Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
Arch Environ Contam Toxicol ; 79(4): 454-460, 2020 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-33140186

RESUMO

Anticoagulant rodenticides (ARs) are commonly used to control rodent pests. However, worldwide, their use is associated with secondary and tertiary poisoning of nontarget species, especially predatory and scavenging birds. No medical device can rapidly test for AR exposure of avian wildlife. Prothrombin time (PT) is a useful biomarker for AR exposure, and multiple commercially available point-of-care (POC) devices measure PT of humans, and domestic and companion mammals. We evaluated the potential of one commercially available POC device, the Coag-Sense® PT/INR Monitoring System, to rapidly detect AR exposure of living birds of prey. The Coag-Sense device delivered repeatable PT measurements on avian blood samples collected from four species of raptors trapped during migration (Intraclass Correlation Coefficient > 0.9; overall intra-sample variation CV: 5.7%). However, PT measurements reported by the Coag-Sense system from 81 ferruginous hawk (Buteo regalis) nestlings were not correlated to those measured by a one-stage laboratory avian PT assay (r = - 0.017, p = 0.88). Although precise, the lack of agreement in PT estimates from the Coag-Sense device and the laboratory assay indicates that this device is not suitable for detecting potential AR exposure of birds of prey. The lack of suitability may be related to the use of a mammalian reagent in the clotting reaction, suggesting that the device may perform better in testing mammalian wildlife.


Assuntos
Anticoagulantes/metabolismo , Monitoramento Ambiental , Aves Predatórias/metabolismo , Rodenticidas/metabolismo , Animais , Anticoagulantes/intoxicação , Aves , Humanos , Fígado , Comportamento Predatório , Rodenticidas/intoxicação
2.
Int J Mol Sci ; 21(6)2020 Mar 20.
Artigo em Inglês | MEDLINE | ID: mdl-32244989

RESUMO

The Center of Biomedical Research Excellence in Matrix Biology strives to improve our understanding of extracellular matrix at molecular, cellular, tissue, and organismal levels to generate new knowledge about pathophysiology, normal development, and regenerative medicine. The primary goals of the Center are to i) support junior investigators, ii) enhance the productivity of established scientists, iii) facilitate collaboration between both junior and established researchers, and iv) build biomedical research infrastructure that will support research relevant to cell-matrix interactions in disease progression, tissue repair and regeneration, and v) provide access to instrumentation and technical support. A Pilot Project program provides funding to investigators who propose applying their expertise to matrix biology questions. Support from the National Institute of General Medical Sciences at the National Institutes of Health that established the Center of Biomedical Research Excellence in Matrix Biology has significantly enhanced the infrastructure and the capabilities of researchers at Boise State University, leading to new approaches that address disease diagnosis, prevention, and treatment. New multidisciplinary collaborations have been formed with investigators who may not have previously considered how their biomedical research programs addressed fundamental and applied questions involving the extracellular matrix. Collaborations with the broader matrix biology community are encouraged.


Assuntos
Pesquisa Biomédica , Comportamento Cooperativo , Matriz Extracelular/metabolismo , Pesquisadores , Comitês Consultivos , Escolha da Profissão , Humanos , Estudantes
3.
J Appl Toxicol ; 39(6): 888-898, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30690755

RESUMO

Electronic cigarettes (e-cigarettes) are nicotine delivery devices advertised as a healthier alternative to conventional tobacco products, but their rapid rise in popularity outpaces research on potential health consequences. As conventional tobacco use is a risk factor for osteoporosis, this study examines whether exposure to electronic liquid (e-liquid) used in e-cigarettes affects bone-forming osteoblasts. Human MG-63 and Saos-2 osteoblast-like cells were treated for 48 hours with 0.004%-4.0% dilutions of commercially available e-liquids of various flavors with or without nicotine. Changes in cell viability and key osteoblast markers, runt-related transcription factor 2 and Col1a1, were assessed. With all e-liquids tested, cell viability decreased in a dose-dependent manner, which was least pronounced in flavorless e-liquids, most pronounced in cinnamon-flavored e-liquids and occurred independently of nicotine. Col1a1, but not runt-related transcription factor 2, mRNA expression was upregulated in response to coffee-flavored and fruit-flavored e-liquids. Cells treated with a non-cytotoxic concentration of fruit-flavored Mango Blast e-liquid with or without nicotine showed significantly increased collagen type I protein expression compared to culture medium only. We conclude that the degree of osteotoxicity is flavor-dependent and occurs independently of nicotine and that flavored e-liquids reveal collagen type I as a potential target in osteoblasts. This study elucidates potential consequences of e-cigarette use in bone.


Assuntos
Colágeno Tipo I/genética , Sistemas Eletrônicos de Liberação de Nicotina , Aromatizantes/farmacologia , Nicotina/efeitos adversos , Osteoblastos/efeitos dos fármacos , Biomarcadores , Linhagem Celular Tumoral , Cadeia alfa 1 do Colágeno Tipo I , Subunidade alfa 1 de Fator de Ligação ao Core/genética , Humanos , RNA Mensageiro/análise
4.
Toxicol Appl Pharmacol ; 311: 42-51, 2016 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-27693115

RESUMO

2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD) is a persistent environmental contaminant and high-affinity ligand for the aryl hydrocarbon receptor (AhR). Increasing evidence indicates that AhR signaling contributes to wound healing, which involves the coordinated deposition and remodeling of the extracellular matrix. In the liver, wound healing is attributed to the activation of hepatic stellate cells (HSCs), which mediate fibrogenesis through the production of soluble mediators and collagen type I. We recently reported that TCDD treatment increases the activation of human HSCs in vitro. The goal of this study was to determine how TCDD impacts HSC activation in vivo using a mouse model of experimental liver fibrosis. To elicit fibrosis, C57BL6/male mice were treated twice weekly for 8weeks with 0.5ml/kg carbon tetrachloride (CCl4). TCDD (20µg/kg) or peanut oil (vehicle) was administered once a week during the last 2weeks. Results indicate that TCDD increased liver-body-weight ratios, serum alanine aminotransferase activity, and hepatic necroinflammation in CCl4-treated mice. Likewise, TCDD treatment increased mRNA expression of HSC activation and fibrogenesis genes, namely α-smooth muscle actin, desmin, delta-like homolog-1, TGF-ß1, and collagen type I. However, TCDD treatment did not exacerbate fibrosis, nor did it increase the collagen content of the liver. Instead, TCDD increased hepatic collagenase activity and increased expression of matrix metalloproteinase (MMP)-13 and the matrix regulatory proteins, TIMP-1 and PAI-1. These results support the conclusion that TCDD increases CCl4-induced liver damage and exacerbates HSC activation, yet collagen deposition and the development of fibrosis may be limited by TCDD-mediated changes in extracellular matrix remodeling.


Assuntos
Células Estreladas do Fígado/efeitos dos fármacos , Inflamação/induzido quimicamente , Cirrose Hepática/induzido quimicamente , Dibenzodioxinas Policloradas/toxicidade , Animais , Tetracloreto de Carbono/toxicidade , Colágeno Tipo I/metabolismo , Colagenases/metabolismo , Células Estreladas do Fígado/metabolismo , Fígado/efeitos dos fármacos , Fígado/enzimologia , Fígado/metabolismo , Cirrose Hepática/metabolismo , Masculino , Metaloproteinase 13 da Matriz/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Índice de Gravidade de Doença
5.
Mol Pharmacol ; 85(4): 533-41, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24431146

RESUMO

Previous studies in hepatocyte-derived cell lines and the whole liver established that the aryl hydrocarbon receptor (AhR) can disrupt G1-phase cell cycle progression following exposure to persistent AhR agonists, such as TCDD (dioxin, 2,3,7,8-tetrachlorodibenzo-p-dioxin). Growth arrest was attributed to inhibition of G1-phase cyclin-dependent kinase 2 (CDK2) activity. The present study examined the effect of TCDD exposure on liver regeneration following 70% partial hepatectomy in mice lacking the Cip/Kip inhibitors p21(Cip1) or p27(Kip1) responsible for regulating CDK2 activity. Assessment of the regenerative process in wild-type, p21(Cip1) knockout, and p27(Kip1) knockout mice confirmed that TCDD-induced inhibition of liver regeneration is entirely dependent on p21(Cip1) expression. Compared with wild-type mice, the absence of p21(Cip1) expression completely abrogated the TCDD inhibition, and accelerated hepatocyte progression through G1 phase during the regenerative process. Analysis of the transcriptional response determined that increased p21(Cip1) expression during liver regeneration involved an AhR-dependent mechanism. Chromatin immunoprecipitation studies revealed that p21(Cip1) induction required AhR binding to the newly characterized nonconsensus xenobiotic response element, in conjunction with the tumor suppressor protein Kruppel-like factor 6 functioning as an AhR binding partner. The evidence also suggests that AhR functionality following partial hepatectomy is dependent on a p21(Cip1)-regulated signaling process, intimately linking AhR biology to the G1-phase cell cycle program.


Assuntos
Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Regeneração Hepática , Receptores de Hidrocarboneto Arílico/metabolismo , Elementos de Resposta , Animais , Inibidor de Quinase Dependente de Ciclina p21/genética , Inibidor de Quinase Dependente de Ciclina p27/genética , Inibidor de Quinase Dependente de Ciclina p27/metabolismo , Hepatectomia , Fator 6 Semelhante a Kruppel , Fatores de Transcrição Kruppel-Like/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Dibenzodioxinas Policloradas/toxicidade , Proteínas Proto-Oncogênicas/metabolismo , Receptores de Hidrocarboneto Arílico/agonistas
6.
Biochim Biophys Acta Gen Subj ; 1864(1): 129455, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31669585

RESUMO

BACKGROUND: Borrelia burgdorferi causes Lyme disease, the most common tick-borne illness in the United States. The Center for Disease Control and Prevention estimates that the occurrence of Lyme disease in the U.S. has now reached approximately 300,000 cases annually. Early stage Borrelia burgdorferi infections are generally treatable with oral antibiotics, but late stage disease is more difficult to treat and more likely to lead to post-treatment Lyme disease syndrome. METHODS: Here we examine three unique 5'-methylthioadenosine/S-adenosylhomocysteine (MTA/SAH) nucleosidases (MTNs or MTANs, EC 3.2.2.9) responsible for salvage of adenine and methionine in B. burgdorferi and explore their potential as antibiotic targets to treat Lyme disease. Recombinant Borrelia MTNs were expressed and purified from E. coli. The enzymes were extensively characterized for activity, specificity, and inhibition using a UV spectrophotometric assay. In vitro antibiotic activities of MTN inhibitors were assessed using a bioluminescent BacTiter-Glo™ assay. RESULTS: The three Borrelia MTNs showed unique activities against the native substrates MTA, SAH, and 5'-deoxyadenosine. Analysis of substrate analogs revealed that specific activity rapidly dropped as the length of the 5'-alkylthio substitution increased. Non-hydrolysable nucleoside transition state analogs demonstrated sub-nanomolar enzyme inhibition constants. Lastly, two late stage transition state analogs exerted in vitro IC50 values of 0.3-0.4 µg/mL against cultured B. burgdorferi cells. CONCLUSION: B. burgdorferi is unusual in that it expresses three distinct MTNs (cytoplasmic, membrane bound, and secreted) that are effectively inactivated by nucleoside analogs. GENERAL SIGNIFICANCE: The Borrelia MTNs appear to be promising targets for developing new antibiotics to treat Lyme disease.


Assuntos
Antibacterianos/uso terapêutico , Borrelia burgdorferi/enzimologia , Doença de Lyme/tratamento farmacológico , N-Glicosil Hidrolases/genética , Borrelia burgdorferi/efeitos dos fármacos , Borrelia burgdorferi/patogenicidade , Desoxiadenosinas/metabolismo , Escherichia coli/genética , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Humanos , Doença de Lyme/enzimologia , Doença de Lyme/microbiologia , N-Glicosil Hidrolases/antagonistas & inibidores , S-Adenosil-Homocisteína/metabolismo , Tionucleosídeos/metabolismo
7.
J Inflamm (Lond) ; 13(1): 28, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27555804

RESUMO

BACKGROUND: Liver regeneration following 70 % partial hepatectomy (PH) requires the coordinated expression of soluble mediators produced by macrophages. Monocyte chemoattractant protein-1 (MCP-1) is a potent stimulus of monocyte recruitment and macrophage activation. The goal of this study was to determine how MCP-1 contributes to liver regeneration. METHODS: PH was performed on anesthetized C57Bl/6 (wild type) and MCP-1 knockout mice, and macrophage-produced cytokines and hepatocyte proliferation were measured. RESULTS: In wild type mice, hepatic MCP-1 protein levels increased 4-6 h after PH, and elevated plasma MCP-1 levels were detected 12 h after PH. Hepatocyte proliferation was comparable in MCP-1 knockout and wild type mice, as was the expression of macrophage-derived cytokines, TNFα and IL-6, and levels of phosphorylated STAT3. The number of CCR2(+) cells in the liver was similar in MCP-1 knockout and wild type mice, which suggests that other chemokines may recruit CCR2(+) cells in the absence of MCP-1. Studies with CCR2 knockout mice revealed that hepatocyte proliferation was suppressed ~40 % compared to wild type mice 36 h after PH, but proliferation and liver-body-weight ratios were similar at 48 h. CONCLUSION: These findings suggest that MCP-1 is not required for PH-induced liver regeneration, yet the role of CCR2 warrants further study.

8.
Toxicology ; 344-346: 26-33, 2016 Feb 17.
Artigo em Inglês | MEDLINE | ID: mdl-26860701

RESUMO

2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD) is a halogenated aromatic hydrocarbon that elicits toxicity through the aryl hydrocarbon receptor (AhR). In the liver, gross markers of TCDD toxicity are attributed to AhR activation in parenchymal hepatocytes. However, less is known regarding the consequences of TCDD treatment on non-parenchymal cells in the liver. Hepatic stellate cells (HSCs) are non-parenchymal cells that store vitamin A when quiescent. Upon liver injury, activated HSCs lose this storage ability and instead function in the development and maintenance of inflammation and fibrosis through the production of pro-inflammatory mediators and collagen type I. Reports that TCDD exposure disrupts hepatic retinoid homeostasis and dysregulates extracellular matrix remodeling in the liver led us to speculate that TCDD treatment may disrupt HSC activity. The human HSC line LX-2 was used to test the hypothesis that TCDD treatment directly activates HSCs. Results indicate that exposure to 10nM TCDD almost completely inhibited lipid droplet storage in LX-2 cells cultured with retinol and palmitic acid. TCDD treatment also increased LX-2 cell proliferation, expression of α-smooth muscle actin, and production of monocyte chemoattractant protein-1 (MCP-1), all of which are characteristics of activated HSCs. However, TCDD treatment had no effect on Col1a1 mRNA levels in LX-2 cells stimulated with the potent profibrogenic mediator, transforming growth factor-ß. The TCDD-mediated increase in LX-2 cell proliferation, but not MCP-1 production, was abolished when phosphoinositide 3-kinase was inhibited. These results indicate that HSCs are susceptible to direct modulation by TCDD and that TCDD likely increases HSC activation through a multi-faceted mechanism.


Assuntos
Proliferação de Células/efeitos dos fármacos , Células Estreladas do Fígado/efeitos dos fármacos , Células Estreladas do Fígado/metabolismo , Dibenzodioxinas Policloradas/toxicidade , Linhagem Celular , Proliferação de Células/fisiologia , Humanos , Receptores de Hidrocarboneto Arílico/agonistas , Receptores de Hidrocarboneto Arílico/metabolismo
9.
Biomed Res Int ; 2016: 5309328, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27672655

RESUMO

The aryl hydrocarbon receptor (AhR) is a soluble, ligand-activated transcription factor that mediates the toxicity of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD). Increasing evidence implicates the AhR in regulating extracellular matrix (ECM) homeostasis. We recently reported that TCDD increased necroinflammation and myofibroblast activation during liver injury elicited by carbon tetrachloride (CCl4). However, TCDD did not increase collagen deposition or exacerbate fibrosis in CCl4-treated mice, which raises the possibility that TCDD may enhance ECM turnover. The goal of this study was to determine how TCDD impacts ECM remodeling gene expression in the liver. Male C57BL/6 mice were treated for 8 weeks with 0.5 mL/kg CCl4, and TCDD (20 µg/kg) was administered during the last two weeks. Results indicate that TCDD increased mRNA levels of procollagen types I, III, IV, and VI and the collagen processing molecules HSP47 and lysyl oxidase. TCDD also increased gelatinase activity and mRNA levels of matrix metalloproteinase- (MMP-) 3, MMP-8, MMP-9, and MMP-13. Furthermore, TCDD modulated expression of genes in the plasminogen activator/plasmin system, which regulates MMP activation, and it also increased TIMP1 gene expression. These findings support the notion that AhR activation by TCDD dysregulates ECM remodeling gene expression and may facilitate ECM metabolism despite increased liver injury.

10.
Toxicol Sci ; 74(1): 74-84, 2003 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-12730613

RESUMO

While considerable evidence indicates that exposure to the pollutant 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) impairs T cell function, the precise mechanism underlying this effect is not well understood. Furthermore, relatively little is known about the effects of TCDD on the fate of activated, antigen-specific T cells in vivo. In the present study, we took advantage of major histocompatibility complex (MHC) class I-restricted tetramers and clonotypic anti-T cell receptor (TCR) antibodies to follow the fate of influenza virus-specific CD8+ T cells in mice treated with TCDD. Exposure to TCDD suppressed the clonal expansion of influenza virus-specific CD8+ T cells, resulting in a three- to five-fold reduction in the number of cytotoxic T lymphocytes (CTL) in the lymph node, as compared to vehicle-treated mice. Studies to address possible mechanisms for the diminished CTL response failed to show evidence for increased apoptosis in virus-specific CD8+ T cells from TCDD-exposed mice. However, treatment with TCDD reduced the number of proliferating virus-specific CD8+ T cells by as much as 70% on day 7 post infection. Moreover, ex vivo restimulation of lymph node cells with influenza virus nucleoprotein (NP366-374) peptide and exogenous interleukin-2 (IL-2) only partially restored the proliferation of influenza virus-specific CD8+ T cells from TCDD-exposed mice and failed to stimulate interferon-gamma (IFNgamma) production by these cells. The observation that neither proliferation nor IFNgamma production by CD8+ T cells could be completely restored, even when cells were provided with optimal stimulation, suggests that exposure to TCDD drives antigen-specific CD8+ T cells into a state of unresponsiveness similar to anergy.


Assuntos
Antígenos Virais/farmacologia , Linfócitos T CD8-Positivos/imunologia , Poluentes Ambientais/toxicidade , Vírus da Influenza A/imunologia , Dibenzodioxinas Policloradas/toxicidade , Animais , Antígenos Virais/metabolismo , Apoptose/efeitos dos fármacos , Antígenos CD8/farmacologia , Linfócitos T CD8-Positivos/efeitos dos fármacos , Linfócitos T CD8-Positivos/metabolismo , Divisão Celular/efeitos dos fármacos , Feminino , Citometria de Fluxo , Genes MHC Classe I/genética , Interferon gama/biossíntese , Linfonodos/patologia , Linfócitos/efeitos dos fármacos , Linfócitos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Fragmentos de Peptídeos/imunologia , Estimulação Química , Proteínas do Core Viral/imunologia
11.
J Immunotoxicol ; 9(4): 359-67, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22530936

RESUMO

Increasing evidence demonstrates a physiological role for the aryl hydrocarbon receptor (AhR) in regulating hepatocyte cell cycle progression. Previous studies have used a murine model of liver regeneration to show that exposure to the potent exogenous AhR ligand, 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD), suppresses hepatocyte proliferation in vivo. Based on recent reports that natural killer (NK) cells negatively regulate liver regeneration, coupled with the well-established immunomodulatory effects of TCDD, it was hypothesized that alterations in lymphocyte activation contribute to the suppression of liver regeneration in TCDD-treated mice. To test this, mice were treated with TCDD (20 µg/kg) 1 day prior to 70% partial hepatectomy (PH), in which two-thirds of the liver was surgically resected. Lymphocytes were collected from the remnant liver and analyzed by flow cytometry. Whereas exposure to TCDD did not alter the number of NK cells or CD3(+) T-cells recovered from the regenerating liver, it reduced the percentage and number of intra-hepatic NKT cells 42 h after PH. With regard to lymphocyte activation, TCDD treatment transiently increased CD69 expression on NK and NKT cells 12 h after PH, but had no effect on intracellular levels of IFNγ in NK, NKT, or CD3(+) T-cells. To determine the relevance of NK cells to the suppression of liver regeneration by TCDD, mice were treated with anti-Asialo GM-1 (ASGM-1) antibody to deplete NK cells prior to TCDD treatment and PH, and hepatocyte proliferation was measured using bromodeoxyuridine incorporation. Exposure to TCDD was found to inhibit hepatocyte proliferation in the regenerating liver of NK cell-depleted mice and control mice to the same extent. Hence, it is unlikely that enhanced numbers or increased activation of NK cells contribute to the suppression of liver regeneration in TCDD-treated mice.


Assuntos
Células Matadoras Naturais/efeitos dos fármacos , Regeneração Hepática/efeitos dos fármacos , Fígado/efeitos dos fármacos , Subpopulações de Linfócitos/efeitos dos fármacos , Dibenzodioxinas Policloradas/administração & dosagem , Animais , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Feminino , Hepatectomia , Células Matadoras Naturais/imunologia , Fígado/imunologia , Regeneração Hepática/imunologia , Ativação Linfocitária/efeitos dos fármacos , Depleção Linfocítica , Subpopulações de Linfócitos/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Dibenzodioxinas Policloradas/farmacologia , Receptores de Hidrocarboneto Arílico/agonistas
12.
Cytokine Growth Factor Rev ; 22(1): 35-43, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21334249

RESUMO

Interferon (IFN)-γ is a cytokine known for its immunomodulatory and anti-proliferative action. In the liver, IFN-γ can induce hepatocyte apoptosis or inhibit hepatocyte cell cycle progression. This article reviews recent mechanistic reports that describe how IFN-γ may direct the fate of hepatocytes either towards apoptosis or a cell cycle arrest. This review also describes a probable role for IFN-γ in modulating hepatocyte fate during liver regeneration, transplantation, hepatitis, fibrosis and hepatocellular carcinoma, and highlights promising areas of research that may lead to the development of IFN-γ as a therapy to enhance recovery from liver disease.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Hepatócitos/efeitos dos fármacos , Hepatócitos/fisiologia , Interferon gama/farmacologia , Animais , Apoptose/efeitos dos fármacos , Apoptose/genética , Apoptose/fisiologia , Diferenciação Celular/genética , Proliferação de Células/efeitos dos fármacos , Hepatócitos/citologia , Hepatócitos/metabolismo , Humanos , Interferon gama/genética , Interferon gama/fisiologia , Interferon gama/uso terapêutico , Fígado/efeitos dos fármacos , Fígado/metabolismo , Fígado/patologia , Fígado/fisiologia , Hepatopatias/genética , Hepatopatias/patologia , Hepatopatias/terapia , Regeneração Hepática/efeitos dos fármacos , Regeneração Hepática/genética , Regeneração Hepática/fisiologia , Transplante de Fígado/métodos , Transplante de Fígado/fisiologia , Modelos Biológicos
13.
Toxicology ; 276(2): 103-9, 2010 Oct 09.
Artigo em Inglês | MEDLINE | ID: mdl-20637255

RESUMO

Mechanisms of hepatocyte proliferation triggered by tissue loss are distinguishable from those that promote proliferation in the intact liver in response to mitogens. Previous studies demonstrate that exogenous activation of the aryl hydrocarbon receptor (AhR), a soluble ligand-activated transcription factor in the basic helix-loop-helix family of proteins, suppresses compensatory liver regeneration elicited by surgical partial hepatectomy. The goal of the present study was to determine how AhR activation modulates hepatocyte cell cycle progression in the intact liver following treatment with the hepatomitogen, 1,4-bis[2-(3,5-dichloropyridyloxy)] benzene (TCPOBOP). Mice were pretreated with the exogenous AhR agonist 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) 24h prior to treatment with TCPOBOP (3 mg/kg).). In contrast to the suppressive effects of AhR activation observed during compensatory regeneration, TCDD pretreatment resulted in a 30-50% increase in hepatocyte proliferation in the intact liver of TCPOBOP-treated mice. Although pretreatment with TCDD suppressed CDK2 kinase activity and increased the association of CDK2 with negative regulatory proteins p21Cip1 and p27Kip1, a corresponding increase in CDK4/cyclin D1 association and CDK4 activity which culminated in enhanced phosphorylation of retinoblastoma protein, consistent with the increased proliferative response. These findings are in stark contrast to previous observations that the activated AhR can suppress hepatocyte proliferation in vivo and reveal a new complexity to AhR-mediated cell cycle control.


Assuntos
Hiperplasia/induzido quimicamente , Fígado/efeitos dos fármacos , Dibenzodioxinas Policloradas/farmacologia , Piridinas/toxicidade , Receptores de Hidrocarboneto Arílico/efeitos dos fármacos , Animais , Ciclo Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Quinase 2 Dependente de Ciclina/efeitos dos fármacos , Quinase 2 Dependente de Ciclina/metabolismo , Quinase 4 Dependente de Ciclina/efeitos dos fármacos , Quinase 4 Dependente de Ciclina/metabolismo , Feminino , Hepatócitos/efeitos dos fármacos , Hepatócitos/metabolismo , Fígado/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fosforilação/efeitos dos fármacos , Receptores de Hidrocarboneto Arílico/genética , Receptores de Hidrocarboneto Arílico/metabolismo , Proteína do Retinoblastoma/efeitos dos fármacos , Proteína do Retinoblastoma/metabolismo
14.
Biochem Pharmacol ; 77(6): 947-56, 2009 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-19027718

RESUMO

The aryl hydrocarbon receptor (AhR) was implicated as a mediator of xenobiotic toxicity over three decades ago. Although a complete picture continues to elude us, investigations by many laboratories during the ensuing period have revealed much about AhR biology in normal physiological processes, as well as the toxicities induced by the dioxins and related polychlorinated aromatic hydrocarbons. The findings are captured in numerous excellent reviews. This commentary attempts to inject a new perspective on some new as well as frequently overlooked observations in the context of established receptor properties. Specifically, we examine the impact of transient versus sustained receptor activation on AhR biology, and explore the potential role for cytochrome P450 expression in regulating AhR activity amongst various tissues. The growing recognition that AhR action functions through multiple mechanisms serves to further highlight the importance of limiting prolonged receptor activation.


Assuntos
Receptores de Hidrocarboneto Arílico/fisiologia , Animais , Hidrocarboneto de Aril Hidroxilases/metabolismo , Hidrocarboneto de Aril Hidroxilases/fisiologia , Humanos , Especificidade de Órgãos/efeitos dos fármacos , Especificidade de Órgãos/fisiologia , Receptores de Hidrocarboneto Arílico/agonistas , Receptores de Hidrocarboneto Arílico/química , Receptores de Hidrocarboneto Arílico/metabolismo , Fatores de Tempo , Xenobióticos/metabolismo , Xenobióticos/toxicidade
15.
Mol Pharmacol ; 70(1): 163-70, 2006 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16636136

RESUMO

In hepatocyte-derived cell lines, either loss of aryl hydrocarbon receptor (AhR) function or treatment with a persistent AhR agonist such as 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) can disrupt G1 phase cell cycle progression. The present study used liver regeneration to explore mechanistically how AhR activity modulates hepatocyte proliferation in vivo. Treatment of mice with 20 mug/kg TCDD 1 day before 70% partial hepatectomy (PH) resulted in a 50 to 75% suppression in liver regeneration. Impaired proliferation was not associated with changes in levels of interleukin-6 or tumor necrosis factor-alpha, which prime quiescent hepatocytes to enter G1 phase. In fact, administration of TCDD 12 h after PH, a period well beyond the priming phase, still induced the G1 arrest. Decreased proliferation in TCDD-treated mice correlated with reduced cyclin-dependent kinase-2 (CDK2) activity, a pivotal regulator of G1/S phase transition. In contrast to observations made in cell culture, suppressed CDK2 activity was not strictly associated with increased binding of the CDK2 inhibitors p21Cip1 or p27Kip1. However, TCDD decreased levels of cyclin E binding to CDK2, despite normal cyclin E expression. The evidence also suggests that TCDD-induced hepatic growth arrest depends upon sustained AhR activity because transient AhR activation in response to endogenous queues failed to suppress the regenerative response. These findings establish a functional role for the AhR in regulating normal cell cycle control during liver regeneration.


Assuntos
Regeneração Hepática/fisiologia , Fígado/fisiopatologia , Receptores de Hidrocarboneto Arílico/fisiologia , Animais , Western Blotting , Proteínas de Ciclo Celular/metabolismo , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Quinase 2 Dependente de Ciclina/metabolismo , Citocromo P-450 CYP1A1/genética , Citocromo P-450 CYP1A1/metabolismo , Poluentes Ambientais/toxicidade , Feminino , Fase G1/efeitos dos fármacos , Expressão Gênica/efeitos dos fármacos , Hepatectomia , Imunoprecipitação , Interleucina-6/biossíntese , Fígado/efeitos dos fármacos , Fígado/metabolismo , Regeneração Hepática/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Dibenzodioxinas Policloradas/toxicidade , Receptores de Hidrocarboneto Arílico/agonistas , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Tempo , Fator de Necrose Tumoral alfa/biossíntese
16.
Mol Pharmacol ; 67(3): 612-22, 2005 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15550680

RESUMO

Liver homeostasis is achieved by the removal of diseased and damaged hepatocytes and their coordinated replacement to maintain a constant liver cell mass. Cirrhosis, viral hepatitis, and toxic drug effects can all trigger apoptosis in the liver as a means of removing the unwanted cells, and the Fas "death receptor" pathway comprises a major physiological mechanism by which this occurs. The susceptibility to Fas-mediated apoptosis is, in part, a function of the hepatocyte's proteome. The aryl hydrocarbon receptor (AhR) is a ligand-activated transcription factor known to influence apoptosis, conceivably by regulating the expression of genes involved in apoptotic signaling. In this article, we present evidence demonstrating that AhR expression and function promote apoptosis in liver cells in response to Fas stimulation. Reintroduction of the AhR into the AhR-negative BP8 hepatoma cells as well as into primary hepatocytes from AhR knockout mice increases the magnitude of cell death in response to Fas ligand. Enhanced apoptosis correlates with increased caspase activity and mitochondrial cytochrome c release but not with the expression of several Bcl-2 family proteins. In vivo studies showed that in contrast to wild-type mice, AhR knockout mice are protected from the lethal effects of the anti-Fas Jo2 antibody. Moreover, down-regulation of the aryl hydrocarbon receptor nuclear translocator protein in vivo by adenovirus-mediated RNA interference to suppress AhR activity provided wild-type mice partial protection from Jo2-induced lethality.


Assuntos
Apoptose/fisiologia , Hepatócitos/citologia , Receptores de Hidrocarboneto Arílico/fisiologia , Receptor fas/fisiologia , Animais , Carcinoma Hepatocelular , Linhagem Celular Tumoral , Proteína Ligante Fas , Hepatócitos/fisiologia , Neoplasias Hepáticas , Glicoproteínas de Membrana/deficiência , Glicoproteínas de Membrana/fisiologia , Camundongos , Camundongos Knockout , Receptores de Hidrocarboneto Arílico/deficiência , Receptores de Hidrocarboneto Arílico/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa
17.
Toxicol Appl Pharmacol ; 192(3): 275-86, 2003 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-14575645

RESUMO

Although exposure to 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) clearly impairs T cell-dependent immune responses, the mechanisms underlying TCDD-induced T cell dysfunction are unclear. With the goal of determining precisely how exposure to TCDD impairs the activation of CD8(+) T cells in vivo, we used a well-defined T cell receptor (TCR) transgenic system. Greater than 95% of the CD8(+) T cells in F5 transgenic mice possess TCR specific for a peptide from influenza A virus expressed in the context of H-2D(b). Unexpectedly, we discovered that exposure to TCDD did not alter CD8(+) T cell function in the transgenic mice. Specifically, treatment of F5 mice with TCDD did not affect the recruitment of virus-specific CD8(+) T cells to the lung, nor did it impair the ability of CD8(+) T cells in the lymph node to produce cytokines, or to clonally expand or differentiate. This is in direct contrast to the suppressive effects of TCDD on the response of CD8(+) T cells in wild-type mice. Exposure of F5 mice to TCDD induced CYP1A1 and suppressed the production of virus-specific antibodies. Likewise, upon adoptive transfer into wild-type mice, TCDD suppressed the expansion and differentiation of F5-derived CD8(+) T cells. This indicates that the F5 mice and lymphocytes derived from them are not inherently resistant to the immunosuppressive effects of TCDD. Rather, our data suggest that in the context of a supraphysiological number of antigen-specific CD8(+) T cells, the function of these cells was not affected by exposure to TCDD. Given that antibody production in the F5 mice was sensitive to suppression by TCDD, while the CD8 response was resistant, our data provide a new perspective on the ways in which exposure to TCDD adversely affects B and T lymphocyte function.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Poluentes Ambientais/toxicidade , Vírus da Influenza A/imunologia , Infecções por Orthomyxoviridae/imunologia , Dibenzodioxinas Policloradas/toxicidade , Receptores de Antígenos de Linfócitos T/imunologia , Transferência Adotiva , Animais , Anticorpos Antivirais/sangue , Anticorpos Antivirais/efeitos dos fármacos , Formação de Anticorpos/efeitos dos fármacos , Linfócitos T CD8-Positivos/virologia , Divisão Celular , Citocromo P-450 CYP1A1/biossíntese , Indução Enzimática , Feminino , Vírus da Influenza A/patogenicidade , Interferon gama/metabolismo , Interleucina-2/metabolismo , Pulmão/enzimologia , Pulmão/virologia , Linfonodos/efeitos dos fármacos , Linfonodos/imunologia , Linfonodos/metabolismo , Camundongos , Camundongos Transgênicos , Receptores de Antígenos de Linfócitos T/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA