Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
1.
Comp Med ; 71(5): 411-432, 2021 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-34548126

RESUMEN

COVID-19, the disease caused by the SARS-CoV-2 betacoronavirus, was declared a pandemic by the World Health Organization on March 11, 2020. Since then, SARS-CoV-2 has triggered a devastating global health and economic emergency. In response, a broad range of preclinical animal models have been used to identify effective therapies and vaccines. Current animal models do not express the full spectrum of human COVID-19 disease and pathology, with most exhibiting mild to moderate disease without mortality. NHPs are physiologically, genetically, and immunologically more closely related to humans than other animal species; thus, they provide a relevant model for SARS-CoV-2 investigations. This overview summarizes NHP models of SARS-CoV-2 and their role in vaccine and therapeutic development.


Asunto(s)
COVID-19 , SARS-CoV-2 , Animales , Vacunas contra la COVID-19 , Humanos , Pandemias , Primates
2.
PLoS One ; 15(6): e0232381, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32584818

RESUMEN

Alphaviruses such as Venezuelan equine encephalitis virus (VEEV) and Eastern equine encephalitis virus (EEEV) are arboviruses that can cause severe zoonotic disease in humans. Both VEEV and EEEV are highly infectious when aerosolized and can be used as biological weapons. Vaccines and therapeutics are urgently needed, but efficacy determination requires animal models. The cynomolgus macaque (Macaca fascicularis) provides a relevant model of human disease, but questions remain whether vaccines or therapeutics can mitigate CNS infection or disease in this model. The documentation of alphavirus encephalitis in animals relies on traditional physiological biomarkers and behavioral/neurological observations by veterinary staff; quantitative measurements such as electroencephalography (EEG) and intracranial pressure (ICP) can recapitulate underlying encephalitic processes. We detail a telemetry implantation method suitable for continuous monitoring of both EEG and ICP in awake macaques, as well as methods for collection and analysis of such data. We sought to evaluate whether changes in EEG/ICP suggestive of CNS penetration by virus would be seen after aerosol exposure of naïve macaques to VEEV IC INH9813 or EEEV V105 strains compared to mock-infection in a cohort of twelve adult cynomolgus macaques. Data collection ran continuously from at least four days preceding aerosol exposure and up to 50 days thereafter. EEG signals were processed into frequency spectrum bands (delta: [0.4 - 4Hz); theta: [4 - 8Hz); alpha: [8-12Hz); beta: [12-30] Hz) and assessed for viral encephalitis-associated changes against robust background circadian variation while ICP data was assessed for signal fidelity, circadian variability, and for meaningful differences during encephalitis. Results indicated differences in delta, alpha, and beta band magnitude in infected macaques, disrupted circadian rhythm, and proportional increases in ICP in response to alphavirus infection. This novel enhancement of the cynomolgus macaque model offers utility for timely determination of onset, severity, and resolution of encephalitic disease and for the evaluation of vaccine and therapeutic candidates.


Asunto(s)
Infecciones por Alphavirus/patología , Encéfalo/fisiología , Encefalitis Viral/patología , Presión Intracraneal/fisiología , Alphavirus/aislamiento & purificación , Alphavirus/patogenicidad , Infecciones por Alphavirus/metabolismo , Animales , Biomarcadores/metabolismo , Ritmo Circadiano , Modelos Animales de Enfermedad , Electroencefalografía/métodos , Encefalitis Viral/metabolismo , Femenino , Macaca , Masculino , Índice de Severidad de la Enfermedad , Telemetría
3.
PLoS Pathog ; 16(3): e1008333, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-32119719

RESUMEN

Unlike HIV infection, SIV infection is generally nonpathogenic in natural hosts, such as African green monkeys (AGMs), despite life-long high viral replication. Lack of disease progression was reportedly based on the ability of SIV-infected AGMs to prevent gut dysfunction, avoiding microbial translocation and the associated systemic immune activation and chronic inflammation. Yet, the maintenance of gut integrity has never been documented, and the mechanism(s) by which gut integrity is preserved are unknown. We sought to investigate the early events of SIV infection in AGMs, specifically examining the impact of SIVsab infection on the gut mucosa. Twenty-nine adult male AGMs were intrarectally infected with SIVsab92018 and serially sacrificed at well-defined stages of SIV infection, preramp-up (1-3 days post-infection (dpi)), ramp-up (4-6 dpi), peak viremia (9-12 dpi), and early chronic SIV infection (46-55 dpi), to assess the levels of immune activation, apoptosis, epithelial damage and microbial translocation in the GI tract and peripheral lymph nodes. Tissue viral loads, plasma cytokines and plasma markers of gut dysfunction were also measured throughout the course of early infection. While a strong, but transient, interferon-based inflammatory response was observed, the levels of plasma markers linked to enteropathy did not increase. Accordingly, no significant increases in apoptosis of either mucosal enterocytes or lymphocytes, and no damage to the mucosal epithelium were documented during early SIVsab infection of AGMs. These findings were supported by RNAseq of the gut tissue, which found no significant alterations in gene expression that would indicate microbial translocation. Thus, for the first time, we confirmed that gut epithelial integrity is preserved, with no evidence of microbial translocation, in AGMs throughout early SIVsab infection. This might protect AGMs from developing intestinal dysfunction and the subsequent chronic inflammation that drives both HIV disease progression and HIV-associated comorbidities.


Asunto(s)
Mucosa Intestinal/inmunología , Síndrome de Inmunodeficiencia Adquirida del Simio/inmunología , Animales , Traslocación Bacteriana , Chlorocebus aethiops , Progresión de la Enfermedad , Microbioma Gastrointestinal , Infecciones por VIH/inmunología , Infecciones por VIH/microbiología , Infecciones por VIH/patología , Infecciones por VIH/virología , VIH-1/fisiología , Humanos , Mucosa Intestinal/microbiología , Masculino , Síndrome de Inmunodeficiencia Adquirida del Simio/microbiología , Síndrome de Inmunodeficiencia Adquirida del Simio/patología , Síndrome de Inmunodeficiencia Adquirida del Simio/virología , Virus de la Inmunodeficiencia de los Simios/fisiología
5.
Nat Commun ; 10(1): 5101, 2019 11 08.
Artículo en Inglés | MEDLINE | ID: mdl-31704931

RESUMEN

Natural hosts of simian immunodeficiency virus (SIV) avoid AIDS despite lifelong infection. Here, we examined how this outcome is achieved by comparing a natural SIV host, African green monkey (AGM) to an AIDS susceptible species, rhesus macaque (RM). To asses gene expression profiles from acutely SIV infected AGMs and RMs, we developed a systems biology approach termed Conserved Gene Signature Analysis (CGSA), which compared RNA sequencing data from rectal AGM and RM tissues to various other species. We found that AGMs rapidly activate, and then maintain, evolutionarily conserved regenerative wound healing mechanisms in mucosal tissue. The wound healing protein fibronectin shows distinct tissue distribution and abundance kinetics in AGMs. Furthermore, AGM monocytes exhibit an embryonic development and repair/regeneration signature featuring TGF-ß and concomitant reduced expression of inflammatory genes compared to RMs. This regenerative wound healing process likely preserves mucosal integrity and prevents inflammatory insults that underlie immune exhaustion in RMs.


Asunto(s)
Fibronectinas/inmunología , Mucosa Intestinal/inmunología , Macrófagos/inmunología , Síndrome de Inmunodeficiencia Adquirida del Simio/inmunología , Factor de Crecimiento Transformador beta/inmunología , Cicatrización de Heridas/inmunología , Animales , Chlorocebus aethiops/genética , Chlorocebus aethiops/inmunología , Progresión de la Enfermedad , Fibronectinas/metabolismo , Mucosa Intestinal/metabolismo , Macaca mulatta/genética , Macaca mulatta/inmunología , Macrófagos/metabolismo , Recto/inmunología , Recto/metabolismo , Virus de la Inmunodeficiencia de los Simios , Biología de Sistemas , Transcriptoma , Factor de Crecimiento Transformador beta/genética , Cicatrización de Heridas/genética
6.
PLoS Pathog ; 12(9): e1005879, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27632364

RESUMEN

Viruses that persist despite seemingly effective antiretroviral treatment (ART) and can reinitiate infection if treatment is stopped preclude definitive treatment of HIV-1 infected individuals, requiring lifelong ART. Among strategies proposed for targeting these viral reservoirs, the premise of the "shock and kill" strategy is to induce expression of latent proviruses [for example with histone deacetylase inhibitors (HDACis)] resulting in elimination of the affected cells through viral cytolysis or immune clearance mechanisms. Yet, ex vivo studies reported that HDACis have variable efficacy for reactivating latent proviruses, and hinder immune functions. We developed a nonhuman primate model of post-treatment control of SIV through early and prolonged administration of ART and performed in vivo reactivation experiments in controller RMs, evaluating the ability of the HDACi romidepsin (RMD) to reactivate SIV and the impact of RMD treatment on SIV-specific T cell responses. Ten RMs were IV-infected with a SIVsmmFTq transmitted-founder infectious molecular clone. Four RMs received conventional ART for >9 months, starting from 65 days post-infection. SIVsmmFTq plasma viremia was robustly controlled to <10 SIV RNA copies/mL with ART, without viral blips. At ART cessation, initial rebound viremia to ~106 copies/mL was followed by a decline to < 10 copies/mL, suggesting effective immune control. Three post-treatment controller RMs received three doses of RMD every 35-50 days, followed by in vivo experimental depletion of CD8+ cells using monoclonal antibody M-T807R1. RMD was well-tolerated and resulted in a rapid and massive surge in T cell activation, as well as significant virus rebounds (~104 copies/ml) peaking at 5-12 days post-treatment. CD8+ cell depletion resulted in a more robust viral rebound (107 copies/ml) that was controlled upon CD8+ T cell recovery. Our results show that RMD can reactivate SIV in vivo in the setting of post-ART viral control. Comparison of the patterns of virus rebound after RMD administration and CD8+ cell depletion suggested that RMD impact on T cells is only transient and does not irreversibly alter the ability of SIV-specific T cells to control the reactivated virus.


Asunto(s)
Antirretrovirales/farmacología , Depsipéptidos/farmacología , Síndrome de Inmunodeficiencia Adquirida del Simio/tratamiento farmacológico , Virus de la Inmunodeficiencia de los Simios/fisiología , Replicación Viral/efectos de los fármacos , Animales , Linfocitos T CD8-positivos/metabolismo , Macaca mulatta , ARN Viral/sangre , Síndrome de Inmunodeficiencia Adquirida del Simio/sangre , Factores de Tiempo
7.
PLoS Pathog ; 12(1): e1005384, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26764484

RESUMEN

Increased chronic immune activation and inflammation are hallmarks of HIV/SIV infection and are highly correlated with progression to AIDS and development of non-AIDS comorbidities, such as hypercoagulability and cardiovascular disease. Intestinal dysfunction resulting in microbial translocation has been proposed as a lead cause of systemic immune activation and hypercoagulability in HIV/SIV infection. Our goal was to assess the biological and clinical impact of a therapeutic strategy designed to reduce microbial translocation through reduction of the microbial content of the intestine (Rifaximin-RFX) and of gut inflammation (Sulfasalazine-SFZ). RFX is an intraluminal antibiotic that was successfully used in patients with hepatic encephalopathy. SFZ is an antiinflammatory drug successfully used in patients with mild to moderate inflammatory bowel disease. Both these clinical conditions are associated with increased microbial translocation, similar to HIV-infected patients. Treatment was administered for 90 days to five acutely SIV-infected pigtailed macaques (PTMs) starting at the time of infection; seven untreated SIVsab-infected PTMs were used as controls. RFX+SFZ were also administered for 90 days to three chronically SIVsab-infected PTMs. RFX+SFZ administration during acute SIVsab infection of PTMs resulted in: significantly lower microbial translocation, lower systemic immune activation, lower viral replication, better preservation of mucosal CD4+ T cells and significantly lower levels of hypercoagulation biomarkers. This effect was clear during the first 40 days of treatment and was lost during the last stages of treatment. Administration of RFX+SFZ to chronically SIVsab-infected PTMs had no discernible effect on infection. Our data thus indicate that early RFX+SFZ administration transiently improves the natural history of acute and postacute SIV infection, but has no effect during chronic infection.


Asunto(s)
Antiinflamatorios/farmacología , Inflamación/tratamiento farmacológico , Síndrome de Inmunodeficiencia Adquirida del Simio/tratamiento farmacológico , Trombofilia/tratamiento farmacológico , Animales , Antibacterianos/farmacología , Citometría de Flujo , Inmunohistoquímica , Inflamación/etiología , Intestinos/efectos de los fármacos , Intestinos/microbiología , Macaca nemestrina , Masculino , Rifamicinas/farmacología , Rifaximina , Síndrome de Inmunodeficiencia Adquirida del Simio/complicaciones , Virus de la Inmunodeficiencia de los Simios , Sulfasalazina/farmacología , Trombofilia/etiología
8.
J Clin Invest ; 124(6): 2802-6, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24837437

RESUMEN

Damage to the intestinal mucosa results in the translocation of microbes from the intestinal lumen into the circulation. Microbial translocation has been proposed to trigger immune activation, inflammation, and coagulopathy, all of which are key factors that drive HIV disease progression and non-HIV comorbidities; however, direct proof of a causal link is still lacking. Here, we have demonstrated that treatment of acutely SIV-infected pigtailed macaques with the drug sevelamer, which binds microbial lipopolysaccharide in the gut, dramatically reduces immune activation and inflammation and slightly reduces viral replication. Furthermore, sevelamer administration reduced coagulation biomarkers, confirming the contribution of microbial translocation in the development of cardiovascular comorbidities in SIV-infected nonhuman primates. Together, our data suggest that early control of microbial translocation may improve the outcome of HIV infection and limit noninfectious comorbidities associated with AIDS.


Asunto(s)
Traslocación Bacteriana/efectos de los fármacos , Poliaminas/farmacología , Síndrome de Inmunodeficiencia Adquirida del Simio/tratamiento farmacológico , Síndrome de Inmunodeficiencia Adquirida del Simio/microbiología , Animales , Linfocitos T CD4-Positivos/efectos de los fármacos , Linfocitos T CD4-Positivos/inmunología , Infecciones por VIH/microbiología , Infecciones por VIH/terapia , Humanos , Mucosa Intestinal/inmunología , Mucosa Intestinal/patología , Mucosa Intestinal/virología , Macaca nemestrina , Sevelamer , Síndrome de Inmunodeficiencia Adquirida del Simio/virología , Virus de la Inmunodeficiencia de los Simios/fisiología , Carga Viral/efectos de los fármacos , Replicación Viral
9.
J Virol ; 88(12): 6778-92, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24696477

RESUMEN

UNLABELLED: While simian immunodeficiency viruses (SIVs) are generally nonpathogenic in their natural hosts, dramatic increases in pathogenicity may occur upon cross-species transmission to new hosts. Deciphering the drivers of these increases in virulence is of major interest for understanding the emergence of new human immunodeficiency viruses (HIVs). We transmitted SIVsab from the sabaeus species of African green monkeys (AGMs) to pigtailed macaques (PTMs). High acute viral replication occurred in all SIVsab-infected PTMs, yet the outcome of chronic infection was highly variable, ranging from rapid progression to controlled infection, which was independent of the dynamics of acute viral replication, CD4(+) T cell depletion, or preinfection levels of microbial translocation. Infection of seven PTMs with plasma collected at necropsy from a rapid-progressor PTM was consistently highly pathogenic, with high acute and chronic viral replication, massive depletion of memory CD4(+) T cells, and disease progression in all PTMs. The plasma inoculum used for the serial passage did not contain adventitious bacterial or viral contaminants. Single-genome amplification showed that this inoculum was significantly more homogenous than the inoculum directly derived from AGMs, pointing to a strain selection in PTMs. In spite of similar peak plasma viral loads between the monkeys in the two passages, immune activation/inflammation levels dramatically increased in PTMs infected with the passaged virus. These results suggest that strain selection and a massive cytokine storm are major factors behind increased pathogenicity of SIV upon serial passage and adaptation of SIVs to new hosts following cross-species transmission. IMPORTANCE: We report here that upon cross-species transmission and serial passage of SIVsab from its natural host, the sabaeus African green monkey (AGM), to a new host, the pigtailed macaque (PTM), viral adaptation and increased pathogenicity involve strain selection and a massive cytokine storm. These results permit the design of strategies aimed at preventing cross-species transmission from natural hosts of SIVs to humans in areas of endemicity. Furthermore, our study describes a new animal model for SIV infection. As the outcomes of SIVsab infection in PTMs, African green monkeys, and rhesus macaques are different, the use of these systems enables comparative studies between pathogenic, nonpathogenic, and elite-controlled infections, to gain insight into the mechanisms of SIV immunodeficiency and comorbidities.


Asunto(s)
Especificidad del Huésped , Síndrome de Inmunodeficiencia Adquirida del Simio/transmisión , Síndrome de Inmunodeficiencia Adquirida del Simio/virología , Virus de la Inmunodeficiencia de los Simios/patogenicidad , Animales , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/virología , Chlorocebus aethiops , VIH/genética , VIH/patogenicidad , VIH/fisiología , Infecciones por VIH/inmunología , Infecciones por VIH/transmisión , Infecciones por VIH/virología , Humanos , Macaca nemestrina , Pase Seriado , Síndrome de Inmunodeficiencia Adquirida del Simio/inmunología , Virus de la Inmunodeficiencia de los Simios/genética , Virus de la Inmunodeficiencia de los Simios/fisiología , Virulencia , Replicación Viral
10.
Pathog Dis ; 71(2): 219-26, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24453160

RESUMEN

The use of common marmosets as an alternative non-human primate model for infectious disease research using BSL-3 viruses such as Rift Valley fever virus (RVFV) presents unique challenges with respect to housing, handling, and safety. Subject matter experts from veterinary care, animal husbandry, biosafety, engineering, and research were consulted to design a pilot experiment using marmosets infected with RVFV. This paper reviews the caging, handling, and safety-related adaptations and modifications that were required to humanely utilize marmosets as a model for high-hazard BSL-3 viral diseases.


Asunto(s)
Experimentación Animal , Animales de Laboratorio , Callithrix/fisiología , Control de Enfermedades Transmisibles/métodos , Contención de Riesgos Biológicos , Fiebre del Valle del Rift/diagnóstico , Fiebre del Valle del Rift/terapia , Animales , Investigación Biomédica/métodos , Modelos Animales de Enfermedad , Vivienda para Animales , Humanos , Masculino , Salud Laboral
11.
PLoS Pathog ; 9(10): e1003600, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24098110

RESUMEN

We assessed the role of myeloid dendritic cells (mDCs) in the outcome of SIV infection by comparing and contrasting their frequency, mobilization, phenotype, cytokine production and apoptosis in pathogenic (pigtailed macaques, PTMs), nonpathogenic (African green monkeys, AGMs) and controlled (rhesus macaques, RMs) SIVagmSab infection. Through the identification of recently replicating cells, we demonstrated that mDC mobilization from the bone marrow occurred in all species postinfection, being most prominent in RMs. Circulating mDCs were depleted with disease progression in PTMs, recovered to baseline values after the viral peak in AGMs, and significantly increased at the time of virus control in RMs. Rapid disease progression in PTMs was associated with low baseline levels and incomplete recovery of circulating mDCs during chronic infection. mDC recruitment to the intestine occurred in all pathogenic scenarios, but loss of mucosal mDCs was associated only with progressive infection. Sustained mDC immune activation occurred throughout infection in PTMs and was associated with increased bystander apoptosis in blood and intestine. Conversely, mDC activation occurred only during acute infection in nonprogressive and controlled infections. Postinfection, circulating mDCs rapidly became unresponsive to TLR7/8 stimulation in all species. Yet, stimulation with LPS, a bacterial product translocated in circulation only in SIV-infected PTMs, induced mDC hyperactivation, apoptosis and excessive production of proinflammatory cytokines. After infection, spontaneous production of proinflammatory cytokines by mucosal mDCs increased only in progressor PTMs. We thus propose that mDCs promote tolerance to SIV in the biological systems that lack intestinal dysfunction. In progressive infections, mDC loss and excessive activation of residual mDCs by SIV and additional stimuli, such as translocated microbial products, enhance generalized immune activation and inflammation. Our results thus provide a mechanistic basis for the role of mDCs in the pathogenesis of AIDS and elucidate the causes of mDC loss during progressive HIV/SIV infections.


Asunto(s)
Movimiento Celular/inmunología , Células Dendríticas/inmunología , Células Mieloides/inmunología , Síndrome de Inmunodeficiencia Adquirida del Simio/inmunología , Virus de la Inmunodeficiencia de los Simios/inmunología , Animales , Apoptosis/inmunología , Médula Ósea/inmunología , Médula Ósea/patología , Efecto Espectador/inmunología , Chlorocebus aethiops , Células Dendríticas/patología , Células Mieloides/patología , Síndrome de Inmunodeficiencia Adquirida del Simio/patología , Receptor Toll-Like 7/inmunología , Receptor Toll-Like 8/inmunología
12.
J Acquir Immune Defic Syndr ; 63(4): 428-37, 2013 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-23429503

RESUMEN

BACKGROUND: Chemokines provide critical immune cell homing and activation signals that if altered could affect the inflammatory milieu and cellular composition of lymphoid tissues. During HIV-1 and simian immunodeficiency virus (SIV)-infection, the virus triggers an increase in inflammation or activation, leading to immunodeficiency and development of opportunistic infections, such as in the lungs-a massive interface between the host and the environment. METHODS: Chemokine, cytokine, and chemokine receptor expression profiles were determined using real-time reverse transcriptase-polymerase chain reaction and in situ hybridization in hilar lymph nodes (HiLNs) from cynomolgus macaques at different stages after infection with SIV/DeltaB670. Immunostaining of tissue sections and flow cytometric analysis of cryopreserved cells were used to examine cellular compositions of lymph nodes. RESULTS: Interferon-gamma, type 1 chemokine, and cognate chemokine receptor mRNAs were upregulated, whereas type 2 and homeostatic chemokine and chemokine receptor mRNAs were down-regulated in HiLNs after SIV infection. Local SIV and interferon-gamma levels were positively correlated with type 1 chemokine levels but negatively correlated with type 2 and homeostatic chemokine levels. Using in situ hybridization, Pneumocystis carinii rRNA was detected in lung-draining lymph nodes from animals with P. carinii pneumonia. Changes in the cellular composition of HiLNs included decreased proportions of CD4 cells and dendritic cells and increased proportions of CD8, CXCR3, and CCR5 cells. CONCLUSIONS: SIV infection of cynomolgus macaques dramatically alters the cellular homing signals of lung-draining lymph nodes, which correlated with changes in the immune cellular composition. These changes could contribute to the loss of immune function that defines AIDS.


Asunto(s)
Quimiocinas/metabolismo , Ganglios Linfáticos/inmunología , Ganglios Linfáticos/metabolismo , Receptores de Quimiocina/metabolismo , Síndrome de Inmunodeficiencia Adquirida del Simio/inmunología , Virus de la Inmunodeficiencia de los Simios/inmunología , Animales , Recuento de Linfocito CD4 , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD8-positivos/inmunología , Células Dendríticas/inmunología , Factores de Transcripción Forkhead/metabolismo , Interferón gamma/metabolismo , Pulmón , Macaca fascicularis , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/metabolismo , Pneumocystis carinii/genética , ARN Bacteriano/metabolismo , ARN Mensajero/metabolismo , ARN Ribosómico/metabolismo , Subgrupos de Linfocitos T/inmunología
13.
Blood ; 120(7): 1357-66, 2012 Aug 16.
Artículo en Inglés | MEDLINE | ID: mdl-22653975

RESUMEN

HIV infection is associated with increased risk of cardiovascular complications, the underlying mechanism of which remains unclear. Plasma levels of the coagulation biomarker D-dimer (DD) correlate with increased mortality and cardiovascular events in HIV-infected patients. We compared the incidence of cardiovascular lesions and the levels of the coagulation markers DD and thrombin antithrombin in pathogenic SIV infections of rhesus and pigtailed macaques (PTMs) and in nonpathogenic SIV infection of African green monkeys (AGMs) and sooty mangabeys. Hypercoagulability and cardiovascular pathology were only observed in pathogenic SIV infections. In PTMs infected with SIV from AGMs (SIVagm), DD levels were highly indicative of AIDS progression and increased mortality and were associated with cardiovascular lesions, pointing to SIVagm-infected PTMs as an ideal animal model for the study of HIV-associated cardiovascular disease. In pathogenic SIV infection, DD increased early after infection, was strongly correlated with markers of immune activation/inflammation and microbial translocation (MT), and was only peripherally associated with viral loads. Endotoxin administration to SIVagm-infected AGMs (which lack chronic SIV-induced MT and immune activation) resulted in significant increases of DD. Our results demonstrate that hypercoagulation and cardiovascular pathology are at least in part a consequence of excessive immune activation and MT in SIV infection.


Asunto(s)
Coagulación Sanguínea , Progresión de la Enfermedad , Primates/sangre , Primates/virología , Síndrome de Inmunodeficiencia Adquirida del Simio/sangre , Síndrome de Inmunodeficiencia Adquirida del Simio/patología , Virus de la Inmunodeficiencia de los Simios/fisiología , Animales , Antígenos CD/metabolismo , Antígenos de Diferenciación Mielomonocítica/metabolismo , Antitrombinas/metabolismo , Biomarcadores/sangre , Coagulación Sanguínea/efectos de los fármacos , Enfermedades Cardiovasculares/patología , Cercocebus/sangre , Cercocebus/virología , Chlorocebus aethiops , Enfermedad Crónica , Productos de Degradación de Fibrina-Fibrinógeno/metabolismo , Lipopolisacáridos/administración & dosificación , Lipopolisacáridos/farmacología , Macaca/sangre , Macaca/virología , Receptores de Superficie Celular/metabolismo , Síndrome de Inmunodeficiencia Adquirida del Simio/inmunología , Síndrome de Inmunodeficiencia Adquirida del Simio/virología , Virus de la Inmunodeficiencia de los Simios/efectos de los fármacos , Solubilidad/efectos de los fármacos , Trombina/metabolismo , Factores de Tiempo
14.
J Neuroinflammation ; 9: 84, 2012 May 03.
Artículo en Inglés | MEDLINE | ID: mdl-22554253

RESUMEN

BACKGROUND: Immunization against beta-amyloid (Aß) is a promising approach for the treatment of Alzheimer's disease, but the optimal timing for the vaccination remains to be determined. Preventive immunization approaches may be more efficacious and associated with fewer side-effects; however, there is only limited information available from primate models about the effects of preclinical vaccination on brain amyloid composition and the neuroinflammatory milieu. METHODS: Ten non-human primates (NHP) of advanced age (18-26 years) and eight 2-year-old juvenile NHPs were immunized at 0, 2, 6, 10 and 14 weeks with aggregated Aß42 admixed with monophosphoryl lipid A as adjuvant, and monitored for up to 6 months. Anti-Aß antibody levels and immune activation markers were assessed in plasma and cerebrospinal fluid samples before and at several time-points after immunization. Microglial activity was determined by [(11)C]PK11195 PET scans acquired before and after immunization, and by post-mortem immunohistochemical and real-time PCR evaluation. Aß oligomer composition was assessed by immunoblot analysis in the frontal cortex of aged immunized and non-immunized control animals. RESULTS: All juvenile animals developed a strong and sustained serum anti-Aß IgG antibody response, whereas only 80 % of aged animals developed detectable antibodies. The immune response in aged monkeys was more delayed and significantly weaker, and was also more variable between animals. Pre- and post-immunization [(11)C]PK11195 PET scans showed no evidence of vaccine-related microglial activation. Post-mortem brain tissue analysis indicated a low overall amyloid burden, but revealed a significant shift in oligomer size with an increase in the dimer:pentamer ratio in aged immunized animals compared with non-immunized controls (P < 0.01). No differences were seen in microglial density or expression of classical and alternative microglial activation markers between immunized and control animals. CONCLUSIONS: Our results indicate that preventive Aß immunization is a safe therapeutic approach lacking adverse CNS immune system activation or other serious side-effects in both aged and juvenile NHP cohorts. A significant shift in the composition of soluble oligomers towards smaller species might facilitate removal of toxic Aß species from the brain.


Asunto(s)
Envejecimiento/inmunología , Vacunas contra el Alzheimer/administración & dosificación , Vacunas contra el Alzheimer/inmunología , Péptidos beta-Amiloides/administración & dosificación , Péptidos beta-Amiloides/inmunología , Encéfalo/inmunología , Inmunización/métodos , Fragmentos de Péptidos/administración & dosificación , Fragmentos de Péptidos/inmunología , Factores de Edad , Envejecimiento/metabolismo , Vacunas contra el Alzheimer/uso terapéutico , Péptidos beta-Amiloides/metabolismo , Animales , Encéfalo/metabolismo , Femenino , Macaca fascicularis , Macaca mulatta , Macaca nemestrina , Masculino , Fragmentos de Péptidos/metabolismo
15.
J Virol ; 86(8): 4158-68, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22318138

RESUMEN

African green monkeys (AGMs) are naturally infected with a simian immunodeficiency virus (SIVagm) that is nonpathogenic in its host. Although SIVagm is common and widespread, little is known about the mechanisms that govern its transmission. Since the earliest virus-host interactions may provide key insights into the nonpathogenic phenotype of SIVagm, we developed a mucosal transmission model for this virus. Using plasma from an acutely infected AGM as the virus inoculum, we exposed adult and juvenile AGMs, as well as pigtailed macaques (PTMs) as a nonnatural host control, by mucosal routes to increasing titers of virus and compared the doses needed to establish a productive infection. Four juvenile and four adult AGMs as well as two PTMs were intrarectally (IR) exposed, while two additional adult female AGMs were intravaginally (IVAG) exposed. No animal became infected following exposure to 10(5) RNA copies. Both PTMs but none of the AGMs became infected following exposure to 10(6) RNA copies. Finally, all adult AGMs and two of the four juvenile AGMs became infected following exposure to 10(7) RNA copies, acquiring either one (2 IR infected juveniles, 1 IR infected adult, 2 IVAG infected adults) or two (3 IR infected adults) transmitted founder viruses. These results were consistent with immunophenotypic data, which revealed a significant correlation between the percentage of CD4(+) T cells expressing CCR5 in the mucosa and the susceptibility to infection, in terms of both the viral dose and the numbers of transmitted founder viruses. Moreover, studies of uninfected AGMs showed that the fraction of CCR5-expressing CD4(+) T cells increased significantly with age. These results indicate that (i) AGMs are readily infected with SIVagm by both intrarectal and intravaginal routes, (ii) susceptibility to infection is proportional to the number of available CCR5(+) CD4(+) target cells in the mucosa, and (iii) the paucity of CCR5(+) CD4(+) target cells in infant and juvenile AGMs may explain the near absence of vertical transmission.


Asunto(s)
Membrana Mucosa/inmunología , Membrana Mucosa/virología , Síndrome de Inmunodeficiencia Adquirida del Simio/inmunología , Síndrome de Inmunodeficiencia Adquirida del Simio/transmisión , Virus de la Inmunodeficiencia de los Simios/inmunología , Animales , Linfocitos T CD4-Positivos/inmunología , Chlorocebus aethiops , Susceptibilidad a Enfermedades/inmunología , Evolución Molecular , Femenino , Macaca nemestrina , Masculino , Filogenia , Receptores CCR5/inmunología , Síndrome de Inmunodeficiencia Adquirida del Simio/genética , Virus de la Inmunodeficiencia de los Simios/clasificación , Virus de la Inmunodeficiencia de los Simios/genética , Proteínas del Envoltorio Viral/genética
16.
PLoS One ; 6(9): e24654, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21931798

RESUMEN

BACKGROUND: Pneumonic tularemia is caused by inhalation of the gram negative bacterium, Francisella tularensis. Because of concerns that tularemia could be used as a bioterrorism agent, vaccines and therapeutics are urgently needed. Animal models of pneumonic tularemia with a pathophysiology similar to the human disease are needed to evaluate the efficacy of these potential medical countermeasures. PRINCIPAL FINDINGS: Rabbits exposed to aerosols containing Francisella tularensis strain SCHU S4 developed a rapidly progressive fatal pneumonic disease. Clinical signs became evident on the third day after exposure with development of a fever (>40.5°C) and a sharp decline in both food and water intake. Blood samples collected on day 4 found lymphopenia and a decrease in platelet counts coupled with elevations in erythrocyte sedimentation rate, alanine aminotransferase, cholesterol, granulocytes and monocytes. Radiographs demonstrated the development of pneumonia and abnormalities of intestinal gas consistent with ileus. On average, rabbits were moribund 5.1 days after exposure; no rabbits survived exposure at any dose (190-54,000 cfu). Gross evaluation of tissues taken at necropsy showed evidence of pathology in the lungs, spleen, liver, kidney and intestines. Bacterial counts confirmed bacterial dissemination from the lungs to the liver and spleen. CONCLUSIONS/SIGNIFICANCE: The pathophysiology of pneumonic tularemia in rabbits resembles what has been reported for humans. Rabbits therefore are a relevant model of the human disease caused by type A strains of F. tularensis.


Asunto(s)
Neumonía/diagnóstico por imagen , Neumonía/microbiología , Tularemia/diagnóstico por imagen , Tularemia/microbiología , Animales , Antígenos CD13/sangre , Ingestión de Alimentos , Humanos , Ileus/microbiología , Intestinos/microbiología , Riñón/microbiología , Hígado/microbiología , Pulmón/microbiología , Linfopenia/microbiología , Recuento de Plaquetas , Neumonía/sangre , Conejos , Radiografía , Bazo/microbiología , Tularemia/sangre
17.
Am J Pathol ; 177(3): 1274-85, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20671263

RESUMEN

Infection by HIV-1 frequently leads to pulmonary complications, including alterations to local immune environments. To better understand these alterations, we have examined in detail the patterns and levels of expression of chemokine, cytokine, and chemokine receptor mRNAs in lung tissues from 16 uninfected or simian immunodeficiency virus (SIV)/DeltaB670 infected cynomolgus macaques at different stages of infection. Among the most up-regulated immune genes were interferon (IFN)-gamma, IFN-gamma-inducible CXCR3 ligands, and CCR5 ligands, as well as the cognate chemokine receptors. These changes were greatest in animals with clear Pneumocystis carinii coinfection. Immunohistochemistry and in situ hybridization revealed monocytes/macrophages to be the predominant type of cell infiltrating into lung tissues and serving as the major cellular source of chemokines. To explore the causes of chemokine alterations, we treated macaque lung cells with IFN-gamma, lipopolysaccharide, Poly(I:C), and P. carinii in vitro, and results revealed that these stimuli can induce the expression of CXCR3 ligand and/or CCR5 ligand mRNAs. Taken together, these studies provide a comprehensive definition of the chemokine networks available to modulate cellular recruitment to lung tissues during SIV infection and implicate both cytokines (IFN-gamma) and pathogens (SIV and P. carinii) as contributors to increased expression of pro-inflammatory chemokines.


Asunto(s)
Quimiocinas/inmunología , Pulmón/inmunología , Pneumocystis carinii/inmunología , Neumonía por Pneumocystis/inmunología , Receptores de Quimiocina/inmunología , Síndrome de Inmunodeficiencia Adquirida del Simio/inmunología , Virus de la Inmunodeficiencia de los Simios/inmunología , Animales , Quimiocinas/metabolismo , Inmunohistoquímica , Hibridación in Situ , Pulmón/metabolismo , Pulmón/virología , Macaca fascicularis , Neumonía por Pneumocystis/complicaciones , Neumonía por Pneumocystis/metabolismo , Neumonía por Pneumocystis/virología , Receptores de Quimiocina/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Síndrome de Inmunodeficiencia Adquirida del Simio/complicaciones , Síndrome de Inmunodeficiencia Adquirida del Simio/metabolismo , Síndrome de Inmunodeficiencia Adquirida del Simio/virología , Estadísticas no Paramétricas
18.
J Virol ; 82(10): 5031-42, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18337567

RESUMEN

The brains of individuals with lentiviral-associated encephalitis contain an abundance of infected and activated macrophages. It has been hypothesized that encephalitis develops when increased numbers of infected monocytes traffic into the central nervous system (CNS) during the end stages of immunosuppression. The relationships between the infection of brain and systemic macrophages and circulating monocytes and the development of lentiviral encephalitis are unknown. We longitudinally examined the extent of monocyte/macrophage infection in blood and lymph nodes of pigtailed macaques that did or did not develop simian immunodeficiency virus encephalitis (SIVE). Compared to levels in macaques that did not develop SIVE, more ex vivo virus production was detected from monocyte-derived macrophages and nonadherent peripheral blood mononuclear cells (PBMCs) from macaques that did develop SIVE. Prior to death, there was an increase in the number of circulating PBMCs following a rise in cerebrospinal fluid viral load in macaques that did develop SIVE but not in nonencephalitic macaques. At necropsy, macaques with SIVE had more infected macrophages in peripheral organs, with the exception of lymph nodes. T cells and NK cells with cytotoxic potential were more abundant in brains with encephalitis; however, T-cell and NK-cell infiltration in SIVE and human immunodeficiency virus encephalitis was more modest than that observed in classical acute herpes simplex virus encephalitis. These findings support the hypothesis that inherent differences in host systemic and CNS monocyte/macrophage viral production are associated with the development of encephalitis.


Asunto(s)
Encefalitis/inmunología , Encefalitis/virología , Macrófagos/virología , Síndrome de Inmunodeficiencia Adquirida del Simio/complicaciones , Síndrome de Inmunodeficiencia Adquirida del Simio/inmunología , Animales , Encéfalo/inmunología , Encéfalo/patología , Recuento de Linfocito CD4 , Líquido Cefalorraquídeo/virología , Productos del Gen gag/biosíntesis , Células Asesinas Naturales/inmunología , Leucocitos Mononucleares/virología , Estudios Longitudinales , Ganglios Linfáticos/inmunología , Ganglios Linfáticos/virología , Macaca nemestrina , ARN Viral/líquido cefalorraquídeo , Virus de la Inmunodeficiencia de los Simios/crecimiento & desarrollo , Linfocitos T/inmunología , Carga Viral
19.
J Immunol ; 178(11): 6958-67, 2007 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-17513745

RESUMEN

The loss of myeloid (mDC) and plasmacytoid dendritic cells (pDC) from the blood of HIV-infected individuals is associated with progressive disease. It has been proposed that DC loss is due to increased recruitment to lymph nodes, although this has not been directly tested. Similarly as in HIV-infected humans, we found that lineage-negative (Lin(-)) HLA-DR(+)CD11c(+)CD123(-) mDC and Lin(-)HLA-DR(+)CD11c(-)CD123(+) pDC were lost from the blood of SIV-infected rhesus macaques with AIDS. In the peripheral lymph nodes of SIV-naive monkeys the majority of mDC were mature cells derived from skin that expressed high levels of HLA-DR, CD83, costimulatory molecules, and the Langerhans cell marker CD1a, whereas pDC expressed low levels of HLA-DR and CD40 and lacked costimulatory molecules, similar to pDC in blood. Surprisingly, both DC subsets were depleted from peripheral and mesenteric lymph nodes and spleens in monkeys with AIDS, although the activation status of the remaining DC subsets was similar to that of DC in health. In peripheral and mesenteric lymph nodes from animals with AIDS there was an accumulation of Lin(-)HLA-DR(moderate)CD11c(-)CD123(-) cells that resembled monocytoid cells but failed to acquire a DC phenotype upon culture, suggesting they were not DC precursors. mDC and pDC from the lymphoid tissues of monkeys with AIDS were prone to spontaneous death in culture, indicating that apoptosis may be a mechanism for their loss in disease. These findings demonstrate that DC are lost from rather than recruited to lymphoid tissue in advanced SIV infection, suggesting that systemic DC depletion plays a direct role in the pathophysiology of AIDS.


Asunto(s)
Células Dendríticas/patología , Síndromes de Inmunodeficiencia/patología , Tejido Linfoide/patología , Células Mieloides/patología , Síndrome de Inmunodeficiencia Adquirida del Simio/patología , Animales , Muerte Celular/inmunología , Células Cultivadas , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Femenino , Síndromes de Inmunodeficiencia/sangre , Síndromes de Inmunodeficiencia/inmunología , Inmunofenotipificación , Tejido Linfoide/inmunología , Tejido Linfoide/metabolismo , Macaca mulatta , Masculino , Células Mieloides/inmunología , Células Mieloides/metabolismo , Síndrome de Inmunodeficiencia Adquirida del Simio/sangre , Síndrome de Inmunodeficiencia Adquirida del Simio/inmunología , Virus de la Inmunodeficiencia de los Simios
20.
Virology ; 354(1): 116-31, 2006 Oct 10.
Artículo en Inglés | MEDLINE | ID: mdl-16884757

RESUMEN

Determining the impact of antiretroviral therapy on virus evolution could advance the development of improved therapeutics/vaccines against HIV. Toward this goal, we analyzed virus burden, quasispecies complexity, and T cell responses in SIV/DeltaB670-infected rhesus macaques+/-treatment for 7 months with PMPA (2-30 weeks postinfection). Treatment divided the animals into two groups: poor responders (a reduction of < or =1 log) and responders (> or =2 log reduction) in virus burden. Virus evolution in poor responders and untreated controls was characterized by expression of a complex quasispecies that evolved as the disease progressed. This included the universal loss of a viral genotype selected against by in vitro passage in monkey cells and selected for by propagation in human cells. In contrast, a good response to PMPA was characterized by infection with a less complex quasispecies that evolved more slowly. Interestingly, in 2 of the best responders, the human-preferred genotype persisted until the study was discontinued (89 weeks p.i.). Neither virus burden nor the magnitude of the T cell response at 2 weeks postinfection predicted PMPA responsiveness. However, responders expressed a less complex quasispecies than nonresponders prior to treatment. These data suggest a role for intrinsic host factors in treatment responsiveness, and lend support for therapeutic vaccination as an adjunct to effective therapy.


Asunto(s)
Adenina/análogos & derivados , Antivirales/uso terapéutico , Organofosfonatos/uso terapéutico , Síndrome de Inmunodeficiencia Adquirida del Simio/tratamiento farmacológico , Síndrome de Inmunodeficiencia Adquirida del Simio/virología , Virus de la Inmunodeficiencia de los Simios/efectos de los fármacos , Virus de la Inmunodeficiencia de los Simios/genética , Adenina/farmacología , Adenina/uso terapéutico , Secuencia de Aminoácidos , Animales , Antivirales/farmacología , Farmacorresistencia Viral , Evolución Molecular , Genoma Viral , Genotipo , Glicosilación , Análisis Heterodúplex , Humanos , Macaca mulatta , Datos de Secuencia Molecular , Organofosfonatos/farmacología , Homología de Secuencia , Síndrome de Inmunodeficiencia Adquirida del Simio/inmunología , Virus de la Inmunodeficiencia de los Simios/clasificación , Virus de la Inmunodeficiencia de los Simios/fisiología , Linfocitos T/inmunología , Tenofovir , Carga Viral , Replicación Viral
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA