Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 273
Filter
1.
J Neuroimmune Pharmacol ; 4(2): 244-8, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19214751

ABSTRACT

CX3CL1 (fractalkine) has been shown not only to be neuroprotective but also may play a role in HIV-1-associated neuropathogenesis. In this study, we found that production of CX3CL1 by human astrocytes stimulated with interleukin (IL)-1beta was inhibited in a concentration-dependent manner following pretreatment with the synthetic cannabinoid WIN55,212-2. The CB(2) receptor selective antagonist SR144528 significantly inhibited WIN55,212-2-mediated suppression of CX3CL1, suggesting a CB(2)-receptor-related mechanism. IL-1beta triggered the activation of p38 and ERK1/2 (p44/42) MAP kinase (MAPK) signaling pathways, but WIN55,212-2 mainly inhibited p38 MAPK phosphorylation. This finding was mirrored in experiments using known inhibitors of these MAPKs, suggesting that the suppression of CX3CL1 production by WIN55,212-2 involves inhibition of signaling via p38 MAPK. Our results support the concept that synthetic cannabinoids have anti-inflammatory properties and that these agents may have therapeutic potential for certain neuroinflammatory disorders.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Astrocytes/drug effects , Benzoxazines/pharmacology , Chemokine CX3CL1/drug effects , Morpholines/pharmacology , Naphthalenes/pharmacology , p38 Mitogen-Activated Protein Kinases/drug effects , Astrocytes/metabolism , Blotting, Western , Camphanes/pharmacology , Cells, Cultured , Chemokine CX3CL1/biosynthesis , Enzyme-Linked Immunosorbent Assay , Humans , MAP Kinase Signaling System/drug effects , Pyrazoles/pharmacology , Receptor, Cannabinoid, CB2/drug effects , Receptor, Cannabinoid, CB2/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
2.
J Neurosci Res ; 83(8): 1471-84, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16547971

ABSTRACT

Microglia, the resident macrophages of the CNS, are responsible for the innate immune response in the brain and participate in the pathogenesis of certain neurodegenerative disorders. Chemokines initiate activation and migration of microglia. The beta-chemokine CCL5 induces an elevation in intracellular calcium concentration ([Ca(2+)](i)) in human microglia. Here, we examined the signal transduction pathway linking activation of chemokine receptor CCR5 to an elevation in [Ca(2+)](i) in cultured microglia by using pharmacological approaches in combination with Fura-2-based digital imaging. The CCL5-induced response required Janus kinase (Jak) activity and the stimulation of an inhibitory G protein. Multiple downstream signaling pathways were involved, including phosphatidylinositol 3-kinase (PI3K), Bruton's tyrosine kinase (Btk), and phospholipase C (PLC)-mediated release of Ca(2+) from inositol 1,4,5-trisphosphate (IP(3))-sensitive stores. Activation of both the kinase and the lipase pathways was required for eliciting the Ca(2+) response. However, the majority of the [Ca(2+)](i) increase was derived from sources activated by NAD metabolites. Cyclic ADP-ribose (cADPR) evoked Ca(2+) release from intracellular stores, and ADPR evoked Ca(2+) influx via a nimodipine-sensitive channel. Thus, a multistep cascade couples CCR5 activation to Ca(2+) increases in human microglia. Because changes in [Ca(2+)](i) affect chemotaxis, secretion, and gene expression, pharmacologic modulation of this pathway may alter inflammatory and degenerative processes in the CNS.


Subject(s)
Calcium Signaling/immunology , Chemokines, CC/immunology , Immunity, Innate/immunology , Immunologic Surveillance/immunology , Microglia/immunology , Receptors, CCR5/immunology , Agammaglobulinaemia Tyrosine Kinase , Calcium/metabolism , Calcium Channels/drug effects , Calcium Channels/metabolism , Calcium Signaling/drug effects , Cells, Cultured , Chemokine CCL5 , Chemokines, CC/metabolism , Chemokines, CC/pharmacology , Cyclic ADP-Ribose/metabolism , Encephalitis/immunology , Encephalitis/metabolism , Encephalitis/physiopathology , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Humans , Inositol 1,4,5-Trisphosphate/metabolism , Janus Kinase 1 , Microglia/drug effects , Microglia/metabolism , NAD/metabolism , Nerve Degeneration/immunology , Nerve Degeneration/metabolism , Nerve Degeneration/physiopathology , Phosphatidylinositol 3-Kinases/metabolism , Protein-Tyrosine Kinases/metabolism , Receptors, CCR5/agonists , Receptors, CCR5/metabolism , Signal Transduction/drug effects , Signal Transduction/immunology , Signal Transduction/physiology , Type C Phospholipases/metabolism
3.
Genes Immun ; 6(8): 712-9, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16163375

ABSTRACT

Microglia, the resident macrophages in the central nervous system (CNS), play a pivotal role in innate and adaptive immune responses in the brain. The immune functions of microglia are regulated by cytokines, including interferon (IFN)-gamma, which is a major mediator of macrophage activation. We describe the transcriptional profile of human fetal microglial cells at 1, 6, and 24 h after IFN-gamma treatment. The results show a change in the expression of 405 genes including transcriptionally induced chemokines, IFN-gamma signaling factors, and major histocompatibility complex genes. Our results demonstrate that activation of microglia by IFN-gamma induces proinflammatory T-lymphocyte-related chemokine genes as well as genes involved in antigen presentation. As a result, signals for T-cell infiltration and antigen presentation are produced to allow for microglia-T-cell interactions that likely contribute to defense against invading pathogens. In sum, our results provide a foundation for the molecular mechanisms of the microglial response to IFN-gamma-a key to understanding cell-mediated immunity of the CNS.


Subject(s)
Interferon-gamma/pharmacology , Microglia/drug effects , Transcription, Genetic/drug effects , Cells, Cultured , Chemokine CCL2/metabolism , Chemokine CCL5 , Chemokine CXCL10 , Chemokine CXCL11 , Chemokines/analysis , Chemokines, CC/metabolism , Chemokines, CXC/metabolism , Culture Media/chemistry , Cytokines/analysis , Fetus , Gene Expression , Humans , Interferon-gamma/genetics , Kinetics , Microglia/immunology , Models, Genetic , RNA, Messenger/analysis , Recombinant Proteins/pharmacology , Time Factors
4.
J Neuroimmunol ; 147(1-2): 123-6, 2004 Feb.
Article in English | MEDLINE | ID: mdl-14741442

ABSTRACT

The influence of substances of abuse on the progression of HIV-1 infection is controversial, and pharmacologic factors have been postulated as a potential explanation for conflicting data arising from epidemiological studies and animal models. In the present study, cell culture models of HIV-1 infection were used to test this hypothesis. The synthetic cannabinoid WIN 55,212-2 was found to potently inhibit HIV-1 expression in a concentration- and time-dependent manner in CD4(+) lymphocyte and microglial cell cultures. In sharp contrast, morphine either inhibited or stimulated viral expression, depending upon the time of drug exposure, and marked differences were observed between CD4(+) and microglial cells. Also, WIN 55,212-2 inhibited the stimulatory effect of morphine in HIV-1 infected CD4(+) cells. These in vitro findings support the notion that pharmacologic factors need to be considered in epidemiological studies and animal models that pertain to HIV-1 infection.


Subject(s)
CD4-Positive T-Lymphocytes/drug effects , Cannabinoids/pharmacology , HIV-1/drug effects , Microglia/drug effects , Morphine/pharmacology , Analysis of Variance , Benzoxazines , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/virology , Cells, Cultured , Dose-Response Relationship, Drug , Drug Interactions , HIV Core Protein p24/metabolism , HIV Infections , HIV-1/physiology , Humans , Microglia/metabolism , Microglia/virology , Morpholines/pharmacology , Naphthalenes/pharmacology , Narcotics/pharmacology , Time Factors , Virus Replication/drug effects
7.
Pediatr Emerg Care ; 17(6): 425-9, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11753186

ABSTRACT

BACKGROUND: To characterize the host response to venom from snakes of the family Viperidae in Costa Rica, we investigated the release of cytokines: IL-1, IL-6, IL-8, TNF-alpha, MIP-1beta, and RANTES in pediatric patients who were bitten by a snake. METHODS: Patients were included in this study if they were admitted to the hospital within 24 hours of the snakebite. Blood samples were taken immediately on admission to the hospital, and then at intervals of 3, 12, and 24 hours, and on days 3, 5, and 7 after the accident. Patients received gentamicin plus clindamycin or gentamicin plus penicillin intravenously for a minimum of 3 days or longer if necessary. IL-1, IL-8, TNF-alpha, MIP-1beta, and RANTES were determined by monoclonal antibody-based ELISAs, while IL-6 was determined by bioassay. RESULTS: Eighteen patients were included in this study; 15 were bitten by Bothrops asper and three by B. lateralis. Eleven patients were male. Median (range) age was 9 (1-12) years. Nine patients had detectable serum concentrations of IL-6 (200 pg/ mL) and IL-8 (51 pg/mL) on admission, increasing to 500 pg/mL and 115 pg/mL for IL-6 and IL-8, respectively, during the first 12-24 hours. Cytokine concentrations returned to normal or undetectable ranges by 72 hours. TNF-alpha concentrations peaked at 12 hours (mean: 48 pg/mL). Low, but detectable concentrations of MIP-1beta were observed in some patients at various time intervals (48 pg/mL), whereas IL-1 was not detectable at any time point. Regulated on Activation Normal T cell Expressed and Secreted (RANTES) concentrations were evaluated in only five patients, being elevated in all of them. Patients with elevated cytokine concentrations required early fasciotomy (<24 hours after the accident) more often than those who had normal or undetectable cytokine concentrations (P < 0.05). There were no statistically significant associations between severity of envenomation, or outcome, and elevated serum cytokine concentrations (P > 0.05). CONCLUSIONS: Bothrops sp snake venoms induce clinical and pathophysiologic alterations similar to acute trauma, with release of proinflammatory cytokines. A better understanding of the role of the inflammatory response could lead to the development of new therapeutic strategies to improve the outcome in snakebitten patients.


Subject(s)
Bothrops , Cytokines/blood , Snake Bites/drug therapy , Snake Bites/immunology , Animals , Anti-Bacterial Agents/therapeutic use , Chemokine CCL5/blood , Child , Child, Preschool , Clindamycin/therapeutic use , Costa Rica , Fasciitis/etiology , Fasciitis/prevention & control , Fasciitis/surgery , Female , Humans , Infant , Interleukin-6/blood , Likelihood Functions , Male , Penicillins/therapeutic use , Severity of Illness Index , Snake Bites/complications
8.
J Neurovirol ; 7(5): 481-6, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11582521

ABSTRACT

During HIV-1 encephalitis, the chemotaxis-inducing activity of Tat may enhance the viral life cycle through recruitment of additional susceptible microglial cells to foci of infection. Benzodiazepines (BDZs) readily penetrate the blood-brain barrier and are known to possess anti-inflammatory properties. Pretreatment of human microglial cells with peripheral (Ro5-4864) and mixed (diazepam), but not central (clonazepam), benzodiazepine receptor ligands was found to potently suppress HIV-1 Tat-induced chemotaxis. Application of Tat to microglial cells evokes an increase in intracellular calcium concentration ([Ca(2+)]i) that rapidly desensitizes the cells. Diazepam's inhibitory effect was associated with its ability to block Tat-induced [Ca(2+)]i mobilization. These data support the notion that through their effects on microglia, peripheral BDZ receptor ligands could alter the neuropathogenesis of HIV-1.


Subject(s)
Adjuvants, Immunologic/pharmacology , Calcium Signaling/drug effects , Chemotaxis/drug effects , Diazepam/pharmacology , Gene Products, tat/antagonists & inhibitors , HIV-1/drug effects , Microglia/drug effects , Nerve Tissue Proteins/drug effects , Receptors, GABA-A/drug effects , Benzodiazepinones/pharmacology , Clonazepam/pharmacology , Gene Products, tat/physiology , Humans , Microglia/virology , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/physiology , Receptors, CCR5/biosynthesis , Receptors, CCR5/genetics , Receptors, GABA-A/physiology , Up-Regulation/drug effects , tat Gene Products, Human Immunodeficiency Virus
9.
Drug Alcohol Depend ; 64(3): 257-63, 2001 Nov 01.
Article in English | MEDLINE | ID: mdl-11672940

ABSTRACT

CD4(+) T lymphocytes are the primary cell target for human immunodeficiency virus-1 (HIV-1), and these cells are known to express opioid receptors. Due to the need for new treatment approaches to HIV-1 infection, we sought to determine whether the non-selective opioid receptor antagonist naltrexone would affect HIV-1 expression in CD4(+) lymphocyte cultures and whether naltrexone would alter the antiviral properties of zidovudine (AZT) or indinavir. Activated CD4(+) lymphocytes were infected with a monocytotropic or T-cell tropic HIV-1 isolate, and p24 antigen levels were measured in supernatants of drug-treated or untreated (control) cultures. While naltrexone alone did not affect HIV-1 expression, at a concentration of 10(-12)-10(-10) M naltrexone increased the antiviral activity of AZT and indinavir 2-3-fold. Similar findings with a kappa-opioid receptor (KOR) selective antagonist supported the possible involvement of KOR in naltrexone's potentiation of the antiretroviral drugs. The results of this in vitro study suggest that treatment of alcohol or opiate dependent HIV-1-infected patients with naltrexone is unlikely to interfere with the activity of antiretroviral drugs. Also, based upon naltrexone's safety profile and its synergistic activity in vitro, these findings suggest clinical trials should be considered of naltrexone as an adjunctive therapy of HIV-1 infection.


Subject(s)
Anti-HIV Agents/pharmacology , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/virology , HIV-1/drug effects , Naltrexone/pharmacology , Narcotic Antagonists/pharmacology , Anti-HIV Agents/therapeutic use , Cells, Cultured , Drug Combinations , Drug Synergism , HIV-1/metabolism , Humans , Indinavir/pharmacology , Naloxone/pharmacology , Naltrexone/therapeutic use , Narcotic Antagonists/therapeutic use , Zidovudine/pharmacology
10.
J Neurovirol ; 7(2): 135-47, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11517386

ABSTRACT

Glial cells function as sensors for infection within the brain and produce cytokines to limit viral replication and spread. We examined both cytokine (TNF-alpha, IL-1beta, and IL-6) and chemokine (MCP-1, MIP-1alpha, RANTES, and IL-8) production by primary human glial cells in response to cytomegalovirus (CMV). Although CMV-infected astrocytes did not produce antiviral cytokines, they generated significant quantities of the chemokines MCP-1 and IL-8 in response to viral infection. On the other hand, supernatants from CMV-stimulated purified microglial cell cultures showed a marked increase in the production of TNF-alpha and IL-6, as well as chemokines. Supernatants from CMV-infected astrocyte cultures induced the migration of microglia towards chemotactic signals generated from infected astrocytes. Antibodies to MCP-1, but not to MIP-1alpha, RANTES, or IL-8, inhibited this migratory activity. These findings suggest that infected astrocytes may use MCP-1 to recruit antiviral cytokine-producing microglial cells to foci of infection. To test this hypothesis, cocultures of astrocytes and microglial cells were infected with CMV. Viral gene expression in these cocultures was 60% lower than in CMV infected purified astrocyte cultures lacking microglia. These results support the hypothesis that microglia play an important antiviral role in defense of the brain against CMV. The host defense function of microglial cells may be directed in part by chemokines, such as MCP-1, produced by infected astrocytes.


Subject(s)
Astrocytes/virology , Cytomegalovirus Infections/immunology , Cytomegalovirus/immunology , Encephalitis, Viral/immunology , Microglia/virology , Astrocytes/cytology , Astrocytes/immunology , Brain/cytology , Brain/virology , Cells, Cultured , Chemokine CCL3 , Chemokine CCL4 , Chemokine CCL5/genetics , Chemokine CCL5/immunology , Chemotaxis/immunology , Coculture Techniques , Cytomegalovirus/growth & development , Encephalitis, Viral/virology , Fetus/cytology , Gene Expression Regulation, Viral/immunology , Humans , Interleukin-1/genetics , Interleukin-1/immunology , Interleukin-6/genetics , Interleukin-6/immunology , Interleukin-8/genetics , Interleukin-8/immunology , Macrophage Inflammatory Proteins/genetics , Macrophage Inflammatory Proteins/immunology , Microglia/cytology , Microglia/immunology , RNA, Messenger/analysis , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology , Virus Replication
11.
J Neurovirol ; 7(3): 208-19, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11517395

ABSTRACT

Cytokine (TNF-alpha/beta, IL-1beta, IL-6, IL-18, IL-10, and IFN-alpha/beta/gamma) and chemokine (IL-8, IP-10, MCP-1, MIP-1alpha/beta, and RANTES) production during herpes simplex virus (HSV) 1 infection of human brain cells was examined. Primary astrocytes as well as neurons were found to support HSV replication, but neither of these fully permissive cell types produced cytokines or chemokines in response to HSV. In contrast, microglia did not support extensive viral replication; however, ICP4 was detected by immunochemical staining, demonstrating these cells were infected. Late viral protein (nucleocapsid antigen) was detected in <10% of infected microglial cells. Microglia responded to nonpermissive viral infection by producing considerable amounts of TNF-alpha, IL-1beta, IP-10, and RANTES, together with smaller amounts of IL-6, IL-8, and MIP-1alpha as detected by RPA and ELISA. Surprisingly, no interferons (alpha, beta, or gamma) were detected in response to viral infection. Pretreatment of fully permissive astrocytes with TNF-alpha prior to infection with HSV was found to dramatically inhibit replication, resulting in a 14-fold reduction of viral titer. In contrast, pretreatment of astrocytes with IL-1beta had little effect on viral replication. When added to neuronal cultures, exogenous TNF-alpha or IL-1beta did not suppress subsequent HSV replication. Exogenously added IP-10 inhibited HSV replication in neurons (with a 32-fold reduction in viral titer), however, similar IP-10 treatment did not affect viral replication in astrocytes. These results suggest that IP-10 possesses direct antiviral activity in neurons and support a role for microglia in both antiviral defense of the brain as well as amplification of immune responses during neuroinflammation.


Subject(s)
Cytokines/immunology , Encephalitis, Herpes Simplex/immunology , Herpesvirus 1, Human/growth & development , Microglia/virology , Astrocytes/cytology , Astrocytes/virology , Brain/cytology , Brain/virology , Butadienes/pharmacology , Cell Death/immunology , Cells, Cultured , Chemokine CCL5/biosynthesis , Chemokine CCL5/immunology , Chemokine CXCL10 , Chemokines, CXC/biosynthesis , Chemokines, CXC/immunology , Cytokines/biosynthesis , Enzyme Inhibitors/pharmacology , Fetus/cytology , Humans , Imidazoles/pharmacology , Interleukin-1/biosynthesis , Interleukin-1/immunology , Microglia/cytology , Microglia/metabolism , Neurons/cytology , Neurons/virology , Nitriles/pharmacology , Pyridines/pharmacology , Signal Transduction/drug effects , Signal Transduction/immunology , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Necrosis Factor-alpha/immunology , Virus Replication
12.
J Immunol ; 167(2): 1097-102, 2001 Jul 15.
Article in English | MEDLINE | ID: mdl-11441121

ABSTRACT

The delta opioid receptors (DORs) modulate T cell proliferation, IL-2 production, chemotaxis, and intracellular signaling. Moreover, in DOR-transfected Jurkat cells, delta opioids have been shown to suppress HIV-1 p24 Ag expression. These observations led us to characterize the expression of DORs by human peripheral blood T cells and to determine whether a specific DOR agonist, benzamide,4-([2,5-dimethyl-4-(2-propenyl)-1-piperazinyl](3-methoxyphenyl)methyl]-N,-,(2S[1(S*),2alpha,5beta])-(9Cl) (SNC-80), can suppress p24 Ag expression by HIV-1-infected CD4+ T cells obtained from normal donors. By immunofluorescence flow cytometry, PHA stimulated the expression of DOR from 1.94 +/- 0.70 (mean +/- SEM) to 20.70 +/- 1.88% of the PBMC population by 48 h (p < 0.0001). DOR expression was approximately 40% of both the PHA-stimulated CD4+ and CD8+ T cell subsets, and virtually all DORs were found on these subsets. To determine whether activated DORs suppress HIV-1 expression, PBMC were prestimulated with PHA, and then CD4+ T cells were purified, pretreated with SNC-80, and infected with HIV-1. In a concentration-dependent manner, SNC-80 inhibited production of p24 Ag. SNC-80 10(-10) M maximally suppressed (approximately 50%) both lymphocytotropic (HIV-1 MN) and monocytotropic (SF162) strains; higher concentrations were less effective. Naltrindole, a selective DOR antagonist, abolished the inhibitory effects of SNC-80. Kinetic studies indicated that 24-h pre- or postincubation with SNC-80, relative to infection with HIV-1, eliminated its suppressive effects. Thus, stimulating the DORs expressed by activated CD4+ T cells significantly suppressed the expression of HIV-1. These findings suggest that opioid immunomodulation directed at host T cells may be adjunctive to standard antiviral approaches to HIV-1 infection.


Subject(s)
Anti-HIV Agents/blood , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/virology , HIV-1/immunology , Immunosuppressive Agents/blood , Receptors, Opioid, delta/blood , Receptors, Opioid, delta/physiology , Virus Replication/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cells, Cultured , Flow Cytometry , Fluorescent Antibody Technique, Direct/methods , Humans , Immunologic Memory , Interphase/immunology , Lymphocyte Activation , Male , Phytohemagglutinins/pharmacology , Receptors, Opioid, delta/biosynthesis , Signal Transduction/immunology
13.
Biochem Pharmacol ; 61(9): 1145-51, 2001 May 01.
Article in English | MEDLINE | ID: mdl-11301048

ABSTRACT

Synthetic kappa-opioid receptor (KOR) agonists have been shown to suppress HIV-1 expression in acutely infected macrophages. In the present study, we examined the effects of the KOR ligand trans-3,4-dichloro-N-methyl-N[2-(1-pyrolidinyl)cyclohexyl]benzeneaceamide methanesulfonate (U50,488) on HIV-1 expression in CD4+ lymphocytes, the main target cell of this virus. When U50,488 was added to activated CD4+ lymphocytes, HIV-1 expression was inhibited in a concentration- and time-dependent manner with maximal suppression (approximately 60%) at 10(-7) M U50,488. The KOR selective antagonist nor-binaltorphimine (nor-BNI) had no effect by itself on viral expression but blocked the antiviral property of U50,488, suggesting that U50,488 was acting via a KOR-related mechanism. Support for the involvement of KOR was provided by the findings that 34% of activated CD4+ lymphocytes were positive for KOR, using an immunofluorescence technique, and that seven additional synthetic KOR ligands also inhibited HIV-1 expression. The results of this study broaden understanding of the antiviral properties of KOR ligands to include cells outside of the nervous system and suggest a potential role for these agents in the treatment of HIV-1 infection.


Subject(s)
3,4-Dichloro-N-methyl-N-(2-(1-pyrrolidinyl)-cyclohexyl)-benzeneacetamide, (trans)-Isomer/pharmacology , CD4-Positive T-Lymphocytes/drug effects , HIV-1/drug effects , Receptors, Opioid, kappa/agonists , 3,4-Dichloro-N-methyl-N-(2-(1-pyrrolidinyl)-cyclohexyl)-benzeneacetamide, (trans)-Isomer/chemistry , Analgesics, Non-Narcotic/chemistry , Analgesics, Non-Narcotic/pharmacology , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/virology , Flow Cytometry , Humans , In Vitro Techniques
14.
Drug Alcohol Depend ; 62(2): 149-54, 2001 Apr 01.
Article in English | MEDLINE | ID: mdl-11245971

ABSTRACT

Opioids may play an immunomodulatory role in the pathogenesis of human immunodeficiency virus-1 (HIV-1) infection. Recently, synthetic kappa-opioid receptor (KOR) ligands have been found to have anti-human immunodeficiency virus type 1 activity in acutely infected brain macrophages. In the present study, we investigated whether the selective KOR ligand U50488 would exert such an anti-HIV-1 effect in acutely infected blood monocyte-derived macrophages (MDM). Treatment of acutely infected MDM with U50488 induced a concentration-dependent inhibition of HIV-1 expression. The dose--response relationship of U50488 was U-shaped with a peak effect observed at 10(-13) M, which was evident at both 7 and 14 days post-infection. The KOR antagonist nor-binaltorphimine blocked the anti-HIV-1 effect of U50488 by 73%, indicating involvement of a KOR-mediated mechanism. Also, expression of KOR mRNA and binding activity with a fluorescence-labeled KOR ligand supported the existence of KOR on MDM. Antibodies to the beta-chemokine, RANTES (regulated on activation normal T-cell expressed and secreted), but not to various other cytokines, blocked U50488 inhibition by 56% suggesting that the anti-HIV-1 effect of U50488 involved, in part, the production of RANTES by MDM. Taken together, these in vitro findings support the anti-HIV-1 property of U50488, and suggest that KOR ligands may have therapeutic potential for treating patients with acquired immunodeficiency syndrome.


Subject(s)
3,4-Dichloro-N-methyl-N-(2-(1-pyrrolidinyl)-cyclohexyl)-benzeneacetamide, (trans)-Isomer/pharmacology , Acquired Immunodeficiency Syndrome/metabolism , Analgesics, Non-Narcotic/pharmacology , HIV-1/drug effects , Macrophages/drug effects , Macrophages/virology , Monocytes/drug effects , Monocytes/virology , 3,4-Dichloro-N-methyl-N-(2-(1-pyrrolidinyl)-cyclohexyl)-benzeneacetamide, (trans)-Isomer/administration & dosage , Acute Disease , Analgesics, Non-Narcotic/administration & dosage , Cells, Cultured , Chemokine CCL5/pharmacology , Cytokines/metabolism , Dose-Response Relationship, Drug , Fluorescence , Humans , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Receptors, Opioid, kappa/metabolism , Reverse Transcriptase Polymerase Chain Reaction
15.
16.
J Psychopharmacol ; 14(3): 238-43, 2000.
Article in English | MEDLINE | ID: mdl-11106302

ABSTRACT

The beta-chemokine RANTES has recently been implicated in the neuropathogenesis of the human immunodeficiency virus. Based upon previous studies of the effects of morphine on microglial cell production of cytokines and chemotaxis towards the activated complement component C5a, we tested the hypothesis that this opiate would alter the production of and migration towards RANTES by human microglia. Treatment of highly purified microglial cell cultures with morphine (10(-8)-10(-6) M) potently inhibited RANTES production by lipopolysaccharide- and interleukin-1beta-stimulated cells. Using a chemotaxis chamber to assess directed migration towards RANTES, treatment of microglial cells with morphine (10(-10)-10(-6) M) was found to suppress chemotaxis. The inhibitory effects of morphine on RANTES production and on chemotaxis were blocked by naloxone and beta-funaltrexamine, indicating that morphine mediated its suppressive effects via activation of microglial p-opioid receptors. Morphine's inhibitory effect on chemotaxis did not appear to be associated with an alteration in RANTES-induced [Ca2+]i mobilization. While the clinical significance of these in-vitro findings is unknown, they suggest that mu-opioid receptor agonists could alter certain neurodegenerative and inflammatory processes within the brain.


Subject(s)
Chemokine CCL5/physiology , Chemotaxis/physiology , Microglia/physiology , Morphine/pharmacology , Brain/cytology , Brain/physiology , Calcium/pharmacology , Calcium/physiology , Cells, Cultured , Chemokine CCL5/biosynthesis , Chemokine CCL5/pharmacology , Chemotaxis/drug effects , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/pharmacology , Fetus , Humans , Interleukin-1/pharmacology , Lipopolysaccharides/pharmacology , Microglia/drug effects , Naloxone/pharmacology , Naltrexone/analogs & derivatives , Naltrexone/pharmacology , Narcotic Antagonists/pharmacology , Recombinant Proteins/pharmacology
17.
J Infect Dis ; 182(3): 983-7, 2000 Sep.
Article in English | MEDLINE | ID: mdl-10950803

ABSTRACT

Thalidomide, a psychoactive drug that readily crosses the blood-brain barrier, has been shown to possess immunomodulatory attributes, including the inhibition of cytokine production by monocytes and microglia. In this study, we investigated the effect of thalidomide on chemokine production by human microglial cells. Microglial cells were stimulated with lipopolysaccharide, a key cell-wall component of gram-negative bacteria responsible for meningitis, and production of chemokines (regulated upon activation normally T cell expressed and secreted [RANTES], monocyte chemoattractant protein [MCP]-1, macrophage inflammatory protein [MIP]-1beta, and interleukin [IL]-8) was examined by ELISA. Thalidomide treatment was found to cause potent and selective inhibition of IL-8 production in a dose-responsive manner. This inhibition was associated with decreased intracellular IL-8 staining as well as reduced transcription of IL-8 mRNA. In addition, thalidomide treatment of lipopolysaccharide-stimulated microglia inhibited the activation of protein NF-kappaB, a transcription factor known to be important for IL-8 production. These results suggest thalidomide could have a therapeutic role in acute bacterial meningitis through inhibition of IL-8-mediated neutrophil chemotaxis.


Subject(s)
Chemokines/biosynthesis , Immunosuppressive Agents/pharmacology , Microglia/metabolism , Thalidomide/pharmacology , Cells, Cultured , Chemokine CCL2/metabolism , Chemokine CCL5/metabolism , Enzyme-Linked Immunosorbent Assay , Humans , Interleukin-8/genetics , Interleukin-8/metabolism , Lipopolysaccharides/pharmacology , RNA, Messenger/metabolism
18.
Clin Immunol ; 96(3): 243-51, 2000 Sep.
Article in English | MEDLINE | ID: mdl-10964543

ABSTRACT

The viral proteins, Tat (HIV-1 nuclear protein) and gp41 (HIV-1 coat protein), detected in the brains of HIV-1-infected patients have been shown to be neurotoxic. We investigated the effects of HIV-1 Tat and gp41 proteins on cytokine, chemokine, and superoxide anion (O(-)(2)) production by microglia, the resident macrophages of the brain. Tat and gp41 dose-dependently stimulated cytokine and chemokine production by microglia. Peak production of these cytokines and chemokines differed in microglial cells treated with gp41 and Tat. Expression of cytokine and chemokine mRNA was also stimulated in gp41- and Tat-treated microglia. Neither gp41 nor Tat alone stimulated O(-)(2) production by microglia. Treatment of microglial cells with Tat but not with gp41 evoked an increase in intracellular Ca(2+). The results of this study suggest that HIV-1 Tat and gp41 proteins impact several key functions of microglial cells which could contribute to the neuropathogenesis of HIV-1.


Subject(s)
Gene Products, tat/pharmacology , HIV Envelope Protein gp41/pharmacology , Microglia/cytology , Calcium/metabolism , Cells, Cultured , Chemokines/biosynthesis , Cytokines/biosynthesis , Cytokines/drug effects , Fetus/cytology , Humans , Microglia/physiology , Superoxides/metabolism
19.
Clin Diagn Lab Immunol ; 7(4): 710-3, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10882679

ABSTRACT

Cytomegalovirus-stimulated CD4(+) lymphocytes from seropositive but not seronegative donors suppressed viral gene expression in primary human astrocytes. This suppressive activity was mediated through soluble factors. These findings suggest that CD4(+) lymphocytes play a role in defense of the brain against cytomegalovirus.


Subject(s)
Astrocytes/virology , CD4-Positive T-Lymphocytes/immunology , Cytomegalovirus/physiology , Astrocytes/immunology , CD4-Positive T-Lymphocytes/virology , Humans , Virus Replication/immunology
20.
Neuroscience ; 98(1): 191-9, 2000.
Article in English | MEDLINE | ID: mdl-10858625

ABSTRACT

Activation of beta-chemokine receptors, co-receptors for human immunodeficiency virus type-1 (HIV-1), stimulates movement and secretion in microglia, possibly through a Ca(2+)-dependent mechanism. We studied chemokine activation of Ca(2+) signaling processes in microglia. Human fetal microglia were grown in primary culture and chemokine-induced increases in intracellular calcium concentration ([Ca(2+)](i)) were measured in single cells using indo-1-based microfluorimetry. Application of 50 ng/ml regulated on activation, normal T expressed and secreted (RANTES; 120 s) evoked responses in 26% of the microglia (187/719 cells). [Ca(2+)](i) increased from a basal level of 66+/-6 nM to peak at 268+/-23 nM (n=187). Chemokine-evoked responses rapidly desensitized as indicated by the rapid return to basal [Ca(2+)](i) levels in the maintained presence of RANTES. The removal of extracellular Ca(2+) or stimulation in the presence of Ni(2+) (2mM) or La(3+) (100 microM) blocked the RANTES-elicited [Ca(2+)](i) increase. The L-type calcium channel antagonist nimodipine (10 microM) inhibited the RANTES-mediated increase in [Ca(2+)](i) by 80+/-16%. Thus, the RANTES-evoked calcium transient appears to result from Ca(2+) influx with little if any release from intracellular stores. Application of gp120(clade) (E) and gp120(CM235) (50 ng/ml) neither mimicked nor antagonized the RANTES-evoked response. Application of 50 ng/ml eotaxin (120 s) evoked an increase in [Ca(2+)](i) in 13% of the human microglia in culture (61/469 cells). The HIV-1 regulatory protein Tat (50 ng/ml) increased the [Ca(2+)](i) in a subset of eotaxin-responsive cells (16/30). The L-type calcium channel antagonist nimodipine (3 microM) inhibited eotaxin- and Tat-mediated increases in [Ca(2+)](i) by 88+/-6% and 93+/-6%, respectively. Thus, activation of CCR3 appears to evoke Ca(2+) influx through L-type Ca(2+) channels.These results indicate that beta-chemokines, RANTES and eotaxin, activate a nimodipine sensitive Ca(2+) influx pathway in human fetal microglia. HIV-1 Tat protein mimicked chemokine-mediated Ca(2+) signaling and may modulate the migratory and secretory responses of microglia.


Subject(s)
Calcium/metabolism , Chemokines, CC/pharmacology , HIV Envelope Protein gp120/pharmacology , Microglia/metabolism , Microglia/virology , Acquired Immunodeficiency Syndrome/immunology , Acquired Immunodeficiency Syndrome/metabolism , Brain/cytology , Calcium Channel Blockers/pharmacology , Calcium Channels, L-Type/physiology , Calcium Signaling/drug effects , Calcium Signaling/immunology , Cells, Cultured , Chemokine CCL11 , Chemokine CCL5/pharmacology , Cytokines/pharmacology , Fetus/cytology , Gene Products, tat/pharmacology , Humans , Microglia/cytology , Nimodipine/pharmacology , tat Gene Products, Human Immunodeficiency Virus
SELECTION OF CITATIONS
SEARCH DETAIL
...