Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
1.
J Neuroinflammation ; 11: 91, 2014 May 16.
Article in English | MEDLINE | ID: mdl-24884548

ABSTRACT

BACKGROUND: Neuroinflammation is a common immune response associated with brain human immunodeficiency virus-1 (HIV-1) infection. Identifying therapeutic compounds that exhibit better brain permeability and can target signaling pathways involved in inflammation may benefit treatment of HIV-associated neurological complications. The objective of this study was to implement an in vivo model of brain inflammation by intracerebroventricular administration of the HIV-1 viral coat protein gp120 in rats and to examine anti-inflammatory properties of HIV adjuvant therapies such as minocycline, chloroquine and simvastatin. METHODS: Male Wistar rats were administered a single dose of gp120ADA (500 ng) daily for seven consecutive days, intracerebroventricularly, with or without prior intraperitoneal administration of minocycline, chloroquine or simvastatin. Maraviroc, a CCR5 antagonist, was administered intracerebroventricularly prior to gp120 administration for seven days as control. Real-time qPCR was used to assess gene expression of inflammatory markers in the frontal cortex, hippocampus and striatum. Interleukin-1ß (IL-1ß) and tumor necrosis factor-α (TNF-α) secretion in cerebrospinal fluid (CSF) was measured applying ELISA. Protein expression of mitogen-activated protein kinases (MAPKs) (extracellular signal-related kinase 1/2 (ERK1/2), c-Jun N-terminal kinases (JNKs) and P38 kinases (P38Ks)) was detected using immunoblot analysis. Student's t-test and ANOVA were applied to determine statistical significance. RESULTS: In gp120ADA-injected rats, mRNA transcripts of interleukin-1ß (IL-1ß) and inducible nitric oxide synthase (iNOS) were significantly elevated in the frontal cortex, striatum and hippocampus compared to saline or heat-inactivated gp120-injected controls. In CSF, a significant increase in TNF-α and IL-1ß was detected. Maraviroc reduced upregulation of these markers suggesting that the interaction of R5-tropic gp120 to CCR5 chemokine receptor is critical for induction of an inflammatory response. Minocycline, chloroquine or simvastatin attenuated upregulation of IL-1ß and iNOS transcripts in different brain regions. In CSF, minocycline suppressed TNF-α and IL-1ß secretion, whereas chloroquine attenuated IL-1ß secretion. In gp120-injected animals, activation of ERK1/2 and JNKs was observed in the hippocampus and ERK1/2 activation was significantly reduced by the anti-inflammatory agents. CONCLUSIONS: Our data demonstrate that anti-inflammatory compounds can completely or partially reverse gp120-associated brain inflammation through an interaction with MAPK signaling pathways and suggest their potential role in contributing towards the prevention and treatment of HIV-associated neurological complications.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Encephalitis/chemically induced , Encephalitis/drug therapy , HIV Envelope Protein gp120/toxicity , Animals , Brain/drug effects , Brain/metabolism , Brain/pathology , Cytokines/genetics , Cytokines/metabolism , Disease Models, Animal , Encephalitis/pathology , Enzyme-Linked Immunosorbent Assay , Gene Expression Regulation/drug effects , Humans , Male , Rats , Rats, Wistar , Time Factors
2.
Adv Exp Med Biol ; 763: 20-69, 2012.
Article in English | MEDLINE | ID: mdl-23397618

ABSTRACT

Drug transport in the central nervous system can be highly regulated by the expression of numerous influx and efflux transport proteins not only at the blood-brain barrier and blood-cerebrospinal fluid barrier but also in brain parenchymal cellular compartments (i.e., astrocytes, microglia, neurons). In particular, members of the ATP-Binding Cassette membrane-associated transporter superfamily and Solute Carrier family are known to be involved in the traffic of several endobiotics and xenobiotics (including drugs) into and out ofthe brain. These transport proteins have also been implicated in a number of neurological disorders including HIV-encephalitis, Alzheimer's disease, Parkinson's disease and neoplasia. This chapter summarizes recent knowledge on the role of drug transporters in the brain.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Blood-Brain Barrier/metabolism , Carrier Proteins/metabolism , Nervous System Diseases/pathology , Transcytosis , AIDS Dementia Complex/metabolism , AIDS Dementia Complex/pathology , AIDS Dementia Complex/virology , ATP Binding Cassette Transporter, Subfamily G, Member 2 , ATP-Binding Cassette Transporters/genetics , Animals , Astrocytes/metabolism , Blood-Brain Barrier/pathology , Carrier Proteins/genetics , Cell Line , Cell Membrane/metabolism , Cerebrospinal Fluid/metabolism , Choroid Plexus/metabolism , Choroid Plexus/pathology , Diffusion , Endothelial Cells/metabolism , Endothelial Cells/pathology , HIV-1/pathogenicity , Humans , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Nervous System Diseases/metabolism , Neuroglia/metabolism , Neuroglia/pathology , Organic Anion Transporters/genetics , Organic Anion Transporters/metabolism , Organic Cation Transport Proteins/genetics , Organic Cation Transport Proteins/metabolism , Xenobiotics/metabolism
3.
J Neurosci Res ; 89(11): 1773-82, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21826700

ABSTRACT

P-glycoprotein (P-gp), a drug efflux pump, is known to alter the bioavailability of antiretroviral drugs at several sites, including the brain. We have previously shown that human immunodeficiency virus-1 (HIV-1) glycoprotein 120 (gp120) induces proinflammatory cytokine secretion and decreases P-gp functional expression in rat astrocytes, a cellular reservoir of HIV-1. However, whether P-gp is regulated in a similar way in human astrocytes is unknown. This study investigates the regulation of P-gp in an in vitro model of gp120-triggered human fetal astrocytes (HFAs). In this system, elevated levels of interleukin-6 (IL-6), IL-1ß, and tumor necrosis factor-α were detected in culture supernatants. Pretreatment with CCR5 neutralizing antibody attenuated cytokine secretion, suggesting that gp120-CCR5 interaction mediated cytokine production. Treatment with gp120 (R5-tropic) resulted in reduced P-gp expression (64%) and function as determined by increased (1.6-fold) cellular accumulation of [(3) H]digoxin, a P-gp substrate. Exposure to R5 or R5/X4-tropic viral isolates led to a downregulation in P-gp expression (75% or 90%, respectively), and treatment with IL-6 also showed lower P-gp expression (50%). Moreover, IL-6 neutralizing antibody blocked gp120-mediated P-gp downregulation, suggesting that IL-6 is a key modulator of P-gp. Gp120- or IL-6-mediated downregulation of P-gp was attenuated by SN50 (a nuclear factor-κB [NF-κB] inhibitor), suggesting involvement of NF-κB signaling in P-gp regulation. Our results suggest that, similarly to the case with rodent astrocytes, pathophysiological stressors associated with brain HIV-1 infection have a downregulatory effect on P-gp functional expression in human astrocytes, which may ultimately result in altered antiretroviral drug accumulation within brain parenchyma.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Astrocytes/metabolism , HIV-1/metabolism , ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , Astrocytes/virology , Cells, Cultured , HIV Envelope Protein gp120/genetics , HIV Envelope Protein gp120/metabolism , HIV-1/genetics , HIV-1/isolation & purification , Humans , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Interleukin-6/genetics , Interleukin-6/metabolism , NF-kappa B/genetics , NF-kappa B/metabolism , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism , Up-Regulation
4.
Mol Pharmacol ; 77(4): 644-59, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20051532

ABSTRACT

Pharmacotherapy of brain HIV-1 infection may be limited by ABC transporters [i.e., P-glycoprotein (P-gp), multidrug resistance protein 1 (Mrp1)] that export antiretroviral drugs from HIV-1 brain cellular targets (i.e., astrocytes, microglia). Using an in vitro astrocyte model of an HIV-1 associated inflammatory response, our laboratory has shown that cytokines [i.e., tumor necrosis factor alpha (TNF-alpha), interleukin (IL)-1 beta, IL-6], which are secreted in response to HIV-1 envelope glycoprotein gp120 exposure, can decrease P-gp functional expression; however, it is unknown whether these same cytokines can alter expression and/or activity of other ABC transporters (i.e., Mrp1). In primary cultures of rat astrocytes, Mrp1 expression was increased by TNF-alpha (2.7-fold) but was not altered by IL-1 beta or IL-6. Cellular retention of 2',7'-bis-(2-carboxyethyl)-5-(and-6)-carboxyfluorescein, an Mrp substrate, was reduced in TNF-alpha-treated astrocytes, suggesting increased Mrp-mediated transport. Pharmacologic inhibition of nuclear factor-kappaB (NF-kappaB) signaling with SN50 prevented both TNF-alpha release and Mrp1 expression changes in astrocytes triggered with gp120; however, SN50 did not attenuate Mrp1 expression in cells triggered with TNF-alpha. In contrast, Mrp1 functional expression was not altered in the presence of gp120 or TNF-alpha when astrocyte cultures were pretreated with 1,9-pyrazoloanthrone (SP600125), an established c-Jun N-terminal kinase (JNK) inhibitor. SP600125 did not affect TNF-alpha release from cultured astrocytes triggered with gp120. Mrp1 mRNA expression was increased after treatment with gp120 (1.6-fold) or TNF-alpha (1.7-fold), suggesting altered Mrp1 gene transcription. These data suggest that gp120 and TNF-alpha can up-regulate Mrp1 expression in cultured astrocytes. Furthermore, our results imply that both NF-kappaB and JNK signaling are involved in Mrp1 regulation during an HIV-1 associated inflammatory response.


Subject(s)
Astrocytes/metabolism , JNK Mitogen-Activated Protein Kinases/physiology , MAP Kinase Signaling System/physiology , Multidrug Resistance-Associated Proteins/genetics , NF-kappa B/physiology , Tumor Necrosis Factor-alpha/pharmacology , Animals , Cells, Cultured , HIV Envelope Protein gp120/pharmacology , HIV-1/pathogenicity , HeLa Cells , Humans , Interleukin-1beta/pharmacology , Interleukin-6/pharmacology , Multidrug Resistance-Associated Proteins/analysis , RNA, Messenger/analysis , Rats , Rats, Wistar , Tumor Necrosis Factor-alpha/metabolism
5.
Adv Pharmacol ; 71: 45-111, 2014.
Article in English | MEDLINE | ID: mdl-25307214

ABSTRACT

Drug permeability in the central nervous system (CNS) across blood-brain and blood-cerebrospinal fluid barriers is an important determinant of neurological disorders therapeutic efficacy and is highly regulated by the expression of membrane-associated transporters belonging to the ATP-binding cassette (ABC) and solute carrier (SLC) superfamilies. Functional expression of ABC efflux transporters exists not only at the brain barriers (primary biochemical barrier) but also in astrocytes, microglia, neurons, and oligodendrocytes can significantly restrict drug penetration into these cells, thus creating a secondary biochemical barrier to permeability in brain parenchyma. In contrast, SLC members primarily contribute to the uptake of endogenous substrates (i.e., hormones, neurotransmitters) and pharmacological agents and can play a critical role in maintaining CNS homeostasis and drug response. In this chapter, we review the functional expression and localization of drug transporters in the brain, their role in CNS drug delivery, and their regulation in neuropathological conditions.


Subject(s)
Brain/metabolism , Membrane Transport Proteins/metabolism , Neuroglia/metabolism , Animals , Brain Diseases/metabolism , Humans , Pharmaceutical Preparations/metabolism
6.
Curr Pharm Des ; 20(10): 1543-63, 2014.
Article in English | MEDLINE | ID: mdl-23789949

ABSTRACT

Membrane-associated drug transporters are important determinants of antiretroviral drug disposition in the central nervous system during HIV-1 infection. A number of influx and efflux transport proteins expressed at the blood-brain barrier, blood-cerebrospinal fluid barrier and in brain parenchyma cellular compartments (i.e., astrocytes, microglia) have been implicated in the traffic of many antiretroviral drugs into and out of the brain. In particular, members of the ATP-binding cassette membrane associated transporter superfamily and Solute Carrier family are known to be involved in the efflux and/or influx of drugs, respectively. As a result, changes in the functional expression of these transporters can alter the disposition and distribution of drugs in the brain. Moreover, antiretroviral therapy itself and/or pathological events (i.e., inflammation, oxidative stress) associated with viral infection may affect the functional expression of these transporters. This review summarizes recent knowledge on the role of drug transporters in regulating brain antiretroviral drug transport in the context of HIV-1 infection.


Subject(s)
Anti-HIV Agents/therapeutic use , Blood-Brain Barrier/metabolism , Drug Delivery Systems , HIV Infections/virology , HIV-1 , Membrane Transport Proteins/metabolism , Nervous System Diseases/drug therapy , Nervous System Diseases/metabolism , Animals , Blood-Brain Barrier/drug effects , HIV Infections/complications , HIV Infections/drug therapy , Humans , Nervous System Diseases/etiology
SELECTION OF CITATIONS
SEARCH DETAIL