidative stress, and triggered the production of cytokines and chemokines that regulate inflammatory influx by leukocytes in response to infection. Understanding how leukocytes interact with NiV and cause ALI in human lung xenografts is crucial for identifying therapeutic targets.


Asunto(s)
Lesión Pulmonar Aguda/patología , Infecciones por Henipavirus/patología , Leucocitos/inmunología , Pulmón/patología , Virus Nipah/crecimiento & desarrollo , Estrés Oxidativo , Animales , Citocinas/análisis , Modelos Animales de Enfermedad , Humanos , Ratones , Ratones SCID
5.
Microb Pathog ; 80: 27-38, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25697665

RESUMEN

We recently characterized the Δlpp Δpla double in-frame deletion mutant of Yersinia pestis CO92 molecularly, biologically, and immunologically. While Braun lipoprotein (Lpp) activates toll-like receptor-2 to initiate an inflammatory cascade, plasminogen activator (Pla) protease facilitates bacterial dissemination in the host. The Δlpp Δpla double mutant was highly attenuated in evoking bubonic and pneumonic plague, was rapidly cleared from mouse organs, and generated humoral and cell-mediated immune responses to provide subsequent protection to mice against a lethal challenge dose of wild-type (WT) CO92. Here, we further characterized the Δlpp Δpla double mutant in two murine macrophage cell lines as well as in primary human monocyte-derived macrophages to gauge its potential as a live-attenuated vaccine candidate. We first demonstrated that the Δpla single and the Δlpp Δpla double mutant were unable to survive efficiently in murine and human macrophages, unlike WT CO92. We observed that the levels of Pla and its associated protease activity were not affected in the Δlpp single mutant, and, likewise, deletion of the pla gene from WT CO92 did not alter Lpp levels. Further, our study revealed that both Lpp and Pla contributed to the intracellular survival of WT CO92 via different mechanisms. Importantly, the ability of the Δlpp Δpla double mutant to be phagocytized by macrophages, to stimulate production of tumor necrosis factor-α and interleukin-6, and to activate the nitric oxide killing pathways of the host cells remained unaltered when compared to the WT CO92-infected macrophages. Finally, macrophages infected with either the WT CO92 or the Δlpp Δpla double mutant were equally efficient in their uptake of zymosan particles as determined by flow cytometric analysis. Overall, our data indicated that although the Δlpp Δpla double mutant of Y. pestis CO92 was highly attenuated, it retained the ability to elicit innate and subsequent acquired immune responses in the host similar to that of WT CO92, which are highly desirable in a live-attenuated vaccine candidate.


Asunto(s)
Eliminación de Gen , Lipoproteínas/deficiencia , Macrófagos Alveolares/microbiología , Macrófagos/microbiología , Péptido Hidrolasas/deficiencia , Activadores Plasminogénicos/deficiencia , Yersinia pestis/crecimiento & desarrollo , Animales , Células Cultivadas , Humanos , Inmunidad Innata , Ratones , Viabilidad Microbiana , Vacuna contra la Peste , Vacunas Atenuadas , Virulencia , Yersinia pestis/genética
6.
Trop Med Infect Dis ; 9(6)2024 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-38922041

RESUMEN

L-arginine metabolism is strongly linked with immunity to mycobacteria, primarily through the antimicrobial activity of nitric oxide (NO). The potential to modulate tuberculosis (TB) outcomes through interventions that target L-arginine pathways are limited by an incomplete understanding of mechanisms and inadequate in vivo modeling. These gaps in knowledge are compounded for HIV and Mtb co-infections, where activation of arginase-1 due to HIV infection may promote survival and replication of both Mtb and HIV. We utilized in vitro and in vivo systems to determine how arginase inhibition using Nω-hydroxy-nor-L-arginine (nor-NOHA) alters L-arginine pathway metabolism relative to immune responses and disease outcomes following Mtb infection. Treatment with nor-NOHA polarized murine macrophages (RAW 264.7) towards M1 phenotype, increased NO, and reduced Mtb in RAW macrophages. In Balb/c mice, nor-NOHA reduced pulmonary arginase and increased the antimicrobial metabolite spermine in association with a trend towards reduced Mtb CFU in lung. In humanized immune system (HIS) mice, HIV infection increased plasma arginase and heightened the pulmonary arginase response to Mtb. Treatment with nor-NOHA increased cytokine responses to Mtb and Mtb/HIV in lung tissue but did not significantly alter bacterial burden or viral load. Our results suggest that L-arginine pathway modulators may have potential as host-directed therapies to augment antibiotics in TB chemotherapy.

7.
NPJ Vaccines ; 8(1): 165, 2023 Oct 28.
Artículo en Inglés | MEDLINE | ID: mdl-37898618

RESUMEN

Therapeutic vaccines have promise as adjunctive treatment for tuberculosis (TB) or as preventives against TB relapse. An important development challenge is the limited understanding of T helper (Th) cell roles during these stages of disease. A murine model of TB relapse was used to identify changes in Th populations and cytokine microenvironment. Active TB promoted expansion of Th1, Th2, Th17, and Th22 cells and cytokines in the lung. Following drug therapy, pulmonary Th17 and Th22 cells contracted, Th1 cells remained elevated, while Th cells producing IL-4 or IL-10 expanded. At relapse, Th22 cells failed to re-expand in the lung despite a moderate re-expansion of Th1 and Th17 cells and an increase in Th cytokine polyfunctionality. The dynamics of Th populations further differed by tissue compartment and disease presentation. These outcomes identify immune bias by Th subpopulations during TB relapse as candidate mechanisms for pathogenesis and targets for therapeutic vaccination.

8.
Infect Immun ; 80(1): 234-42, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22006566

RESUMEN

Natural killer (NK) cells have innate antibacterial activity that could be targeted for clinical interventions for infectious disease caused by naturally occurring or weaponized bacterial pathogens. To determine a potential role for NK cells in immunity to Bacillus anthracis, we utilized primary human and murine NK cells, in vitro assays, and in vivo NK cell depletion in a murine model of inhalational anthrax. Our results demonstrate potent antibacterial activity by human NK cells against B. anthracis bacilli within infected autologous monocytes. Surprisingly, NK cells also mediate moderate antibacterial effects on extracellular vegetative bacilli but do not have activity against extracellular or intracellular spores. The immunosuppressive anthrax lethal toxin impairs NK gamma interferon (IFN-γ) expression, but neither lethal nor edema toxin significantly alters the viability or cytotoxic effector function of NK cells. Compared to human NK cells, murine NK cells have a similar, though less potent, activity against intracellular and extracellular B. anthracis. The in vivo depletion of murine NK cells does not alter animal survival following intranasal infection with B. anthracis spores in our studies but significantly increases the bacterial load in the blood of infected animals. Our studies demonstrate that NK cells participate in the innate immune response against B. anthracis and suggest that immune modulation to augment NK cell function in early stages of anthrax should be further explored in animal models as a clinical intervention strategy.


Asunto(s)
Carbunco/inmunología , Bacillus anthracis/inmunología , Células Asesinas Naturales/inmunología , Adulto , Animales , Antígenos Bacterianos/toxicidad , Toxinas Bacterianas/toxicidad , Células Cultivadas , Modelos Animales de Enfermedad , Femenino , Humanos , Interferón gamma/antagonistas & inhibidores , Células Asesinas Naturales/microbiología , Procedimientos de Reducción del Leucocitos , Ratones , Persona de Mediana Edad , Esporas Bacterianas/inmunología , Análisis de Supervivencia
9.
J Med Chem ; 65(4): 2866-2879, 2022 02 24.
Artículo en Inglés | MEDLINE | ID: mdl-34570513

RESUMEN

The emergence of a new coronavirus, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), presents an urgent public health crisis. Without available targeted therapies, treatment options remain limited for COVID-19 patients. Using medicinal chemistry and rational drug design strategies, we identify a 2-phenyl-1,2-benzoselenazol-3-one class of compounds targeting the SARS-CoV-2 main protease (Mpro). FRET-based screening against recombinant SARS-CoV-2 Mpro identified six compounds that inhibit proteolysis with nanomolar IC50 values. Preincubation dilution experiments and molecular docking determined that the inhibition of SARS-CoV-2 Mpro can occur by either covalent or noncovalent mechanisms, and lead E04 was determined to inhibit Mpro competitively. Lead E24 inhibited viral replication with a nanomolar EC50 value (844 nM) in SARS-CoV-2-infected Vero E6 cells and was further confirmed to impair SARS-CoV-2 replication in human lung epithelial cells and human-induced pluripotent stem cell-derived 3D lung organoids. Altogether, these studies provide a structural framework and mechanism of Mpro inhibition that should facilitate the design of future COVID-19 treatments.


Asunto(s)
Antivirales/farmacología , Benzotiazoles/farmacología , Proteasas 3C de Coronavirus/antagonistas & inhibidores , Inhibidores de Cisteína Proteinasa/farmacología , Descubrimiento de Drogas , SARS-CoV-2/efectos de los fármacos , Animales , Antivirales/síntesis química , Antivirales/química , Benzotiazoles/química , COVID-19/metabolismo , Chlorocebus aethiops , Proteasas 3C de Coronavirus/aislamiento & purificación , Proteasas 3C de Coronavirus/metabolismo , Cristalografía por Rayos X , Inhibidores de Cisteína Proteinasa/síntesis química , Inhibidores de Cisteína Proteinasa/química , Relación Dosis-Respuesta a Droga , Transferencia Resonante de Energía de Fluorescencia , Humanos , Pruebas de Sensibilidad Microbiana , Simulación del Acoplamiento Molecular , Estructura Molecular , SARS-CoV-2/enzimología , Células Vero , Replicación Viral/efectos de los fármacos , Tratamiento Farmacológico de COVID-19
10.
J Clin Microbiol ; 49(5): 1708-15, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21367990

RESUMEN

We evaluated two commercial F1 antigen capture-based immunochromatographic dipsticks, Yersinia Pestis (F1) Smart II and Plague BioThreat Alert test strips, in detecting plague bacilli by using whole-blood samples from mice experimentally infected with Yersinia pestis CO92. To assess the specificities of these dipsticks, an in-frame F1-deficient mutant of CO92 (Δcaf) was generated by homologous recombination and used as a negative control. Based on genetic, antigenic/immunologic, and electron microscopic analyses, the Δcaf mutant was devoid of a capsule. The growth rate of the Δcaf mutant generally was similar to that of the wild-type (WT) bacterium at both 26 and 37 °C, although the mutant's growth dropped slightly during the late phase at 37 °C. The Δcaf mutant was as virulent as WT CO92 in the pneumonic plague mouse model; however, it was attenuated in developing bubonic plague. Both dipsticks had similar sensitivities, requiring a minimum of 0.5 µg/ml of purified F1 antigen or 1 × 10(5) to 5 × 10(5) CFU/ml of WT CO92 for positive results, while the blood samples were negative for up to 1 × 10(8) CFU/ml of the Δcaf mutant. Our studies demonstrated the diagnostic potential of two plague dipsticks in detecting capsular-positive strains of Y. pestis in bubonic and pneumonic plague.


Asunto(s)
Proteínas Bacterianas/análisis , Técnicas de Laboratorio Clínico/métodos , Eliminación de Gen , Peste/diagnóstico , Peste/patología , Factores de Virulencia/genética , Yersinia pestis/patogenicidad , Animales , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Modelos Animales de Enfermedad , Femenino , Inmunoensayo , Ratones , Ratones Endogámicos BALB C , Peste/microbiología , Peste/mortalidad , Juego de Reactivos para Diagnóstico , Sensibilidad y Especificidad , Análisis de Supervivencia , Virulencia , Factores de Virulencia/metabolismo , Yersinia pestis/genética , Yersinia pestis/crecimiento & desarrollo
11.
Artículo en Inglés | MEDLINE | ID: mdl-32373548

RESUMEN

Tuberculosis relapse following drug treatment of active disease is an important global public health problem due to the poorer clinical outcomes and increased risk of drug resistance development. Concurrent infection with HIV, including in those receiving anti-retroviral therapy (ART), is an important risk factor for relapse and expansion of drug resistant Mycobacterium tuberculosis (Mtb) isolates. A greater understanding of the HIV-associated factors driving TB relapse is important for development of interventions that support immune containment and complement drug therapy. We employed the humanized mouse to develop a new model of post-chemotherapy TB relapse in the setting of HIV infection. Paucibacillary TB infection was observed following treatment with Rifampin and Isoniazid and subsequent infection with HIV-1 was associated with increased Mtb burden in the post-drug phase. Organized granulomas were observed during development of acute TB and appeared to resolve following TB drug therapy. At relapse, granulomatous pathology in the lung was infrequent and mycobacteria were most often observed in the interstitium and at sites of diffuse inflammation. Compared to animals with HIV mono-infection, higher viral replication was observed in the lung and liver, but not in the periphery, of animals with post-drug TB relapse. The results demonstrate a potential role for the humanized mouse as an experimental model of TB relapse in the setting of HIV. Long term, the model could facilitate discovery of disease mechanisms and development of clinical interventions.


Asunto(s)
Coinfección , Infecciones por VIH , Mycobacterium tuberculosis , Tuberculosis , Animales , Antituberculosos/uso terapéutico , Coinfección/tratamiento farmacológico , Modelos Animales de Enfermedad , Infecciones por VIH/complicaciones , Infecciones por VIH/tratamiento farmacológico , Ratones , Recurrencia , Tuberculosis/complicaciones , Tuberculosis/tratamiento farmacológico
12.
Viruses ; 8(5)2016 05 23.
Artículo en Inglés | MEDLINE | ID: mdl-27223297

RESUMEN

Rift Valley fever is a mosquito-transmitted, zoonotic disease that infects humans and ruminants. Dendritic cell specific intercellular adhesion molecule 3 (ICAM-3) grabbing non-integrin (DC-SIGN) acts as a receptor for members of the phlebovirus genus. The Rift Valley fever virus (RVFV) glycoproteins (Gn/Gc) encode five putative N-glycan sequons (asparagine (N)-any amino acid (X)-serine (S)/threonine (T)) at positions: N438 (Gn), and N794, N829, N1035, and N1077 (Gc). The N-glycosylation profile and significance in viral infection via DC-SIGN have not been elucidated. Gc N-glycosylation was first evaluated by using Gc asparagine (N) to glutamine (Q) mutants. Subsequently, we generated a series of recombinant RVFV MP-12 strain mutants, which encode N-to-Q mutations, and the infectivity of each mutant in Jurkat cells stably expressing DC-SIGN was evaluated. Results showed that Gc N794, N1035, and N1077 were N-glycosylated but N829 was not. Gc N1077 was heterogeneously N-glycosylated. RVFV Gc made two distinct N-glycoforms: "Gc-large" and "Gc-small", and N1077 was responsible for "Gc-large" band. RVFV showed increased infection of cells expressing DC-SIGN compared to cells lacking DC-SIGN. Infection via DC-SIGN was increased in the presence of either Gn N438 or Gc N1077. Our study showed that N-glycans on the Gc and Gn surface glycoproteins redundantly support RVFV infection via DC-SIGN.


Asunto(s)
Moléculas de Adhesión Celular/metabolismo , Glicoproteínas/metabolismo , Lectinas Tipo C/metabolismo , Polisacáridos/metabolismo , Receptores de Superficie Celular/metabolismo , Receptores Virales/metabolismo , Virus de la Fiebre del Valle del Rift/fisiología , Proteínas Estructurales Virales/metabolismo , Acoplamiento Viral , Sustitución de Aminoácidos , Glicoproteínas/genética , Humanos , Células Jurkat , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Virus de la Fiebre del Valle del Rift/genética , Proteínas Estructurales Virales/genética
13.
Sci Rep ; 6: 21522, 2016 Feb 24.
Artículo en Inglés | MEDLINE | ID: mdl-26908312

RESUMEN

Co-infection with HIV increases the morbidity and mortality associated with tuberculosis due to multiple factors including a poorly understood microbial synergy. We developed a novel small animal model of co-infection in the humanized mouse to investigate how HIV infection disrupts pulmonary containment of Mtb. Following dual infection, HIV-infected cells were localized to sites of Mtb-driven inflammation and mycobacterial replication in the lung. Consistent with disease in human subjects, we observed increased mycobacterial burden, loss of granuloma structure, and increased progression of TB disease, due to HIV co-infection. Importantly, we observed an HIV-dependent pro-inflammatory cytokine signature (IL-1ß, IL-6, TNFα, and IL-8), neutrophil accumulation, and greater lung pathology in the Mtb-co-infected lung. These results suggest that in the early stages of acute co-infection in the humanized mouse, infection with HIV exacerbates the pro-inflammatory response to pulmonary Mtb, leading to poorly formed granulomas, more severe lung pathology, and increased mycobacterial burden and dissemination.


Asunto(s)
Coinfección/inmunología , Infecciones por VIH/inmunología , VIH-1/inmunología , Huésped Inmunocomprometido , Tuberculosis Pulmonar/inmunología , Animales , Modelos Animales de Enfermedad , Infecciones por VIH/virología , Pulmón/inmunología , Pulmón/microbiología , Pulmón/patología , Ratones , Infiltración Neutrófila
14.
Tuberculosis (Edinb) ; 93 Suppl: S60-5, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24388651

RESUMEN

Memory T cell populations recover following phase I chemotherapy for tuberculosis (TB) and augment the effectiveness of antibiotics during the continuation phase of treatment. For those with human immunodeficiency virus (HIV), the CD8(+)T cells may have an especially important role in host defense to Mycobacterium tuberculosis (M.tb) as CD4(+)T cell function and/or numbers decline. Here we performed a preliminary study to investigate the impact of HIV infection status on CD8(+)T cell effector function during the convalescent TB period. Peripheral blood samples from convalescent HIV(+) and HIV(-) TB subjects were used to determine CD4(+)T cell count and monitor antigen-specific CD8(+) T cell activation of effector function (lymphoproliferation, IFN-γ, granulysin) in response to M.tb antigen. Our preliminary results suggest that HIV co-infection is associated with moderate suppression of the M.tb-specific memory CD8(+)T cell compartment in many subjects convalescent for TB. Interestingly, highly activated CD8(+)T cells were observed in recall experiments using peripheral blood from several HIV+ subjects that had low (<200 cells/mm(3)) CD4(+)T cell counts. Further investigation may provide important information for development of novel approaches to target M.tb-specific CD8(+)T cell memory to protect against TB in HIV-endemic regions.


Asunto(s)
Infecciones Oportunistas Relacionadas con el SIDA/inmunología , Linfocitos T CD8-positivos/inmunología , Coinfección/inmunología , Infecciones por VIH/inmunología , Activación de Linfocitos/inmunología , Mycobacterium tuberculosis/fisiología , Tuberculosis/inmunología , Inmunidad Adaptativa , Adulto , Anticuerpos Monoclonales/inmunología , Antígenos Bacterianos/inmunología , Recuento de Linfocito CD4 , Coinfección/patología , Convalecencia , Femenino , Citometría de Flujo , Infecciones por VIH/patología , Interacciones Huésped-Patógeno , Humanos , Inmunidad Celular , Kenia , Masculino , Persona de Mediana Edad , Mycobacterium tuberculosis/inmunología , Tuberculosis/patología
15.
PLoS One ; 8(5): e63331, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23691024

RESUMEN

Mycobacterium tuberculosis (M.tb) is the second leading infectious cause of death worldwide and the primary cause of death in people living with HIV/AIDS. There are several excellent animal models employed to study tuberculosis (TB), but many have limitations for reproducing human pathology and none are amenable to the direct study of HIV/M.tb co-infection. The humanized mouse has been increasingly employed to explore HIV infection and other pathogens where animal models are limiting. Our goal was to develop a small animal model of M.tb infection using the bone marrow, liver, thymus (BLT) humanized mouse. NOD-SCID/γc(null) mice were engrafted with human fetal liver and thymus tissue, and supplemented with CD34(+) fetal liver cells. Excellent reconstitution, as measured by expression of the human CD45 pan leukocyte marker by peripheral blood populations, was observed at 12 weeks after engraftment. Human T cells (CD3, CD4, CD8), as well as natural killer cells and monocyte/macrophages were all observed within the human leukocyte (CD45(+)) population. Importantly, human T cells were functionally competent as determined by proliferative capacity and effector molecule (e.g. IFN-γ, granulysin, perforin) expression in response to positive stimuli. Animals infected intranasally with M.tb had progressive bacterial infection in the lung and dissemination to spleen and liver from 2-8 weeks post infection. Sites of infection in the lung were characterized by the formation of organized granulomatous lesions, caseous necrosis, bronchial obstruction, and crystallization of cholesterol deposits. Human T cells were distributed throughout the lung, liver, and spleen at sites of inflammation and bacterial growth and were organized to the periphery of granulomas. These preliminary results demonstrate the potential to use the humanized mouse as a model of experimental TB.


Asunto(s)
Modelos Animales de Enfermedad , Tuberculosis/fisiopatología , Animales , Trasplante de Médula Ósea/métodos , Humanos , Hígado/citología , Hígado/patología , Pulmón/patología , Ratones , Ratones Endogámicos NOD , Ratones SCID , Linfocitos T/inmunología , Timo/citología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA