Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
J Neurosci ; 32(29): 9917-30, 2012 Jul 18.
Artículo en Inglés | MEDLINE | ID: mdl-22815507

RESUMEN

Chronic drug users account for a third of all cases of AIDS in the United States and the progression to AIDS dementia is accelerated in opiate drug abusers. Clinically, microglial activation better correlates with HIV-associated neurocognitive disorders (HAND) than productive HIV-1 infection in the CNS. Moreover, pneumococcal pneumonia is the most common opportunistic infection in individuals with HAND. We show that coinfection with Streptococcus pneumoniae may be a contributing factor in the increased prevalence of HAND in the opioid-dependent population. To date, there have been no studies published implicating the Toll-like receptors (TLR) in the neurocognitive disorders associated with NeuroAIDS in the context of opportunistic infection. Our studies show for the first time, in a morphine-dependent model, synergistic increase and activation of TLR expression in the presence of HIV-1 protein TAT and S. pneumoniae with a significant increase in proinflammatory cytokines (IL-6, TNF-α) levels. Furthermore, concurrent increases in reactive oxygen species and nitric oxide production leading to increased caspase 3 activation are also observed in both murine and human microglial cells. These effects are recapitulated with TLR 2, 4, and 9 cognate ligands (Pam3CSK4, LPS, and CpG) and significantly attenuated in TLR 2 and 4 knock-out mice and TLR2/4 double knock-out mice. Therefor, our findings clearly suggest for the first time that activation of TLRs on microglia cells by morphine and TAT in the context of S. pneumoniae infection may be a potential mechanism for the increased prevalence of HAND in HIV-infected opioid-dependent patients.


Asunto(s)
Coinfección/metabolismo , Infecciones por VIH/metabolismo , Microglía/efectos de los fármacos , Morfina/farmacología , Neumonía Neumocócica/metabolismo , Receptores Toll-Like/metabolismo , Animales , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Citocinas/metabolismo , Infecciones por VIH/complicaciones , VIH-1 , Masculino , Ratones , Ratones Noqueados , Microglía/metabolismo , Neumonía Neumocócica/complicaciones , Receptores Opioides mu/genética , Receptores Opioides mu/metabolismo , Streptococcus pneumoniae , Receptores Toll-Like/genética
2.
J Biol Chem ; 286(34): 29806-17, 2011 Aug 26.
Artículo en Inglés | MEDLINE | ID: mdl-21730055

RESUMEN

Withdrawal stress is a common occurrence in opioid users, yet very few studies have examined the effects of morphine withdrawal (MW) on immune functioning or the role of glucocorticoids in MW-induced immunomodulation. This study investigated for the first time the role of glucocorticoids in MW modulation of LPS-induced IL-12p40, a key cytokine playing a pivotal role in immunoprotection. Using WT and µ-opioid receptor knock-out mice, we show that MW in vivo significantly attenuated LPS-induced IL-12p40 mRNA and protein expression. The role of glucocorticoids in MW modulation of IL-12p40 was investigated using a murine macrophage cell line, CRL2019, in an in vitro MW model. Interestingly, MW alone in the absence of glucocorticoids resulted in a significant reduction in IL-12p40 promoter activity and mRNA and protein expression. EMSA revealed a concurrent decrease in consensus binding to transcription factors NFκB, Activator Protein-1, and CCAAT/enhancer-binding protein and Western blot analysis demonstrated a significant activation of LPS-induced ERK1/2 phosphorylation. Interestingly, although glucocorticoid treatment alone also modulated these transcription factors and ERK1/2 activation, the addition of glucocorticoids to MW samples resulted in a greater than additive reduction in the transcription factors and significant hyperactivation of LPS-induced ERK1/2 phosphorylation. ERK inhibitors reversed MW and MW plus corticosterone inhibition of LPS-induced IL-12p40. The potentiating effects of glucocorticoids were non-genomic because nuclear translocation of glucocorticoid receptor was not significantly different between MW and corticosterone treatment. This study demonstrates for the first time that MW and glucocorticoids independently modulate IL-12p40 production through a mechanism involving ERK1/2 hyperactivation and that glucocorticoids can significantly augment MW-induced inhibition of IL-12p40.


Asunto(s)
Regulación de la Expresión Génica/efectos de los fármacos , Glucocorticoides/farmacología , Subunidad p40 de la Interleucina-12/biosíntesis , Lipopolisacáridos/farmacología , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Morfina , Estrés Fisiológico , Síndrome de Abstinencia a Sustancias/metabolismo , Animales , Factor de Unión a CCAAT/genética , Factor de Unión a CCAAT/metabolismo , Línea Celular , Activación Enzimática , Inhibidores Enzimáticos/farmacología , Subunidad p40 de la Interleucina-12/genética , Masculino , Ratones , Proteína Quinasa 3 Activada por Mitógenos/genética , Fosforilación/efectos de los fármacos , Fosforilación/genética , Receptores de Glucocorticoides/genética , Receptores de Glucocorticoides/metabolismo , Síndrome de Abstinencia a Sustancias/genética , Factor de Transcripción AP-1/genética , Factor de Transcripción AP-1/metabolismo
3.
Am J Pathol ; 176(2): 786-99, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20042674

RESUMEN

Patients prescribed morphine for the management of chronic pain, and chronic heroin abusers, often present with complications such as increased susceptibility to opportunistic infections and inadequate healing of wounds. We investigated the effect of morphine on wound-healing events in the presence of an infection in an in vivo murine model that mimics the clinical manifestations seen in opioid user and abuser populations. We show for the first time that in the presence of an inflammatory inducer, lipopolysaccharide, chronic morphine treatment results in a marked decrease in wound closure, compromised wound integrity, and increased bacterial sepsis. Morphine treatment resulted in a significant delay and reduction in both neutrophil and macrophage recruitment to the wound site. The delay and reduction in neutrophil reduction was attributed to altered early expression of keratinocyte derived cytokine and was independent of macrophage inflammatory protein 2 expression, whereas suppression of macrophage infiltration was attributed to suppressed levels of the potent macrophage chemoattractant monocyte chemotactic protein-1. When the effects of chronic morphine on later wound healing events were investigated, a significant suppression in angiogenesis and myofibroblast recruitment were observed in animals that received chronic morphine administration. Taken together, our findings indicate that morphine treatment results in a delay in the recruitment of cellular events following wounding, resulting in a lack of bacterial clearance and delayed wound closure.


Asunto(s)
Quimiotaxis de Leucocito/efectos de los fármacos , Inmunidad Celular/efectos de los fármacos , Morfina/farmacología , Cicatrización de Heridas/efectos de los fármacos , Heridas y Lesiones/inmunología , Analgésicos Opioides/administración & dosificación , Analgésicos Opioides/farmacología , Animales , Inhibición de Migración Celular/efectos de los fármacos , Células Cultivadas , Regulación hacia Abajo/efectos de los fármacos , Regulación hacia Abajo/inmunología , Esquema de Medicación , Evaluación Preclínica de Medicamentos , Masculino , Ratones , Ratones Endogámicos C57BL , Morfina/administración & dosificación , Factores de Tiempo , Heridas y Lesiones/patología
4.
Infect Immun ; 78(2): 830-7, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19995896

RESUMEN

Streptococcus pneumoniae is a pathogen that causes serious respiratory disease and meningitis in the immunocompromised drug abuse population. However, the precise mechanisms by which drug abuse compromises the host immune defense to pulmonary S. pneumoniae infection is not fully understood. Using a well-established murine model of opiate abuse and S. pneumoniae lung infection, we explored the influence of morphine treatment on the interleukin-23 (IL-23)/IL-17 axis and related innate immunity. Impairment of early IL-23/IL-17 production caused by morphine treatment was associated with delayed neutrophil migration and decreased pneumococcal clearance. Furthermore, morphine treatment impaired MyD88-dependent IL-23 production in alveolar macrophages and dendritic cells in response to in vitro S. pneumoniae cell infection. Moreover, morphine treatment significantly inhibited the S. pneumoniae-induced phosphorylation of interferon response factor 3 (IRF3), ATF2, and NF-kappaBp65. T-cell receptor delta (TCRdelta)-deficient mice showed a decrease in IL-17 production and a severely weakened capacity to clear lung S. pneumoniae infection. Finally, morphine treatment resulted in diminished secretion of antimicrobial proteins S100A9 and S100A8/A9 during early stages of S. pneumoniae infection. In conclusion, morphine treatment causes a dysfunction in IL-23-producing dendritic cells and macrophages and IL-17-producing gammadeltaT lymphocytes in response to S. pneumoniae lung infection. This leads to diminished release of antimicrobial S100A8/A9 proteins, compromised neutrophil recruitment, and more-severe infection.


Asunto(s)
Interleucina-17/biosíntesis , Interleucina-23/efectos de los fármacos , Morfina/farmacología , Narcóticos/farmacología , Mucosa Respiratoria/efectos de los fármacos , Animales , Western Blotting , Líquido del Lavado Bronquioalveolar , Células Dendríticas/efectos de los fármacos , Células Dendríticas/inmunología , Ensayo de Inmunoadsorción Enzimática , Inmunidad Innata/efectos de los fármacos , Interleucina-17/inmunología , Interleucina-23/biosíntesis , Interleucina-23/inmunología , Macrófagos Alveolares/efectos de los fármacos , Macrófagos Alveolares/inmunología , Ratones , Infecciones Neumocócicas/inmunología , ARN Mensajero/análisis , Mucosa Respiratoria/inmunología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Streptococcus pneumoniae
5.
Cell Immunol ; 265(2): 139-45, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20843508

RESUMEN

Delayed wound healing is a chronic problem in opioid drug abusers. We investigated the role chronic morphine plays on later stages of wound healing events using an angiogenesis model. Our results show that morphine treatment resulted in a significant decrease in inflammation induced angiogenesis. To delineate the mechanisms involved we investigate the role of hypoxia inducible factor 1 alpha (HIF-1 alpha), a potent inducer of angiogenic growth factor. Morphine treatment resulted in a significant decrease in the expression and nuclear translocation of HIF-1 alpha with a concurrent suppression in vascular endothelial growth factor (VEGF) synthesis. Cells of the innate immune system play a dominant role in the angiogenic process. Morphine treatment inhibited early recruitment of both neutrophils and monocytes towards an inflammatory signal with a significant decrease in the monocyte chemoattractant MCP-1. Taken together, our studies show that morphine regulates the wound repair process on multiple levels. Morphine acts both directly and indirectly in suppressing angiogenesis.


Asunto(s)
Morfina/farmacología , Neovascularización Patológica/tratamiento farmacológico , Cicatrización de Heridas/efectos de los fármacos , Inductores de la Angiogénesis/farmacología , Animales , Hipoxia de la Célula/efectos de los fármacos , Quimiocina CCL2/metabolismo , Antagonismo de Drogas , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Lipopolisacáridos/farmacología , Ratones , Monocitos/metabolismo , Morfina/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo
6.
J Leukoc Biol ; 71(5): 782-90, 2002 May.
Artículo en Inglés | MEDLINE | ID: mdl-11994502

RESUMEN

Wild-type and mu-opioid receptor knockout (MORKO) mice were used to investigate the role of corticosterone (CORT) and the mu-opioid receptor (MOR) in chronic morphine-mediated immunosuppression. We found that although plasma CORT concentrations in CORT infusion (10 mg/kg/day) and morphine-pellet implantation (75 mg) mice were similar (400-450 ng/ml), chronic morphine treatment resulted in a significantly higher (two- to threefold) inhibition of thymic, splenic, and lymph node cellularity; inhibition of thymic-lymphocyte proliferation; inhibition of IL-2 synthesis; and activation of macrophage nitric oxide (NO) production when compared with CORT infusion. In addition, results show that the inhibition of IFN-gamma synthesis and splenic- and lymph node-lymphocyte proliferation and activation of macrophage TNF-alpha and IL-1beta synthesis occurred only with chronic morphine treatment but not with CORT infusion. These morphine effects were abolished in MORKO mice. The role of the sympathetic nervous system on morphine-mediated effects was investigated by using the ganglionic blocker chlorisondamine. Our results show that chlorisondamine was able to only partially reverse morphine's inhibitory effects. The results clearly show that morphine-induced immunosuppression is mediated by the MOR and that although some functions are amplified in the presence of CORT or sympathetic activation, the inhibition of IFN-gamma synthesis and activation of macrophage-cytokine synthesis is CORT-independent and only partially dependent on sympathetic activation.


Asunto(s)
Corticosterona/farmacología , Inmunosupresores/farmacología , Morfina/farmacología , Receptores Opioides mu/fisiología , Animales , Células Cultivadas , Clorisondamina/farmacología , Corticosterona/sangre , Corticosterona/fisiología , Citocinas/biosíntesis , Implantes de Medicamentos , Bloqueadores Ganglionares/farmacología , Inmunosupresores/administración & dosificación , Inmunosupresores/antagonistas & inhibidores , Activación de Linfocitos/efectos de los fármacos , Linfocitos/efectos de los fármacos , Linfocitos/inmunología , Tejido Linfoide/citología , Tejido Linfoide/efectos de los fármacos , Tejido Linfoide/inmunología , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Morfina/administración & dosificación , Morfina/antagonistas & inhibidores , Óxido Nítrico/biosíntesis , ARN Mensajero/biosíntesis , Receptores Opioides mu/genética
7.
PLoS One ; 10(9): e0138587, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26381529

RESUMEN

OBJECTIVE: Intimal hyperplasia (IH) is a clinical concern leading to failure of up to 50% of vein grafts and 10% of arterial grafts after 10 years with no known current treatment. Recent studies have shown that hypoxia differentially regulates proliferation of vein derived smooth muscle cells (V-SMC) compared to artery derived smooth muscle cells (A-SMC). The objective of this study is to evaluate the effect of hypoxia on cellular migration and the mechanisms underlying the differential effects of hypoxia on A-SMC and V-SMC migration. METHODS AND RESULTS: Hypoxic treatment (3-5% O2) of Smooth Muscle Cells (SMC) resulted in differential migration in scratch wound and electric cell substrate impedance sensing (ECIS) assays. Hypoxia led to greater migration compared to normoxia with venous derived wound closure (V-SMC 30.8% Normoxia to 67% Hypoxia) greater than arterial wound closure (A-SMC 6.2% Normoxia to 24.7% Hypoxia). Paracrine factors secreted by hypoxic endothelial cells induced more migration in SMC compared to factors secreted by normoxic endothelial cells. Migration of V-SMC was greater than A-SMC in the presence of paracrine factors. Neutralizing antibody to Vascular Endothelial Growth Factor Receptor -1 (VEGFR-1) completely inhibited V-SMC migration while there was only partial inhibition of A-SMC migration. A-SMC migration was completely inhibited by Platelet Derived Growth Factor BB (PDGF-BB) neutralizing antibody. p38 Mitogen Activated Protein kinase (p38 MAPK) inhibitor pre-incubation completely inhibited migration induced by paracrine factors in both A-SMC and V-SMC. CONCLUSION: Our study determines that SMC migration under hypoxia occurs via both an autocrine and paracrine mechanism and is dependent on Vascular Endothelial Growth Factor-A (VEGF-A) in V-SMC and PDGF-BB in A-SMC. Migration of both A-SMC and V-SMC is inhibited by p38 MAPK inhibitor. These studies suggest that pharmacotherapeutic strategies directed at modulating p38 MAPK activity can be exploited to prevent IH in vascular grafts.


Asunto(s)
Hipoxia de la Célula/fisiología , Movimiento Celular/fisiología , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Arterias/metabolismo , Humanos , Músculo Liso Vascular/citología , Miocitos del Músculo Liso/citología , Fosforilación , Receptor 1 de Factores de Crecimiento Endotelial Vascular/metabolismo , Venas/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
8.
J Neuroimmunol ; 147(1-2): 78-81, 2004 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-14741432

RESUMEN

Chronic morphine treatment in animal models has been shown to alter a number of immune parameters including suppression of cellular immunity. T helper cell differentiation into Th2 effector cell may be a major contributing factor to impaired cellular immunity following chronic drug abuse. We had previously shown that chronic morphine treatment in vivo and in vitro decreases IL-2 and IFNgamma (Th1) protein levels and increases IL-4 and IL-5 (Th2) protein levels in a time-dependent manner. In addition in this paper, we show that chronic morphine treatment resulted in a decrease in IFNgamma and IL-2 mRNA and an increase in IL-4 and IL-5 mRNA accumulation in murine splenocytes. Furthermore, chronic morphine treatment inhibited IFNgamma promoter activity and increased IL-4 promoter activity in respective promoter transfected primary T cells. In addition, we also demonstrate that chronic morphine treatment resulted in an increase in GATA 3 binding to DNA consensus elements in electromobility shift assays and an increase in GATA 3 protein and mRNA levels. In contrast, chronic morphine treatment resulted in a decrease in T-bet mRNA levels. From these data, we conclude that chronic morphine treatment differentiates T helper cell to Th2 effector cells by modulating key master switches that results in committing T helper cell to a Th2 phenotype.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Morfina/farmacología , Narcóticos/farmacología , Linfocitos T Colaboradores-Inductores/efectos de los fármacos , Células Th2/efectos de los fármacos , Transactivadores/metabolismo , Factores de Transcripción/metabolismo , Animales , Anticuerpos/farmacología , Western Blotting/métodos , Antígenos CD28/inmunología , Complejo CD3/inmunología , Recuento de Células/métodos , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Citocinas/genética , Citocinas/metabolismo , ADN/metabolismo , Proteínas de Unión al ADN/genética , Relación Dosis-Respuesta a Droga , Ensayo de Cambio de Movilidad Electroforética/métodos , Factor de Transcripción GATA3 , Ratones , Ratones Noqueados , Regiones Promotoras Genéticas/efectos de los fármacos , Regiones Promotoras Genéticas/fisiología , Unión Proteica , ARN Mensajero/biosíntesis , ARN Mensajero/efectos de los fármacos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/métodos , Proteínas de Dominio T Box , Linfocitos T Colaboradores-Inductores/metabolismo , Células TH1/efectos de los fármacos , Células TH1/metabolismo , Células Th2/metabolismo , Transactivadores/genética , Factores de Transcripción/genética
9.
PLoS One ; 8(1): e54040, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23349783

RESUMEN

Opiates are among the most prescribed drugs for pain management. However, morphine use or abuse results in significant gut bacterial translocation and predisposes patients to serious infections with gut origin. The mechanism underlying this defect is still unknown. In this report, we investigated the mechanisms underlying compromised gut immune function and bacterial translocation following morphine treatment. We demonstrate significant bacterial translocation to mesenteric lymph node (MLN) and liver following morphine treatment in wild-type (WT) animals that was dramatically and significantly attenuated in Toll-like receptor (TLR2 and 4) knockout mice. We further observed significant disruption of tight junction protein organization only in the ileum but not in the colon of morphine treated WT animals. Inhibition of myosin light chain kinase (MLCK) blocked the effects of both morphine and TLR ligands, suggesting the role of MLCK in tight junction modulation by TLR. This study conclusively demonstrates that morphine induced gut epithelial barrier dysfunction and subsequent bacteria translocation are mediated by TLR signaling and thus TLRs can be exploited as potential therapeutic targets for alleviating infections and even sepsis in morphine-using or abusing populations.


Asunto(s)
Traslocación Bacteriana/efectos de los fármacos , Intestinos/efectos de los fármacos , Morfina/farmacología , Receptor Toll-Like 2/metabolismo , Receptor Toll-Like 4/metabolismo , Analgésicos Opioides/farmacología , Animales , Azepinas/farmacología , Western Blotting , Línea Celular , Colon/efectos de los fármacos , Colon/metabolismo , Colon/microbiología , Epitelio/efectos de los fármacos , Epitelio/metabolismo , Epitelio/microbiología , Íleon/efectos de los fármacos , Íleon/metabolismo , Íleon/microbiología , Mucosa Intestinal/metabolismo , Intestinos/microbiología , Hígado/efectos de los fármacos , Hígado/metabolismo , Hígado/microbiología , Ganglios Linfáticos/efectos de los fármacos , Ganglios Linfáticos/metabolismo , Ganglios Linfáticos/microbiología , Mesenterio , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía Fluorescente , Quinasa de Cadena Ligera de Miosina/antagonistas & inhibidores , Quinasa de Cadena Ligera de Miosina/metabolismo , Naftalenos/farmacología , Receptores Opioides mu/genética , Receptores Opioides mu/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Uniones Estrechas/efectos de los fármacos , Uniones Estrechas/metabolismo , Receptor Toll-Like 2/genética , Receptor Toll-Like 4/genética , Proteína de la Zonula Occludens-1/metabolismo
10.
J Neuroimmune Pharmacol ; 6(4): 442-65, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21789507

RESUMEN

Infection rate among intravenous drug users (IDU) is higher than the general public, and is the major cause of morbidity and hospitalization in the IDU population. Epidemiologic studies provide data on increased prevalence of opportunistic bacterial infections such as TB and pneumonia, and viral infections such as HIV-1 and hepatitis in the IDU population. An important component in the intravenous drug abuse population and in patients receiving medically indicated chronic opioid treatment is opioid withdrawal. Data on bacterial virulence in the context of opioid withdrawal suggest that mice undergoing withdrawal had shortened survival and increased bacterial load in response to Salmonella infection. As the body of evidence in support of opioid dependency and its immunosuppressive effects is growing, it is imperative to understand the mechanisms by which opioids exert these effects and identify the populations at risk that would benefit the most from the interventions to counteract opioid immunosuppressive effects. Thus, it is important to refine the existing animal model to closely match human conditions and to cross-validate these findings through carefully controlled human studies. Better understanding of the mechanisms will facilitate the search for new therapeutic modalities to counteract adverse effects including increased infection rates. This review will summarize the effects of morphine on innate and adaptive immunity, identify the role of the mu opioid receptor in these functions and the signal transduction activated in the process. The role of opioid withdrawal in immunosuppression and the clinical relevance of these findings will also be discussed.


Asunto(s)
Analgésicos Opioides/inmunología , Sistema Inmunológico/efectos de los fármacos , Infecciones Oportunistas/inducido químicamente , Infecciones Oportunistas/inmunología , Trastornos Relacionados con Sustancias/inmunología , Analgésicos Opioides/farmacología , Animales , Humanos , Morfina/farmacología , Dependencia de Morfina/inmunología , Dependencia de Morfina/microbiología , Infecciones Oportunistas/metabolismo , Receptores Opioides mu/metabolismo , Síndrome de Abstinencia a Sustancias/inmunología , Síndrome de Abstinencia a Sustancias/metabolismo , Trastornos Relacionados con Sustancias/metabolismo
11.
Arch Immunol Ther Exp (Warsz) ; 56(5): 299-309, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18836861

RESUMEN

The first line of defense against invading bacteria is provided by the innate immune system. Morphine and other opiates can immediately disrupt the body's first line of defense against harmful external bacteria. Opiate, for example morphine, abuse degrades physical and physiologic barriers, and modulates phagocytic cells (macrophages, neutrophils) and, nonspecific cytotoxic T cells (gammadelta T), natural killer cells, and dendritic cells, that are functionally important for carrying out a rapid immune reaction to invading pathogens. In vitro studies with innate immune cells from experimental animals and humans and in vivo studies with animal models have shown that opiate abuse impairs innate immunity and is responsible for increased susceptibility to bacterial infection. However, to better understand the complex interactions between opiates, innate immunity, and bacterial infection and develop novel approaches to treat and even prevent bacterial infection in the opiate-abuse population, there is an urgent need to fill the numerous gaps in our understanding of the cellular and molecular mechanisms by which opiate abuse increases susceptibility to bacterial infection.


Asunto(s)
Infecciones Bacterianas/inmunología , Inmunidad Innata , Trastornos Relacionados con Opioides/inmunología , Animales , Infecciones Bacterianas/complicaciones , Citocinas/biosíntesis , Citocinas/inmunología , Susceptibilidad a Enfermedades , Humanos , Trastornos Relacionados con Opioides/complicaciones
12.
J Immunol ; 180(5): 3594-600, 2008 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-18292587

RESUMEN

Resident alveolar macrophages and respiratory epithelium constitutes the first line of defense against invading lung pneumococci. Results from our study showed that increased mortality and bacterial outgrowth and dissemination seen in morphine-treated mice were further exaggerated following depletion of alveolar macrophages with liposomal clodronate. Using an in vitro alveolar macrophages and lung epithelial cells infection model, we show significant release of MIP-2 from alveolar macrophages, but not from lung epithelial cells, following 4 h of exposure of cells to pneumococci infection. Morphine treatment reduced MIP-2 release in pneumococci stimulated alveolar macrophages. Furthermore, morphine treatment inhibited Streptococcus pneumoniae-induced NF-kappaB-dependent gene transcription in alveolar macrophages following 2 h of in vitro infection. S. pneumoniae infection resulted in a significant induction of NF-kappaB activity only in TLR9 stably transfected HEK 293 cells, but not in TLR2 and TLR4 transfected HEK 293 cells, and morphine treatment inhibited S. pneumoniae-induced NF-kappaB activity in these cells. Moreover, morphine treatment also decreased bacterial uptake and killing in alveolar macrophages. Taken together, these results suggest that morphine treatment impairs TLR9-NF-kappaB signaling and diminishes bacterial clearance following S. pneumoniae infection in resident macrophages during the early stages of infection, leading to a compromised innate immune response.


Asunto(s)
Macrófagos Alveolares/efectos de los fármacos , Macrófagos Alveolares/microbiología , Morfina/uso terapéutico , FN-kappa B/antagonistas & inhibidores , Neumonía Neumocócica/tratamiento farmacológico , Neumonía Neumocócica/inmunología , Transducción de Señal/efectos de los fármacos , Streptococcus pneumoniae/inmunología , Receptor Toll-Like 9/antagonistas & inhibidores , Animales , Línea Celular , Línea Celular Transformada , Humanos , Macrófagos Alveolares/inmunología , Macrófagos Alveolares/patología , Ratones , Ratones Endogámicos C57BL , Morfina/administración & dosificación , FN-kappa B/fisiología , Infiltración Neutrófila/efectos de los fármacos , Infiltración Neutrófila/inmunología , Neumonía Neumocócica/microbiología , Neumonía Neumocócica/mortalidad , Transducción de Señal/inmunología , Streptococcus pneumoniae/efectos de los fármacos , Factores de Tiempo , Receptor Toll-Like 2/fisiología , Receptor Toll-Like 4/fisiología , Receptor Toll-Like 9/fisiología
13.
J Immunol ; 180(6): 3670-9, 2008 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-18322172

RESUMEN

There are very few studies that examine the effects that morphine withdrawal has on immune functioning, and of these even fewer describe the mechanisms by which withdrawal brings about these changes. Our previous work demonstrated that morphine withdrawal contributed to Th cell differentiation by biasing cells toward the Th2 lineage. A major finding from these studies was that IL-12 was decreased following withdrawal, and it was concluded that this decrease may be a mechanism by which morphine withdrawal is mediating Th2 polarization. Therefore, it was the aim of the current studies to develop an in vitro model to examine the process of morphine withdrawal and to understand the signaling mechanisms that withdrawal may use to effect IL-12 production through the use of this model. It was demonstrated and concluded that morphine withdrawal may be effecting IL-12 production by increasing cAMP levels, which activates protein kinase A. Protein kinase A activation then prevents the phosphorylation and subsequent degradation of IkappaB, which in turn prevents translocation of the NF-kappaB p65 subunit to the nucleus to transactivate the IL-12 p40 gene, ultimately resulting in decreased IL-12 production following LPS stimulation.


Asunto(s)
AMP Cíclico/fisiología , Regulación hacia Abajo/inmunología , Inmunosupresores/toxicidad , Interleucina-12/antagonistas & inhibidores , Interleucina-12/biosíntesis , Macrófagos Alveolares/inmunología , Morfina/toxicidad , Síndrome de Abstinencia a Sustancias/inmunología , Animales , Línea Celular , Células Cultivadas , Modelos Animales de Enfermedad , Regulación hacia Abajo/efectos de los fármacos , Humanos , Inmunosupresores/administración & dosificación , Subunidad p40 de la Interleucina-12/antagonistas & inhibidores , Subunidad p40 de la Interleucina-12/biosíntesis , Lipopolisacáridos/farmacología , Macrófagos Alveolares/efectos de los fármacos , Macrófagos Alveolares/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Morfina/administración & dosificación , Transducción de Señal/efectos de los fármacos , Transducción de Señal/inmunología , Bazo/citología , Bazo/inmunología , Bazo/metabolismo , Síndrome de Abstinencia a Sustancias/metabolismo
14.
J Biol Chem ; 282(10): 7164-71, 2007 Mar 09.
Artículo en Inglés | MEDLINE | ID: mdl-17227776

RESUMEN

Chronic morphine inhibits interleukin-2 (IL-2) at both the transcriptional and protein synthesis levels. The molecular mechanisms by which morphine decreases IL-2 are not fully understood. The production of IL-2 is tightly regulated by several transcription factors that bind to the IL-2 promoter. Herein, we show that chronic morphine treatment results in an increase in cAMP levels with a concurrent up-regulation of the cAMP inducible repressor inducible cAMP early repressor (ICER)/cAMP response element modulator (CREM) and down-regulation of p-cAMP-response element-binding protein (CREB) in activated T cells. Furthermore, ICER competes for p-CREB binding to the cAMP-responsive elements (CREs) site. This leads to the uncoupling of CBP/p300 thereby abrogating IL-2 transcription. Overexpression of either antisense CREM or CREB plasmid rescued morphine-induced inhibition of IL-2 promoter activity and protein production. In addition, we also found that chronic morphine treatment inhibited the acetylation and trimethylation of histones and decreased both DNA demethylation and accessibility of the IL-2 promoter. These findings suggest that chronic morphine treatment may function through both transcriptional and epigenetic mechanisms to inhibit IL-2 production.


Asunto(s)
Epigénesis Genética/efectos de los fármacos , Interleucina-2/genética , Activación de Linfocitos , Morfina/farmacología , Linfocitos T/efectos de los fármacos , Transcripción Genética/efectos de los fármacos , Animales , Modulador del Elemento de Respuesta al AMP Cíclico/genética , Modulador del Elemento de Respuesta al AMP Cíclico/metabolismo , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Metilación de ADN , Regulación de la Expresión Génica/efectos de los fármacos , Interleucina-2/biosíntesis , Ratones , Ratones Endogámicos C57BL , Regiones Promotoras Genéticas , Linfocitos T/metabolismo , Factores de Transcripción p300-CBP/metabolismo
15.
J Immunol ; 175(4): 2655-65, 2005 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-16081842

RESUMEN

The consequences that drug withdrawal has on immune functioning has only recently been appreciated; however, given the wide variety of use and abuse of opiate analgesics, understanding the decrements to immune function that withdrawal from these drugs causes is of crucial importance. In previous work, we have demonstrated that morphine treatment contributes to immunosuppression by polarizing Th cells toward the Th2 lineage. In the current study, it was hypothesized that morphine withdrawal would result in Th2 differentiation and subsequent immune dysfunction. To address this hypothesis, mice were chronically treated with morphine for 72 h followed by a 24-h withdrawal period. It was determined that 24-h morphine withdrawal resulted in a decrease in IFN-gamma, the Th1 signature cytokine, whereas the Th2 cytokine, IL-4, was increased. In addition, Western blot and EMSA experiments revealed that morphine withdrawal-induced Th2 differentiation was mediated through the classical Th2 transcription factors Stat-6 and GATA-3. In addition, the consequence of morphine withdrawal in the presence of an immune stimulation was also examined by treating mice in vivo with LPS before morphine withdrawal. Following withdrawal, it was found that the Th1-polarizing cytokine IL-12 was significantly decreased, providing further support for the observation that withdrawal results in Th2 differentiation by possibly impacting the generation of an appropriate innate immune response which directs subsequent adaptive Th1/Th2 responses.


Asunto(s)
Diferenciación Celular/inmunología , Dependencia de Morfina/inmunología , Síndrome de Abstinencia a Sustancias/inmunología , Células Th2/inmunología , Células Th2/metabolismo , Transporte Activo de Núcleo Celular/efectos de los fármacos , Animales , Diferenciación Celular/efectos de los fármacos , Linaje de la Célula/efectos de los fármacos , Linaje de la Célula/inmunología , Células Cultivadas , Corticosterona/metabolismo , Corticosterona/fisiología , Modelos Animales de Enfermedad , Factor de Transcripción GATA3/genética , Factor de Transcripción GATA3/metabolismo , Interleucina-4/biosíntesis , Lipopolisacáridos/administración & dosificación , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Dependencia de Morfina/metabolismo , Unión Proteica/efectos de los fármacos , ARN Mensajero/biosíntesis , Elementos de Respuesta/efectos de los fármacos , Factor de Transcripción STAT5/metabolismo , Factor de Transcripción STAT6/metabolismo , Síndrome de Abstinencia a Sustancias/metabolismo , Células Th2/citología
16.
J Immunol ; 174(1): 426-34, 2005 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-15611267

RESUMEN

Chronic morphine use impairs host innate immune response and increases susceptibility to bacteria and virus. In this study a novel mouse model of chronic morphine treatment, followed by intranasal inoculation with Streptococcus pneumoniae, was used to investigate microbial events and host innate immune response. Our results show that chronic morphine treatment markedly delayed neutrophil recruitment and increased bacterial burden in the lung, spleen, and blood with a subsequent increase in mortality. In morphine-treated animals, before neutrophil recruitment, a significant decrease in TNF-alpha, IL-1, IL-6, MIP-2, and KC was observed both in bronchoalveolar lavage fluids and in lung tissue. In the early phase of infection, we found that accumulation of galectin-3 in the alveolar space of streptococcus-infected lungs was decreased after morphine treatment. The transcription factor NF-kappaB in lung resident cells was also inhibited after morphine treatment. Taken together, these results suggest that chronic morphine treatment in an S. pneumoniae infection model suppresses NF-kappaB gene transcription in lung resident cells, which, in turn, modulates the transcriptional regulation of MIP-2 and inflammatory cytokines. The decreased synthesis of MIP-2 and inflammatory cytokines coupled with the decreased release of galectin-3 result in reduced migration of neutrophils to the site of infection, thereby increasing susceptibility to S. pneumoniae infection after morphine treatment.


Asunto(s)
Analgésicos Opioides/farmacología , Susceptibilidad a Enfermedades , Inmunidad Innata/efectos de los fármacos , Pulmón/microbiología , Morfina/farmacología , Infecciones Neumocócicas/inmunología , Animales , Western Blotting , Líquido del Lavado Bronquioalveolar/inmunología , Líquido del Lavado Bronquioalveolar/microbiología , Quimiocina CXCL2 , Quimiocinas/metabolismo , Quimiotaxis de Leucocito/efectos de los fármacos , Citocinas/efectos de los fármacos , Citocinas/metabolismo , Modelos Animales de Enfermedad , Galectina 3/efectos de los fármacos , Galectina 3/metabolismo , Pulmón/patología , Masculino , Ratones , FN-kappa B/efectos de los fármacos , FN-kappa B/metabolismo , Neutrófilos/efectos de los fármacos , Receptores de Interleucina-8B/efectos de los fármacos , Receptores de Interleucina-8B/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Bazo/microbiología
17.
J Immunol ; 175(10): 6361-7, 2005 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-16272288

RESUMEN

Impaired host defense mechanisms after major operative procedures and trauma are recognized as important factors in the development of infectious complication. Trauma is associated with impaired cellular immunity and CD4+ T cell Th2 differentiation. We have previously implicated morphine treatment as a possible mechanism for Th2 differentiation after injury. In this investigation we first establish that morphine treatment in vivo results in Th2 differentiation and that this effect is mediated through a naltrexone-sensitive opioid receptor. We investigated the intracellular mechanism by which morphine controls CD4+ T cell differentiation and demonstrate that morphine treatment in vitro 1) increases anti CD3/CD28 Ab-induced CD4+ T cell IL-4 protein synthesis, IL-4 mRNA, and GATA-3 mRNA accumulation through a pertussis toxin-sensitive receptor; 2) results in a dose-dependent increase in anti-CD3/CD28 Ab-induced CD4+ T cell cytoplasmic cAMP concentration; and 3) increases the forskolin-stimulated cytoplasmic cAMP level through a pertussis toxin-sensitive receptor. We also demonstrate that chronic morphine treatment increases anti-CD3/CD28 Ab-induced IL-4 promoter activity and IL-4 immunoprotein expression through a p38 MAPK-dependent, but protein kinase A- and Erk1/Erk2-independent, mechanism.


Asunto(s)
Adenilil Ciclasas/metabolismo , Linfocitos T CD4-Positivos/efectos de los fármacos , Linfocitos T CD4-Positivos/inmunología , Interleucina-4/biosíntesis , Morfina/farmacología , Animales , Antígenos CD28/metabolismo , Complejo CD3/metabolismo , Linfocitos T CD4-Positivos/citología , Linfocitos T CD4-Positivos/enzimología , Diferenciación Celular/efectos de los fármacos , Colforsina/farmacología , AMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Humanos , Técnicas In Vitro , Interleucina-4/genética , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Morfina/administración & dosificación , Toxina del Pertussis/farmacología , Regiones Promotoras Genéticas/efectos de los fármacos , Células Th2/citología , Células Th2/efectos de los fármacos , Células Th2/enzimología , Células Th2/inmunología , Heridas y Lesiones/tratamiento farmacológico , Heridas y Lesiones/inmunología
18.
J Biol Chem ; 278(39): 37622-31, 2003 Sep 26.
Artículo en Inglés | MEDLINE | ID: mdl-12842891

RESUMEN

To explore the mechanism by which morphine promotes the incidence of HIV infection, we evaluated the regulatory role of morphine on the interferon-gamma (IFN-gamma) promoter in activated T cells from wild type and mu-opioid receptor knockout mice. Our results show that morphine inhibited anti-CD3/CD28-stimulated IFN-gamma promoter activity in a dose-dependent manner. Chronic morphine treatment of T cells increased intracellular cAMP. To evaluate the role of cAMP in morphine's modulatory function, the effects of dibutyryl cyclic AMP and forskolin were investigated. Both dibutyryl cyclic AMP and forskolin treatment inhibited IFN-gamma promoter activity. Treatment with pertussis toxin, but not with a protein kinase A inhibitor, antagonized morphine's inhibitory effects. Morphine inhibited phosphorylation of ERK1/2 and p38 MAPK; in addition, morphine treatment in the presence of either ERK1/2 or p38 MAPK inhibitor (PD98059 or SB203580) resulted in an additive inhibition of IFN-gamma promoter activity. The transcription factor activator protein-1, NF-kappaB, and nuclear factor of activated T cells (NFAT) were negatively regulated by morphine. Overexpression of NF-kappaB p65 rescued the inhibitory effect of morphine on IFN-gamma promoter activity. However, only when NFATc1 was co-overexpressed with c-fos was the inhibitory effect of morphine on IFN-gamma promoter counteracted. The inhibitory effects of morphine were not observed in T cells obtained from mu-opioid receptor knockout mice, suggesting that morphine modulation of IFN-gamma promoter activity is mediated through the mu-opioid receptor. In summary, our data indicate that morphine modulation of IFN-gamma promoter activity is mediated through two distinct cAMP-dependent pathways, the NF-kappaB signaling pathway and the ERK1/2, p38 MAPK, AP-1/NFAT pathway.


Asunto(s)
AMP Cíclico/fisiología , Interferón gamma/genética , Morfina/farmacología , Proteínas Nucleares , Regiones Promotoras Genéticas/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Linfocitos T/metabolismo , Animales , Antígenos CD28/fisiología , Proteína Quinasa Tipo II Dependiente de AMP Cíclico , Proteínas Quinasas Dependientes de AMP Cíclico/fisiología , Proteínas de Unión al ADN/metabolismo , Activación de Linfocitos , Ratones , Ratones Endogámicos BALB C , Proteínas Quinasas Activadas por Mitógenos/fisiología , FN-kappa B/metabolismo , Factores de Transcripción NFATC , Proteínas Proto-Oncogénicas c-fos/análisis , Factor de Transcripción AP-1/metabolismo , Factores de Transcripción/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos
19.
J Immunol ; 169(7): 3630-6, 2002 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-12244154

RESUMEN

Psychological stress is associated with immunosuppression in both humans and animals. Although it was well established that psychological stressors stimulate the hypothalamic-pituitary-adrenal axis and the sympathetic nervous system, resulting in the release of various hormones and neurotransmitters, the mechanisms underlying these phenomena are poorly understood. In this study, mu-opioid receptor knockout (MORKO) mice were used to investigate whether the mu-opioid receptor mediates the immunosuppression induced by restraint stress. Our results showed that wild-type (WT) mice subjected to chronic 12-h daily restraint stress for 2 days exhibited a significant decrease in splenocyte number with a substantial increase in apoptosis and CD95 (Fas/APO-1) expression of splenocytes. The effects are essentially abolished in MORKO mice. Furthermore, inhibition of splenic lymphocyte proliferation, IL-2, and IFN-gamma production induced by restraint stress in WT mice was also significantly abolished in MORKO mice. Interestingly, both stressed WT and MORKO mice showed a significant elevation in plasma corticosterone and pituitary proopiomelanocortin mRNA expression, although the increase was significantly lower in MORKO mice. Adrenalectomy did not reverse restraint stress-induced immunosuppression in WT mice. These data clearly established that the mu-opioid receptor is involved in restraint stress-induced immune alterations via a mechanism of apoptotic cell death, and that the effect is not mediated exclusively through the glucocorticoid pathway.


Asunto(s)
Apoptosis/inmunología , Subgrupos Linfocitarios/metabolismo , Receptores Opioides mu/fisiología , Estrés Psicológico/inmunología , Estrés Psicológico/metabolismo , Adrenalectomía , Animales , Corticosterona/sangre , Femenino , Sistema Hipotálamo-Hipofisario/inmunología , Terapia de Inmunosupresión , Interferón gamma/biosíntesis , Interleucina-2/biosíntesis , Activación de Linfocitos/genética , Recuento de Linfocitos , Subgrupos Linfocitarios/citología , Subgrupos Linfocitarios/inmunología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Sistema Hipófiso-Suprarrenal/inmunología , Proopiomelanocortina/biosíntesis , Proopiomelanocortina/genética , ARN Mensajero/biosíntesis , Receptores Opioides delta/fisiología , Receptores Opioides kappa/fisiología , Receptores Opioides mu/deficiencia , Receptores Opioides mu/genética , Restricción Física , Bazo/citología , Bazo/inmunología , Bazo/metabolismo , Estrés Psicológico/genética , Estrés Psicológico/patología , Regulación hacia Arriba/genética , Regulación hacia Arriba/inmunología , Receptor fas/biosíntesis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA