Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 133
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
J Autoimmun ; 147: 103246, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38788540

RESUMEN

OBJECTIVES: Systemic sclerosis (SSc) is a multiorgan disease with a 10-year mortality rate of up to 50 %. B cell-depleting therapy with rituximab (RTX) appears effective in SSc treatment, but data from randomized controlled trials (RCTs) are lacking, and the frequency and dosage of RTX in SSc have no consensus. We aimed to evaluate the long-term efficacy and safety of quarterly RTX administration in SSc. METHODS: This study retrospectively analyzed 40 patients with SSC treated with RTX twice within 14 days every 3 months from 2010 to 2020. The patients fulfilled the LeRoy and the American College of Rheumatology/European League Against Rheumatism Criteria for SSc. Modified Rodnan skin score (mRSS), lung function test results, and serum immunoglobulin (IgG, IgA, and IgM) concentrations were analyzed. RESULTS: A total of 40 patients with SSc received RTX over a median time of 3.9 years (range: 1-10 years). The median mRSS (baseline: 19, 24 months: 16, p < 0.001) demonstrated a significant improvement, and the predicted forced vital capacity was stable. No new or unexpected safety signals, especially regarding treatment-related infectious adverse events, were observed. Immunoglobulin concentrations were within normal range, and specific antibodies to pneumococcal polysaccharides were preserved despite long-term B cell-depleting therapy. None of the patients died during the observation period of up to 10 years. CONCLUSION: SSc was effectively and safely treated with low-dose RTX quarterly. RCTs are warranted to validate the advantage of continuous B cell depletion by quarterly low-dose RTX administration compared to other treatment intervals.


Asunto(s)
Linfocitos B , Depleción Linfocítica , Rituximab , Esclerodermia Sistémica , Humanos , Esclerodermia Sistémica/mortalidad , Esclerodermia Sistémica/inmunología , Esclerodermia Sistémica/terapia , Esclerodermia Sistémica/tratamiento farmacológico , Femenino , Masculino , Persona de Mediana Edad , Linfocitos B/inmunología , Rituximab/uso terapéutico , Estudios Retrospectivos , Adulto , Resultado del Tratamiento , Anciano
2.
Vet Pathol ; 53(2): 277-90, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26869153

RESUMEN

Aging is the biological process of declining physiologic function associated with increasing mortality rate during advancing age. Humans and higher nonhuman primates exhibit unusually longer average life spans as compared with mammals of similar body mass. Furthermore, the population of humans worldwide is growing older as a result of improvements in public health, social services, and health care systems. Comparative studies among a wide range of organisms that include nonhuman primates contribute greatly to our understanding about the basic mechanisms of aging. Based on their genetic and physiologic relatedness to humans, nonhuman primates are especially important for better understanding processes of aging unique to primates, as well as for testing intervention strategies to improve healthy aging and to treat diseases and disabilities in older people. Rhesus and cynomolgus macaques are the predominant monkeys used in studies on aging, but research with lower nonhuman primate species is increasing. One of the priority topics of research about aging in nonhuman primates involves neurologic changes associated with cognitive decline and neurodegenerative diseases. Additional areas of research include osteoporosis, reproductive decline, caloric restriction, and their mimetics, as well as immune senescence and chronic inflammation that affect vaccine efficacy and resistance to infections and cancer. The purpose of this review is to highlight the findings from nonhuman primate research that contribute to our understanding about aging and health span in humans.


Asunto(s)
Envejecimiento/patología , Primates/fisiología , Investigación , Envejecimiento/genética , Animales , Humanos , Macaca fascicularis , Macaca mulatta , Modelos Animales , Primates/genética
3.
Br J Cancer ; 107(12): 1978-86, 2012 Dec 04.
Artículo en Inglés | MEDLINE | ID: mdl-23169291

RESUMEN

BACKGROUND: Activins control the growth of several tumour types including thoracic malignancies. In the present study, we investigated their expression and function in malignant pleural mesothelioma (MPM). METHODS: The expression of activins and activin receptors was analysed by quantitative PCR in a panel of MPM cell lines. Activin A expression was further analysed by immunohistochemistry in MPM tissue specimens (N=53). Subsequently, MPM cells were treated with activin A, activin receptor inhibitors or activin-targeting siRNA and the impact on cell viability, proliferation, migration and signalling was assessed. RESULTS: Concomitant expression of activin subunits and receptors was found in all cell lines, and activin A was overexpressed in most cell lines compared with non-malignant mesothelial cells. Similarly, immunohistochemistry demonstrated intense staining of tumour cells for activin A in a subset of patients. Treatment with activin A induced SMAD2 phosphorylation and stimulated clonogenic growth of mesothelioma cells. In contrast, treatment with kinase inhibitors of activin receptors (SB-431542, A-8301) inhibited MPM cell viability, clonogenicity and migration. Silencing of activin A expression by siRNA oligonucleotides further confirmed these results and led to reduced cyclin D1/3 expression. CONCLUSION: Our study suggests that activin A contributes to the malignant phenotype of MPM cells via regulation of cyclin D and may represent a valuable candidate for therapeutic interference.


Asunto(s)
Activinas/antagonistas & inhibidores , Antineoplásicos/farmacología , Ciclina D/metabolismo , Mesotelioma/metabolismo , Mesotelioma/patología , Neoplasias Pleurales/metabolismo , Neoplasias Pleurales/patología , Western Blotting , Movimiento Celular , Proliferación Celular , Supervivencia Celular , Ciclina D/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica , Silenciador del Gen , Humanos , Inmunohistoquímica , Mesotelioma/tratamiento farmacológico , Fenotipo , Fosforilación/efectos de los fármacos , Neoplasias Pleurales/tratamiento farmacológico , ARN Interferente Pequeño/farmacología , Reacción en Cadena en Tiempo Real de la Polimerasa , Transducción de Señal/efectos de los fármacos , Proteína Smad2/metabolismo , Ensayo de Tumor de Célula Madre , Regulación hacia Arriba
4.
Neuropathol Appl Neurobiol ; 38(2): 162-74, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21696421

RESUMEN

AIMS: The pathogenesis of human/simian immunodeficiency virus encephalitis (HIVE/SIVE) remains incompletely understood, but is associated with alterations in the blood-brain barrier. At present, it is not possible to easily determine if an individual has HIVE/SIVE before post mortem examination. METHODS: We have examined serum levels of the astroglial protein S100ß in SIV-infected macaques and show that it can be used to determine which animals have SIVE. We also checked for correlations with inflammatory markers such as CCL2/MCP-1, IL-6 and C-reactive protein. RESULTS: We found that increased S100ß protein in serum correlated with decreased expression of the tight junction protein zonula occludens-1 on brain microvessels. Furthermore, the decrease in zonula occludens-1 expression was spatially related to SIVE lesions and perivascular deposition of plasma fibrinogen. There was no correlation between encephalitis and plasma levels of IL-6, MCP-1/CCL2 or C-reactive protein. CONCLUSIONS: Together, these data indicate that SIVE lesions are associated with vascular leakage that can be determined by S100ß protein in the periphery. The ability to simply monitor the presence of SIVE will greatly facilitate studies of the neuropathogenesis of AIDS.


Asunto(s)
Síndrome de Inmunodeficiencia Adquirida/sangre , Encefalitis Viral/sangre , Encefalitis Viral/diagnóstico , Monocitos/metabolismo , Factores de Crecimiento Nervioso/sangre , Proteínas S100/sangre , Síndrome de Inmunodeficiencia Adquirida/complicaciones , Animales , Biomarcadores/sangre , Barrera Hematoencefálica/patología , Encéfalo/patología , Proteína C-Reactiva/metabolismo , Quimiocina CCL2/sangre , Encefalitis Viral/complicaciones , Interleucina-6/sangre , Macaca mulatta , Proteínas de la Membrana/metabolismo , Fosfoproteínas/metabolismo , Subunidad beta de la Proteína de Unión al Calcio S100 , Virus de la Inmunodeficiencia de los Simios , Uniones Estrechas/metabolismo , Proteína de la Zonula Occludens-1
5.
J Med Primatol ; 41(3): 191-201, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22429048

RESUMEN

Non-human primates (NHPs) are used to model human disease owing to their remarkably similar genomes, physiology, and immune systems. Recently, there has been an increased interest in modeling tuberculosis (TB) in NHPs. Macaques are susceptible to infection with different strains of Mycobacterium tuberculosis (Mtb), producing the full spectrum of disease conditions, including latent infection, chronic progressive infection, and acute TB, depending on the route and dose of infection. Clearly, NHPs are an excellent model of human TB. While the initial aim of the NHP model was to allow preclinical testing of candidate vaccines and drugs, it is now also being used to study pathogenesis and immune correlates of protection. Recent advances in this field are discussed in this review. Key questions such as the effect of hypoxia on the biology of Mtb and the basis of reactivation of latent TB can now be investigated through the use of this model.


Asunto(s)
Modelos Animales de Enfermedad , Macaca , Enfermedades de los Monos/microbiología , Mycobacterium tuberculosis/clasificación , Tuberculosis/microbiología , Tuberculosis/veterinaria , Animales , Humanos , Mycobacterium tuberculosis/genética , Radiografía , Tuberculosis/diagnóstico por imagen
6.
Br J Cancer ; 104(8): 1303-12, 2011 Apr 12.
Artículo en Inglés | MEDLINE | ID: mdl-21407220

RESUMEN

BACKGROUND: Activins and inhibins belong to the TGFß-superfamily, which controls cell proliferation and differentiation in many organs. Activin A, the dimer of inhibin ßA subunit, acts strongly anti-proliferative in hepatocytes. Little is known on the other activin/inhibin subunits in human liver and hepatocellular carcinoma (HCC). METHODS: We studied the expression of the complete inhibin family α, ßA, ßB, ßC, ßE in normal liver, tumour-adjacent and HCC tissue, 12 additional organs and rodent liver. A total of 16 HCC and 10 disease-free livers were analysed. Expression of inhibin subunits was determined by qRT-PCR, normalised to RNA input and by geNorm algorithm, and confirmed by immunohistochemistry. RESULTS: Remarkably, ßA expression was not decreased in HCC. Similarly, ßC and ßE exhibited no major changes. In contrast, inhibin α, barely detectable in normal liver, was strongly increased in tumour-adjacent liver and dramatically enhanced in HCC. ßB was strongly enhanced in some HCC. At variance with human liver, rodent liver showed higher inhibin α and ßC expression, but ßA was somewhat, and ßB dramatically lower. CONCLUSIONS: Upregulation of inhibin α - and possibly of ßB - may shield HCC cells from anti-proliferative effects of activin A. Dramatic variations between humans and rodents may reflect different functions of some inhibins/activins.


Asunto(s)
Activinas/genética , Carcinoma Hepatocelular/genética , Inhibinas/genética , Neoplasias Hepáticas/genética , Hígado/metabolismo , Roedores/genética , Activinas/metabolismo , Adulto , Anciano , Animales , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patología , Estudios de Casos y Controles , Femenino , Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Inhibinas/metabolismo , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Subunidades de Proteína/genética , Subunidades de Proteína/metabolismo , Ratas , Ratas Wistar , Roedores/metabolismo
7.
Nat Med ; 3(1): 32-6, 1997 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-8986737

RESUMEN

A major safety concern of using live-attenuated vaccine strategies against AIDS is the potential exposure of neonates or fetuses to vaccine virus from the mother. Here we report that high viral loads and disease were observed in only 2 of 18 neonatal monkeys infected with gene-deleted vaccine strains of simian immunodeficiency virus. Pathogenicity was restricted to neonates born to unvaccinated mothers, that is, lacking maternal immunity, and that received extremely high doses of vaccine virus orally. No in utero transmission of vaccine virus was observed in four neonates born to mothers vaccinated during the second trimester. Our results suggest that the live attenuated vaccine approach should remain a viable option for preventing HIV infection and disease in high-risk human populations.


Asunto(s)
Animales Recién Nacidos/inmunología , Animales Recién Nacidos/virología , Inmunidad Materno-Adquirida/inmunología , Macaca mulatta/inmunología , Vacunas contra el SIDAS/inmunología , Animales , Linfocitos T CD4-Positivos , Relación Dosis-Respuesta a Droga , Femenino , Feto/inmunología , Feto/virología , Macaca mulatta/virología , Mutación , Embarazo , Vacunas contra el SIDAS/farmacología , Virus de la Inmunodeficiencia de los Simios/genética , Virus de la Inmunodeficiencia de los Simios/inmunología , Vacunas Atenuadas/inmunología , Vacunas Atenuadas/farmacología
8.
Nat Med ; 4(4): 435-40, 1998 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-9546789

RESUMEN

At a position equivalent to the gene encoding the saimiri transforming protein (STP) of herpesvirus saimiri (HVS), Kaposi's sarcoma-associated herpesvirus (KSHV) contains a distinct open reading frame called K1. Although KSHV and HVS are related members of the rhadinovirus subgroup of gamma herpesviruses, K1 and STP exhibit no similarity in amino acid sequence or in structural organization. Since STP is required for the oncogenic potential of HVS, we investigated the functional consequence of K1 expression. Expression of the K1 gene in rodent fibroblasts produced morphologic changes and focus formation indicative of transformation. A recombinant herpesvirus in which the STP oncogene of HVS was replaced with K1, immortalized primary T lymphocytes to IL-2 independent growth and induced lymphoma in common marmosets. These results demonstrate the transforming potential of the K1 gene of KSHV.


Asunto(s)
Genes Virales , Herpesvirus Humano 8/genética , Sistemas de Lectura Abierta , Sarcoma de Kaposi/virología , Proteínas Virales/biosíntesis , Proteínas Virales/genética , Síndrome de Inmunodeficiencia Adquirida/complicaciones , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Células COS , Línea Celular , Transformación Celular Neoplásica , Cartilla de ADN , ADN Viral/genética , Herpesvirus Saimiriino 2/genética , Herpesvirus Saimiriino 2/patogenicidad , Humanos , Masculino , Datos de Secuencia Molecular , Proteínas Oncogénicas Virales/biosíntesis , Proteínas Oncogénicas Virales/química , Proteínas Oncogénicas Virales/genética , Reacción en Cadena de la Polimerasa , Ratas , Proteínas Recombinantes de Fusión/biosíntesis , Proteínas Recombinantes de Fusión/química , Sarcoma de Kaposi/etiología , Sarcoma de Kaposi/patología , Alineación de Secuencia , Homología de Secuencia de Aminoácido , Transfección , Proteínas Virales/química
9.
Nat Med ; 2(10): 1084-9, 1996 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-8837605

RESUMEN

Simian immunodeficiency virus (SIV) can cross the intact vaginal epithelium to establish a systemic infection in macaques (mac). Using this SIVmac model, we found that subcutaneous progesterone implants, which could mimic hormonally based contraceptives, thinned the vaginal epithelium and enhanced SIV vaginal transmission 7.7-fold over that observed in macaques treated with placebo implants and exposed to SIV in the follicular phase of the menstrual cycle. Progesterone treatment also increased the number of SIV DNA-positive cells in the vaginal lamina propria as detected by in situ polymerase chain reaction analysis. Moreover, plasma viral RNA was elevated for the first three months in macaques with progesterone implants, and three of the progesterone-treated macaques developed relatively rapid disease courses. This study shows that SIV genital infection and disease course are enhanced by subcutaneous implants containing progesterone when compared with the rate of vaginal transmission in the follicular phase.


Asunto(s)
Progesterona/farmacología , Síndrome de Inmunodeficiencia Adquirida del Simio/transmisión , Virus de la Inmunodeficiencia de los Simios/fisiología , Vagina/inmunología , Viremia/virología , Animales , Anticuerpos Antivirales/sangre , ADN Viral/análisis , Progresión de la Enfermedad , Susceptibilidad a Enfermedades , Implantes de Medicamentos , Epitelio/efectos de los fármacos , Epitelio/inmunología , Epitelio/ultraestructura , Femenino , Fase Folicular , Leucocitos Mononucleares/virología , Macaca mulatta , Membrana Mucosa/efectos de los fármacos , Membrana Mucosa/inmunología , Membrana Mucosa/ultraestructura , Progesterona/administración & dosificación , Provirus/aislamiento & purificación , Vagina/efectos de los fármacos , Vagina/ultraestructura
10.
Nat Med ; 5(9): 997-103, 1999 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-10470075

RESUMEN

Infection of macrophage lineage cells is a feature of primate lentivirus replication, and several properties of primate lentiviruses seem to have evolved to promote the infection of macrophages. Here we demonstrate that the accessory gene product Nef induces the production of two CC-chemokines, macrophage inflammatory proteins 1alpha and 1beta, by HIV-1-infected macrophages. Adenovirus-mediated expression of Nef in primary macrophages was sufficient for chemokine induction. Supernatants from Nef-expressing macrophages induced both the chemotaxis and activation of resting T lymphocytes, permitting productive HIV-1 infection. These results indicate a role for Nef in lymphocyte recruitment and activation at sites of virus replication.


Asunto(s)
Quimiotaxis , Productos del Gen nef/fisiología , VIH-1/fisiología , Activación de Linfocitos , Macrófagos/virología , Linfocitos T/inmunología , Adenoviridae/genética , Animales , Línea Celular , Células Cultivadas , Quimiocina CCL4 , Quimiocinas/biosíntesis , Quimiocinas/genética , Quimiotaxis/efectos de los fármacos , Medios de Cultivo Condicionados , Citocinas/biosíntesis , Encefalitis Viral/inmunología , Encefalitis Viral/virología , Productos del Gen nef/genética , VIH-1/efectos de los fármacos , VIH-1/genética , VIH-1/crecimiento & desarrollo , Humanos , Activación de Linfocitos/efectos de los fármacos , Macaca , Proteínas Inflamatorias de Macrófagos/biosíntesis , Proteínas Inflamatorias de Macrófagos/genética , Macrófagos/inmunología , Macrófagos/metabolismo , Mutación , Virus de la Inmunodeficiencia de los Simios/efectos de los fármacos , Virus de la Inmunodeficiencia de los Simios/crecimiento & desarrollo , Virus de la Inmunodeficiencia de los Simios/fisiología , Linfocitos T/efectos de los fármacos , Linfocitos T/virología , Replicación Viral/efectos de los fármacos , Productos del Gen nef del Virus de la Inmunodeficiencia Humana
11.
B-ENT ; 7(2): 121-5, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21838097

RESUMEN

OBJECTIVES: To increase awareness of neuroendocrine adenomas of the middle ear (NAME), rare lesions often mistaken for other entities or chronic otitis media. Histogenesis remains controversial, although the consensus tends toward a pluripotent stem cell of the middle ear mucosa as the origin of the lesion. The tumour is characterised by dual differentiation with exocrine and endocrine components. The most common symptoms are conductive hearing loss, tinnitus and vertigo. The treatment of choice is complete surgical removal of the tumour with no adjuvant radiotherapy being required. CASE REPORT: We report the case of a 23-year-old man presenting with chronic otitis media, conductive hearing loss, vertigo and tinnitus who, some years previously, had suffered from an episode of facial nerve palsy. Conservative therapy failed and so surgery was performed. Tumour-like masses were encountered and histological and immunohistochemical examination revealed a neuroendocrine adenoma of the middle ear. CONCLUSION: This rare entity should be considered as differential diagnosis when treating chronic inflammatory disease not responding to conservative therapy or dealing with unclear expansive processes of the middle ear. MRI scans should be performed since CT scans are inconclusive.


Asunto(s)
Adenoma/diagnóstico , Neoplasias del Oído/diagnóstico , Oído Medio , Parálisis Facial/diagnóstico , Tumores Neuroendocrinos/diagnóstico , Otitis Media/diagnóstico , Adenoma/cirugía , Audiometría , Enfermedad Crónica , Diagnóstico Diferencial , Neoplasias del Oído/cirugía , Estudios de Seguimiento , Humanos , Imagen por Resonancia Magnética , Masculino , Tumores Neuroendocrinos/cirugía , Procedimientos Quirúrgicos Otológicos/métodos , Tomografía Computarizada por Rayos X , Adulto Joven
12.
J Exp Med ; 192(1): 11-21, 2000 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-10880522

RESUMEN

The B cell antigen receptor (BCR) is a large complex that consists of a disulfide-linked tetramer of two transmembrane heavy (mu) chains and two light (lambda or kappa) chains in association with a heterodimer of Igalpha and Igbeta. Kaposi's sarcoma-associated herpesvirus (KSHV) encodes a transforming protein called K1, which has structural and functional similarity to Igalpha and Igbeta. We demonstrate that K1 downregulates the expression of BCR complexes on the surface. The NH(2)-terminal region of K1 specifically interacts with the mu chains of BCR complexes, and this interaction retains BCR complexes in the endoplasmic reticulum, preventing their intracellular transport to the cell surface. Thus, KSHV K1 resembles Igalpha and Igbeta in its ability to induce signaling and to interact with mu chains of the BCR. However, unlike Igalpha and Igbeta, which interact with mu chains to direct BCR complexes to the cell surface, K1 interacts with mu chains to block the intracellular transport of BCR complexes to the cell surface. These results demonstrate a unique feature of the K1 transforming protein, which may confer virus-infected cells with a long-term survival advantage.


Asunto(s)
Herpesvirus Humano 8/fisiología , Proteínas de la Membrana/fisiología , Receptores de Antígenos de Linfocitos B/fisiología , Proteínas del Envoltorio Viral/fisiología , Proteínas Virales/fisiología , Sitios de Unión , Línea Celular , Regulación hacia Abajo , Herpesvirus Humano 8/genética , Humanos , Cadenas kappa de Inmunoglobulina/química , Cadenas kappa de Inmunoglobulina/fisiología , Cadenas lambda de Inmunoglobulina/química , Cadenas lambda de Inmunoglobulina/fisiología , Cadenas mu de Inmunoglobulina/química , Cadenas mu de Inmunoglobulina/fisiología , Proteínas de la Membrana/genética , Sistemas de Lectura Abierta , Receptores de Antígenos de Linfocitos B/química , Receptores de Antígenos de Linfocitos B/genética , Proteínas del Envoltorio Viral/genética , Proteínas Virales/genética
13.
J Exp Med ; 193(8): 905-15, 2001 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-11304551

RESUMEN

The macrophage is well established as a target of HIV and simian immunodeficiency virus (SIV) infection and a major contributor to the neuropathogenesis of AIDS. However, the identification of distinct subpopulations of monocyte/macrophages that carry virus to the brain and that sustain infection within the central nervous system (CNS) has not been examined. We demonstrate that the perivascular macrophage and not the parenchymal microglia is the primary cell productively infected by SIV. We further demonstrate that although productive viral infection of the CNS occurs early, thereafter it is not easily detectable until terminal AIDS. The biology of perivascular macrophages, including their rate of turnover and replacement by peripheral blood monocytes, may explain the timing of neuroinvasion, disappearance, and reappearance of virus in the CNS, and questions the ability of the brain to function as a reservoir for productive infection by HIV/SIV.


Asunto(s)
Encéfalo/virología , Macrófagos/virología , Síndrome de Inmunodeficiencia Adquirida del Simio/patología , Virus de la Inmunodeficiencia de los Simios/fisiología , Complejo SIDA Demencia/patología , Complejo SIDA Demencia/virología , Animales , Antígenos de Diferenciación/análisis , Encéfalo/patología , Circulación Cerebrovascular , ADN Viral/análisis , Humanos , Inmunofenotipificación , Macaca mulatta , Macrófagos/inmunología , Macrófagos/patología , Microglía/patología , Microglía/virología , Microscopía Confocal , ARN Viral/análisis , Virus de la Inmunodeficiencia de los Simios/genética , Virus de la Inmunodeficiencia de los Simios/aislamiento & purificación , Proteínas Virales/análisis
14.
J Exp Med ; 187(11): 1767-78, 1998 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-9607918

RESUMEN

The thymus plays a critical role in the maturation and production of T lymphocytes and is a target of infection by human immunodeficiency virus (HIV) and the related simian immunodeficiency virus (SIV). Using the SIV/macaque model of AIDS, we examined the early effects of SIV on the thymus. We found that thymic infection by SIV resulted in increased apoptosis 7-14 d after infection, followed by depletion of thymocyte progenitors by day 21. A marked rebound in thymocyte progenitors occurred by day 50 and was accompanied by increased levels of cell proliferation in the thymus. Our results demonstrate a marked increase in thymic progenitor activity very early in the course of SIV infection, long before marked declines in peripheral CD4(+) T cell counts.


Asunto(s)
Síndrome de Inmunodeficiencia Adquirida del Simio/patología , Virus de la Inmunodeficiencia de los Simios , Células Madre , Timo/patología , Animales , Apoptosis , División Celular , Macaca mulatta , Masculino , Regeneración , Síndrome de Inmunodeficiencia Adquirida del Simio/inmunología , Síndrome de Inmunodeficiencia Adquirida del Simio/virología , Timo/inmunología , Timo/virología
15.
Science ; 248(4959): 1109-12, 1990 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-2160735

RESUMEN

Better understanding of the pathogenesis of acquired immunodeficiency syndrome (AIDS) would be greatly facilitated by a relevant animal model that uses molecularly cloned virus of defined sequence to induce the disease. Such a system would also be of great value for AIDS vaccine research. An infectious molecular clone of simian immunodeficiency virus (SIV) was identified that induces AIDS in common rhesus monkeys in a time frame suitable for laboratory investigation. These results provide another strong link in the chain of evidence for the viral etiology of AIDS. More importantly, they define a system for molecular dissection of the determinants of AIDS pathogenesis.


Asunto(s)
Síndrome de Inmunodeficiencia Adquirida , Modelos Animales de Enfermedad , Infecciones por Retroviridae , Virus de la Inmunodeficiencia de los Simios , Animales , Anticuerpos Antivirales/biosíntesis , Clonación Molecular , Leucocitos Mononucleares/microbiología , Macaca mulatta , Macrófagos/microbiología , Infecciones Oportunistas/etiología , Infecciones por Retroviridae/complicaciones , Infecciones por Retroviridae/inmunología , Virus de la Inmunodeficiencia de los Simios/genética , Virus de la Inmunodeficiencia de los Simios/inmunología , Virus de la Inmunodeficiencia de los Simios/aislamiento & purificación , Virus de la Inmunodeficiencia de los Simios/patogenicidad , Transfección , Replicación Viral
16.
Science ; 280(5362): 427-31, 1998 Apr 17.
Artículo en Inglés | MEDLINE | ID: mdl-9545219

RESUMEN

Human and simian immunodeficiency virus (HIV and SIV) replicate optimally in activated memory CD4(+) T cells, a cell type that is abundant in the intestine. SIV infection of rhesus monkeys resulted in profound and selective depletion of CD4+ T cells in the intestine within days of infection, before any such changes in peripheral lymphoid tissues. The loss of CD4+ T cells in the intestine occurred coincident with productive infection of large numbers of mononuclear cells at this site. The intestine appears to be a major target for SIV replication and the major site of CD4+ T cell loss in early SIV infection.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Colon/inmunología , Intestino Delgado/inmunología , Síndrome de Inmunodeficiencia Adquirida del Simio/inmunología , Síndrome de Inmunodeficiencia Adquirida del Simio/virología , Virus de la Inmunodeficiencia de los Simios/fisiología , Animales , Recuento de Linfocito CD4 , Linfocitos T CD4-Positivos/virología , Colon/virología , Inmunidad Mucosa , Memoria Inmunológica , Mucosa Intestinal/inmunología , Mucosa Intestinal/virología , Intestino Delgado/virología , Activación de Linfocitos , Linfocitos/inmunología , Linfocitos/virología , Tejido Linfoide/inmunología , Tejido Linfoide/virología , Macaca mulatta , Macrófagos/virología , Masculino , Receptores de Interleucina-2/análisis , Virus de la Inmunodeficiencia de los Simios/inmunología , Virus de la Inmunodeficiencia de los Simios/patogenicidad , Carga Viral , Virulencia , Replicación Viral
17.
Cell Immunol ; 248(2): 69-76, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17980355

RESUMEN

It has been well established that serotonin (5-hydroxytryptamine, 5-HT) plays a key role in neuro-endocrine-immune networks, mostly through its receptors and/or transporters. Although the presence of 5-HT receptor mRNAs in peripheral blood mononuclear cells (PBMCs) of rhesus monkeys has been reported, there is little information about serotonin transporter (SERT) expression by these cells. To examine SERT expression at the transcription and translation level, one-step RT-PCR, confocal microscopy and flow cytometry were used to detect SERT mRNA and protein expression by rhesus monkey PBMCs. It was found that SERT mRNA could be detected by RT-PCR from all of the rhesus macaque PBMC RNA samples and the nucleotide sequence of the amplicons was identical to the published SERT mRNA sequence. Low level SERT immunoreactivity was also demonstrated on the surface of rhesus PBMCs by confocal microscopy. Almost all lymphocytes and most monocytes were positive for SERT by flow cytometry. In the 2 rhesus macaques examined by multicolor flow cytometry, SERT(bright) cells were more than 84%, 94%, and 96% among CD20+, CD3+, and CD3+CD4+ lymphocytes respectively. These data demonstrate expression of SERT by rhesus macaque PBMCs, and indicate that rhesus macaques would be suitable models to test the in vivo immune regulatory effects of 5-HT or drugs targeting SERT.


Asunto(s)
Leucocitos Mononucleares/metabolismo , Proteínas de Transporte de Serotonina en la Membrana Plasmática/biosíntesis , Proteínas de Transporte de Serotonina en la Membrana Plasmática/genética , Animales , Citometría de Flujo , Macaca mulatta , Microscopía Confocal , ARN Mensajero/biosíntesis
18.
Mucosal Immunol ; 9(4): 1089-101, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-26647718

RESUMEN

Innate immune responses have a critical role in the control of early virus replication and dissemination. It remains unknown, however, how severe acute respiratory syndrome coronavirus (SARS-CoV) evades respiratory innate immunity to establish a systemic infection. Here we show in Chinese macaques that SARS-CoV traversed the mucosa through the respiratory tract within 2 days, resulting in extensive mucosal infiltration by T cells, MAC387(+), and CD163(+) monocytes/macrophages followed by limited viral replication in the lung but persistent viral shedding into the upper airway. Mucosal monocytes/macrophages sequestered virions in intracellular vesicles together with infected Langerhans cells and migrated into the tonsils and/or draining lymph nodes within 2 days. In lymphoid tissues, viral RNA and proteins were detected in infected monocytes upon differentiation into dendritic cells (DCs) within 3 days. Systemic viral dissemination was observed within 7 days. This study provides a comprehensive overview of the spatiotemporal interactions of SARS-CoV, monocytes/macrophages, and the DC network in mucosal tissues and highlights the fact that, while these innate cells contribute to viral clearance, they probably also serve as shelters and vehicles to provide a mechanism for the virus to escape host mucosal innate immunity and disseminate systemically.


Asunto(s)
Coronavirus/fisiología , Células Dendríticas/inmunología , Macaca mulatta/inmunología , Macrófagos/inmunología , Mucosa Respiratoria/inmunología , Síndrome Respiratorio Agudo Grave/inmunología , Linfocitos T/inmunología , Animales , Diferenciación Celular , Movimiento Celular , Células Cultivadas , Células Dendríticas/virología , Humanos , Evasión Inmune , Inmunidad Innata , Macrófagos/virología , ARN Viral/inmunología , Mucosa Respiratoria/virología , Linfocitos T/virología , Carga Viral , Replicación Viral , Esparcimiento de Virus
19.
Circulation ; 101(2): 185-93, 2000 Jan 18.
Artículo en Inglés | MEDLINE | ID: mdl-10637207

RESUMEN

BACKGROUND: Cardiomyopathy is being recognized with increasing frequency in patients with AIDS, yet the relationship between HIV infection and cardiac contractile dysfunction remains obscure. The purpose of the present study was to determine if infection with simian immunodeficiency virus (SIV) in nonhuman primates is associated with cardiac dysfunction and myocardial injury. METHODS AND RESULTS: Left ventricular size and function were determined by 2D echocardiography in 16 rhesus macaques before and at weekly intervals following infection with cloned pathogenic SIV(mac) 239 or the highly attenuated SIV(mac) 239 nef deletion mutant. A second group of 15 rhesus macaques chronically infected with pathogenic (n=6) or nonpathogenic (n=9) virus were studied at >2 years following infection. Cardiac tissues from 24 rhesus macaques chronically infected (>2 years) with pathogenic SIV were reviewed for evidence of cardiac pathology. Acute infection (<6 weeks) with either pathogenic or nonpathogenic SIV caused neither contractile dysfunction nor cardiac pathology. However, LV ejection fraction was significantly (P<0.05) depressed (43+/-7%) in rhesus macaques chronically infected with pathogenic SIV compared with rhesus macaques chronically infected with nonpathogenic SIV (61+/-3%). Furthermore, two thirds of rhesus macaques that succumbed to simian AIDS had myocardial pathology including lymphocytic myocarditis (n=9) and coronary arteriopathy (n=6), with complete vessel occlusion (n=4) and associated myocardial infarction and necrosis. CONCLUSIONS: This unique model is valuable in understanding the pathogenesis of cardiac injury associated with retroviral infection in a relevant nonhuman primate model of AIDS.


Asunto(s)
Cardiomiopatía Dilatada/virología , Síndrome de Inmunodeficiencia Adquirida del Simio/complicaciones , Enfermedad Aguda , Animales , Sistema Cardiovascular/patología , Sistema Cardiovascular/fisiopatología , Enfermedad Crónica , Inmunohistoquímica , Macaca mulatta , Miocardio/metabolismo , Miocardio/patología , Síndrome de Inmunodeficiencia Adquirida del Simio/metabolismo , Síndrome de Inmunodeficiencia Adquirida del Simio/mortalidad , Síndrome de Inmunodeficiencia Adquirida del Simio/patología , Síndrome de Inmunodeficiencia Adquirida del Simio/fisiopatología , Función Ventricular Izquierda
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA