Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 95
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
PLoS Pathog ; 19(4): e1010941, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-37115795

RESUMEN

The encapsulated fungus Cryptococcus neoformans is the most common cause of fungal meningitis, with the highest rate of disease in patients with AIDS or immunosuppression. This microbe enters the human body via inhalation of infectious particles. C. neoformans capsular polysaccharide, in which the major component is glucuronoxylomannan (GXM), extensively accumulates in tissues and compromises host immune responses. C. neoformans travels from the lungs to the bloodstream and crosses to the brain via transcytosis, paracytosis, or inside of phagocytes using a "Trojan horse" mechanism. The fungus causes life-threatening meningoencephalitis with high mortality rates. Hence, we investigated the impact of intranasal exogenous GXM administration on C. neoformans infection in C57BL/6 mice. GXM enhances cryptococcal pulmonary infection and facilitates fungal systemic dissemination and brain invasion. Pre-challenge of GXM results in detection of the polysaccharide in lungs, serum, and surprisingly brain, the latter likely reached through the nasal cavity. GXM significantly alters endothelial cell tight junction protein expression in vivo, suggesting significant implications for the C. neoformans mechanisms of brain invasion. Using a microtiter transwell system, we showed that GXM disrupts the trans-endothelial electrical resistance, weakening human brain endothelial cell monolayers co-cultured with pericytes, supportive cells of blood vessels/capillaries found in the blood-brain barrier (BBB) to promote C. neoformans BBB penetration. Our findings should be considered in the development of therapeutics to combat the devastating complications of cryptococcosis that results in an estimated ~200,000 deaths worldwide each year.


Asunto(s)
Criptococosis , Cryptococcus neoformans , Meningitis Criptocócica , Animales , Ratones , Humanos , Cryptococcus neoformans/metabolismo , Roedores , Ratones Endogámicos C57BL , Criptococosis/microbiología , Polisacáridos/metabolismo , Pulmón/metabolismo
2.
Cell Mol Life Sci ; 80(4): 116, 2023 Apr 04.
Artículo en Inglés | MEDLINE | ID: mdl-37016051

RESUMEN

HIV infection has become a chronic and manageable disease due to the effective use of antiretroviral therapies (ART); however, several chronic aging-related comorbidities, including cognitive impairment, remain a major public health issue. However, these mechanisms are unknown. Here, we identified that glial and myeloid viral reservoirs are associated with local myelin damage and the release of several myelin components, including the lipid sulfatide. Soluble sulfatide compromised gap junctional communication and calcium wave coordination, essential for proper cognition. We propose that soluble sulfatide could be a potential biomarker and contributor to white matter compromise observed in HIV-infected individuals even in the current ART era.


Asunto(s)
Infecciones por VIH , Sustancia Blanca , Humanos , Infecciones por VIH/tratamiento farmacológico , Infecciones por VIH/complicaciones , Sulfoglicoesfingolípidos , Daño Encefálico Crónico/complicaciones , Comunicación Celular
3.
Cell Mol Life Sci ; 79(7): 365, 2022 Jun 16.
Artículo en Inglés | MEDLINE | ID: mdl-35708858

RESUMEN

SARS-CoV-2, although not being a circulatory virus, spread from the respiratory tract resulting in multiorgan failures and thrombotic complications, the hallmarks of fatal COVID-19. A convergent contributor could be platelets that beyond hemostatic functions can carry infectious viruses. Here, we profiled 52 patients with severe COVID-19 and demonstrated that circulating platelets of 19 out 20 non-survivor patients contain SARS-CoV-2 in robust correlation with fatal outcome. Platelets containing SARS-CoV-2 might originate from bone marrow and lung megakaryocytes (MKs), the platelet precursors, which were found infected by SARS-CoV-2 in COVID-19 autopsies. Accordingly, MKs undergoing shortened differentiation and expressing anti-viral IFITM1 and IFITM3 RNA as a sign of viral sensing were enriched in the circulation of deadly COVID-19. Infected MKs reach the lung concomitant with a specific MK-related cytokine storm rich in VEGF, PDGF and inflammatory molecules, anticipating fatal outcome. Lung macrophages capture SARS-CoV-2-containing platelets in vivo. The virus contained by platelets is infectious as capture of platelets carrying SARS-CoV-2 propagates infection to macrophages in vitro, in a process blocked by an anti-GPIIbIIIa drug. Altogether, platelets containing infectious SARS-CoV-2  alter COVID-19 pathogenesis and provide a powerful fatality marker. Clinical targeting of platelets might prevent viral spread, thrombus formation and exacerbated inflammation at once and increase survival in COVID-19.


Asunto(s)
COVID-19 , Trombosis , Plaquetas , Humanos , Pulmón , Megacariocitos , Proteínas de la Membrana , Proteínas de Unión al ARN , SARS-CoV-2
4.
PLoS Pathog ; 16(6): e1008381, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32525948

RESUMEN

HIV invades the brain during acute infection. Yet, it is unknown whether long-lived infected brain cells release productive virus that can egress from the brain to re-seed peripheral organs. This understanding has significant implication for the brain as a reservoir for HIV and most importantly HIV interplay between the brain and peripheral organs. Given the sheer number of astrocytes in the human brain and their controversial role in HIV infection, we evaluated their infection in vivo and whether HIV infected astrocytes can support HIV egress to peripheral organs. We developed two novel models of chimeric human astrocyte/human peripheral blood mononuclear cells: NOD/scid-IL-2Rgc null (NSG) mice (huAstro/HuPBMCs) whereby we transplanted HIV (non-pseudotyped or VSVg-pseudotyped) infected or uninfected primary human fetal astrocytes (NHAs) or an astrocytoma cell line (U138MG) into the brain of neonate or adult NSG mice and reconstituted the animals with human peripheral blood mononuclear cells (PBMCs). We also transplanted uninfected astrocytes into the brain of NSG mice and reconstituted with infected PBMCs to mimic a biological infection course. As expected, the xenotransplanted astrocytes did not escape/migrate out of the brain and the blood brain barrier (BBB) was intact in this model. We demonstrate that astrocytes support HIV infection in vivo and egress to peripheral organs, at least in part, through trafficking of infected CD4+ T cells out of the brain. Astrocyte-derived HIV egress persists, albeit at low levels, under combination antiretroviral therapy (cART). Egressed HIV evolved with a pattern and rate typical of acute peripheral infection. Lastly, analysis of human cortical or hippocampal brain regions of donors under cART revealed that astrocytes harbor between 0.4-5.2% integrated HIV gag DNA and 2-7% are HIV gag mRNA positive. These studies establish a paradigm shift in the dynamic interaction between the brain and peripheral organs which can inform eradication of HIV reservoirs.


Asunto(s)
Astrocitos , Barrera Hematoencefálica , Infecciones por VIH , VIH-1/metabolismo , Hipocampo , Liberación del Virus , Animales , Antirretrovirales/farmacología , Astrocitos/metabolismo , Astrocitos/patología , Astrocitos/virología , Barrera Hematoencefálica/metabolismo , Barrera Hematoencefálica/patología , Barrera Hematoencefálica/virología , Línea Celular Tumoral , Infecciones por VIH/genética , Infecciones por VIH/metabolismo , Infecciones por VIH/patología , VIH-1/genética , Hipocampo/metabolismo , Hipocampo/patología , Hipocampo/virología , Humanos , Ratones , Ratones Endogámicos NOD , Ratones Noqueados , Ratones SCID
5.
J Immunol ; 205(10): 2726-2741, 2020 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-33037140

RESUMEN

HIV has become a chronic disease despite the effective use of antiretroviral therapy (ART). However, the mechanisms of tissue colonization, viral evolution, generation of viral reservoirs, and compartmentalization are still a matter of debate due to the challenges involved in examining early events of infection at the cellular and molecular level. Thus, there is still an urgent need to explore these areas to develop effective HIV cure strategies. In this study, we describe the early events of tissue colonization and compartmentalization as well as the role of tunneling nanotube-like structures during viral spread in the presence and absence of effective antiretroviral treatment. To examine these mechanisms, NOD/SCID IL-2 RG-/- humanized mice were either directly infected with HIVADA or with low numbers of HIVADA-infected leukocytes to limit tissue colonization in the presence and absence of TAK779, an effective CCR5 blocker of HIV entry. We identify that viral seeding in tissues occurs early in a tissue- and cell type-specific manner (24-72 h). Reduction in systemic HIV replication by TAK779 treatment did not affect tissue seeding or spreading, despite reduced systemic viral replication. Tissue-associated HIV-infected cells had different properties than cells in the circulation because the virus continues to spread in tissues in a tunneling nanotube-like structure-dependent manner, despite ART. Thus, understanding these mechanisms can provide new approaches to enhance the efficacy of existing ART and HIV infection cure strategies.


Asunto(s)
Antirretrovirales/administración & dosificación , Linfocitos T CD4-Positivos/citología , Linfocitos T CD4-Positivos/virología , Infecciones por VIH/inmunología , VIH-1/patogenicidad , Amidas/administración & dosificación , Animales , Linfocitos T CD4-Positivos/inmunología , Modelos Animales de Enfermedad , Infecciones por VIH/sangre , Infecciones por VIH/tratamiento farmacológico , Infecciones por VIH/virología , VIH-1/inmunología , VIH-1/aislamiento & purificación , Trasplante de Células Madre Hematopoyéticas , Humanos , Subunidad gamma Común de Receptores de Interleucina/genética , Ratones , Ratones Noqueados , Compuestos de Amonio Cuaternario/administración & dosificación , Quimera por Trasplante , Carga Viral , Integración Viral/efectos de los fármacos , Integración Viral/inmunología , Internalización del Virus/efectos de los fármacos , Replicación Viral/efectos de los fármacos , Replicación Viral/inmunología
6.
J Neurochem ; 158(2): 429-443, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33655498

RESUMEN

The major barrier to eradicating Human immunodeficiency virus-1 (HIV) infection is the generation of tissue-associated quiescent long-lasting viral reservoirs refractory to therapy. Upon interruption of anti-retroviral therapy (ART), HIV replication can be reactivated. Within the brain, microglia/macrophages and a small population of astrocytes are infected with HIV. However, the role of astrocytes as a potential viral reservoir is becoming more recognized because of the improved detection and quantification of HIV viral reservoirs. In this report, we examined the infectivity of human primary astrocytes in vivo and in vitro, and their capacity to maintain HIV infection, become latently infected, be reactivated, and transfer new HIV virions into neighboring cells. Analysis of human brain tissue sections obtained from HIV-infected individuals under effective and prolonged ART indicates that a small population of astrocytes has integrated HIV-DNA. In vitro experiments using HIV-infected human primary astrocyte cultures confirmed a low percentage of astrocytes had integrated HIV-DNA, with poor to undetectable replication. Even in the absence of ART, long-term culture results in latency that could be transiently reactivated with histone deacetylase inhibitor, tumor necrosis factor-alpha (TNF-α), or methamphetamine. Reactivation resulted in poor viral production but efficient cell-to-cell viral transfer into cells that support high viral replication. Together, our data provide a new understanding of astrocytes' role as viral reservoirs within the central nervous system (CNS).


Asunto(s)
Astrocitos/virología , Encéfalo/virología , Infecciones por VIH/patología , Infecciones por VIH/virología , VIH , Replicación Viral/efectos de los fármacos , Adulto , Anciano , Terapia Antirretroviral Altamente Activa , Preescolar , ADN Viral/genética , Femenino , Infecciones por VIH/transmisión , Inhibidores de Histona Desacetilasas/farmacología , Humanos , Masculino , Metanfetamina/farmacología , Persona de Mediana Edad , Cultivo Primario de Células , Factor de Necrosis Tumoral alfa/farmacología
7.
Brain Behav Immun ; 91: 159-171, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-32979471

RESUMEN

The human respiratory syncytial virus (hRSV) is the most common infectious agent that affects children before two years of age. hRSV outbreaks cause a significant increase in hospitalizations during the winter season associated with bronchiolitis and pneumonia. Recently, neurologic alterations have been associated with hRSV infection in children, which include seizures, central apnea, and encephalopathy. Also, hRSV RNA has been detected in cerebrospinal fluids (CSF) from patients with neurological symptoms after hRSV infection. Additionally, previous studies have shown that hRSV can be detected in the lungs and brains of mice exposed to the virus, yet the potential effects of hRSV infection within the central nervous system (CNS) remain unknown. Here, using a murine model for hRSV infection, we show a significant behavior alteration in these animals, up to two months after the virus exposure, as shown in marble-burying tests. hRSV infection also produced the expression of cytokines within the brain, such as IL-4, IL-10, and CCL2. We found that hRSV infection alters the permeability of the blood-brain barrier (BBB) in mice, allowing the trespassing of macromolecules and leading to increased infiltration of immune cells into the CNS together with an increased expression of pro-inflammatory cytokines in the brain. Finally, we show that hRSV infects murine astrocytes both, in vitro and in vivo. We identified the presence of hRSV in the brain cortex where it colocalizes with vWF, MAP-2, Iba-1, and GFAP, which are considered markers for endothelial cells, neurons, microglia, and astrocyte, respectively. hRSV-infected murine astrocytes displayed increased production of nitric oxide (NO) and TNF-α. Our results suggest that hRSV infection alters the BBB permeability to macromolecules and immune cells and induces CNS inflammation, which can contribute to the behavioral alterations shown by infected mice. A better understanding of the neuropathy caused by hRSV could help to reduce the potential detrimental effects on the CNS in hRSV-infected patients.


Asunto(s)
Virus Sincitial Respiratorio Humano , Animales , Astrocitos , Barrera Hematoencefálica , Sistema Nervioso Central , Células Endoteliales , Humanos , Inflamación , Pulmón , Ratones , Permeabilidad
8.
Purinergic Signal ; 17(4): 563-576, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34542793

RESUMEN

Only recently, the role of large ionic channels such as Pannexin-1 channels and Connexin hemichannels has been implicated in several physiological and pathological conditions, including HIV infection and associated comorbidities. These channels are in a closed stage in healthy conditions, but in pathological conditions including HIV, Pannexin-1 channels and Connexin hemichannels become open. Our data demonstrate that acute and chronic HIV infection induces channel opening (Pannexin and Connexin channels), ATP release into the extracellular space, and subsequent activation of purinergic receptors in immune and non-immune cells. We demonstrated that Pannexin and Connexin channels contribute to HIV infection and replication, the long-term survival of viral reservoirs, and comorbidities such as NeuroHIV. Here, we discuss the available data to support the participation of these channels in the HIV life cycle and the potential therapeutic approach to prevent HIV-associated comorbidities.


Asunto(s)
Conexinas/metabolismo , Infecciones por VIH/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Receptores Purinérgicos/metabolismo , Animales , Humanos
9.
Int J Mol Sci ; 21(7)2020 Apr 03.
Artículo en Inglés | MEDLINE | ID: mdl-32260308

RESUMEN

At least half of human immunodeficiency virus (HIV)-infected individuals suffer from a wide range of cognitive, behavioral and motor deficits, collectively known as HIV-associated neurocognitive disorders (HAND). The molecular mechanisms that amplify damage within the brain of HIV-infected individuals are unknown. Recently, we described that HIV augments the opening of connexin-43 (Cx43) hemichannels in cultured human astrocytes, which result in the collapse of neuronal processes. Whether HIV soluble viral proteins such as gp120, can regulate hemichannel opening in astrocytes is still ignored. These channels communicate the cytosol with the extracellular space during pathological conditions. We found that gp120 enhances the function of both Cx43 hemichannels and pannexin-1 channels in mouse cortical astrocytes. These effects depended on the activation of IL-1ß/TNF-α, p38 MAP kinase, iNOS, cytoplasmic Ca2+ and purinergic signaling. The gp120-induced channel opening resulted in alterations in Ca2+ dynamics, nitric oxide production and ATP release. Although the channel opening evoked by gp120 in astrocytes was reproduced in ex vivo brain preparations, these responses were heterogeneous depending on the CA1 region analyzed. We speculate that soluble gp120-induced activation of astroglial Cx43 hemichannels and pannexin-1 channels could be crucial for the pathogenesis of HAND.


Asunto(s)
Astrocitos/citología , Conexina 43/metabolismo , Conexinas/metabolismo , Proteína gp120 de Envoltorio del VIH/metabolismo , VIH-1/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Astrocitos/metabolismo , Calcio/metabolismo , Células Cultivadas , Corteza Cerebral/citología , Corteza Cerebral/metabolismo , Ratones , Óxido Nítrico/metabolismo , Transducción de Señal , Imagen de Lapso de Tiempo , Regulación hacia Arriba
10.
J Neurochem ; 151(5): 558-569, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31381153

RESUMEN

Chemical and electrical synapses are the two major communication systems that permit cell-to-cell communication within the nervous system. Although most studies are focused on chemical synapses (glutamate, γ-aminobutyric acid, and other neurotransmitters), clearly both types of synapses interact and cooperate to allow the coordination of several cell functions within the nervous system. The pineal gland has limited independent axonal innervation and not every cell has access to nerve terminals. Thus, additional communication systems, such as gap junctions, have been postulated to coordinate metabolism and signaling. Using acutely isolated glands and dissociated cells, we found that gap junctions spread glycogenolytic signals from cells containing adrenoreceptors to the entire gland lacking these receptors. Our data using glycogen and lactate quantification, electrical stimulation, and high-performance liquid chromatography with electrochemical detection, demonstrate that gap junctional communication between cells of the rat pineal gland allows cell-to-cell propagation of norepinephrine-induced signal that promotes glycogenolysis throughout the entire gland. Thus, the interplay of both synapses is essential for coordinating glycogen metabolism and lactate production in the pineal gland.


Asunto(s)
Comunicación Celular/fisiología , Sinapsis Eléctricas/metabolismo , Glucogenólisis/fisiología , Norepinefrina/metabolismo , Glándula Pineal/metabolismo , Animales , Femenino , Masculino , Ratas , Ratas Sprague-Dawley
11.
Brain Behav Immun ; 80: 328-343, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-30953770

RESUMEN

Increasing evidence indicates that innate immune receptors play important, yet controversial, roles in traumatic central nervous system (CNS) injury. Despite many advances, the contributions of toll-like receptors (TLRs) to spinal cord injury (SCI) remain inadequately defined. We previously reported that a toll-like receptor 9 (TLR9) antagonist, oligodeoxynucleotide 2088 (ODN 2088), administered intrathecally, improves the functional and histopathological outcomes of SCI. However, the molecular and cellular changes that occur at the injury epicenter following ODN 2088 treatment are not completely understood. Following traumatic SCI, a glial scar, consisting primarily of proliferating reactive astrocytes, forms at the injury epicenter and assumes both beneficial and detrimental roles. Increased production of chondroitin sulfate proteoglycans (CSPGs) by reactive astrocytes inhibits the regeneration of injured axons. Astrocytes express TLR9, which can be activated by endogenous ligands released by damaged cells. It is not yet known how TLR9 antagonism modifies astrocyte function at the glial scar and how this affects axonal preservation or re-growth following SCI. The present studies were undertaken to address these issues. We report that in female mice sustaining a severe mid-thoracic (T8) contusion injury, the number of proliferating astrocytes in regions rostral and caudal to the lesion border increased significantly by 30- and 24-fold, respectively, compared to uninjured controls. Intrathecal ODN 2088 treatment significantly reduced the number of proliferating astrocytes by 60% in both regions. This effect appeared to be, at least partly, mediated through the direct actions of ODN 2088 on astrocytes, since the antagonist decreased proliferation in pure SC astrocyte cultures by preventing the activation of the Erk/MAPK signaling pathway. In addition, CSPG immunoreactivity at the lesion border was more pronounced in vehicle-treated injured mice compared to uninjured controls and was significantly reduced following administration of ODN 2088 to injured mice. Moreover, ODN 2088 significantly decreased astrocyte migration in an in vitro scratch-wound assay. Anterograde tracing and quantification of corticospinal tract (CST) axons in injured mice, indicated that ODN 2088 preserves proximal axons. Taken together, these findings suggest that ODN 2088 modifies the glial scar and creates a milieu that fosters axonal protection at the injury site.


Asunto(s)
Astrocitos/metabolismo , Axones/metabolismo , Traumatismos de la Médula Espinal/inmunología , Traumatismos de la Médula Espinal/metabolismo , Receptor Toll-Like 9/metabolismo , Animales , Astrocitos/patología , Axones/inmunología , Axones/patología , Proliferación Celular , Femenino , Ratones Endogámicos C57BL , Oligodesoxirribonucleótidos/administración & dosificación , Traumatismos de la Médula Espinal/patología , Receptor Toll-Like 9/antagonistas & inhibidores
12.
J Immunol ; 199(1): 224-232, 2017 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-28533442

RESUMEN

HIV-1 enters the CNS soon after peripheral infection and causes chronic neuroinflammation and neuronal damage that leads to cognitive impairment in 40-70% of HIV-infected people. The nonpathogenic cellular isoform of the human prion protein (PrPc) is an adhesion molecule constitutively expressed in the CNS. Previously, our laboratory showed that shed PrPc (sPrPc) is increased in the cerebrospinal fluid of HIV-infected people with cognitive deficits as compared with infected people with no impairment. In this article, we demonstrate that CCL2 and TNF-α, inflammatory mediators that are elevated in the CNS of HIV-infected people, increase shedding of PrPc from human astrocytes by increasing the active form of the metalloprotease ADAM10. We show that the consequence of this shedding can be the production of inflammatory mediators, because treatment of astrocytes with rPrPc increased secretion of CCL2, CXCL-12, and IL-8. Supernatants from rPrPc-treated astrocytes containing factors produced in response to this treatment, but not rPrPc by itself, cause increased chemotaxis of both uninfected and HIV-infected human monocytes, suggesting a role for sPrPc in monocyte recruitment into the brain. Furthermore, we examined whether PrPc participates in glutamate uptake and found that rPrPc decreased uptake of this metabolite in astrocytes, which could lead to neurotoxicity and neuronal loss. Collectively, our data characterize mediators involved in PrPc shedding and the effect of this sPrPc on monocyte chemotaxis and glutamate uptake from astrocytes. We propose that shedding of PrPc could be a potential target for therapeutics to limit the cognitive impairment characteristic of neuroAIDS.


Asunto(s)
Astrocitos/metabolismo , Sistema Nervioso Central/fisiopatología , Infecciones por VIH/fisiopatología , VIH/fisiología , Monocitos/inmunología , Monocitos/virología , Proteínas Priónicas/metabolismo , Proteína ADAM10/metabolismo , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Astrocitos/efectos de los fármacos , Astrocitos/inmunología , Células Cultivadas , Sistema Nervioso Central/virología , Quimiocina CCL2/metabolismo , Quimiocina CCL2/farmacología , Quimiocina CXCL1/inmunología , Quimiocina CXCL1/metabolismo , Quimiotaxis de Leucocito , Dipéptidos/farmacología , VIH/inmunología , Infecciones por VIH/virología , Humanos , Ácidos Hidroxámicos/farmacología , Interleucina-8/inmunología , Interleucina-8/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas Priónicas/farmacología , Factor de Necrosis Tumoral alfa/metabolismo , Factor de Necrosis Tumoral alfa/farmacología
13.
Lab Invest ; 98(10): 1347-1359, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-29959417

RESUMEN

The cellular prion protein (PrPc) is a surface adhesion molecule expressed at junctions of various cell types including brain microvascular endothelial cells (BMVEC) that are important components of the blood-brain barrier (BBB). PrPc is involved in several physiological processes including regulation of epithelial cell barrier function and monocyte migration across BMVEC. BBB dysfunction and disruption are significant events in central nervous system (CNS) inflammatory processes including HIV neuropathogenesis. Tumor necrosis factor (TNF)-α and vascular endothelial growth factor (VEGF) are two inflammatory factors that have been implicated in the processes that affect BBB integrity. To examine the effect of inflammation on PrPc expression in BMVEC, we used these mediators and found that TNF-α and VEGF decrease surface PrPc on primary human BMVEC. We also showed that these factors decrease total PrPc protein as well as mRNA, indicating that they regulate expression of this protein by de novo synthesis. To determine the effect of PrPc loss from the surface of BMVEC on barrier integrity, we used small hairpin RNAs to knockdown PrPc. We found that the absence of PrPc from BMVEC causes increased permeability as determined by a fluorescein isothiocyanate (FITC)-dextran permeability assay. This suggests that cell surface PrPc is essential for endothelial monolayer integrity. To determine the mechanism by which PrPc downregulation leads to increased permeability of an endothelial monolayer, we examined changes in expression and localization of tight junction proteins, occludin and claudin-5, and found that decreased PrPc leads to decreased total and membrane-associated occludin and claudin-5. We propose that an additional mechanism by which inflammatory factors affect endothelial monolayer permeability is by decreasing cell-associated PrPc. This increase in permeability may have subsequent consequences that lead to CNS damage.


Asunto(s)
Barrera Hematoencefálica/metabolismo , Células Endoteliales/metabolismo , Proteínas PrPC/metabolismo , Factor de Necrosis Tumoral alfa/fisiología , Factor A de Crecimiento Endotelial Vascular/fisiología , Células Cultivadas , Claudina-5/metabolismo , Humanos , Inflamación/metabolismo , Ocludina/metabolismo
14.
Biochim Biophys Acta Biomembr ; 1860(1): 154-165, 2018 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-28559189

RESUMEN

Connexin (Cx) and pannexin (Panx) containing channels - gap junctions (GJs) and hemichannels (HCs) - are present in virtually all cells and tissues. Currently, the role of these channels under physiological conditions is well defined. However, their role in the immune response and pathological conditions has only recently been explored. Data from several laboratories demonstrates that infectious agents, including HIV, have evolved to take advantage of GJs and HCs to improve viral/bacterial replication, enhance inflammation, and help spread toxicity into neighboring areas. In the current review, we discuss the role of Cx and Panx containing channels in immune activation and the pathogenesis of several infectious diseases. This article is part of a Special Issue entitled: Gap Junction Proteins edited by Jean Claude Herve.


Asunto(s)
Inmunidad Adaptativa , Bacterias/inmunología , Infecciones Bacterianas/inmunología , Conexinas/inmunología , Uniones Comunicantes/inmunología , Infecciones por VIH/inmunología , VIH-1/inmunología , Inmunidad Innata , Animales , Humanos
15.
Artículo en Inglés | MEDLINE | ID: mdl-29203492

RESUMEN

Tuberculosis (TB) recently became the leading infectious cause of death in adults, while attempts to shorten therapy have largely failed. Dormancy, persistence, and drug tolerance are among the factors driving the long therapy duration. Assays to measure in situ drug susceptibility of Mycobacterium tuberculosis bacteria in pulmonary lesions are needed if we are to discover new fast-acting regimens and address the global TB threat. Here we take a first step toward this goal and describe an ex vivo assay developed to measure the cidal activity of anti-TB drugs against M. tuberculosis bacilli present in cavity caseum obtained from rabbits with active TB. We show that caseum M. tuberculosis bacilli are largely nonreplicating, maintain viability over the course of the assay, and exhibit extreme tolerance to many first- and second-line TB drugs. Among the drugs tested, only the rifamycins fully sterilized caseum. A similar trend of phenotypic drug resistance was observed in the hypoxia- and starvation-induced nonreplicating models, but with notable qualitative and quantitative differences: (i) caseum M. tuberculosis exhibits higher drug tolerance than nonreplicating M. tuberculosis in the Wayne and Loebel models, and (ii) pyrazinamide is cidal in caseum but has no detectable activity in these classic nonreplicating assays. Thus, ex vivo caseum constitutes a unique tool to evaluate drug potency against slowly replicating or nonreplicating bacilli in their native caseous environment. Intracaseum cidal concentrations can now be related to the concentrations achieved in the necrotic foci of granulomas and cavities to establish correlations between clinical outcome and lesion-centered pharmacokinetics-pharmacodynamics (PK-PD) parameters.


Asunto(s)
Antituberculosos/farmacología , Mycobacterium tuberculosis/efectos de los fármacos , Tuberculosis/tratamiento farmacológico , Animales , Tolerancia a Medicamentos , Pirazinamida/farmacología , Conejos , Rifamicinas/farmacología
16.
Am J Pathol ; 187(9): 1960-1970, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28688235

RESUMEN

Chronic HIV infection due to effective antiretroviral treatment has resulted in a broad range of clinical complications, including accelerated heart disease. Individuals with HIV infection have a 1.5 to 2 times higher incidence of cardiovascular diseases than their uninfected counterparts; however, the underlying mechanisms are poorly understood. To explore the link between HIV infection and cardiovascular diseases, we used postmortem human heart tissues obtained from HIV-infected and control uninfected individuals to examine connexin 43 (Cx43) expression and distribution and HIV-associated inflammation. Here, we demonstrate that Cx43 is dysregulated in the hearts of HIV-infected individuals. In all HIV heart samples analyzed, there were areas where Cx43 was overexpressed and found along the lateral membrane of the cardiomyocyte and in the intercalated disks. Areas of HIV tissue with anomalous Cx43 expression and localization also showed calcium overload, sarcofilamental atrophy, and accumulation of collagen. All these changes were independent of viral replication, CD4 counts, inflammation, and type of antiretroviral treatment. Overall, we propose that HIV infection increases Cx43 expression in heart, resulting in tissue damage that likely contributes to the high rates of cardiovascular disease in HIV-infected individuals.


Asunto(s)
Enfermedades Cardiovasculares/metabolismo , Conexina 43/metabolismo , Infecciones por VIH/metabolismo , Miocardio/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Recuento de Linfocito CD4 , Enfermedades Cardiovasculares/etiología , Femenino , Infecciones por VIH/complicaciones , Humanos , Inflamación/metabolismo , Masculino , Persona de Mediana Edad , Adulto Joven
17.
J Immunol ; 196(10): 4338-47, 2016 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-27076682

RESUMEN

Pannexin1 (Panx1) channels are large high conductance channels found in all vertebrates that can be activated under several physiological and pathological conditions. Our published data indicate that HIV infection results in the extended opening of Panx1 channels (5-60 min), allowing for the secretion of ATP through the channel pore with subsequent activation of purinergic receptors, which facilitates HIV entry and replication. In this article, we demonstrate that chemokines, which bind CCR5 and CXCR4, especially SDF-1α/CXCL12, result in a transient opening (peak at 5 min) of Panx1 channels found on CD4(+) T lymphocytes, which induces ATP secretion, focal adhesion kinase phosphorylation, cell polarization, and subsequent migration. Increased migration of immune cells is key for the pathogenesis of several inflammatory diseases including multiple sclerosis (MS). In this study, we show that genetic deletion of Panx1 reduces the number of the CD4(+) T lymphocytes migrating into the spinal cord of mice subjected to experimental autoimmune encephalomyelitis, an animal model of MS. Our results indicate that opening of Panx1 channels in response to chemokines is required for CD4(+) T lymphocyte migration, and we propose that targeting Panx1 channels could provide new potential therapeutic approaches to decrease the devastating effects of MS and other inflammatory diseases.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Movimiento Celular/efectos de los fármacos , Quimiocina CXCL12/inmunología , Conexinas/inmunología , Encefalomielitis Autoinmune Experimental/inmunología , Inflamación/inmunología , Proteínas del Tejido Nervioso/inmunología , Adenosina Trifosfato/metabolismo , Animales , Linfocitos T CD4-Positivos/citología , Células Cultivadas , Conexinas/genética , Encefalomielitis Autoinmune Experimental/genética , Femenino , Eliminación de Gen , Humanos , Inflamación/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas del Tejido Nervioso/genética , Médula Espinal
18.
J Immunol ; 194(7): 3246-58, 2015 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-25716997

RESUMEN

Despite successful combined antiretroviral therapy, ∼ 60% of HIV-infected people exhibit HIV-associated neurocognitive disorders (HAND). CCL2 is elevated in the CNS of infected people with HAND and mediates monocyte influx into the CNS, which is critical in neuroAIDS. Many HIV-infected opiate abusers have increased neuroinflammation that may augment HAND. Buprenorphine is used to treat opiate addiction. However, there are few studies that examine its impact on HIV neuropathogenesis. We show that buprenorphine reduces the chemotactic phenotype of monocytes. Buprenorphine decreases the formation of membrane projections in response to CCL2. It also decreases CCL2-induced chemotaxis and mediates a delay in reinsertion of the CCL2 receptor, CCR2, into the cell membrane after CCL2-mediated receptor internalization, suggesting a mechanism of action of buprenorphine. Signaling pathways in CCL2-induced migration include increased phosphorylation of p38 MAPK and of the junctional protein JAM-A. We show that buprenorphine decreases these phosphorylations in CCL2-treated monocytes. Using DAMGO, CTAP, and Nor-BNI, we demonstrate that the effect of buprenorphine on CCL2 signaling is opioid receptor mediated. To identify additional potential mechanisms by which buprenorphine inhibits CCL2-induced monocyte migration, we performed proteomic analyses to characterize additional proteins in monocytes whose phosphorylation after CCL2 treatment was inhibited by buprenorphine. Leukosialin and S100A9 were identified and had not been shown previously to be involved in monocyte migration. We propose that buprenorphine limits CCL2-mediated monocyte transmigration into the CNS, thereby reducing neuroinflammation characteristic of HAND. Our findings underscore the use of buprenorphine as a therapeutic for neuroinflammation as well as for addiction.


Asunto(s)
Quimiocina CCL2/metabolismo , Quimiotaxis de Leucocito/inmunología , Monocitos/inmunología , Monocitos/metabolismo , Analgésicos Opioides/farmacología , Buprenorfina/farmacología , Moléculas de Adhesión Celular/metabolismo , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Quimiotaxis de Leucocito/efectos de los fármacos , Humanos , Monocitos/efectos de los fármacos , Fenotipo , Fosfopéptidos/metabolismo , Fosforilación , Proteoma , Proteómica , Receptores CCR2/metabolismo , Receptores de Superficie Celular/metabolismo , Receptores Opioides/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
19.
J Neurochem ; 137(4): 561-75, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-26953131

RESUMEN

Methamphetamine (meth) is a central nervous system (CNS) stimulant that results in psychological and physical dependency. The long-term effects of meth within the CNS include neuronal plasticity changes, blood-brain barrier compromise, inflammation, electrical dysfunction, neuronal/glial toxicity, and an increased risk to infectious diseases including HIV. Most of the reported meth effects in the CNS are related to dysregulation of chemical synapses by altering the release and uptake of neurotransmitters, especially dopamine, norepinephrine, and epinephrine. However, little is known about the effects of meth on connexin (Cx) containing channels, such as gap junctions (GJ) and hemichannels (HC). We examined the effects of meth on Cx expression, function, and its role in NeuroAIDS. We found that meth altered Cx expression and localization, decreased GJ communication between neurons and astrocytes, and induced the opening of Cx43/Cx36 HC. Furthermore, we found that these changes in GJ and HC induced by meth treatment were mediated by activation of dopamine receptors, suggesting that dysregulation of dopamine signaling induced by meth is essential for GJ and HC compromise. Meth-induced changes in GJ and HC contributed to amplified CNS toxicity by dysregulating glutamate metabolism and increasing the susceptibility of neurons and astrocytes to bystander apoptosis induced by HIV. Together, our results indicate that connexin containing channels, GJ and HC, are essential in the pathogenesis of meth and increase the sensitivity of the CNS to HIV CNS disease. Methamphetamine (meth) is an extremely addictive central nervous system stimulant. Meth reduced gap junctional (GJ) communication by inducing internalization of connexin-43 (Cx43) in astrocytes and reducing expression of Cx36 in neurons by a mechanism involving activation of dopamine receptors (see cartoon). Meth-induced changes in Cx containing channels increased extracellular levels of glutamate and resulted in higher sensitivity of neurons and astrocytes to apoptosis in response to HIV infection.


Asunto(s)
Astrocitos/efectos de los fármacos , Comunicación Celular/efectos de los fármacos , Estimulantes del Sistema Nervioso Central/toxicidad , Uniones Comunicantes/efectos de los fármacos , Metanfetamina/toxicidad , Neuronas/efectos de los fármacos , Animales , Astrocitos/metabolismo , Astrocitos/patología , Comunicación Celular/fisiología , Relación Dosis-Respuesta a Droga , Femenino , Uniones Comunicantes/metabolismo , Uniones Comunicantes/patología , Células HeLa , Humanos , Ratones , Ratones Endogámicos C57BL , Neuronas/metabolismo , Neuronas/patología , Ratas
20.
J Neuroinflammation ; 13(1): 54, 2016 Mar 02.
Artículo en Inglés | MEDLINE | ID: mdl-26934876

RESUMEN

BACKGROUND: HIV-associated neurocognitive disorders (HAND) are a major complication in at least half of the infected population despite effective antiretroviral treatment and immune reconstitution. HIV-associated CNS damage is not correlated with active viral replication but instead is associated with mechanisms that regulate inflammation and neuronal compromise. Our data indicate that one of these mechanisms is mediated by gap junction channels and/or hemichannels. Normally, gap junction channels shutdown under inflammatory conditions, including viral diseases. However, HIV infection upregulates Connexin43 (Cx43) expression and maintains gap junctional communication by unknown mechanism(s). METHODS: Human primary astrocytes were exposed to several HIV proteins as well as to HIV, and expression and function of Connexin43- and Connexin30-containing channels were determined by western blot, immunofluorescence, microinjection of a fluorescent tracer and chromatin immunoprecipitation (ChIP). RESULTS: Here, we demonstrate that HIV infection increases Cx43 expression in vivo. HIV-tat, the transactivator of the virus, and no other HIV proteins tested, increases Cx43 expression and maintains functional gap junctional communication in human astrocytes. Cx43 upregulation is mediated by binding of the HIV-tat protein to the Cx43 promoter, but not to the Cx30 promoter, resulting in increased Cx43 messenger RNA (mRNA) and protein as well as gap junctional communication. CONCLUSIONS: We propose that HIV-tat contributes to the spread of intracellular toxic signals generated in a few HIV-infected cells into surrounding uninfected cells by upregulating gap junctional communication. In the current antiretroviral era, where HIV replication is often completely suppressed, viral factors such as HIV-tat are still produced and released from infected cells. Thus, blocking the effects of HIV-tat could result in new strategies to reduce the damaging consequences of HIV infection of the CNS.


Asunto(s)
Nefropatía Asociada a SIDA/metabolismo , Astrocitos/metabolismo , Conexina 43/biosíntesis , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/farmacología , Nefropatía Asociada a SIDA/patología , Astrocitos/efectos de los fármacos , Encéfalo/patología , Comunicación Celular/efectos de los fármacos , Células Cultivadas , Inmunoprecipitación de Cromatina , Conexina 43/genética , Uniones Comunicantes/efectos de los fármacos , Humanos , ARN Interferente Pequeño/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA