Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Nat Commun ; 10(1): 951, 2019 03 12.
Artículo en Inglés | MEDLINE | ID: mdl-30862842

RESUMEN

Targeted noninvasive control of the nervous system and end-organs may enable safer and more effective treatment of multiple diseases compared to invasive devices or systemic medications. One target is the cholinergic anti-inflammatory pathway that consists of the vagus nerve to spleen circuit, which has been stimulated with implantable devices to improve autoimmune conditions such as rheumatoid arthritis. Here we report that daily noninvasive ultrasound (US) stimulation targeting the spleen significantly reduces disease severity in a mouse model of inflammatory arthritis. Improvements are observed only with specific parameters, in which US can provide both protective and therapeutic effects. Single cell RNA sequencing of splenocytes and experiments in genetically-immunodeficient mice reveal the importance of both T and B cell populations in the anti-inflammatory pathway. These findings demonstrate the potential for US stimulation of the spleen to treat inflammatory diseases.


Asunto(s)
Artritis Experimental/fisiopatología , Artritis Experimental/terapia , Bazo/inervación , Bazo/fisiopatología , Terapia por Ultrasonido/métodos , Animales , Artritis Experimental/inmunología , Artritis Reumatoide/inmunología , Artritis Reumatoide/fisiopatología , Artritis Reumatoide/terapia , Linfocitos B/inmunología , Linfocitos B/metabolismo , Fibras Colinérgicas/inmunología , Fibras Colinérgicas/fisiología , Expresión Génica , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Vías Nerviosas/inmunología , Neuroinmunomodulación/genética , Bazo/inmunología , Linfocitos T/inmunología , Linfocitos T/metabolismo , Estimulación del Nervio Vago/métodos
2.
Arthritis Res Ther ; 20(1): 154, 2018 07 27.
Artículo en Inglés | MEDLINE | ID: mdl-30053832

RESUMEN

BACKGROUND: Pain accompanies rheumatoid arthritis and other chronic inflammatory conditions and is difficult to manage. Although opioids provide potent analgesia, chronic opioid use can cause tolerance and addiction. Recent studies have demonstrated functional interactions between chemokine and opioid receptor signaling pathways. Reported heterodimerization of chemokine and opioid receptors led our group to develop bivalent compounds that bind both types of receptors, with the goal of targeting opioids to sites of inflammation. MCC22 is a novel bivalent compound containing a CCR5 antagonist and mu opioid receptor (MOR) agonist pharmacophores linked through a 22-atom spacer. We evaluated the efficacy of MCC22 in the K/B.g7 T-cell receptor transgenic mouse model of spontaneous inflammatory arthritis. METHODS: MCC22 or morphine was administered intraperitoneally at varying doses to arthritic K/B.g7 mice or nonarthritic control mice. Mechanical pain hypersensitivity was measured each day before and after drug administration, using the electronic von Frey test. The potency of MCC22 relative to that of morphine was calculated. Functional readouts of pain included grip strength and nesting behavior. A separate dosing regimen was used to determine whether the drugs induced pharmacologic tolerance. RESULTS: MCC22 provided ~ 3000-fold more potent analgesia than morphine in this model. Daily treatment with MCC22 also led to a cumulative analgesic effect, reducing the daily baseline pain level. MCC22 produced no observable analgesic effect in nonarthritic control mice. Importantly, repeated administration of MCC22 did not induce pharmacologic tolerance, whereas a similar regimen of morphine did. Both grip strength and nesting behaviors improved among arthritic mice treated with MCC22. Ankle thickness and arthritis scores were not affected by MCC22. The analgesic effect of MCC22 was abolished in K/B.g7 mice genetically lacking CCR5, demonstrating the receptor specificity of the antagonist pharmacophore. CONCLUSIONS: MCC22 is a novel bivalent ligand that targets CCR5 and MOR. Our findings demonstrate that MCC22 provides highly potent analgesia and improved functional outcomes in a model of inflammatory arthritis, without inducing typical opioid tolerance. These findings suggest that MCC22 or similar compounds could be used to treat the pain associated with inflammatory arthritis and related conditions, while minimizing the risks typically associated with chronic opioid use.


Asunto(s)
Analgésicos/farmacología , Artritis Experimental/patología , Artritis Reumatoide/patología , Isoquinolinas/farmacología , Dolor , Piperidinas/farmacología , Receptores CCR5/efectos de los fármacos , Receptores Opioides mu/agonistas , Animales , Artritis Experimental/complicaciones , Artritis Reumatoide/complicaciones , Tolerancia a Medicamentos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Dolor/etiología , Receptores CCR5/agonistas , Receptores CCR5/metabolismo
3.
Gut ; 67(4): 600-602, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-28642332

RESUMEN

BACKGROUND: Opioids such as morphine are widely used for the management of pain associated with acute pancreatitis. Interestingly, opioids are also known to affect the immune system and modulate inflammatory pathways in non-pancreatic diseases. However, the impact of morphine on the progression of acute pancreatitis has never been evaluated. In the current study, we evaluated the impact of morphine on the progression and severity of acute pancreatitis. METHODS: Effect of morphine treatment on acute pancreatitis in caerulein, L-arginine and ethanol-palmitoleic acid models was evaluated after induction of the disease. Inflammatory response, gut permeability and bacterial translocation were compared. Experiments were repeated in mu (µ) opioid receptor knockout mice (MORKO) and in wild-type mice in the presence of opioid receptor antagonist naltrexone to evaluate the role of µ-opioid receptors in morphine's effect on acute pancreatitis. Effect of morphine treatment on pathways activated during pancreatic regeneration like sonic Hedgehog and activation of embryonic transcription factors like pdx-1 and ptf-1 were measured by immunofluorescence and quantitative PCR. RESULTS: Histological data show that treatment with morphine after induction of acute pancreatitis exacerbates the disease with increased pancreatic neutrophilic infiltration and necrosis in all three models of acute pancreatitis. Morphine also exacerbated acute pancreatitis-induced gut permeabilisation and bacteraemia. These effects were antagonised in the MORKO mice or in the presence of naltrexone suggesting that morphine's effect on severity of acute pancreatitis are mediated through the µ-opioid receptors. Morphine treatment delayed macrophage infiltration, sonic Hedgehog pathway activation and expression of pdx-1 and ptf-1. CONCLUSION: Morphine treatment worsens the severity of acute pancreatitis and delays resolution and regeneration. Considering our results, the safety of morphine for analgesia during acute pancreatitis should be re-evaluated in future human studies.


Asunto(s)
Analgésicos Opioides/efectos adversos , Morfina/efectos adversos , Páncreas/patología , Pancreatitis/diagnóstico , Enfermedad Aguda , Analgésicos Opioides/administración & dosificación , Animales , Arginina , Ceruletida , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Ácidos Grasos Monoinsaturados , Ratones , Ratones Noqueados , Morfina/administración & dosificación , Pancreatitis/inducido químicamente , Pancreatitis/tratamiento farmacológico , Índice de Severidad de la Enfermedad , Factores de Tiempo
5.
Sci Rep ; 6: 21094, 2016 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-26891899

RESUMEN

Opioid drug abusers have a greater susceptibility to gram positive (Gram (+)) bacterial infections. However, the mechanism underlying opioid modulation of Gram (+) versus Gram (-) bacterial clearance has not been investigated. In this study, we show that opioid treatment resulted in reduced phagocytosis of Gram (+), when compared to Gram (-) bacteria. We further established that LPS priming of chronic morphine treated macrophages leads to potentiated phagocytosis and killing of both Gram (+) and Gram (-) bacteria in a P-38 MAP kinase dependent signaling pathway. In contrast, LTA priming lead to inhibition of both phagocytosis and bacterial killing. This study demonstrates for the first time the differential effects of TLR4 and TLR2 agonists on morphine induced inhibition of phagocytosis. Our results suggest that the incidence and severity of secondary infections with Gram (+) bacteria would be higher in opioid abusers.


Asunto(s)
Bacterias Gramnegativas/metabolismo , Bacterias Grampositivas/metabolismo , Lipopolisacáridos/farmacología , Morfina/farmacología , Fagocitosis/efectos de los fármacos , Animales , Expresión Génica , Bacterias Gramnegativas/inmunología , Bacterias Grampositivas/inmunología , Inmunomodulación , Ligandos , Lipopolisacáridos/inmunología , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Macrófagos/metabolismo , Ratones , Ratones Noqueados , Viabilidad Microbiana/efectos de los fármacos , Viabilidad Microbiana/inmunología , Modelos Biológicos , Fagocitosis/inmunología , Ácidos Teicoicos/inmunología , Ácidos Teicoicos/farmacología , Receptor Toll-Like 2/genética , Receptor Toll-Like 2/metabolismo , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/metabolismo
6.
J Neuroinflammation ; 12: 120, 2015 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-26087960

RESUMEN

BACKGROUND: Persistent systemic infection results in excessive trafficking of peripheral immune cells into the central nervous system (CNS), thereby contributing to sustained neuroinflammation that leads to neurocognitive deficits. In this study, we explored the role of opportunistic systemic infection with Streptococcus pneumoniae in the recruitment of peripheral leukocytes into the CNS and its contribution to HIV-1-associated neurocognitive disorders in opioid-dependent individuals. METHODS: Wild-type B6CBAF1 (wt), µ-opioid receptor knockout (MORKO), FVB/N luciferase transgenic, and Toll-like receptor 2 and 4 knockout (TLR2KO and TLR4KO) mice were subcutaneously implanted with morphine/placebo pellet followed by HIV-1 Transactivator of transcription (Tat) protein injection intravenously and S. pneumoniae administration intraperitoneally. On postoperative day 5, brains perfused with phosphate-buffered saline were harvested and subjected to immunohistochemistry (for bacterial trafficking and chemokine ligand generation), flow cytometry (for phenotypic characterization of CNS trafficked immune cells), Western blot, and real-time PCR (for ligand expression). RESULTS: Our results show differential leukocyte trafficking of T lymphocytes (CD3+) and inflammatory monocytes (Ly6C+) into the CNS of mice treated with morphine, HIV-1 Tat, and/or S. pneumoniae. In addition, we demonstrate a Trojan horse mechanism for bacterial dissemination across the blood-brain barrier into the CNS by monocytes. Activation of TLRs on microglia induced a chemokine gradient that facilitated receptor-dependent trafficking of peripheral immune cells into the CNS. HIV-1 Tat induced trafficking of Ly6C+ and CD3+ cells into the CNS; infection with S. pneumoniae facilitated infiltration of only T lymphocytes into the CNS. We also observed differential chemokine secretion in the CNS, with CCL5 being the predominant chemokine following HIV-1 Tat treatment, which was potentiated further with morphine. S. pneumoniae alone led to preferential induction of CXCL12. Furthermore, we attributed a regulatory role for TLRs in the chemokine-mediated trafficking of leukocytes into the CNS. Chronic morphine and HIV-1 Tat, in the context of systemic S. pneumoniae co-infection, differentially modulated induction of TLR2/4, which consequently facilitated trafficking of TLR2 → CD3 + CCR5+ and TLR4 → Ly6C+(CCR5+/CXCR4+) immune cells into the CNS. CONCLUSION: Our murine study suggests that secondary infection in opioid-dependent individuals infected with HIV-1 augments peripheral leukocyte trafficking as a consequence of sustained chemokine gradients in the CNS.


Asunto(s)
Antígenos Ly/inmunología , Complejo CD3/inmunología , Monocitos/efectos de los fármacos , Monocitos/inmunología , Morfina/farmacología , Narcóticos/farmacología , Infecciones Neumocócicas/inmunología , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/farmacología , Animales , Proliferación Celular , Quimiocinas/metabolismo , Inmunidad Celular/efectos de los fármacos , Inflamación/patología , Masculino , Ratones , Ratones Noqueados , Microglía/efectos de los fármacos , Microglía/inmunología , Infecciones Neumocócicas/microbiología , Receptores Opioides mu/genética , Receptores Toll-Like/genética
7.
J Neuroimmune Pharmacol ; 10(1): 74-87, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25502600

RESUMEN

Opioids are known to exacerbate HIV pathogenesis, however current studies have been limited by models of HIV infection. Given that HIV causes many systemic effects via direct infection of host cells as well as indirect bystander effects, it is important to establish a systemic infection model in a small animal so that genetic tools can be utilized to elucidate the mechanisms of action. In this study, the systemic effects of EcoHIV infection, a modified HIV which can infect mouse cells, are examined in conjunction with morphine. EcoHIV infection with opioid treatment induced bacterial translocation from the lumen of the gut into systemic compartments such as liver, which is similar to observations in human patients with LPS. Bacterial translocation corresponds with alterations in gut morphology, disorganization of the tight junction protein occludin, and a concurrent increase in systemic inflammation in both IL-6 and TNFα. Long term infection also had increased expression of inflammatory cytokines in the CNS when co-treated with morphine. Overall, this study shows that EcoHIV is an appropriate model to study the effects of opioids on HIV pathogenesis, including the HIV-induced pathology at early stages of pathogenesis in the gut.


Asunto(s)
Analgésicos Opioides/farmacología , Enfermedades Gastrointestinales/patología , Infecciones por VIH/patología , Animales , Traslocación Bacteriana/efectos de los fármacos , Femenino , Enfermedades Gastrointestinales/etiología , Enfermedades Gastrointestinales/microbiología , Infecciones por VIH/complicaciones , Infecciones por VIH/microbiología , VIH-1 , Interleucina-6/metabolismo , Lipopolisacáridos/farmacología , Hígado/microbiología , Hígado/patología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ocludina/metabolismo , Fagocitosis/efectos de los fármacos , Factor de Necrosis Tumoral alfa/metabolismo
8.
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
9.
Curr HIV Res ; 10(5): 469-77, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22591371

RESUMEN

Drug abuse and HIV infection are interlinked. From the onset of the HIV/AIDS epidemic, the impact of illicit drug use on HIV disease progression has been a focus of many investigations. Both laboratory-based and epidemiological studies strongly indicate that drug abuse may exacerbate HIV disease progression and increase mortality and morbidity in these patients. Increase susceptibility to opportunistic infection has been implicated as one of the major causes for this detriment. Furthermore, opioids are known to elicit prevalence of neurodegenerative disorders in HIV-infected patients. Numerous authors have delineated various molecular as well as cellular mechanisms associated with neurological complications in these patients. This review gives an overview of these findings. Understanding the mechanisms will allow for the development of targeted therapies aimed at reducing the progression of neurocognitive decline in the drug abusing HIV infected individuals.


Asunto(s)
Complejo SIDA Demencia/metabolismo , Infecciones Oportunistas Relacionadas con el SIDA/metabolismo , Síndrome de Inmunodeficiencia Adquirida/metabolismo , Infecciones Bacterianas/metabolismo , VIH-1/efectos de los fármacos , Hepatitis Viral Humana/metabolismo , Trastornos Relacionados con Sustancias/metabolismo , Complejo SIDA Demencia/fisiopatología , Infecciones Oportunistas Relacionadas con el SIDA/fisiopatología , Síndrome de Inmunodeficiencia Adquirida/fisiopatología , Animales , Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Infecciones Bacterianas/fisiopatología , Barrera Hematoencefálica/efectos de los fármacos , Progresión de la Enfermedad , Hepatitis Viral Humana/fisiopatología , Humanos , Terapia de Inmunosupresión , Microglía/efectos de los fármacos , Microglía/metabolismo , Trastornos Relacionados con Sustancias/fisiopatología , Receptores Toll-Like/efectos de los fármacos , Receptores Toll-Like/metabolismo
10.
Am J Physiol Heart Circ Physiol ; 302(5): H1173-84, 2012 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-22159994

RESUMEN

Despite intensive research studies, theories have yet to focus on the contribution of hypoxia to patency differences observed clinically between arterial vs. venous grafts. This study investigates the differential hypoxic response of smooth muscle cells (SMC) to hypoxia-derived endothelial cell (EC) growth factors. Initiation of SMC proliferation under hypoxia (<5% O(2)) occurred only after incubation with hypoxic endothelial cell-conditioned media (H-ECM). After the investigation of several possible growth factors in the H-ECM that may be responsible for SMC proliferation, the greatest difference was observed in vascular endothelial growth factor (VEGF-A) and platelet-derived growth factor homodimer B (PDGF-BB) expression. VEGF-A increased (2-fold) significantly (P < 0.05) in arterial-derived smooth muscle cells (ASMC) under hypoxia compared with venous-derived smooth muscle cells (VSMC), which showed no significant change. VSMC showed significant (P < 0.05) increase in VEGFR-2 expression under hypoxia compared with ASMC. Incubation with VEGFR-2-neutralizing antibody/PDGFR antagonist in VSMC before addition of H-ECM resulted in decreased proliferation. ASMC proliferation under hypoxia did not decrease during incubation with VEGFR-2-neutralizing antibody but did decrease upon PDGFR antagonist incubation. Current therapies focusing on treating intimal hyperplasia have negated the fact that combinational therapy might be required to combat induction of SMC proliferation. Clinically, therapy with PDGFR antagonists plus anti-VEGFR-2 may prove to be efficacious in managing SMC proliferation in venous-derived grafts.


Asunto(s)
Proliferación Celular , Músculo Liso Vascular/fisiología , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/biosíntesis , Receptor 2 de Factores de Crecimiento Endotelial Vascular/biosíntesis , Arterias/metabolismo , Becaplermina , Hipoxia de la Célula , Línea Celular , Humanos , Proteínas Proto-Oncogénicas c-sis/biosíntesis , Factor A de Crecimiento Endotelial Vascular/biosíntesis , Receptor 2 de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Venas/metabolismo
11.
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
12.
Toxicology ; 285(3): 126-32, 2011 Jul 29.
Artículo en Inglés | MEDLINE | ID: mdl-21557984

RESUMEN

Fenvalerate, a type II synthetic pyrethroid, has emerged as one of the most potent indoor toxicants. Despite its widespread usage, the adverse effect of this insecticide on immune defense mechanism has not been comprehensively investigated. In this in vitro study we report the effect of fenvalerate on two pivotal components of the immune network, namely the complement system and macrophages. Fenvalerate treated human sera showed serum complement activation as evident by significant (p<0.05) increase in C3b, C3d and C3a levels and a significant (p<0.05) decline in CH50 levels. Further detailed study demonstrates that the activation of complement system is through alternative pathway. This is possibly responsible for various allergic manifestations often reported in subjects exposed to fenvalerate. In addition, fenvalerate induce cellular apoptosis and cytotoxicity, as demonstrated by cytoplasmic vacuolization, heterochromatin condensation, hypodiploid nuclei and DNA fragmentation in macrophages. Considerable deleterious effects on macrophages in conjunction with uncontrolled serum complement activation are probably one of the major mechanisms contributing for the immunosuppressive effects of fenvalerate.


Asunto(s)
Complemento C3/análisis , Insecticidas/farmacología , Macrófagos/efectos de los fármacos , Nitrilos/farmacología , Piretrinas/farmacología , Adulto , Apoptosis/efectos de los fármacos , Western Blotting , Activación de Complemento/efectos de los fármacos , Complemento C3/inmunología , Complemento C3a/análisis , Complemento C3a/inmunología , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Humanos , Inmunidad Celular/efectos de los fármacos , Inmunoelectroforesis Bidimensional , Técnicas In Vitro , Insecticidas/efectos adversos , Macrófagos/inmunología , Microscopía Electrónica de Transmisión , Nitrilos/efectos adversos , Piretrinas/efectos adversos , Adulto Joven
13.
Mediators Inflamm ; 2009: 417658, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19503841

RESUMEN

Chlamydia trachomatis is a leading cause of sexually transmitted infection worldwide and responsible for myriad of immunopathological changes associated with reproductive health. Delayed secretion of proinflammatory chemokine interleukin (IL)-8 is a hallmark of chlamydial infection and is dependent on chlamydial growth. We examined the effect of iron chelators on IL-8 production in HeLa 229 (cervix epitheloid cell, CCL2) cells infected with C. trachomatis. IL-8 production was induced by Iron chelator DFO and Mimosine, however, synergy with chlamydial infection was obtained with DFO only. Temporal expression of proinflammatory secreted cytokines IL-1beta, TNF-alpha, and IL-8 did not show synchrony in Chlamydia trachomatis infected cells. Secretion of IL-8 from Hela cells infected with C. trachomatis was not dependent on IL-1 beta and TNF- alpha induction. These results indicate towards involvement of iron in chlamydia induced IL-8 production.


Asunto(s)
Infecciones por Chlamydia/metabolismo , Chlamydia trachomatis/crecimiento & desarrollo , Interleucina-8/metabolismo , Hierro/metabolismo , Ensayo de Inmunoadsorción Enzimática , Células HeLa , Humanos , Interleucina-1beta/metabolismo , Quelantes del Hierro/farmacología , Factor de Necrosis Tumoral alfa/metabolismo
14.
Toxicology ; 252(1-3): 78-85, 2008 Oct 30.
Artículo en Inglés | MEDLINE | ID: mdl-18755234

RESUMEN

DDT (bis[4-chlorophenyl]-1,1,1-trichloroethane) is responsible for many immuno-dysregulatory functions in exposed animals, but data particularly on complement system and macrophages are limited. In this study we have shown that DDT activates the complement system through the alternative pathway in the absence of any pathogen. A significant (p<0.05) increase in C3b, C3d and C3a generation, and decline in complement hemolytic activity was observed in insecticide exposed sera. The uncontrolled complement consumption reduces the lytic activity of the complement, which enhances the susceptibility to pyogenic infection if the exposure to DDT remains unabated. Further, DDT induced the significant (p<0.05) production of tumor necrosis factor-alpha (TNF-alpha) and nitric oxide (NO) in macrophages and thus contributes inflammatory reactions, cytokine imbalance and immune-dysregulation. These molecular changes in macrophages lead to structural aberrations like heterochromatin condensation, loss of pseudopodia, cytoplasmic vacuolization, DNA fragmentation and hypodiploid nuclei as seen in our study, suggesting apoptosis. However, in presence lipopolysaccharide, DDT induced significant (p<0.05) suppression of TNF-alpha and NO generation, suggestive of impairment of macrophage microbiocidal effects. This study concludes that the functional and structural derangements of macrophages in association with uncontrolled and excessive complement consumption by DDT are perhaps one of the major mechanisms contributing to the immunosuppressive effects of insecticide.


Asunto(s)
Proteínas del Sistema Complemento/fisiología , DDT/toxicidad , Insecticidas/toxicidad , Macrófagos Alveolares/inmunología , Animales , Western Blotting , Complemento C3/fisiología , Complemento C3d/fisiología , Ensayo de Actividad Hemolítica de Complemento , Vía Clásica del Complemento/efectos de los fármacos , ADN/biosíntesis , ADN/genética , Ácido Edético/farmacología , Ácido Egtácico/farmacología , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Inmunoelectroforesis Bidimensional , Macrófagos Alveolares/efectos de los fármacos , Macrófagos Alveolares/ultraestructura , Masculino , Microscopía Electrónica de Transmisión , Óxido Nítrico/metabolismo , Ratas , Ratas Wistar , Factor de Necrosis Tumoral alfa/metabolismo
15.
Mol Immunol ; 41(4): 449-56, 2004 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15163541

RESUMEN

Systemic lupus erythematosus (SLE) is a chronic systemic autoimmune disease characterized by the production of a broad spectrum of autoantibodies against nuclear, cytoplasmic and cell surface antigens and immune complex overload. Complement receptor 1 (CR1, CD 35), a transmembrane glycoprotein found on the surface of erythrocytes, leukocytes and glomerular podocytes plays a key role in the clearance of immune complexes and regulation of complement cascade. A drastic decline in the level of cell surface CR1 appears to be an important event in pathology of SLE. However, the etiology of lower than normal expression of cell surface CR1 in this disease is poorly understood. We studied the level of leukocyte CR1 transcription in 30 patients with active SLE and 30 controls by reverse transcriptase-polymerase chain reaction (RT-PCR) and related the same with the level of CR1 protein expression monitored by Western blotting. For RT-PCR, ratio of CR1/beta-actin was considered for semiquantitation of the level of CR1 transcription. Despite individual variation at the level of transcription, 70% (21 out of 30) of the patients expressed CR1 transcript at the lowest range of 0-15% as compared to the controls wherein only 30% (9 out of 30 individuals) demonstrated CR1 transcript in this range. Majority of the controls (70%) expressed CR1 transcript at the level above 15%. Mean level of CR1 transcript in patients (mean +/- S.D. = 21.09 +/- 14.3) was significantly lower than the controls (mean +/- S.D. = 48.91 +/- 26.34) (P < 0.001). The level of CR1 transcription correlated inversely with circulating immune complexes (CIC) (r = 0.52, P < 0.01). This may suggest that although erythrocyte CR1 is the chief vehicle for CIC clearance, drastic decline in leukocyte CR1 expression may impair the phagocyte mediated immune complex clearance and contribute to increased complement consumption in SLE. Total leukocyte CR1 protein expression was also significantly reduced in patients (P < 0.001) as compared to controls. This decline at the protein level gave a very significant positive correlation with CR1 transcript (r = 0.67, P < 0.01). A marginal increase in soluble CR1 (sCR1) was observed in the plasma (ELISA) of SLE patients compared to the controls but was insignificant. This paper for the first time brings evidence to suggest that reduced synthesis of CR1 contributes substantially to the low cell surface CR1 expression in SLE. Our findings also suggest increased proteolytic cleavage of leukocyte cell surface CR1 in these patients. However, evidence for the latter is indirect.


Asunto(s)
Leucocitos/metabolismo , Lupus Eritematoso Sistémico/inmunología , Receptores de Complemento/deficiencia , Adulto , Complejo Antígeno-Anticuerpo/sangre , Western Blotting , Proteínas del Sistema Complemento/metabolismo , Femenino , Regulación de la Expresión Génica , Humanos , Lupus Eritematoso Sistémico/genética , Lupus Eritematoso Sistémico/metabolismo , Receptores de Complemento/biosíntesis , Receptores de Complemento/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transcripción Genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...