Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Bioorg Chem ; 138: 106659, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37336104

RESUMEN

A library of 4-substituted quinolines was synthesised based on the structural features of the privileged 4-(benzylthio)-6-methoxy-2-methylquinoline scaffold. Quinoline-based chemical probes have proven to be effective anti-tuberculosis agents with the ability of inhibiting components of Mycobacterium tuberculosis (MTB) respiratory chain including the b subunit of the cytochrome bc1 complex. Novel 4-(arylalkyl)-thio, -oxy and sulfoxy-quinoline analogues were tested for their ability to inhibit the growth of MTB H37Rv and QcrB mutant strains, and the compounds mode of action was investigated. Members of the 4-subtituted thio- and sulfoxyquinoline series exhibited significant growth inhibitory activity in the high nanomolar range against wild-type MTB and induced depletion of intracellular ATP. These probes also showed reduced potency in the QcrB T313I mutant strain, thus indicating the cytochrome bc1 oxidase complex as the molecular target. Interestingly, new 4-(quinolin-2-yl)oxy-quinoline 4i was more selective for the QcrB T313I strain compared to the wild-type strain.


Asunto(s)
Mycobacterium tuberculosis , Quinolinas , Antituberculosos/química , Complejo III de Transporte de Electrones/farmacología , Quinolinas/farmacología , Citocromos/farmacología , Pruebas de Sensibilidad Microbiana
2.
Infect Immun ; 90(6): e0013122, 2022 06 16.
Artículo en Inglés | MEDLINE | ID: mdl-35583346

RESUMEN

Mycoplasma genitalium is a sexually transmitted bacterial pathogen that causes urogenital disease in men and women. M. genitalium infections can persist for months to years and can ascend to the upper reproductive tract in women where it is associated with serious sequelae including pelvic inflammatory disease, tubal factor infertility, and preterm birth. An animal model is needed to understand immune evasion strategies that allow persistence, mechanisms of ascending infection, and factors associated with clearance. In earlier studies, we determined that pig-tailed macaques are susceptible to cervical infection; however, not all primates were successfully infected, persistence varied between animals, and ascension to the upper reproductive tract was not observed after 4 or 8 weeks of follow-up. Building on our previous findings, we refined our inoculation methods to increase infection rates, extended observation to 18 weeks, and comprehensively sampled the upper reproductive tract to detect ascending infection. With these improvements, we established infection in all (3/3) primates inoculated with M. genitalium and demonstrated lower tract persistence for 16 to 18 weeks. Ascension to the upper reproductive tract at endpoint was observed in two out of three primates. All three primates developed serum and local antibodies reacting primarily to the MgpB and MgpC adherence proteins. Elevated genital polymorphonuclear leukocytes (PMNs) and inflammatory cytokines and chemokines, erythema of the ectocervix in one primate, and histologic evidence of vaginitis and endocervicitis in two primates suggest a mild to moderate inflammatory response to infection. This model will be valuable to understand the natural history of M. genitalium infection including mechanisms of persistence, immune evasion, and ascension to the upper reproductive tract.


Asunto(s)
Infecciones por Mycoplasma , Mycoplasma genitalium , Nacimiento Prematuro , Infecciones del Sistema Genital , Animales , Femenino , Humanos , Recién Nacido , Macaca nemestrina , Infecciones por Mycoplasma/microbiología
3.
J Virol ; 88(14): 7962-72, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24807713

RESUMEN

Pathology resulting from human immunodeficiency virus (HIV) infection is driven by protracted inflammation; the primary loss of CD4(+) T cells is caused by activation-driven apoptosis. Recent studies of nonhuman primates (NHPs) have suggested that during the acute phase of infection, antiviral mucosal immunity restricts viral replication in the primary infection compartment. These studies imply that HIV achieves systemic infection as a consequence of a failure in host antiviral immunity. Here, we used high-dose intrarectal inoculation of rhesus macaques with simian immunodeficiency virus (SIV) SIVmac251 to examine how the mucosal immune system is overcome by SIV during acute infection. The host response in rectal mucosa was characterized by deep mRNA sequencing (mRNA-seq) at 3 and 12 days postinoculation (dpi) in 4 animals for each time point. While we observed a strong host transcriptional response at 3 dpi, functions relating to antiviral immunity were absent. Instead, we observed a significant number of differentially expressed genes relating to cell adhesion and reorganization of the cytoskeleton. We also observed downregulation of genes encoding members of the claudin family of cell adhesion molecules, which are coexpressed with genes associated with pathology in the colorectal mucosa, and a large number of noncoding transcripts. In contrast, at 12 dpi the differentially expressed genes were enriched in those involved with immune system functions, in particular, functions relating to T cells, B cells, and NK cells. Our findings indicate that host responses that negatively affect mucosal integrity occur before inflammation. Consequently, when inflammation is activated at peak viremia, mucosal integrity is already compromised, potentially enabling rapid tissue damage, driving further inflammation. Importance: The HIV pandemic is one of the major threats to human health, causing over a million deaths per year. Recent studies have suggested that mucosal antiviral immune responses play an important role in preventing systemic infection after exposure to the virus. Yet, despite their potential role in decreasing transmission rates between individuals, these antiviral mechanisms are poorly understood. Here, we carried out the first deep mRNA sequencing analysis of mucosal host responses in the primary infection compartment during acute SIV infection. We found that during acute infection, a significant host response was mounted in the mucosa before inflammation was triggered. Our analysis indicated that the response has a detrimental effect on tissue integrity, causing increased permeability, tissue damage, and recruitment of SIV target cells. These results emphasize the importance of mucosal host responses preceding immune activation in preventing systemic SIV infection.


Asunto(s)
Adhesión Celular , Interacciones Huésped-Patógeno , Mucosa Intestinal/inmunología , Mucosa Intestinal/virología , Recto/inmunología , Recto/virología , Virus de la Inmunodeficiencia de los Simios/inmunología , Animales , Linfocitos B/inmunología , Claudinas/metabolismo , Citoesqueleto/metabolismo , Perfilación de la Expresión Génica , Mucosa Intestinal/fisiología , Células Asesinas Naturales/inmunología , Macaca mulatta , Masculino , Linfocitos T/inmunología , Factores de Tiempo
4.
PLoS Pathog ; 8(10): e1002962, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23055934

RESUMEN

Two gammaherpesviruses, Epstein-Barr virus (EBV) (Lymphocryptovirus genus) and Kaposi's sarcoma-associated herpesvirus (KSHV) (Rhadinovirus genus) have been implicated in the etiology of AIDS-associated lymphomas. Homologs of these viruses have been identified in macaques and other non-human primates. In order to assess the association of these viruses with non-human primate disease, archived lymphoma samples were screened for the presence of macaque lymphocryptovirus (LCV) homologs of EBV, and macaque rhadinoviruses belonging to the RV1 lineage of KSHV homologs or the more distant RV2 lineage of Old World primate rhadinoviruses. Viral loads were determined by QPCR and infected cells were identified by immunolabeling for different viral proteins. The lymphomas segregated into three groups. The first group (n = 6) was associated with SIV/SHIV infections, contained high levels of LCV (1-25 genomes/cell) and expressed the B-cell antigens CD20 or BLA.36. A strong EBNA-2 signal was detected in the nuclei of the neoplastic cells in one of the LCV-high lymphomas, indicative of a type III latency stage. None of the lymphomas in this group stained for the LCV viral capsid antigen (VCA) lytic marker. The second group (n = 5) was associated with D-type simian retrovirus-2 (SRV-2) infections, contained high levels of RV2 rhadinovirus (9-790 genomes/cell) and expressed the CD3 T-cell marker. The third group (n = 3) was associated with SIV/SHIV infections, contained high levels of RV2 rhadinovirus (2-260 genomes/cell) and was negative for both CD20 and CD3. In both the CD3-positive and CD3/CD20-negative lymphomas, the neoplastic cells stained strongly for markers of RV2 lytic replication. None of the lymphomas had detectable levels of retroperitoneal fibromatosis herpesvirus (RFHV), the macaque RV1 homolog of KSHV. Our data suggest etiological roles for both lymphocryptoviruses and RV2 rhadinoviruses in the development of simian AIDS-associated lymphomas and indicate that the virus-infected neoplastic lymphoid cells are derived from different lymphocyte lineages and differentiation stages.


Asunto(s)
Herpesvirus Humano 4 , Herpesvirus Humano 8 , Lymphocryptovirus/aislamiento & purificación , Linfoma Relacionado con SIDA/virología , Síndrome de Inmunodeficiencia Adquirida del Simio/virología , Animales , Antígenos CD20/biosíntesis , Antígenos de Neoplasias/biosíntesis , Complejo CD3/biosíntesis , Antígenos Nucleares del Virus de Epstein-Barr/biosíntesis , Herpesvirus Humano 4/clasificación , Herpesvirus Humano 4/genética , Herpesvirus Humano 8/clasificación , Herpesvirus Humano 8/genética , Lymphocryptovirus/genética , Macaca , Virus del Mono Mason-Pfizer/genética , Virus del Mono Mason-Pfizer/aislamiento & purificación , Rhadinovirus/aislamiento & purificación , Síndrome de Inmunodeficiencia Adquirida del Simio/complicaciones , Virus de la Inmunodeficiencia de los Simios/genética , Virus de la Inmunodeficiencia de los Simios/aislamiento & purificación , Células Tumorales Cultivadas , Carga Viral , Proteínas Virales/biosíntesis , Proteínas Virales/genética
5.
Mol Cell Proteomics ; 11(9): 605-18, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22580588

RESUMEN

Morphine has long been known to have immunosuppressive properties in vivo, but the molecular and immunologic changes induced by it are incompletely understood. To explore how these changes interact with lentiviral infections in vivo, animals from two nonhuman primate species (African green monkeys and pigtailed macaques) were provided morphine and studied using a systems biology approach. Biological specimens were obtained from multiple sources (e.g. lymph node, colon, cerebrospinal fluid, and peripheral blood) before and after the administration of morphine (titrated up to a maximum dose of 5 mg/kg over a period of 20 days). Cellular immune, plasma cytokine, and proteome changes were measured and morphine-induced changes in these parameters were assessed on an interorgan, interindividual, and interspecies basis. In both species, morphine was associated with decreased levels of Ki-67(+) T-cell activation but with only minimal changes in overall T-cell counts, neutrophil counts, and NK cell counts. Although changes in T-cell maturation were observed, these varied across the various tissue/fluid compartments studied. Proteomic analysis revealed a morphine-induced suppressive effect in lymph nodes, with decreased abundance of protein mediators involved in the functional categories of energy metabolism, signaling, and maintenance of cell structure. These findings have direct relevance for understanding the impact of heroin addiction and the opioids used to treat addiction as well as on the potential interplay between opioid abuse and the immunological response to an infective agent.


Asunto(s)
Tolerancia Inmunológica , Inmunosupresores/farmacología , Activación de Linfocitos/efectos de los fármacos , Morfina/farmacología , Proteómica , Animales , Chlorocebus aethiops , Colon/efectos de los fármacos , Citocinas/sangre , Metabolismo Energético/efectos de los fármacos , Antígeno Ki-67 , Células Asesinas Naturales/efectos de los fármacos , Ganglios Linfáticos/efectos de los fármacos , Recuento de Linfocitos , Macaca nemestrina , Morfina/sangre , Morfina/líquido cefalorraquídeo , Neutrófilos/efectos de los fármacos , Proteoma/análisis , Transducción de Señal/efectos de los fármacos , Trastornos Relacionados con Sustancias , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología
6.
J Clin Invest ; 134(7)2024 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-38557496

RESUMEN

Programmed cell death protein 1 (PD-1) is an immune checkpoint marker commonly expressed on memory T cells and enriched in latently HIV-infected CD4+ T cells. We engineered an anti-PD-1 chimeric antigen receptor (CAR) to assess the impact of PD-1 depletion on viral reservoirs and rebound dynamics in SIVmac239-infected rhesus macaques (RMs). Adoptive transfer of anti-PD-1 CAR T cells was done in 2 SIV-naive and 4 SIV-infected RMs on antiretroviral therapy (ART). In 3 of 6 RMs, anti-PD-1 CAR T cells expanded and persisted for up to 100 days concomitant with the depletion of PD-1+ memory T cells in blood and tissues, including lymph node CD4+ follicular helper T (TFH) cells. Loss of TFH cells was associated with depletion of detectable SIV RNA from the germinal center (GC). However, following CAR T infusion and ART interruption, there was a marked increase in SIV replication in extrafollicular portions of lymph nodes, a 2-log higher plasma viremia relative to controls, and accelerated disease progression associated with the depletion of CD8+ memory T cells. These data indicate anti-PD-1 CAR T cells depleted PD-1+ T cells, including GC TFH cells, and eradicated SIV from this immunological sanctuary.


Asunto(s)
Linfocitos T CD4-Positivos , Receptores Quiméricos de Antígenos , Síndrome de Inmunodeficiencia Adquirida del Simio , Virus de la Inmunodeficiencia de los Simios , Animales , Linfocitos T CD4-Positivos/inmunología , Centro Germinal/inmunología , Infecciones por VIH/terapia , Macaca mulatta/metabolismo , Receptor de Muerte Celular Programada 1 , Receptores Quiméricos de Antígenos/genética , Síndrome de Inmunodeficiencia Adquirida del Simio/terapia
7.
Front Virol ; 32023.
Artículo en Inglés | MEDLINE | ID: mdl-37383986

RESUMEN

Zika virus (ZIKV) is a mosquito-borne flavivirus that causes an acute febrile illness. ZIKV can be transmitted between sexual partners and from mother to fetus. Infection is strongly associated with neurologic complications in adults, including Guillain-Barré syndrome and myelitis, and congenital ZIKV infection can result in fetal injury and congenital Zika syndrome (CZS). Development of an effective vaccine is imperative to protect against ZIKV vertical transmission and CZS. Recombinant Vesicular Stomatitis virus (rVSV) is a highly effective and safe vector for the delivery of foreign immunogens for vaccine purposes. Here, we evaluate an rVSV vaccine expressing the full length pre-membrane (prM) and ZIKV envelope (E) proteins (VSV-ZprME), shown to be immunogenic in murine models of ZIKV infection, for its capacity to induce immune responses in nonhuman primates. Moreover, we assess the efficacy of the rVSVΔM-ZprME vaccine in the protection of pigtail macaques against ZIKV infection. Administration of the rVSVΔM-ZprME vaccine was safe, but it did not induce robust anti-ZIKV T-cell responses, IgM or IgG antibodies, or neutralizing antibodies in most animals. Post ZIKV challenge, animals that received the rVSVΔM control vaccine lacking ZIKV antigen had higher levels of plasma viremia compared to animals that received the rVSVΔM-ZprME vaccine. Anti-ZIKV neutralizing Ab titers were detected in a single animal that received the rVSVΔM-ZprME vaccine that was associated with reduced plasma viremia. The overall suboptimal ZIKV-specific cellular and humoral responses post-immunization indicates the rVSVΔM-ZprME vaccine did not elicit an immune response in this pilot study. However, recall antibody response to the rVSVΔM-ZprME vaccine indicates it may be immunogenic and further developments to the vaccine construct could enhance its potential as a vaccine candidate in a nonhuman primate pre-clinical model.

8.
Hepatol Commun ; 7(2): e0009, 2023 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-37074875

RESUMEN

BACKGROUND AND AIMS: Adeno-associated virus (AAV) vectors are widely used to deliver therapeutic transgenes to distinct tissues, including the liver. Vectors based on naturally occurring AAV serotypes as well as vectors using engineered capsids have shown variations in tissue tropism and level of transduction between different mouse models. Moreover, results obtained in rodents frequently lack translatability into large animal studies. In light of the increasing interest in AAV vectors for human gene therapy, an increasing number of studies are being performed in nonhuman primates. To keep animal numbers to a minimum and thus optimize the process of AAV capsid selection, we developed a multiplex barcoding approach to simultaneously evaluate the in vivo vector performance for a set of serotypes and capsid-engineered AAV vectors across multiple organs. APPROACH AND RESULTS: Vector biodistribution and transgene expression were assessed by quantitative PCR, quantitative reverse transcription PCR, vector DNA amplicon Illumina sequencing and vRNAseq in male and female rhesus macaques simultaneously dosed with a mixture of barcoded naturally occurring or engineered AAV vectors encoding the same transgene. As expected, our findings show animal-to-animal variation in both the biodistribution and tissue transduction pattern, which was partly influenced by each animal's distinctive serological status. CONCLUSIONS: This method offers a robust approach to AAV vector optimization that can be used to identify and validate AAV vectors for gene delivery to potentially any anatomical site or cell type.


Asunto(s)
Cápside , Dependovirus , Animales , Ratones , Femenino , Masculino , Humanos , Cápside/metabolismo , Dependovirus/genética , Dependovirus/metabolismo , Distribución Tisular , Macaca mulatta/genética , Macaca mulatta/metabolismo , Proteínas de la Cápside/genética , Proteínas de la Cápside/metabolismo , Terapia Genética/métodos
9.
AIDS Res Hum Retroviruses ; 37(7): 505-509, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33356854

RESUMEN

Coccidioidomycosis is a common fungal infection in people living with HIV-1, particularly in southwest regions of the United States where the Coccidioides sp. is endemic, but rates of infection have significantly declined in the era of potent combination antiretroviral therapy (cART). Natural coccidioidomycosis also occurs in outdoor-housed macaques residing in the southwestern states that are utilized in biomedical research. Here, we report on a recrudescent case of previously treated, naturally occurring coccidioidomycosis in a pigtail macaque that was experimentally infected with simian immunodeficiency virus (SIV) and virally suppressed on cART. Coccidioides IgG antibody titer became detectable before discontinuation of cART, but symptomatic coccidioidomycosis developed subsequent to cART withdrawal. This animal was screened and treated in accordance with the guidelines for the prevention and treatment of coccidioidomycosis, suggesting that macaques with a history of coccidioidomycosis should be excluded from enrollment in HIV studies. Continual monitoring for known endemic pathogens based on the colony of origin is also recommended for animals utilized for HIV/AIDS research.


Asunto(s)
Coccidioidomicosis , Infecciones por VIH , Síndrome de Inmunodeficiencia Adquirida del Simio , Virus de la Inmunodeficiencia de los Simios , Animales , Coccidioidomicosis/tratamiento farmacológico , Infecciones por VIH/complicaciones , Infecciones por VIH/tratamiento farmacológico , Humanos , Macaca nemestrina , Recurrencia , Síndrome de Inmunodeficiencia Adquirida del Simio/complicaciones , Síndrome de Inmunodeficiencia Adquirida del Simio/tratamiento farmacológico , Carga Viral
10.
Hum Gene Ther ; 32(1-2): 96-112, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-32998579

RESUMEN

Adeno-associated virus (AAV) vectors such as AAV6, which shows tropism for primary human CD4+ T cells in vitro, are being explored for delivery of anti-HIV therapeutic modalities in vivo. However, pre-existing immunity and sequestration in nontarget organs can significantly hinder their performance. To overcome these challenges, we investigated whether immunosuppression would allow gene delivery by AAV6 or targeted AAV6 derivatives in seropositive rhesus macaques. Animals were immune suppressed with rapamycin before intravenous (IV) or subcutaneous (SC) delivery of AAV, and we monitored vector biodistribution, gene transfer, and safety. Macaques received phosphate-buffered saline, AAV6 alone, or an equal dose of AAV6 and an AAV6-55.2 vector retargeted to CD4 through a direct ankyrin repeat protein (DARPin). AAV6 and AAV6-55.2 vector genomes were found in peripheral blood mononuclear cells and most organs up to 28 days postadministration, with the highest levels seen in liver, spleen, lymph nodes (LNs), and muscle, suggesting that retargeting did not prevent vector sequestration. Despite vector genome detection, gene expression from AAV6-55.2 was not detected in any tissue. SC injection of AAV6 facilitated efficient gene expression in muscle adjacent to the injection site, plus low-level gene expression in spleen, LNs, and liver, whereas gene expression following IV injection of AAV6 was predominantly seen in the spleen. AAV vectors were well tolerated, although elevated liver enzymes were detected in three of four AAV-treated animals 14 days after rapamycin withdrawal. One SC-injected animal had muscle inflammation proximal to the injection site, plus detectable T cell responses against transgene and AAV6 capsid at study finish. Overall, our data suggest that rapamycin treatment may offer a possible strategy to express anti-HIV therapeutics such as broadly neutralizing antibodies from muscle. This study provides important safety and efficacy data that will aid study design for future anti-HIV gene therapies.


Asunto(s)
Dependovirus , Vectores Genéticos , Animales , Dependovirus/genética , Proteínas de Repetición de Anquirina Diseñadas , Vectores Genéticos/genética , Humanos , Leucocitos Mononucleares , Macaca mulatta , Sirolimus/uso terapéutico , Distribución Tisular
11.
Toxicol Pathol ; 37(6): 799-804, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19773593

RESUMEN

The systemic inflammatory response syndrome (SIRS) is a clinicopathological manifestation of overexuberant acute-phase inflammation caused by infectious or noninfectious etiologies. The systemic release of pro-inflammatory cytokines, chemokines, and lipid and vasoactive mediators induces endothelial damage and microvascular thrombosis, potentially culminating in disseminated intravascular coagulation (DIC), acute respiratory distress syndrome (ARDS), and multiple organ dysfunction (MOD) or failure (MOF). We present five cases in the pig-tailed macaque and olive baboon where SIRS resulted in MOF, ARDS, DIC, and the Waterhouse-Friderichsen syndrome; each with gross and histological elements manifested as edema, deposition of fibrin, hemorrhage, and thrombosis. In the described cases, SIRS was the end-common pathway for multiple risk factors that parallel those documented in humans: major surgery, obstetric complications, and infection. The diagnosis of SIRS should be considered when evaluating nonhuman primate (NHP) cases of MOF manifesting with histological evidence of vascular leakage. Experimental manipulation of NHP models may be complicated by SIRS and accompanying rapid clinical decompensation. Such adverse events may compromise toxicological studies and should be avoided when possible.


Asunto(s)
Lesión Pulmonar Aguda/veterinaria , Coagulación Intravascular Diseminada/veterinaria , Insuficiencia Multiorgánica/veterinaria , Síndrome de Respuesta Inflamatoria Sistémica/veterinaria , Lesión Pulmonar Aguda/sangre , Lesión Pulmonar Aguda/patología , Animales , Citocinas/metabolismo , Coagulación Intravascular Diseminada/sangre , Coagulación Intravascular Diseminada/patología , Femenino , Macaca nemestrina , Masculino , Insuficiencia Multiorgánica/sangre , Insuficiencia Multiorgánica/patología , Papio anubis , Factores de Riesgo , Síndrome de Respuesta Inflamatoria Sistémica/sangre , Síndrome de Respuesta Inflamatoria Sistémica/patología
12.
Stem Cell Reports ; 13(1): 91-104, 2019 07 09.
Artículo en Inglés | MEDLINE | ID: mdl-31204301

RESUMEN

Myeloid-differentiated hematopoietic stem cells (HSCs) have contributed to a number of novel treatment approaches for lysosomal storage diseases of the central nervous system (CNS), and may also be applied to patients infected with HIV. We quantified hematopoietic stem and progenitor cell (HSPC) trafficking to 20 tissues including lymph nodes, spleen, liver, gastrointestinal tract, CNS, and reproductive tissues. We observed efficient marking of multiple macrophage subsets, including CNS-associated myeloid cells, suggesting that HSPC-derived macrophages are a viable approach to target gene-modified cells to tissues. Gene-marked cells in the CNS were unique from gene-marked cells at any other physiological sites including peripheral blood. This novel finding suggests that these cells were derived from HSPCs, migrated to the brain, were compartmentalized, established myeloid progeny, and could be targeted for lifelong delivery of therapeutic molecules. Our findings have highly relevant implications for the development of novel therapies for genetic and infectious diseases of the CNS.


Asunto(s)
Sistema Nervioso Central/citología , Trasplante de Células Madre Hematopoyéticas , Células Mieloides/citología , Animales , Diferenciación Celular , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Terapia Genética/métodos , Células Madre Hematopoyéticas , Estudios Longitudinales , Enfermedades por Almacenamiento Lisosomal/patología , Enfermedades por Almacenamiento Lisosomal/terapia , Macaca nemestrina , Macrófagos/citología
14.
PLoS One ; 13(6): e0198154, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29874260

RESUMEN

A critical issue in transgene delivery studies is immune reactivity to the transgene- encoded protein and its impact on sustained gene expression. Here, we test the hypothesis that immunomodulation by rapamycin can decrease immune reactivity after intrathecal AAV9 delivery of a transgene (GFP) in non-human primates, resulting in sustained GFP expression in the CNS. We show that rapamycin treatment clearly reduced the overall immunogenicity of the AAV9/GFP vector by lowering GFP- and AAV9-specific antibody responses, and decreasing T cell responses including cytokine and cytolytic effector responses. Spinal cord GFP protein expression was sustained for twelve weeks, with no toxicity. Immune correlates of robust transgene expression include negligible GFP-specific CD4 and CD8 T cell responses, absence of GFP-specific IFN-γ producing T cells, and absence of GFP-specific cytotoxic T cells, which support the hypothesis that decreased T cell reactivity results in sustained transgene expression. These data strongly support the use of modest doses of rapamycin to modulate immune responses for intrathecal gene therapies, and potentially a much wider range of viral vector-based therapeutics.


Asunto(s)
Sistema Nervioso Central/metabolismo , Dependovirus/genética , Vectores Genéticos , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/inmunología , Inmunomodulación/fisiología , Primates/genética , Primates/inmunología , Animales , Animales Modificados Genéticamente , Autoantígenos/inmunología , Sistema Nervioso Central/inmunología , Dependovirus/inmunología , Expresión Génica , Técnicas de Transferencia de Gen , Vectores Genéticos/inmunología , Macaca fascicularis , Distribución Aleatoria , Transducción Genética , Transgenes/inmunología
15.
Nat Commun ; 9(1): 4438, 2018 10 25.
Artículo en Inglés | MEDLINE | ID: mdl-30361514

RESUMEN

Allogeneic transplantation (allo-HCT) has led to the cure of HIV in one individual, raising the question of whether transplantation can eradicate the HIV reservoir. To test this, we here present a model of allo-HCT in SHIV-infected, cART-suppressed nonhuman primates. We infect rhesus macaques with SHIV-1157ipd3N4, suppress them with cART, then transplant them using MHC-haploidentical allogeneic donors during continuous cART. Transplant results in ~100% myeloid donor chimerism, and up to 100% T-cell chimerism. Between 9 and 47 days post-transplant, terminal analysis shows that while cell-associated SHIV DNA levels are reduced in the blood and in lymphoid organs post-transplant, the SHIV reservoir persists in multiple organs, including the brain. Sorting of donor-vs.-recipient cells reveals that this reservoir resides in recipient cells. Moreover, tetramer analysis indicates a lack of virus-specific donor immunity post-transplant during continuous cART. These results suggest that early post-transplant, allo-HCT is insufficient for recipient reservoir eradication despite high-level donor chimerism and GVHD.


Asunto(s)
Reservorios de Enfermedades/virología , Trasplante de Células Madre Hematopoyéticas , Complejo Mayor de Histocompatibilidad , Virus de la Inmunodeficiencia de los Simios/fisiología , Trasplante Haploidéntico , Animales , Terapia Antirretroviral Altamente Activa , Linfocitos T CD8-positivos/inmunología , ADN Viral/metabolismo , Macaca mulatta , ARN Viral/metabolismo , Síndrome de Inmunodeficiencia Adquirida del Simio/tratamiento farmacológico , Síndrome de Inmunodeficiencia Adquirida del Simio/inmunología , Síndrome de Inmunodeficiencia Adquirida del Simio/virología , Trasplante Homólogo
16.
JCI Insight ; 2(4): e91230, 2017 02 23.
Artículo en Inglés | MEDLINE | ID: mdl-28239658

RESUMEN

The conditioning regimen used as part of the Berlin patient's hematopoietic cell transplant likely contributed to his eradication of HIV infection. We studied the impact of conditioning in simian-human immunodeficiency virus-infected (SHIV-infected) macaques suppressed by combination antiretroviral therapy (cART). The conditioning regimen resulted in a dramatic, but incomplete depletion of CD4+ and CD8+ T cells and CD20+ B cells, increased T cell activation and exhaustion, and a significant loss of SHIV-specific Abs. The disrupted T cell homeostasis and markers of microbial translocation positively correlated with an increased viral rebound after cART interruption. Quantitative viral outgrowth and Tat/rev-induced limiting dilution assays showed that the size of the latent SHIV reservoir did not correlate with viral rebound. These findings identify perturbations of the immune system as a mechanism for the failure of autologous transplantation to eradicate HIV. Thus, transplantation strategies may be improved by incorporating immune modulators to prevent disrupted homeostasis, and gene therapy to protect transplanted cells.


Asunto(s)
Linfocitos T CD4-Positivos/efectos de la radiación , Linfocitos T CD8-positivos/efectos de la radiación , Infecciones por VIH/inmunología , VIH-1/efectos de la radiación , Trasplante de Células Madre Hematopoyéticas/métodos , Síndrome de Inmunodeficiencia Adquirida del Simio/inmunología , Virus de la Inmunodeficiencia de los Simios/efectos de la radiación , Acondicionamiento Pretrasplante/métodos , Irradiación Corporal Total , Animales , Antirretrovirales/uso terapéutico , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Infecciones por VIH/tratamiento farmacológico , Homeostasis/efectos de la radiación , Infecciones por Lentivirus/tratamiento farmacológico , Infecciones por Lentivirus/inmunología , Macaca nemestrina , Síndrome de Inmunodeficiencia Adquirida del Simio/tratamiento farmacológico , Trasplante Autólogo , Carga Viral/efectos de la radiación
17.
PLoS One ; 12(12): e0189780, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29267331

RESUMEN

Recent avian and swine-origin influenza virus outbreaks illustrate the ongoing threat of influenza pandemics. We investigated immunogenicity and protective efficacy of a multi-antigen (MA) universal influenza DNA vaccine consisting of HA, M2, and NP antigens in cynomolgus macaques. Following challenge with a heterologous pandemic H1N1 strain, vaccinated animals exhibited significantly lower viral loads and more rapid viral clearance when compared to unvaccinated controls. The MA DNA vaccine induced robust serum and mucosal antibody responses but these high antibody titers were not broadly neutralizing. In contrast, the vaccine induced broadly-reactive NP specific T cell responses that cross-reacted with the challenge virus and inversely correlated with lower viral loads and inflammation. These results demonstrate that a MA DNA vaccine that induces strong cross-reactive T cell responses can, independent of neutralizing antibody, mediate significant cross-protection in a nonhuman primate model and further supports development as an effective approach to induce broad protection against circulating and emerging influenza strains.


Asunto(s)
Reacciones Cruzadas , Vacunas contra la Influenza/inmunología , Linfocitos T/inmunología , Vacunas de ADN/inmunología , Animales , Anticuerpos Neutralizantes/inmunología , Subtipo H1N1 del Virus de la Influenza A/inmunología , Vacunas contra la Influenza/genética , Macaca fascicularis , Vacunas de ADN/genética
18.
J Vet Diagn Invest ; 14(4): 326-8, 2002 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-12152813

RESUMEN

A 2.5-year-old captive female mandrill (Papio sphinx) died following a protracted course of intermittent abdominal bloat, diarrhea, and severe weight loss. Necropsy revealed emaciation and marked gastrointestinal distention with gas and ingesta. Histologic evaluation revealed severe diffuse granulomatous enterocolitis and mesenteric lymphadenitis with massive numbers of 1-2-microm acid-fast bacilli within macrophages. Additionally, there was moderate to severe multifocal myocardial and vascular amyloidosis, moderate multifocal pyogranulomatous interstitial pneumonia with no acid-fast bacteria, and moderate multifocal glossal candidiasis. Samples of feces, ileum, and colon were positive for Mycobacterium avium subsp. paratuberculosis by radiometric culture and a polymerase chain reaction-amplified DNA probe specific for the insertion sequence IS900 of this organism.


Asunto(s)
Mycobacterium avium subsp. paratuberculosis/patogenicidad , Papio/microbiología , Paratuberculosis/diagnóstico , Animales , Autopsia/veterinaria , ADN Bacteriano/análisis , Diarrea/etiología , Diarrea/veterinaria , Femenino , Mycobacterium avium subsp. paratuberculosis/genética , Mycobacterium avium subsp. paratuberculosis/aislamiento & purificación , Paratuberculosis/genética , Paratuberculosis/patología , Reacción en Cadena de la Polimerasa/veterinaria , Pérdida de Peso
19.
J Zoo Wildl Med ; 35(4): 525-9, 2004 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-15732595

RESUMEN

Three juvenile male Micronesian kingfishers (Halcyon cinnamomina cinnamomina) housed in the same enclosure presented with rapid weight gain and coelomic distension. Physical examination and radiography revealed marked enlargement of the ventriculus and a single, large foreign body within the ventriculus in each individual. Surgical removal by ventriculotomy was attempted in one individual, which died during the procedure. A second individual was treated with natural peanut butter 0.5 ml p.o. b.i.d. for 14 days and recovered, casting the foreign material. The third bird was similarly treated without success and subsequently died during attempts at endoscopic removal of the foreign body. In all three birds, the foreign bodies proved to be phytobezoars. The birds had been observed stripping leaf fragments from live corn plants (Dracaena fragrans) used in the enclosure. Plant fibers from the phytobezoars were compared with D. fragrans leaves and were considered identical. Medical treatment of phytobezoars with peanut butter or similar oil-containing substances in birds should be considered as an alternative to surgical extraction.


Asunto(s)
Arachis , Bezoares/veterinaria , Enfermedades de las Aves/terapia , Estómago de Aves , Animales , Arachis/química , Bezoares/cirugía , Bezoares/terapia , Enfermedades de las Aves/cirugía , Aves , Resultado Fatal , Masculino , Aceite de Cacahuete , Aceites de Plantas/uso terapéutico , Estómago de Aves/cirugía , Resultado del Tratamiento , Aumento de Peso
20.
Comp Med ; 64(1): 63-7, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24512963

RESUMEN

A 2.25-y-old male pigtailed macaque (Macaca nemestrina) was experimentally irradiated and received a bone marrow transplant. After transplantation and engraftment, the macaque had unexpected recurring pancytopenia and dependent edema of the prepuce, scrotum, and legs. The diagnostic work-up included a blood smear, which revealed a trypomastigote consistent with Trypanosoma cruzi, the causative agent of Chagas disease (CD). We initially hypothesized that the macaque had acquired the infection when it lived in Georgia. However, because the animal had received multiple blood transfusions, all blood donors were screened for CD. One male pigtailed macaque blood donor, which was previously housed in Louisiana, was positive for T. cruzi antibodies via serology. Due to the low prevalence of infection in Georgia, the blood transfusion was hypothesized to be the source of T. cruzi infection. The transfusion was confirmed as the mechanism of transmission when screening of archived serum revealed seroconversion after blood transfusion from the seropositive blood donor. The macaque made a full clinical recovery, and further follow-up including thoracic radiography, echocardiography, and gross necropsy did not show any abnormalities associated with CD. Other animals that received blood transfusions from the positive blood donor were tested, and one additional pigtailed macaque on the same research protocol was positive for T. cruzi. Although CD has been reported to occur in many nonhuman primate species, especially pigtailed macaques, the transmission of CD via blood transfusion in nonhuman primates has not been reported previously.


Asunto(s)
Transfusión Sanguínea/veterinaria , Enfermedad de Chagas/veterinaria , Huésped Inmunocomprometido , Macaca nemestrina/parasitología , Enfermedades de los Monos/parasitología , Trypanosoma cruzi/aislamiento & purificación , Animales , Anticuerpos Antiprotozoarios/sangre , Biomarcadores/sangre , Enfermedad de Chagas/sangre , Enfermedad de Chagas/inmunología , Enfermedad de Chagas/transmisión , Fraccionamiento de la Dosis de Radiación , Terapia Genética , Macaca nemestrina/sangre , Macaca nemestrina/inmunología , Masculino , Modelos Animales , Enfermedades de los Monos/sangre , Enfermedades de los Monos/inmunología , Trasplante de Células Madre , Reacción a la Transfusión , Trypanosoma cruzi/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA