Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
1.
Sci Rep ; 13(1): 11414, 2023 07 14.
Artículo en Inglés | MEDLINE | ID: mdl-37452120

RESUMEN

Our study utilizes a longitudinal isotopic metabolic labeling approach in vivo in combination with organelle fraction proteomics to address the role of parkin in mitochondrial protein turnover in mice. The use of metabolic labeling provides a method to quantitatively determine the global changes in protein half-lives whilst simultaneously assessing protein expression. Studying two diverse mitochondrial populations, we demonstrated the median half-life of brain striatal synaptic mitochondrial proteins is significantly greater than that of hepatic mitochondrial proteins (25.7 vs. 3.5 days). Furthermore, loss of parkin resulted in an overall, albeit modest, increase in both mitochondrial protein abundance and half-life. Pathway and functional analysis of our proteomics data identified both known and novel pathways affected by loss of parkin that are consistent with its role in both mitochondrial quality control and neurodegeneration. Our study therefore adds to a growing body of evidence suggesting dependence on parkin is low for basal mitophagy in vivo and provides a foundation for the investigation of novel parkin targets.


Asunto(s)
Proteoma , Ubiquitina-Proteína Ligasas , Animales , Ratones , Proteoma/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Mitocondrias/metabolismo , Mitofagia , Proteínas Mitocondriales/metabolismo
2.
iScience ; 25(5): 104207, 2022 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-35494221

RESUMEN

In the era of antiretroviral therapy, inflammation is a central factor in numerous HIV-associated comorbidities, such as cardiovascular disease, cognitive impairment, and neuropsychiatric disorders. This highlights the value of developing therapeutics that both reduce HIV-associated inflammation and treat associated comorbidities. Previous research on monoamine oxidase inhibitors (MAOIs) suggests this class of drugs has anti-inflammatory properties in addition to neuropsychiatric effects. Therefore, we examined the impact of deprenyl, an MAOI, on SIV-associated inflammation during acute SIV infection using the rhesus macaque model of HIV infection. Our results show deprenyl decreased both peripheral and CNS inflammation but had no effect on viral load in either the periphery or CNS. These data show that the MAOI deprenyl may have broad anti-inflammatory effects when given during the acute stage of SIV infection, suggesting more research into the anti-inflammatory effects of this drug could result in a beneficial adjuvant for antiretroviral therapy.

3.
Cell Death Dis ; 5: e982, 2014 Jan 09.
Artículo en Inglés | MEDLINE | ID: mdl-24407235

RESUMEN

Regulation of cellular proliferation and differentiation during brain development results from processes requiring several regulatory networks to function in synchrony. MicroRNAs are part of this regulatory system. Although many microRNAs are evolutionarily conserved, recent evolution of such regulatory molecules can enable the acquisition of new means of attaining specialized functions. Here we identify and report the novel expression and functions of a human and higher primate-specific microRNA, miR-1290, in neurons. Using human fetal-derived neural progenitors, SH-SY5Y neuroblastoma cell line and H9-ESC-derived neural progenitors (H9-NPC), we found miR-1290 to be upregulated during neuronal differentiation, using microarray, northern blotting and qRT-PCR. We then conducted knockdown and overexpression experiments to look at the functional consequences of perturbed miR-1290 levels. Knockdown of miR-1290 inhibited differentiation and induced proliferation in differentiated neurons; correspondingly, miR-1290 overexpression in progenitors led to a slowing down of the cell cycle and differentiation to neuronal phenotypes. Consequently, we identified that crucial cell cycle proteins were aberrantly changed in expression level. Therefore, we conclude that miR-1290 is required for maintaining neurons in a differentiated state.


Asunto(s)
Proteínas de Ciclo Celular/genética , Evolución Molecular , MicroARNs/metabolismo , Mitosis , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Neurogénesis , Proteínas de Ciclo Celular/metabolismo , Proliferación Celular , Regulación del Desarrollo de la Expresión Génica , Humanos , MicroARNs/genética
4.
Cell Death Dis ; 3: e381, 2012 Aug 30.
Artículo en Inglés | MEDLINE | ID: mdl-22932723

RESUMEN

Neuronal damage is a hallmark feature of HIV-associated neurological disorders (HANDs). Opiate drug abuse accelerates the incidence and progression of HAND; however, the mechanisms underlying the potentiation of neuropathogenesis by these drugs remain elusive. Opiates such as morphine have been shown to enhance HIV transactivation protein Tat-mediated toxicity in both human neurons and neuroblastoma cells. In the present study, we demonstrate reduced expression of the tropic factor platelet-derived growth factor (PDGF)-B with a concomitant increase in miR-29b in the basal ganglia region of the brains of morphine-dependent simian immunodeficiency virus (SIV)-infected macaques compared with the SIV-infected controls. In vitro relevance of these findings was corroborated in cultures of astrocytes exposed to morphine and HIV Tat that led to increased release of miR-29b in exosomes. Subsequent treatment of neuronal SH-SY5Y cell line with exosomes from treated astrocytes resulted in decreased expression of PDGF-B, with a concomitant decrease in viability of neurons. Furthermore, it was shown that PDGF-B was a target for miR-29b as evidenced by the fact that binding of miR-29 to the 3'-untranslated region of PDGF-B mRNA resulted in its translational repression in SH-SY5Y cells. Understanding the regulation of PDGF-B expression may provide insights into the development of potential therapeutic targets for neuronal loss in HIV-1-infected opiate abusers.


Asunto(s)
Analgésicos Opioides/farmacología , Exosomas/metabolismo , Regulación de la Expresión Génica , MicroARNs/metabolismo , Morfina/farmacología , Neuronas/metabolismo , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/metabolismo , Regiones no Traducidas 3' , Animales , Astrocitos/metabolismo , Células Cultivadas , Modelos Animales de Enfermedad , Regulación hacia Abajo/efectos de los fármacos , Ganglios/efectos de los fármacos , Ganglios/metabolismo , Humanos , Macaca mulatta , Neuronas/efectos de los fármacos , Proteínas Proto-Oncogénicas c-sis/genética , Proteínas Proto-Oncogénicas c-sis/metabolismo , Ratas , Virus de la Inmunodeficiencia de los Simios/patogenicidad , Transfección , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/genética
5.
J Neurovirol ; 18(2): 113-9, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22403026

RESUMEN

Plasma gelsolin (pGSN), an isoform 1, is secreted by various types of cells in the central nervous system (CNS) and periphery, but not by the liver. pGSN circulates in blood and cerebrospinal fluid (CSF); however, its concentration in CSF is approximately twenty times lower than in plasma. It has been shown that several types of cells such as oligodendrocytes, neurons, and/or astrocytes contribute to the overall pool of pGSN in the CNS. Further, it has been postulated that pGSN plays multiple roles during microbial infection and modulates inflammatory responses; however, the exact mechanism of regulation is not known. We previously showed that levels of pGSN in CSF of individuals with advanced neurocognitive impairment due to HIV infection of the brain are decreased. Here, we show that macrophages express significant amounts of pGSN in response to HIV infection in vitro. Using immunohistochemistry of simian immunodeficiency virus infected rhesus monkey brains, we show that increased levels of pGSN are present in macrophage nodules creating locally a high level of this protein within the brain. This may not be reflected by the overall decreased level in the distinct CSF compartment.


Asunto(s)
Encéfalo/metabolismo , Gelsolina/sangre , Macrófagos/metabolismo , Síndrome de Inmunodeficiencia Adquirida del Simio/sangre , Virus de la Inmunodeficiencia de los Simios , Animales , Encéfalo/patología , Encéfalo/virología , Femenino , Inmunohistoquímica , Macaca mulatta , Macrófagos/patología , Macrófagos/virología , Masculino , Síndrome de Inmunodeficiencia Adquirida del Simio/patología , Síndrome de Inmunodeficiencia Adquirida del Simio/virología , Replicación Viral
6.
Cell Death Dis ; 2: e246, 2011 Dec 22.
Artículo en Inglés | MEDLINE | ID: mdl-22190005

RESUMEN

Mitochondrial dysfunction is implicated in a majority of neurodegenerative disorders and much study of neurodegenerative disease is done on cultured neurons. In traditional tissue culture, the oxygen level that cells experience is dramatically higher (21%) than in vivo conditions (1-11%). These differences can alter experimental results, especially, pertaining to mitochondria and oxidative metabolism. Our results show that primary neurons cultured at physiological oxygen levels found in the brain showed higher polarization, lower rates of ROS production, larger mitochondrial networks, greater cytoplasmic fractions of mitochondria and larger mitochondrial perimeters than those cultured at higher oxygen levels. Although neurons cultured in either physiological oxygen or atmospheric oxygen exhibit significant increases in mitochondrial reactive oxygen species (ROS) production when treated with the human immunodeficiency virus (HIV) virotoxin trans-activator of transcription, mitochondria of neurons cultured at physiological oxygen underwent depolarization with dramatically increased cell death, whereas those cultured at atmospheric oxygen became hyperpolarized with no increase in cell death. Studies with a second HIV virotoxin, negative regulation factor (Nef), revealed that Nef treatment also increased mitochondrial ROS production for both the oxygen conditions, but resulted in mitochondrial depolarization and increased death only in neurons cultured in physiological oxygen. These results indicate a role for oxidative metabolism in a mechanism of neurotoxicity during HIV infection and demonstrate the importance of choosing the correct, physiological, culture oxygen in mitochondrial studies performed in neurons.


Asunto(s)
VIH/metabolismo , Mitocondrias/metabolismo , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Oxígeno/metabolismo , Péptidos Cíclicos/farmacología , Adenosina Trifosfato/metabolismo , Apoptosis , Células Cultivadas , Productos del Gen tat/farmacología , Humanos , Microscopía Confocal , Mitocondrias/química , Neuronas/citología , Especies Reactivas de Oxígeno/metabolismo , Productos del Gen nef del Virus de la Inmunodeficiencia Humana/farmacología
7.
Cell Death Dis ; 1: e77, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-21170291

RESUMEN

MicroRNAs (miRNAs) play important roles in regulating a plethora of physiological and pathophysiogical processes including neurodegeneration. In both HIV associated dementia in humans and its monkey model SIV encephalitis we find miR-21, a miRNA largely known for its link to oncogenesis, to be significantly upregulated in the brain. In situ hybridization of the diseased brain sections revealed induction of miR-21 in neurons. MiR-21 can be induced in neurons by prolonged N-methyl-D-aspartic acid receptor stimulation, an excitotoxic process active in HIV and other neurodegenerative diseases. Introduction of miR-21 into human neurons leads to pathological functional defects. Furthermore, we show that miR-21 specifically targets the mRNA of myocyte enhancer factor 2C (MEF2C), a transcription factor crucial for neuronal function, and reduces its expression. MEF2C is dramatically downregulated in neurons of HIV-associated dementia patients as well as monkeys with SIVE. Together, this study elucidates a novel role for miR-21 in the brain, not only as a potential signature of neurological disease but also as a crucial effector of HIV induced neuronal dysfunction and neurodegeneration.


Asunto(s)
Infecciones por VIH/genética , MicroARNs/metabolismo , Factores Reguladores Miogénicos/genética , Enfermedades Neurodegenerativas/genética , Neuronas/metabolismo , Síndrome de Inmunodeficiencia Adquirida del Simio/genética , Animales , Regulación hacia Abajo , Infecciones por VIH/metabolismo , Haplorrinos , Humanos , Factores de Transcripción MEF2 , Factores Reguladores Miogénicos/metabolismo , Enfermedades Neurodegenerativas/metabolismo , Enfermedades Neurodegenerativas/virología , ARN Mensajero/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Síndrome de Inmunodeficiencia Adquirida del Simio/metabolismo , Síndrome de Inmunodeficiencia Adquirida del Simio/virología
8.
Rheumatol Int ; 25(1): 28-32, 2005 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-13680143

RESUMEN

We planned to investigate the expression of protein kinase C (PKC) isoforms in acinar epithelial cells of salivary glands in the non-obese diabetic (NOD) mouse to find out if they develop changes of the PKC system like those seen in the human counterpart, i.e. in Sjögren's syndrome. Parotid, submandibular, and sublingual glands from NOD and control BALB/c mice were stained with a panel of monoclonal antibodies directed against conventional (alpha, beta, and gamma), novel (delta, epsilon, and theta), and atypical (lambda and iota) PKC isoforms using the streptavidin/HRP method. Similarly to human labial salivary glands, acinar epithelial cells of the healthy control BALB/c mice contained two of the conventional PKC isoforms, alpha and beta. Acinar and ductal epithelial cells also contained the atypical PKC isoforms lambda and iota. PKC isoforms gamma, delta, epsilon, and theta were not found. NOD mice which displayed focal sialadenitis contained the same conventional and atypical PKC isoforms. The acinar cells in NOD mice, in contrast to the Sjögren's syndrome patients, did not lack PKC alpha or beta. On the contrary, PKC alpha and beta staining was stronger than in the control BALB/c mice. The present results demonstrate that both conventional and atypical PKC isoforms participate in the salivary epithelial cell biology and that there are mouse strain-associated and/or disease state-associated changes in their expression. The lack of PKC alpha and beta isoforms found in Sjögren's syndrome was not reproduced in NOD mice, which discloses one more difference between the human disease and its NOD mouse model.


Asunto(s)
Diabetes Mellitus Tipo 1/enzimología , Células Epiteliales/enzimología , Proteína Quinasa C/metabolismo , Glándulas Salivales/enzimología , Síndrome de Sjögren/enzimología , Animales , Diabetes Mellitus Tipo 1/patología , Modelos Animales de Enfermedad , Técnicas para Inmunoenzimas , Isoenzimas/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos NOD , Transducción de Señal , Síndrome de Sjögren/patología
9.
Biotechniques ; 33(3): 516-8, 520, 522 passim, 2002 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-12238761

RESUMEN

There exists a significant limitation in the variety of organismsfor which microarrays have been developed because of a lack of genomic sequence data. A near-term solution to this limitation is to use microarrays designed for one species to analyze RNA samples from closely related species. The assumption is that conservation of gene sequences between species will be sufficient to generate a reasonable amount of good-quality data. While there have been relatively few published reports describing the use of microarrays for cross-species hybridizations, this technique is potentially a powerful tool for understanding genomics in model organisms such as nonhuman primates. Here we describe the analysis and comparison of hybridization characteristics and data variability from a set of cross-species (rhesus macaque) and same-species (human) hybridization experiments using human high-density Affymetrix oligonucleotide arrays. The data reveal that a large fraction of probe sets are effective at transcript detection in the cross-species hybridization, validating the application of cross-species hybridizations for nonhuman primate genomics research.


Asunto(s)
Sondas de ADN/genética , Variación Genética , Hibridación Genética/genética , Análisis de Secuencia por Matrices de Oligonucleótidos/métodos , ARN/análisis , ARN/genética , Especificidad de la Especie , Análisis de Varianza , Animales , Humanos , Macaca mulatta , Análisis de Secuencia por Matrices de Oligonucleótidos/instrumentación , Reproducibilidad de los Resultados , Sensibilidad y Especificidad
11.
J Immunol ; 167(9): 5429-38, 2001 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-11673562

RESUMEN

One of the consequences of HIV infection is damage to the CNS. To characterize the virologic, immunologic, and functional factors involved in HIV-induced CNS disease, we analyzed the viral loads and T cell infiltrates in the brains of SIV-infected rhesus monkeys whose CNS function (sensory evoked potential) was impaired. Following infection, CNS evoked potentials were abnormal, indicating early CNS disease. Upon autopsy at 11 wk post-SIV inoculation, the brains of infected animals contained over 5-fold more CD8(+) T cells than did uninfected controls. In both infected and uninfected groups, these CD8(+) T cells presented distinct levels of activation markers (CD11a and CD95) at different sites: brain > CSF > spleen = blood > lymph nodes. The CD8(+) cells obtained from the brains of infected monkeys expressed mRNA for cytolytic and proinflammatory molecules, such as granzymes A and B, perforin, and IFN-gamma. Therefore, the neurological dysfunctions correlated with increased numbers of CD8(+) T cells of an activated phenotype in the brain, suggesting that virus-host interactions contributed to the related CNS functional defects.


Asunto(s)
Complejo SIDA Demencia/etiología , Encefalopatías/etiología , Encéfalo/inmunología , Linfocitos T CD8-positivos/inmunología , Activación de Linfocitos , Síndrome de Inmunodeficiencia Adquirida del Simio/inmunología , Animales , Antígenos CD28/análisis , Inmunofenotipificación , Antígeno-1 Asociado a Función de Linfocito/análisis , Macaca mulatta , Síndrome de Inmunodeficiencia Adquirida del Simio/complicaciones , Receptor fas/análisis
13.
J Virol ; 75(15): 7067-77, 2001 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-11435587

RESUMEN

The CXC chemokine gamma interferon (IFN-gamma)-inducible protein CXCL10/IP-10 is markedly elevated in cerebrospinal fluid and brain of individuals infected with human immunodeficiency virus type 1 (HIV-1) and is implicated in the pathogenesis of HIV-associated dementia (HAD). To explore the possible role of CXCL10/IP-10 in HAD, we examined the expression of this and other chemokines in the central nervous system (CNS) of transgenic mice with astrocyte-targeted expression of HIV gp120 under the control of the glial fibrillary acidic protein (GFAP) promoter, a murine model for HIV-1 encephalopathy. Compared with wild-type controls, CNS expression of the CC chemokine gene CCL2/MCP-1 and the CXC chemokine genes CXCL10/IP-10 and CXCL9/Mig was induced in the GFAP-HIV gp120 mice. CXCL10/IP-10 RNA expression was increased most and overlapped the expression of the transgene-encoded HIV gp120 gene. Astrocytes and to a lesser extent microglia were identified as the major cellular sites for CXCL10/IP-10 gene expression. There was no detectable expression of any class of IFN or their responsive genes. In astrocyte cultures, soluble recombinant HIV gp120 protein was capable of directly inducing CXCL10/IP-10 gene expression a process that was independent of STAT1. These findings highlight a novel IFN- and STAT1-independent mechanism for the regulation of CXCL10/IP-10 expression and directly link expression of HIV gp120 to the induction of CXCL10/IP-10 that is found in HIV infection of the CNS. Finally, one function of IP-10 expression may be the recruitment of leukocytes to the CNS, since the brain of GFAP-HIV gp120 mice had increased numbers of CD3(+) T cells that were found in close proximity to sites of CXCL10/IP-10 RNA expression.


Asunto(s)
Astrocitos/metabolismo , Quimiocinas CXC/genética , Proteína gp120 de Envoltorio del VIH/metabolismo , VIH-1/metabolismo , Animales , Astrocitos/citología , Encéfalo/metabolismo , Encéfalo/patología , Células Cultivadas , Quimiocina CXCL10 , Quimiocinas CXC/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/fisiología , Expresión Génica , Proteína Ácida Fibrilar de la Glía/genética , Proteína gp120 de Envoltorio del VIH/genética , VIH-1/genética , Humanos , Interferón-alfa/metabolismo , Interferón-alfa/farmacología , Interferón gamma/metabolismo , Interferón gamma/farmacología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Receptores CXCR3 , Receptores de Quimiocina/genética , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Factor de Transcripción STAT1 , Solubilidad , Linfocitos T/citología , Transactivadores/genética , Transactivadores/fisiología
14.
J Clin Invest ; 106(1): 37-45, 2000 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-10880046

RESUMEN

Simian immunodeficiency virus (SIV) infection of rhesus monkeys provides an excellent model of the central nervous system (CNS) consequences of HIV infection. To discern the relationship between viral load and abnormalities induced in the CNS by the virus, we infected animals with SIV and later instituted antiviral treatment to lower peripheral viral load. Measurement of sensory-evoked potentials, assessing CNS neuronal circuitry, revealed delayed latencies after infection that could be reversed by lowering viral load. Cessation of treatment led to the reappearance of these abnormalities. In contrast, the decline in general motor activity induced by SIV infection was unaffected by antiviral treatment. An acute increase in the level of the chemokine monocyte chemoattractant protein-1 (MCP-1) was found in the cerebrospinal fluid (CSF) relative to plasma in the infected animals at the peak of acute viremia, likely contributing to an early influx of immune cells into the CNS. Examination of the brains of the infected animals after return of the electrophysiological abnormalities revealed diverse viral and inflammatory findings. Although some of the physiological abnormalities resulting from SIV infection can be at least temporarily reversed by lowering viral load, the viral-host interactions initiated by infection may result in long-lasting changes in CNS-mediated functions.


Asunto(s)
Antivirales/uso terapéutico , Encéfalo/fisiopatología , Trastornos del Movimiento/tratamiento farmacológico , Síndrome de Inmunodeficiencia Adquirida del Simio/tratamiento farmacológico , Animales , Barrera Hematoencefálica , Encéfalo/efectos de los fármacos , Encéfalo/virología , Potenciales Evocados Auditivos del Tronco Encefálico/efectos de los fármacos , Macaca mulatta , Actividad Motora/efectos de los fármacos , Síndrome de Inmunodeficiencia Adquirida del Simio/fisiopatología , Síndrome de Inmunodeficiencia Adquirida del Simio/virología , Virus de la Inmunodeficiencia de los Simios/aislamiento & purificación
15.
J Virol ; 74(3): 1415-24, 2000 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-10627552

RESUMEN

Infection of C57BL/6 mice with mouse hepatitis virus (MHV) results in a demyelinating encephalomyelitis characterized by mononuclear cell infiltration and white matter destruction similar to the pathology of the human demyelinating disease multiple sclerosis. The contributions of CD4(+) and CD8(+) T cells in the pathogenesis of the disease were investigated. Significantly less severe inflammation and demyelination were observed in CD4(-/-) mice than in CD8(-/-) and C57BL/6 mice (P < or = 0.002 and P < or = 0.001, respectively). Immunophenotyping of central nervous system (CNS) infiltrates revealed that CD4(-/-) mice had a significant reduction in numbers of activated macrophages/microglial cells in the brain compared to the numbers in CD8(-/-) and C57BL/6 mice, indicating a role for these cells in myelin destruction. Furthermore, CD4(-/-) mice displayed lower levels of RANTES (a C-C chemokine) mRNA transcripts and protein, suggesting a role for this molecule in the pathogenesis of MHV-induced neurologic disease. Administration of RANTES antisera to MHV-infected C57BL/6 mice resulted in a significant reduction in macrophage infiltration and demyelination (P < or = 0.001) compared to those in control mice. These data indicate that CD4(+) T cells have a pivotal role in accelerating CNS inflammation and demyelination within infected mice, possibly by regulating RANTES expression, which in turn coordinates the trafficking of macrophages into the CNS, leading to myelin destruction.


Asunto(s)
Linfocitos T CD4-Positivos/fisiología , Quimiocina CCL5/fisiología , Infecciones por Coronavirus/patología , Enfermedades Desmielinizantes/patología , Encefalomielitis/patología , Virus de la Hepatitis Murina/patogenicidad , Animales , Encéfalo/patología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Quimiocina CCL5/biosíntesis , Quimiocina CCL5/inmunología , Infecciones por Coronavirus/inmunología , Infecciones por Coronavirus/virología , Enfermedades Desmielinizantes/inmunología , Enfermedades Desmielinizantes/virología , Encefalomielitis/inmunología , Encefalomielitis/virología , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Sueros Inmunes , Inmunohistoquímica , Hibridación in Situ , Masculino , Ratones , Ratones Endogámicos C57BL , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Médula Espinal/patología
16.
J Neurovirol ; 5(1): 48-54, 1999 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-10190690

RESUMEN

Infection of C57BL/6 mice with mouse hepatitis virus strain V5A13.1 (MHV-V5A13.1) results in an acute encephalitis followed by a chronic, progressive demyelinating disease with clinical and histological similarities to the human demyelinating disease Multiple Sclerosis (MS). Studies were undertaken to evaluate the contribution of NOS2 generated NO in demyelination in MHV-infected mice. MHV-infected animals were treated daily with either 8 mg of aminoguanidine (AG), a selective inhibitor of NOS2 activity, or PBS by intraperitoneal (i.p.) injection. MHV-infection of mice resulted in 20% mortality in both groups with surviving mice clearing virus below levels of detection, as measured by plaque assay, by day 12 postinfection (p.i.). A significant decrease in the severity of clinical disease was observed in AG-treated animals as compared to mice receiving PBS at days 7 and 12 p.i. (P< or =0.001 and 0.003, respectively) however, by day 21 p.i. AG-treated mice exhibited the same severity of clinical disease as control animals. Examination of brain and spinal cords from infected mice revealed a pronounced reduction in the severity of inflammation at day 7 p.i. in mice treated with AG as compared to control mice. By day 12 p.i. there was a significant decrease (P< or =0.02) in the severity of demyelination in AG-treated mice as compared to control animals yet both PBS and AG treated mice had a similar degree of demyelination by day 21 p.i. Analysis of chemokine mRNA transcripts by RNase protection assay revealed that AG-treated mice had significantly lower levels (P < or = 0.007) of transcripts for the C-C chemokine monocyte chemoattractant protein-1 (MCP-1) at day 7 p.i. as compared to control animals. By day 12 p.i., AG-treated mice and control mice had similar levels of chemokine transcripts. Together, these data suggest that inhibition of NOS2/NO slows the progression of MHV-induced demyelination. One potential mechanism by which this may occur is through controlling inflammation through modulation of chemokine expression in the CNS.


Asunto(s)
Quimiocinas/metabolismo , Infecciones por Coronavirus/metabolismo , Enfermedades Desmielinizantes/metabolismo , Virus de la Hepatitis Murina , Óxido Nítrico Sintasa/antagonistas & inhibidores , Óxido Nítrico/metabolismo , Animales , Encéfalo/metabolismo , Quimiocina CCL2/genética , Quimiocina CCL2/metabolismo , Quimiocinas/genética , Infecciones por Coronavirus/complicaciones , Enfermedades Desmielinizantes/etiología , Inhibidores Enzimáticos/farmacología , Guanidinas/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Óxido Nítrico Sintasa/metabolismo , ARN Mensajero/análisis , Factores de Tiempo
17.
Lab Invest ; 79(12): 1719-26, 1999 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-10616220

RESUMEN

By inducing both adhesion and migration of lymphocytes, chemokines play an important role in immune and inflammatory responses. To learn how these processes promote disease, we have examined the activities of chemokines in the lacrimal glands (LG) of nonobese diabetic (NOD) mice, an animal model of Sjogren's syndrome (SS). The expression of three molecules in the chemokine superfamily, RANTES, IP-10 and lymphotactin, correlated with the local recruitment of lymphocytes into the LG of NOD mice. Both RANTES and IP-10 gene transcripts were first detected in these LG when the mice were 8 weeks of age and amounts increased markedly during the course of active disease; lymphotactin mRNA was also expressed but at lower levels. In situ hybridization of LG indicated that lymphocytic cells in the inflammatory infiltrates were responsible for the production of RANTES and IP-10. Concomitant with the induction of chemokine expression was the appearance of cellular receptors for RANTES (CCR1, CCR5) and IP-10 (CXCR3). Furthermore, anti-RANTES treatment significantly reduced inflammation in the LG from NOD mice. In the SS-like disease of NOD mice, this distinct pattern of activity provides evidence for the contribution of these components to site- and time-specific recruitment of lymphocytes in the characteristic destruction of glandular structures.


Asunto(s)
Quimiocina CCL5/metabolismo , Quimiocinas C , Aparato Lagrimal/patología , Linfocinas/metabolismo , Factor Plaquetario 4/metabolismo , Sialoglicoproteínas/metabolismo , Animales , Secuencia de Bases , Cartilla de ADN , Expresión Génica , Ratones , Ratones Endogámicos NOD , Receptores de Quimiocina/genética , Receptores de Quimiocina/metabolismo
18.
J Gend Specif Med ; 2(5): 33-40, 1999.
Artículo en Inglés | MEDLINE | ID: mdl-11252833

RESUMEN

OBJECTIVE: To identify the targets of estrogen in immune system lymphocytes and to examine gender differences in autoimmunity. DESIGN: RNA samples from purified lymphocyte subsets were analyzed for the presence of mRNA for estrogen receptor alpha and beta (ER alpha and ER beta). Groups of male, female, and testicular-feminized mice were compared for autoantibody production. SUBJECTS: Autoimmune-prone lpr (Fas-deficient), testicular-feminized (Tfm, androgen receptor-deficient) and wild-type mice were studied. METHOD: Lymphocyte subsets were purified by fluorescence-activated cell sorting (FACS) and RNA was assessed for the presence of estrogen receptor sequences using specific oligonucleotide primers and the reverse transcription-polymerase chain reaction (RT-PCR). Spontaneous and induced antibody production in mice was determined by enzyme-linked immunosorbent assay (ELISA). RESULTS: ER alpha was expressed in all lymphocyte subsets examined. ER beta was expressed at low levels in thymic CD4/CD8- T cells in wild-type mice and at high levels in the peripheral CD4-/CD8- T cells in lpr mice. Both spontaneous and induced autoantibody production was higher in female lpr mice than in male lpr mice. CONCLUSIONS: The presence of ERs in lymphocytes indicates that estrogen may affect immune cells during their development and mature function. The selective expression of ER beta may help explain some of the physiological effects of estrogen and its pharmacologic analogues and may lead to means to direct estrogen analogues to such cells. Such effects may be explored in lpr mice, given the enhanced capacity of female lpr mice for autoantibody production.


Asunto(s)
Enfermedades Autoinmunes/metabolismo , Autoinmunidad , Receptores de Estrógenos/metabolismo , Caracteres Sexuales , Linfocitos T/metabolismo , Síndrome de Resistencia Androgénica/inmunología , Síndrome de Resistencia Androgénica/metabolismo , Animales , Autoanticuerpos/biosíntesis , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD8-positivos/metabolismo , Ensayo de Inmunoadsorción Enzimática , Receptor alfa de Estrógeno , Receptor beta de Estrógeno , Estrógenos/metabolismo , Femenino , Masculino , Ratones , Ratones Mutantes , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Linfocitos T/inmunología
19.
Proc Natl Acad Sci U S A ; 95(25): 15072-7, 1998 Dec 08.
Artículo en Inglés | MEDLINE | ID: mdl-9844017

RESUMEN

Central nervous system (CNS) damage and dysfunction are devastating consequences of HIV infection. Although the CNS is one of the initial targets for HIV infection, little is known about early viral-induced abnormalities that can affect CNS function. Here we report the detection of early physiological abnormalities in simian immunodeficiency virus-infected monkeys. The acute infection caused a disruption of the circadian rhythm manifested by rises in body temperature, observed in all five individuals between 1 and 2 weeks postinoculation (p.i.), accompanied by a reduction in daily motor activity to 50% of control levels. Animals remained hyperthermic at 1 and 2 months p.i. and returned to preinoculation temperatures at 3 months after viral inoculation. Although motor activity recovered to baseline values at 1 month p.i., activity levels then decreased to approximately 50% of preinoculation values over the next 2 months. Analysis of sensory-evoked responses 1 month p.i. revealed distinct infection-induced changes in auditory-evoked potential peak latencies that persisted at 3 months after viral inoculation. These early physiological abnormalities may precede the development of observable cognitive or motor deficiencies and can provide an assay to evaluate agents to prevent or alleviate neuronal dysfunction.


Asunto(s)
Encéfalo/fisiopatología , Síndrome de Inmunodeficiencia Adquirida del Simio/fisiopatología , Virus de la Inmunodeficiencia de los Simios/aislamiento & purificación , Animales , Temperatura Corporal , Ritmo Circadiano , Macaca mulatta , Masculino , Actividad Motora
20.
J Med Primatol ; 27(2-3): 104-12, 1998.
Artículo en Inglés | MEDLINE | ID: mdl-9747951

RESUMEN

A model is proposed in which a neurovirulent, microglial-passaged, simian immunodeficiency virus (SIV) is used to produce central nervous system (CNS) pathology and behavioral deficits in rhesus monkeys reminiscent of those seen in humans infected with human immunodeficiency virus (HIV). The time course of disease progression was characterized by using functional measures of cognition and motor skill, as well as neurophysiologic monitoring. Concomitant assessment of immunological and virological parameters illustrated correspondence between impaired behavioral performance and viral pathogenesis. Convergent results were obtained from neuropathological findings indicative of significant CNS disease. In ongoing studies, this SIV model is being used to explore the behavioral sequelae of immunodeficiency virus infection, the viral and host factors leading to neurologic dysfunction, and to begin testing potential therapeutic agents.


Asunto(s)
Síndrome de Inmunodeficiencia Adquirida del Simio/fisiopatología , Síndrome de Inmunodeficiencia Adquirida del Simio/psicología , Virus de la Inmunodeficiencia de los Simios/fisiología , Animales , Cognición , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Infecciones por VIH/inmunología , Infecciones por VIH/fisiopatología , Infecciones por VIH/psicología , Humanos , Estudios Longitudinales , Macaca mulatta , Actividad Motora , Pruebas Neuropsicológicas , Síndrome de Inmunodeficiencia Adquirida del Simio/inmunología , Virus de la Inmunodeficiencia de los Simios/aislamiento & purificación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...