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

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
J Immunol ; 203(1): 58-75, 2019 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-31109957

RESUMEN

CD40 is a costimulatory receptor on APCs that is critical for the induction and maintenance of humoral and cell-mediated immunity. Accordingly, CD40 and its ligand, CD40L, have long been considered targets for the treatment of autoimmune diseases. We developed a rat/mouse chimeric anti-mouse CD40 antagonist mAb, 201A3, and evaluated its ability to alleviate murine lupus. Treatment of NZB/W-F1 mice with 201A3 after the onset of severe proteinuria rapidly reversed established severe proteinuria and nephritis and largely restored normal glomerular and tubular morphology. This coincided with a normalization of the expression of genes associated with proteinuria and injury by kidney parenchymal cells. Anti-CD40 treatment also prevented and reversed loss of saliva production and sialadenitis. These effects on kidney and salivary gland function were confirmed using mice of a second strain, MRL/Mp-lpr/lpr, and extended to alleviating joint inflammation. Immunologically, anti-CD40 treatment disrupted multiple processes that contribute to the pathogenesis of systemic lupus erythematosus (SLE), including autoreactive B cell activation, T effector cell function in target tissues, and type I IFN production. This ability to disrupt disease-critical immunological mechanisms, to reverse glomerular and tubular injury at the cellular and gene expression levels, and to confer exceptional therapeutic efficacy suggests that CD40 is a central disease pathway in murine SLE. Thus, a CD40 antagonist Ab could be an effective therapeutic in the treatment of SLE.


Asunto(s)
Anticuerpos Bloqueadores/uso terapéutico , Linfocitos B/inmunología , Antígenos CD40/inmunología , Inmunoterapia/métodos , Glomérulos Renales/patología , Lupus Eritematoso Sistémico/terapia , Proteínas Recombinantes de Fusión/uso terapéutico , Linfocitos T/inmunología , Animales , Autoantígenos/inmunología , Células Cultivadas , Modelos Animales de Enfermedad , Humanos , Interferón Tipo I/metabolismo , Lupus Eritematoso Sistémico/inmunología , Ratones , Ratones Endogámicos MRL lpr , Ratones Endogámicos NZB , Proteinuria , Ratas , Eliminación Salival
2.
FASEB J ; 29(4): 1165-75, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25466897

RESUMEN

There are no approved therapies for muscle wasting in children infected with human immunodeficiency virus (HIV), which portends poor disease outcomes. To determine whether a soluble ActRIIb receptor Fc fusion protein (ActRIIB.Fc), a ligand trap for TGF-ß/activin family members including myostatin, can prevent or restore loss of lean body mass and body weight in simian immunodeficiency virus (SIV)-infected juvenile rhesus macaques (Macaca mulatta). Fourteen pair-housed, juvenile male rhesus macaques were inoculated with SIVmac239 and, 4 wk postinoculation (WPI) treated with intramuscular injections of 10 mg ⋅ kg(-1) ⋅ wk(-1) ActRIIB.Fc or saline placebo. Body weight, lean body mass, SIV titers, and somatometric measurements were assessed monthly for 16 wk. Age-matched SIV-infected rhesus macaques were injected with saline. Intervention groups did not differ at baseline. Gains in lean mass were significantly greater in the ActRIIB.Fc group than in the placebo group (P < 0.001). Administration of ActRIIB.Fc was associated with greater gains in body weight (P = 0.01) and upper arm circumference than placebo. Serum CD4(+) T-lymphocyte counts and SIV copy numbers did not differ between groups. Administration of ActRIIB.Fc was associated with higher muscle expression of myostatin than placebo. ActRIIB.Fc effectively blocked and reversed loss of body weight, lean mass, and fat mass in juvenile SIV-infected rhesus macaques.


Asunto(s)
Receptores de Activinas Tipo II/uso terapéutico , Fragmentos Fc de Inmunoglobulinas/uso terapéutico , Síndrome de Inmunodeficiencia Adquirida del Simio/terapia , Virus de la Inmunodeficiencia de los Simios , Animales , Modelos Animales de Enfermedad , Síndrome de Emaciación por VIH/prevención & control , Hematócrito , Humanos , Ligandos , Macaca mulatta , Masculino , Músculo Esquelético/patología , Miostatina/antagonistas & inhibidores , Miostatina/genética , Miostatina/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas Recombinantes de Fusión/uso terapéutico , Síndrome de Inmunodeficiencia Adquirida del Simio/patología , Síndrome de Inmunodeficiencia Adquirida del Simio/fisiopatología , Factor de Crecimiento Transformador beta/antagonistas & inhibidores , Regulación hacia Arriba , Aumento de Peso
3.
J Virol ; 88(16): 9310-20, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24899204

RESUMEN

UNLABELLED: The UL128 complex of human cytomegalovirus (CMV) is a major determinant of viral entry into epithelial and endothelial cells and a target for vaccine development. The UL/b' region of rhesus CMV contains several open reading frames, including orthologs of the UL128 complex. We recently showed that the coding content of the rhesus CMV (RhCMV) UL/b' region predicts acute endothelial tropism and long-term shedding in vivo in the rhesus macaque model of CMV infection. The laboratory-passaged RhCMV 180.92 strain has a truncated UL/b' region but an intact UL128 complex. To investigate whether the presence of the UL128 complex alone was sufficient to confer endothelial and epithelial tropism in vivo, we investigated tissue dissemination and viral excretion following experimental RhCMV 180.92 inoculation of RhCMV-seronegative rhesus macaques. We show the presence of at least two virus variants in the RhCMV 180.92 infectious virus stock. A rare variant noted for a nontruncated wild-type-virus-like UL/b' region, rapidly emerged during in vivo replication and showed high-level replication in blood and tissues and excretion in urine and saliva, features similar to those previously reported in naturally occurring wild-type RhCMV infection. In contrast, the predominant truncated version of RhCMV 180.92 showed significantly lower plasma DNAemia and limited tissue dissemination and viral shedding. These data demonstrate that the truncated RhCMV 180.92 variant is attenuated in vivo and suggest that additional UL/b' genes, besides the UL128 complex, are required for optimal in vivo CMV replication and dissemination. IMPORTANCE: An effective vaccine against human CMV infection will need to target genes that are essential for virus propagation and transmission. The human CMV UL128 complex represents one such candidate antigen since it is essential for endothelial and epithelial cell tropism, and is a target for neutralizing antibodies in CMV-infected individuals. In this study, we used the rhesus macaque animal model of CMV infection to investigate the in vivo function of the UL128 complex. Using experimental infection of rhesus macaques with a rhesus CMV virus variant that contained an intact UL128 complex but was missing several other genes, we show that the presence of the UL128 complex alone is not sufficient for widespread tissue dissemination and virus excretion. These data highlight the importance of in vivo studies in evaluating human CMV gene function and suggest that additional UL/b' genes are required for optimal CMV dissemination and transmission.


Asunto(s)
Citomegalovirus/genética , Citomegalovirus/metabolismo , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Proteínas del Envoltorio Viral/genética , Proteínas del Envoltorio Viral/metabolismo , Proteínas Virales/genética , Proteínas Virales/metabolismo , Esparcimiento de Virus/genética , Animales , Línea Celular , Infecciones por Citomegalovirus/virología , Modelos Animales de Enfermedad , Endotelio/virología , Epitelio/virología , Fibroblastos/virología , Humanos , Macaca mulatta/virología , Glicoproteínas de Membrana/orina , Sistemas de Lectura Abierta , Proteínas del Envoltorio Viral/orina , Proteínas Virales/orina , Tropismo Viral/genética , Replicación Viral/genética
4.
BMC Neurosci ; 16: 91, 2015 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-26666889

RESUMEN

BACKGROUND: Spatial and temporal changes in brain tissue after acute ischemic stroke are still poorly understood. Aims of this study were three-fold: (1) to determine unique temporal magnetic resonance imaging (MRI) patterns at the acute, subacute and chronic stages after stroke in macaques by combining quantitative T2 and diffusion MRI indices into MRI 'tissue signatures', (2) to evaluate temporal differences in these signatures between transient (n = 2) and permanent (n = 2) middle cerebral artery occlusion, and (3) to correlate histopathology findings in the chronic stroke period to the acute and subacute MRI derived tissue signatures. RESULTS: An improved iterative self-organizing data analysis algorithm was used to combine T2, apparent diffusion coefficient (ADC), and fractional anisotropy (FA) maps across seven successive timepoints (1, 2, 3, 24, 72, 144, 240 h) which revealed five temporal MRI signatures, that were different from the normal tissue pattern (P < 0.001). The distribution of signatures between brains with permanent and transient occlusions varied significantly between groups (P < 0.001). Qualitative comparisons with histopathology revealed that these signatures represented regions with different histopathology. Two signatures identified areas of progressive injury marked by severe necrosis and the presence of gitter cells. Another signature identified less severe but pronounced neuronal and axonal degeneration, while the other signatures depicted tissue remodeling with vascular proliferation and astrogliosis. CONCLUSION: These exploratory results demonstrate the potential of temporally and spatially combined voxel-based methods to generate tissue signatures that may correlate with distinct histopathological features. The identification of distinct ischemic MRI signatures associated with specific tissue fates may further aid in assessing and monitoring the efficacy of novel pharmaceutical treatments for stroke in a pre-clinical and clinical setting.


Asunto(s)
Algoritmos , Encéfalo/patología , Procesamiento de Imagen Asistido por Computador/métodos , Imagen por Resonancia Magnética/métodos , Accidente Cerebrovascular/patología , Enfermedad Aguda , Animales , Enfermedad Crónica , Imagen de Difusión Tensora , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Infarto de la Arteria Cerebral Media , Macaca fascicularis , Masculino , Estudios Retrospectivos
5.
Am J Pathol ; 183(6): 1910-1917, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24095925

RESUMEN

Simian virus 40 (SV40), family Polyomaviridae, in immunocompromised macaques can cause fatal demyelinating central nervous system disease analogous to progressive multifocal leukoencephalopathy caused by John Cunningham (JC) virus in immunocompromised humans. Recently, we have demonstrated that JC virus can infect cerebellar granule cell neurons and cortical pyramidal neurons in immunosuppressed people. To examine whether SV40 neuronal infection occurs spontaneously in immunosuppressed macaques, we analyzed archival brain specimens from 20 simian immunodeficiency virus-infected rhesus with AIDS and 1 cynomolgus post-transplant selected with SV40 brain infection from archival records from 1991 to 2012. In addition to white matter SV40 distribution in classic demyelinating progressive multifocal leukoencephalopathy, some of the 21 monkeys exhibited meningeal, subpial neocortical, and periventricular virus. This distribution pattern corresponded to broader viral tropism with neuronal infection in 14 (66.7%) of 21 cases. In all 14 cases, identified neurons were positive for early SV40 transcript large T antigen, but only 4 of the 14 cases exhibited late viral transcript viral protein 1-positive neurons. SV40-infected neurons were detected in frontal, parietal, occipital, and temporal cortices, hippocampus, thalamus, and brain stem. These observations confirm that spontaneous SV40 neuronal infection occurs in immunosuppressed macaques, which parallels JC virus-neuronal infection in immunosuppressed patients. Neuronal infection may be an important aspect of both SV40 and JC virus neuropathogenesis in their respective hosts.


Asunto(s)
Encéfalo , Coinfección , Leucoencefalopatías , Meningoencefalitis , Infecciones por Polyomavirus , Síndrome de Inmunodeficiencia Adquirida del Simio , Virus de la Inmunodeficiencia de los Simios , Virus 40 de los Simios , Infecciones Tumorales por Virus , Animales , Encéfalo/metabolismo , Encéfalo/patología , Coinfección/metabolismo , Coinfección/patología , Leucoencefalopatías/metabolismo , Leucoencefalopatías/patología , Macaca fascicularis , Macaca mulatta , Meningoencefalitis/metabolismo , Meningoencefalitis/patología , Infecciones por Polyomavirus/metabolismo , Infecciones por Polyomavirus/patología , Síndrome de Inmunodeficiencia Adquirida del Simio/metabolismo , Síndrome de Inmunodeficiencia Adquirida del Simio/patología , Infecciones Tumorales por Virus/metabolismo , Infecciones Tumorales por Virus/patología
6.
J Med Primatol ; 43(6): 468-76, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25082291

RESUMEN

BACKGROUND: Simian immunodeficiency virus (SIV), a model for HIV pathogenesis, is associated with neuropathology. METHODS: Five SIV-infected animals were selected following a database search of 1206 SIV-infected animals for nodular or astrocytic lesions. Two of five had neurologic dysfunction, and 3 of 5 were incidental findings. RESULTS: Histologic examination revealed multifocal nodular foci in the gray and white matter formed by interlacing astrocytes with abundant cytoplasm and large, reactive nuclei. Nodules were often enmeshed with small capillaries. Immunohistochemistry revealed variable immunoreactivity for a panel of markers: GFAP (4/5), vimentin (5/5), Glut-1 (1/5), CNPase (0/5), S100 (5/5), Iba1 (0/5), Ki67 (0/5), and p53 (4/4). In situ hybridization failed to detect any SIV RNA (0/5). Immunohistochemistry for simian virus 40, rhesus cytomegalovirus, and rhesus lymphocryptovirus failed to detect any antigen within the lesions. CONCLUSION: The immunoreactivity of p53 in the lesions compared with adjacent tissue suggests a local derangement in astrocyte proliferation and function.


Asunto(s)
Gliosis/veterinaria , Macaca mulatta , Enfermedades de los Monos/patología , Síndrome de Inmunodeficiencia Adquirida del Simio/complicaciones , Virus de la Inmunodeficiencia de los Simios/fisiología , Animales , Femenino , Gliosis/inmunología , Gliosis/patología , Gliosis/virología , Inmunohistoquímica/veterinaria , Masculino , Enfermedades de los Monos/inmunología , Enfermedades de los Monos/virología , Estudios Retrospectivos , Síndrome de Inmunodeficiencia Adquirida del Simio/inmunología , Síndrome de Inmunodeficiencia Adquirida del Simio/virología , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo
7.
J Virol ; 86(11): 6354-7, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22491451

RESUMEN

We previously reported that long-term rhesus cytomegalovirus (RhCMV) excretion in infected macaques was related to UL/b' coding content. Acute biopsy specimens of the inoculation sites from the previous study have now been analyzed to determine whether there were acute phenotypic predictors of long-term RhCMV infection. Only in animals displaying acute endothelial tropism and neutrophilic inflammation was RhCMV excretion detected. The results imply that vaccinating against these early viral determinants would significantly impede long-term RhCMV infection.


Asunto(s)
Infecciones por Citomegalovirus/veterinaria , Citomegalovirus/patogenicidad , Enfermedades de los Primates/patología , Enfermedades de los Primates/virología , Animales , Enfermedad Crónica , Infecciones por Citomegalovirus/patología , Células Endoteliales/virología , Macaca , Neutrófilos/inmunología
8.
Toxicol Pathol ; 41(7): 1016-27, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23427274

RESUMEN

Nonhuman primates, particularly rhesus macaques (Macaca mulatta), provide important model systems for studying human reproductive infectious diseases such as human immunodeficiency virus, human papillomavirus, and Chlamydia spp. An understanding of the spectrum of spontaneous cervical disease provides essential context for interpreting experimental disease outcomes in the female reproductive tract. This retrospective study characterizes the incidence of inflammatory and/or proliferative cervicovaginal lesions seen over a 14-year period in a multispecies nonhuman primate colony, focusing on rhesus macaques. The most common observations included a spectrum of lymphocytic accumulation from within normal limits to lymphoplasmacytic cervicitis, and suppurative inflammation with occasional squamous metaplasia or polyp formation. These inflammatory spectra frequently occurred in the context of immunosuppression following experimental simian immunodeficiency virus (SIV) infection. Cervical neoplasias were uncommon and included leiomyomas and carcinomas. Cervical sections from 13 representative cases, with an emphasis on proliferative and dysplastic lesions, were surveyed for leukocyte infiltration, abnormal epithelial proliferation, and the presence of papillomavirus antigens. Proliferative lesions showed sporadic evidence of spontaneous papillomavirus infection and variable immune cell responses. These results underscore the importance of pre screening potential experimental animals for the presence of preexisting reproductive tract disease, and the consideration of normal variability within cycling reproductive tracts in interpretation of cervical lesions.


Asunto(s)
Enfermedades de los Primates/patología , Neoplasias del Cuello Uterino/veterinaria , Neoplasias Vaginales/veterinaria , Animales , Callitrichinae , Femenino , Inmunohistoquímica , Macaca mulatta , Papillomaviridae/inmunología , Enfermedades de los Primates/inmunología , Estudios Retrospectivos , Virus de la Inmunodeficiencia de los Simios/inmunología , Neoplasias del Cuello Uterino/inmunología , Neoplasias del Cuello Uterino/patología , Neoplasias Vaginales/inmunología , Neoplasias Vaginales/patología
9.
BMC Genomics ; 13: 98, 2012 Mar 19.
Artículo en Inglés | MEDLINE | ID: mdl-22429831

RESUMEN

BACKGROUND: Marmosets are playing an increasingly large and important role in biomedical research. They share genetic, anatomical, and physiological similarities with humans and other primate model species, but their smaller sizes, reproductive efficiency, and amenability to genetic manipulation offer an added practicality. While their unique biology can be exploited to provide insights into disease and function, it is also important that researchers are aware of the differences that exist between marmosets and other species. The New World monkey family Callitrichidae, containing both marmoset and tamarin species, typically produces dizygotic twins that show chimerism in the blood and other cells from the hematopoietic lineage. Recently, a study extended these findings to identify chimerism in many tissues, including somatic tissues from other lineages and germ cells. This has raised the intriguing possibility that chimerism may play an increasingly pervasive role in marmoset biology, ranging from natural behavioral implications to increased variability and complexity in biomedical studies. RESULTS: Using a quantitative PCR based methodology, Y-chromosomes can be reliably detected in the females with male fraternal twins allowing for a relative quantification of chimerism levels between individuals and tissues. With this approach in common marmosets (Callithrix jacchus) and cotton-top tamarins (Saguinus oedipus), chimerism was detected across a broad array of tissues. Chimerism levels were significantly higher in tissues primarily derived from the hematopoietic lineage, while they were lower, though still detectable, in tissues with other origins. Interestingly, animals with a characteristic marmoset wasting disease show higher levels of chimerism in those tissues affected. Fibroblast cell lines from chimeric individuals, however, are not found to be chimeric themselves. CONCLUSION: Taken together, the levels of chimerism in tissues of different origins coupled with other lines of evidence suggest that indeed only hematopoietic cell lineages are chimeric in callitrichids. The chimerism detected in other tissues is likely the result of blood or lymphocytic infiltration. Using molecular methods to detect chimerism in a tissue sample seems to have allowed a substantial increase in the ability to detect these minor cell populations.


Asunto(s)
Callithrix/genética , Quimerismo , Saguinus/genética , Animales , Células Sanguíneas/metabolismo , Femenino , Fibroblastos/metabolismo , Masculino , Cromosoma Y
10.
Magn Reson Med ; 66(3): 625-34, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21381104

RESUMEN

Proton magnetic resonance spectroscopy has emerged as one of the most informative neuroimaging modalities for studying the effect of HIV infection in the brain, providing surrogate markers by which to assess disease progression and monitor treatment. Reductions in the level of N-Acetylaspartate and N-Acetylaspartate/creatine are established markers of neuronal injury or loss. However, the biochemical basis of altered creatine levels in neuroAIDS is not well understood. This study used a rapid progression macaque model of neuroAIDS to elucidate the changes in creatine. As the disease progressed, proton magnetic resonance spectroscopy revealed a decrease in N-Acetylaspartate, indicative of neuronal injury, and an increase in creatine yet to be elucidated. Subsequently, immunohistochemistry and stereology measures of decreased synaptophysin, microtubule-associated protein 2, and neuronal density confirmed neuronal injury. Furthermore, increases in ionized calcium binding adaptor molecule 1 and glial fibrillary acidic protein indicated microglial and astroglial activation, respectively. Given these data, elevated creatine may reflect enhanced high-energy phosphate turnover in highly metabolizing activated astrocytes and microglia.


Asunto(s)
Encéfalo/metabolismo , Creatina/metabolismo , Metabolismo Energético , Imagen por Resonancia Magnética/métodos , Espectroscopía de Resonancia Magnética/métodos , Neuronas/metabolismo , Síndrome de Inmunodeficiencia Adquirida del Simio/metabolismo , Análisis de Varianza , Animales , Ácido Aspártico/análogos & derivados , Ácido Aspártico/metabolismo , Encéfalo/patología , Linfocitos T CD8-positivos , Colina/metabolismo , Citometría de Flujo , Inmunohistoquímica , Inositol/metabolismo , Macaca , Masculino , Neuronas/patología , Síndrome de Inmunodeficiencia Adquirida del Simio/patología , Carga Viral
11.
Am J Pathol ; 177(2): 777-91, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20595631

RESUMEN

Antiretroviral drugs suppress virus burden in the cerebrospinal fluid of HIV-infected individuals; however, the direct effect of antiretrovirals on virus replication in brain parenchyma is poorly understood. We investigated the effect of short-term combined antiretroviral therapy (CART) on brain virus burden in rhesus monkeys using the CD8-depletion model of accelerated simian immunodeficiency virus (SIV) encephalitis. Four monkeys received CART (consisting of the nonpenetrating agents PMPA and RCV) for four weeks, beginning 28 days after SIV inoculation. Lower virus burdens were measured by real-time RT-PCR in four of four regions of brain from monkeys that received CART as compared with four SIV-infected, untreated controls; however, the difference was only significant for the frontal cortex (P < 0.05). In contrast, significantly lower virus burdens were measured in plasma and four of five lymphoid compartments from animals that received CART. Surprisingly, despite normalization of neuronal function in treated animals, the numbers of activated macrophages/microglia and the magnitude of TNF-alpha mRNA expression in brain were similar between treated animals and controls. These results suggest that short-term therapy with antiretrovirals that fail to penetrate the blood-cerebrospinal fluid barrier can reduce brain virus burden provided systemic virus burden is suppressed; however, longer treatment may be required to completely resolve encephalitic lesions and microglial activation, which may reflect the longer half-life of the principal target cells of HIV/SIV in the brain (macrophages) versus lymphoid tissues (T lymphocytes).


Asunto(s)
Antirretrovirales/farmacología , Encéfalo , Linfocitos T CD8-positivos/virología , Macaca mulatta/virología , Virus de la Inmunodeficiencia de los Simios/efectos de los fármacos , Carga Viral , Animales , Antirretrovirales/uso terapéutico , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Encéfalo/virología , Linfocitos T CD8-positivos/inmunología , Productos del Gen gag/genética , Productos del Gen gag/metabolismo , Humanos , Tejido Linfoide/metabolismo , Tejido Linfoide/patología , Tejido Linfoide/virología , Macrófagos/metabolismo , Síndrome de Inmunodeficiencia Adquirida del Simio/tratamiento farmacológico , Síndrome de Inmunodeficiencia Adquirida del Simio/patología , Virus de la Inmunodeficiencia de los Simios/genética , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo , Replicación Viral/efectos de los fármacos
12.
J Neurovirol ; 17(5): 455-68, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21789725

RESUMEN

Understanding the mechanisms of neuronal regeneration and repair in the adult central nervous system is a vital area of research. Using a rhesus lentiviral encephalitis model, we sought to determine whether recovery of neuronal metabolism after injury coincides with the induction of two important markers of synaptodendritic repair: growth-associated protein-43 (GAP-43) and ephrin B3. We examined whether the improvement of neuronal metabolism with combined anti-retroviral therapy (cART) after simian immunodeficiency virus (SIV) infection in rhesus macaques involved induction of GAP-43, also known as neuromodulin, and ephrin B3, both implicated in axonal pathfinding during neurodevelopment and regulation of synapse formation, neuronal plasticity, and repair in adult brain. We utilized magnetic resonance spectroscopy to demonstrate improved neuronal metabolism in vivo in adult SIV-infected cART animals compared to untreated and uninfected controls. We then assessed levels of GAP-43, ephrin B3, and synaptophysin, a pre-synaptic marker, in three brain regions important for cognitive function, cortex, hippocampus, and putamen, by quantitative real-time RT-PCR and immunohistochemistry. Here we demonstrate that (1) GAP-43 mRNA and protein are induced with SIV infection, (2) GAP-43 protein is higher in the hippocampus outer molecular layer in SIV-infected animals that received cART compared to those that did not, and (3) activated microglia and infiltrating SIV-infected macrophages express abundant ephrin B3, an important axonal guidance molecule. We propose a model whereby SIV infection triggers events that lead to induction of GAP-43 and ephrin B3, and that short-term cART results in increased magnitude of repair mechanisms especially in the hippocampus, a region known for high levels of adult plasticity.


Asunto(s)
Encéfalo/metabolismo , Efrina-B3/metabolismo , Proteína GAP-43/metabolismo , Síndrome de Inmunodeficiencia Adquirida del Simio/patología , Animales , Antirretrovirales/uso terapéutico , Encéfalo/patología , Encéfalo/virología , Hipocampo/metabolismo , Macaca mulatta/metabolismo , Macaca mulatta/virología , Macrófagos/metabolismo , Microglía/metabolismo , Plasticidad Neuronal , ARN Mensajero/metabolismo , Síndrome de Inmunodeficiencia Adquirida del Simio/tratamiento farmacológico , Síndrome de Inmunodeficiencia Adquirida del Simio/virología , Virus de la Inmunodeficiencia de los Simios/patogenicidad , Sinaptofisina/metabolismo
13.
BMC Evol Biol ; 10: 51, 2010 Feb 18.
Artículo en Inglés | MEDLINE | ID: mdl-20167089

RESUMEN

BACKGROUND: The trace amine associated receptor family is a diverse array of GPCRs that arose before the first vertebrates walked on land. Trace amine associated receptor 1 (TAAR1) is a wide spectrum aminergic receptor that acts as a modulator in brain monoaminergic systems. Other trace amine associated receptors appear to relate to environmental perception and show a birth-and-death pattern in mammals similar to olfactory receptors. RESULTS: Across mammals, avians, and amphibians, the TAAR1 gene is intact and appears to be under strong purifying selection based on rates of amino acid fixation compared to neutral mutations. We have found that in dogs it has become a pseudogene. Our analyses using a comparative genetics approach revealed that the pseudogenization event predated the emergence of the Canini tribe rather than being coincident with canine domestication. By assessing the effects of the TAAR1 agonist beta-phenylethylamine on [3H]dopamine uptake in canine striatal synaptosomes and comparing the degree and pattern of uptake inhibition to that seen in other mammals, including TAAR1 knockout mice, wild type mice and rhesus monkey, we found that the TAAR1 pseudogenization event resulted in an uncompensated loss of function. CONCLUSION: The gene family has seen expansions among certain mammals, notably rodents, and reductions in others, including primates. By placing the trace amine associated receptors in an evolutionary context we can better understand their function and their potential associations with behavior and neurological disease.


Asunto(s)
Perros/genética , Evolución Molecular , Seudogenes , Receptores Acoplados a Proteínas G/genética , Animales , Duplicación de Gen , Humanos , Mamíferos/genética
14.
J Virol ; 83(14): 7305-21, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19420076

RESUMEN

The generation of humanized BLT mice by the cotransplantation of human fetal thymus and liver tissues and CD34(+) fetal liver cells into nonobese diabetic/severe combined immunodeficiency mice allows for the long-term reconstitution of a functional human immune system, with human T cells, B cells, dendritic cells, and monocytes/macrophages repopulating mouse tissues. Here, we show that humanized BLT mice sustained high-level disseminated human immunodeficiency virus (HIV) infection, resulting in CD4(+) T-cell depletion and generalized immune activation. Following infection, HIV-specific humoral responses were present in all mice by 3 months, and HIV-specific CD4(+) and CD8(+) T-cell responses were detected in the majority of mice tested after 9 weeks of infection. Despite robust HIV-specific responses, however, viral loads remained elevated in infected BLT mice, raising the possibility that these responses are dysfunctional. The increased T-cell expression of the negative costimulator PD-1 recently has been postulated to contribute to T-cell dysfunction in chronic HIV infection. As seen in human infection, both CD4(+) and CD8(+) T cells demonstrated increased PD-1 expression in HIV-infected BLT mice, and PD-1 levels in these cells correlated positively with viral load and inversely with CD4(+) cell levels. The ability of humanized BLT mice to generate both cellular and humoral immune responses to HIV will allow the further investigation of human HIV-specific immune responses in vivo and suggests that these mice are able to provide a platform to assess candidate HIV vaccines and other immunotherapeutic strategies.


Asunto(s)
Formación de Anticuerpos , Modelos Animales de Enfermedad , Infecciones por VIH/inmunología , VIH/inmunología , Animales , Antígenos de Superficie/genética , Antígenos de Superficie/inmunología , Proteínas Reguladoras de la Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/inmunología , Linfocitos B/inmunología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Células Dendríticas/inmunología , Anticuerpos Anti-VIH/sangre , Infecciones por VIH/genética , Infecciones por VIH/virología , Humanos , Inmunidad Celular , Ratones , Ratones Endogámicos NOD , Ratones SCID , Análisis de Secuencia por Matrices de Oligonucleótidos , Receptor de Muerte Celular Programada 1
15.
Cytokine ; 51(3): 232-9, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20663688

RESUMEN

IL-18 is a proinflammatory cytokine that is important for host defense, but is also involved in the pathogenesis of a number of disease processes, ranging from autoimmune disorders to atherosclerosis. IL-18 binding protein (IL-18BP) is a constitutively expressed glycoprotein that specifically neutralizes the effects of IL-18, resulting in decreased production of IFN-gamma and reduction in Th1 immune responses. In this study we cloned and sequenced a full-length cDNA of the rhesus IL-18BP (RhIL-18BP) from the spleen of rhesus macaques (Macaca mulatta) and compared its nucleotide and amino acid sequences to the functional murine and human IL-18BP orthologues. In addition, we fused RhIL-18BP to the Fc portion of human IgG1 to make recombinant RhIL-18BP x Fcgamma1 in order to facilitate its detection by Western blot analysis and determined the approximate molecular weight of RhIL-18BP x Fcgamma1 to be 66 kD. With this fusion protein, we showed that RhIL-18BP was functional and could significantly reduce murine IL-18 and LPS-induced IFN-gamma production by murine splenocytes. Furthermore, we demonstrated the expression of IL-18BP in atherosclerotic lesions in a rhesus model of atherosclerosis, underscoring the need to fully understand the role of this protein as a primary negative regulator of IL-18 in multiple disease processes.


Asunto(s)
Péptidos y Proteínas de Señalización Intercelular/genética , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Interleucina-18/antagonistas & inhibidores , Macaca mulatta/genética , Secuencia de Aminoácidos , Animales , Aterosclerosis/metabolismo , Secuencia de Bases , Clonación Molecular , Humanos , Inmunoglobulina G/metabolismo , Inmunohistoquímica , Péptidos y Proteínas de Señalización Intercelular/química , Interferón gamma/biosíntesis , Interleucina-18/farmacología , Lipopolisacáridos/farmacología , Ratones , Datos de Secuencia Molecular , Peso Molecular , Transporte de Proteínas/efectos de los fármacos , Proteínas Recombinantes de Fusión/metabolismo , Análisis de Secuencia de Proteína , Bazo/citología , Bazo/efectos de los fármacos , Bazo/metabolismo
16.
Psychopharmacology (Berl) ; 236(5): 1531-1544, 2019 May.
Artículo en Inglés | MEDLINE | ID: mdl-30903211

RESUMEN

RATIONALE: Increasing evidence has demonstrated that changes in the gut microbiome, including those associated with dietary influences, are associated with alterations in many physiological processes. Alcohol consumption is common across human cultures and is likely to have a major effect on the gut microbiome, but there remains a paucity of information on its effects in primates. OBJECTIVES: The effects of chronic alcohol consumption on the primate gut microbiome and metabolome were studied in rhesus macaques that were freely drinking alcohol. The objectives of the study were to determine what changes occurred in the gut microbiome following long-term exposure to alcohol and if these changes were reversible following a period of abstinence. METHODS: Animals consuming alcohol were compared to age-matched controls without access to alcohol and were studied before and after a period of abstinence. Fecal samples from rhesus macaques were used for 16S rRNA sequencing to profile the gut microbiome and for metabolomic profiling using mass spectrometry. RESULTS: Alcohol consumption resulted in a loss of alpha-diversity in rhesus macaques, though this was partially ameliorated by a period of abstinence. Higher levels of Firmicutes were observed in alcohol-drinking animals at the expense of a number of other microbial taxa, again normalizing in part with a period of abstinence. Metabolomic changes were primarily associated with differences in glycolysis when animals were consuming alcohol and differences in fatty acids when alcohol-drinking animals became abstinent. CONCLUSIONS: The consumption of alcohol has specific effects on the microbiome and metabolome of rhesus macaques independent of secondary influences. Many of these changes are reversed by a relatively short period of abstinence.


Asunto(s)
Consumo de Bebidas Alcohólicas/efectos adversos , Etanol/toxicidad , Microbioma Gastrointestinal/efectos de los fármacos , Metaboloma/efectos de los fármacos , Consumo de Bebidas Alcohólicas/genética , Consumo de Bebidas Alcohólicas/metabolismo , Animales , Etanol/administración & dosificación , Heces/microbiología , Microbioma Gastrointestinal/fisiología , Humanos , Macaca mulatta , Masculino , Metaboloma/fisiología , ARN Ribosómico 16S/genética , ARN Ribosómico 16S/metabolismo
17.
J Neurosci Res ; 86(15): 3435-46, 2008 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-18627029

RESUMEN

Several recent studies report an association between trace amine-associated receptor 6 (TAAR6) and susceptibility to schizophrenia and bipolar affective disorder in humans. However, endogenous TAAR6 agonists and the receptor signaling profile and brain distribution remain unclear. Here, we clone TAAR6 from the rhesus monkey and use transfected cells to investigate whether this receptor interacts with brain monoamines and a psychostimulant drug to trigger cAMP signaling or extracellular signal-regulated kinase (ERK) phosphorylation, while investigating its expression profile in the rhesus monkey brain. Unlike TAAR1, rhesus monkey TAAR6 did not alter cAMP levels in response to 10 microM of monoamines (dopamine, norepinephrine, serotonin, beta-phenylethylamine (beta-PEA), octopamine, tryptamine, and tyramine) or methamphetamine in stably transfected cells in vitro. Real-time cell electronic sensing analysis indicated that the receptor did not alter cell impedance or change the effect of forskolin on cell impedance at exposure to 20 microM of each monoamine, suggesting a lack of either Gs or Gi-linked signaling. Whereas kappa opioid receptor activation led to ERK phosphorylation at exposure to 1 microM U69593, rhesus monkey TAAR6 had no such effect at exposure to 10 microM of monoamines or methamphetamine. Membrane and cell surface localization of TAAR6 was confirmed by immunocytochemistry, biotinylation, and Western blot testing with a TAAR6 antibody in the transfected cells. Real-time reverse transcriptase-polymerase chain reaction amplification showed that TAAR6 mRNA was undetectable in selected rhesus monkey brain regions. Together, the data reveal that TAAR6 is unresponsive to brain monoamines and is not expressed in rhesus monkey brain monoaminergic nuclei, suggesting TAAR6 lacks direct association with brain monoaminergic neuronal function.


Asunto(s)
Encéfalo/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal/fisiología , Secuencia de Aminoácidos , Animales , Monoaminas Biogénicas/farmacología , Western Blotting , Encéfalo/efectos de los fármacos , Clonación Molecular , AMP Cíclico/metabolismo , Dopaminérgicos/farmacología , Expresión Génica , Perfilación de la Expresión Génica , Humanos , Inmunohistoquímica , Macaca mulatta , Metanfetamina/farmacología , Datos de Secuencia Molecular , Receptores Acoplados a Proteínas G/efectos de los fármacos , Receptores Acoplados a Proteínas G/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transfección
18.
J Pharmacol Exp Ther ; 325(2): 629-40, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18310473

RESUMEN

In brain monoaminergic systems, common biogenic amines, including dopamine, norepinephrine, and serotonin, serve as neurotransmitters. Monoamine autoreceptors provide feedback regulation in neurotransmitter release, and monoamine transporters clear the released neurotransmitters to control synaptic signaling. Recently, trace amine-associated receptor 1 (TAAR1) has been found to be expressed in brain monoaminergic nuclei and activated by common biogenic amines in vitro. This study used transfected cells and brain synaptosomes to evaluate the interaction of common biogenic amines with TAAR1 and monoamine autoreceptors and explore their modulatory effects on monoamine transporters. We confirmed that TAAR1 was activated by dopamine, norepinephrine, and serotonin and demonstrated that TAAR1 signaling was attenuated by monoamine autoreceptors at exposure to dopamine, norepinephrine, and serotonin. In transfected cells, TAAR1 in response to dopamine, norepinephrine, and serotonin significantly inhibited uptake and promoted efflux of [3H]dopamine, [3H]norepinephrine, and [3H]serotonin, respectively, whereas the monoamine autoreceptors, D2s, alpha(2A), and 5-HT(1B) enhanced the uptake function under the same condition. In brain synaptosomes, dopamine, norepinephrine, and serotonin significantly altered the uptake and efflux of [3H]dopamine, [3H]norepinephrine, and [3H]serotonin, respectively, when the monoamine autoreceptors were blocked. By comparing the effects of dopamine, norepinephrine, and serotonin in monkey and wild-type mouse synaptosomes to their effects in TAAR1 knockout mouse synaptosomes, we deduced that TAAR1 activity inhibited uptake and promoted efflux by monoamine transporters and that monoamine autoreceptors exerted opposite effects. These data provide the first evidence that common biogenic amines modulate monoamine transporter function via both TAAR1 and monoamine autoreceptors, which may balance monoaminergic activity.


Asunto(s)
Monoaminas Biogénicas/farmacología , Encéfalo/metabolismo , Proteínas de Transporte de Dopamina a través de la Membrana Plasmática/metabolismo , Proteínas de Transporte de Noradrenalina a través de la Membrana Plasmática/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Proteínas de Transporte de Serotonina en la Membrana Plasmática/metabolismo , Animales , Encéfalo/efectos de los fármacos , Línea Celular , Humanos , Macaca mulatta , Ratones , Ratones Noqueados , Saguinus , Sinaptosomas/efectos de los fármacos , Sinaptosomas/metabolismo
19.
Transplantation ; 85(6): 893-902, 2008 Mar 27.
Artículo en Inglés | MEDLINE | ID: mdl-18360273

RESUMEN

BACKGROUND: A regimen consisting of polyclonal anti-T-cell antibody, sirolimus (SRL), and donor bone marrow (DBM) infusion induces robust transplantation tolerance to skin allografts in mice. We investigated the effect of a similar regimen in a nonhuman primate (NHP) model. METHODS: Cynomolgus macaques (Macaca fascicularis) were transplanted with mismatched kidney allografts. Recipients were treated with 7 doses of antithymocyte globulin (Thymoglobulin, day 1 to 9), sirolimus, and DBM infusion (day 14). Anti-CD20 antibody, rituximab, was given on days 0 and 5. RESULTS: A regimen of Thymoglobulin, 30 days of SRL, and DBM infusion induced significantly greater prolongation of graft survival with a mean survival time of 88 days compared with the control regimen (no DBM) with an mean survival time of 53 days (P=0.022). Unlike the murine skin allograft model, all grafts were rejected within 111 days. A combination of Thymoglobulin, continuous SRL, and rituximab caused graft and systemic SV40 infection and failed to achieve further extension of graft survival. C4d deposition was observed in 50% of recipients as early as 18 days, suggesting antidonor antibody production. A transient, low-to-moderate degrees of multilineage chimerism was observed after DBM infusion. Treatment with Thymoglobulin resulted in profound depletion of CD4+ and CD8+ T cells, whereas addition of rituximab achieved prolonged (up to 3 months) depletion of CD20+ B cells. CONCLUSION: The Thymoglobulin, SRL, and DBM protocol is simple and produces long-term kidney allograft survival in NHP although additional treatment modalities may be necessary for induction of long-term tolerance.


Asunto(s)
Anticuerpos Monoclonales/efectos adversos , Anticuerpos Monoclonales/uso terapéutico , Trasplante de Riñón/inmunología , Infecciones por Polyomavirus/inducido químicamente , Infecciones Tumorales por Virus/inducido químicamente , Animales , Anticuerpos Monoclonales de Origen Murino , Linfocitos B/inmunología , Trasplante de Médula Ósea , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Complemento C4b/análisis , Factores Inmunológicos/efectos adversos , Factores Inmunológicos/uso terapéutico , Trasplante de Riñón/patología , Recuento de Linfocitos , Macaca fascicularis , Fragmentos de Péptidos/análisis , Infecciones por Polyomavirus/patología , Rituximab , Virus 40 de los Simios , Donantes de Tejidos , Trasplante Homólogo , Infecciones Tumorales por Virus/patología
20.
Rejuvenation Res ; 11(2): 321-32, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18341428

RESUMEN

Alzheimer's disease (AD) is the most common progressive form of dementia in the elderly. Two major neuropathological hallmarks of AD include cerebral deposition of amyloid-beta protein (Abeta) into plaques and blood vessels, and the presence of neurofibrillary tangles in brain. In addition, activated microglia and reactive astrocytes are often associated with plaques and tangles. Numerous other proteins are associated with plaques in human AD brain, including Apo E and ubiquitin. The amyloid precursor protein and its shorter fragment, Abeta, are homologous between humans and non-human primates. Cerebral Abeta deposition has been reported previously for rhesus monkeys, vervets, squirrel monkeys, marmosets, lemurs, cynomologous monkeys, chimpanzees, and orangutans. Here we report, for the first time, age-related neuropathological changes in cotton-top tamarins (CTT, Saguinus oedipus), an endangered non-human primate native to the rainforests of Colombia and Costa Rica. Typical lifespan is 13-14 years of age in the wild and 15-20+ years in captivity. We performed detailed immunohistochemical analyses of Abeta deposition and associated pathogenesis in archived brain sections from 36 tamarins ranging in age from 6-21 years. Abeta plaque deposition was observed in 16 of the 20 oldest tamarins (>12 years). Plaques contained mainly Abeta42, and in the oldest animals, were associated with reactive astrocytes, activated microglia, Apo E, and ubiquitin-positive dystrophic neurites, similar to human plaques. Vascular Abeta was detected in 14 of the 20 aged tamarins; Abeta42 preceded Abeta40 deposition. Phospho-tau labeled dystrophic neurites and tangles, typically present in human AD, were absent in the tamarins. In conclusion, tamarins may represent a model of early AD pathology.


Asunto(s)
Envejecimiento/patología , Péptidos beta-Amiloides/metabolismo , Encéfalo/patología , Modelos Animales de Enfermedad , Saguinus/metabolismo , Enfermedad de Alzheimer/patología , Amiloide/metabolismo , Péptidos beta-Amiloides/sangre , Péptidos beta-Amiloides/inmunología , Animales , Encéfalo/irrigación sanguínea , Colitis/metabolismo , Femenino , Humanos , Masculino , Fragmentos de Péptidos/metabolismo , Placa Amiloide/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA