Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
Proc Natl Acad Sci U S A ; 119(28): e2202256119, 2022 07 12.
Artigo em Inglês | MEDLINE | ID: mdl-35867766

RESUMO

Phenotypic variations in the retinal pigment epithelial (RPE) layer are often a predecessor and driver of ocular degenerative diseases, such as age-related macular degeneration (AMD), the leading cause of vision loss in the elderly. We previously identified the orphan nuclear receptor-related 1 (NURR1), from a nuclear receptor atlas of human RPE cells, as a candidate transcription factor potentially involved in AMD development and progression. In the present study we characterized the expression of NURR1 as a function of age in RPE cells harvested from human donor eyes and in donor tissue from AMD patients. Mechanistically, we found an age-dependent shift in NURR1 dimerization from NURR1-RXRα heterodimers toward NURR1-NURR1 homodimers in primary human RPE cells. Additionally, overexpression and activation of NURR1 attenuated TNF-α-induced epithelial-to-mesenchymal transition (EMT) and migration, and modulated EMT-associated gene and protein expression in human RPE cells independent of age. In vivo, oral administration of IP7e, a potent NURR1 activator, ameliorated EMT in an experimental model of wet AMD and improved retinal function in a mouse model that presents with dry AMD features, impacting AMD phenotype, structure, and function of RPE cells, inhibiting accumulation of immune cells, and diminishing lipid accumulation. These results provide insight into the mechanisms of action of NURR1 in the aging eye, and demonstrate that the relative expression levels and activity of NURR1 is critical for both physiological and pathological functions of human RPE cells through RXRα-dependent regulation, and that targeting NURR1 may have therapeutic potential for AMD by modulating EMT, inflammation, and lipid homeostasis.


Assuntos
Transição Epitelial-Mesenquimal , Degeneração Macular , Membro 2 do Grupo A da Subfamília 4 de Receptores Nucleares , Epitélio Pigmentado da Retina , Idoso , Animais , Humanos , Lipídeos , Degeneração Macular/metabolismo , Degeneração Macular/patologia , Camundongos , Membro 2 do Grupo A da Subfamília 4 de Receptores Nucleares/química , Membro 2 do Grupo A da Subfamília 4 de Receptores Nucleares/metabolismo , Fenótipo , Multimerização Proteica , Epitélio Pigmentado da Retina/metabolismo
2.
Inhal Toxicol ; 35(7-8): 214-229, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37339372

RESUMO

BACKGROUND: Exposure to asbestos is associated with malignant and nonmalignant respiratory disease. To strengthen the scientific basis for risk assessment on fibers, the National Institute of Environmental Health Sciences (NIEHS) has initiated a series of studies to address fundamental questions on the toxicology of naturally occurring asbestos and related mineral fibers after inhalation exposure. A prototype nose-only exposure system was previously developed and validated. The prototype system was expanded to a large-scale exposure system in this study for conducting subsequent in vivo rodent inhalation studies of Libby amphibole (LA) 2007, selected as a model fiber. RESULTS: The exposure system consisting of six exposure carousels was able to independently deliver stable LA 2007 aerosol to individual carousels at target concentrations of 0 (control group), 0.1, 0.3, 1, 3, or 10 mg/m3. A single aerosol generator was used to provide aerosol to all carousels to ensure that exposure atmospheres were chemically and physically similar, with aerosol concentration as the only major variable among the carousels. Transmission electron microscopy (TEM) coupled with energy dispersive spectrometry (EDS) and selected area electron diffraction (SAED) analysis of aerosol samples collected at the exposure ports indicated the fiber dimensions, chemical composition, and mineralogy were equivalent across exposure carousels and were comparable to the bulk LA 2007 material. CONCLUSION: The exposure system developed is ready for use in conducting nose-only inhalation toxicity studies of LA 2007 in rats. The exposure system is anticipated to have applicability for the inhalation toxicity evaluation of other natural mineral fibers of concern.


Assuntos
Amiantos Anfibólicos , Amianto , Ratos , Animais , Amiantos Anfibólicos/toxicidade , Fibras Minerais , Aerossóis , Exposição por Inalação/efeitos adversos
3.
Inhal Toxicol ; 35(7-8): 201-213, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37339371

RESUMO

BACKGROUND: Asbestos has been classified as a human carcinogen, and exposure may increase the risk of diseases associated with impaired respiratory function. As the range of health effects and airborne concentrations that result in health effects across asbestos-related natural mineral fiber types are not fully understood, the National Institute of Environmental Health Sciences has established a series of research studies to characterize hazards of natural mineral fibers after inhalation exposure. This paper presents the method development work of this research project. RESULTS: A prototype nose-only exposure system was fabricated to explore the feasibility of generating natural mineral fiber aerosol for in vivo inhalation toxicity studies. The prototype system consisted of a slide bar aerosol generator, a distribution/delivery system and an exposure carousel. Characterization tests conducted using Libby Amphibole 2007 (LA 2007) demonstrated the prototype system delivered stable and controllable aerosol concentration to the exposure carousel. Transmission electron microscopy (TEM) analysis of aerosol samples collected at the exposure port showed the average fiber length and width were comparable to the bulk LA 2007. TEM coupled with energy dispersive spectrometry (EDS) and selected area electron diffraction (SAED) analysis further confirmed fibers from the aerosol samples were consistent with the bulk LA 2007 chemically and physically. CONCLUSIONS: Characterization of the prototype system demonstrated feasibility of generating LA 2007 fiber aerosols appropriate for in vivo inhalation toxicity studies. The methods developed in this study are suitable to apply to a multiple-carousel exposure system for a rat inhalation toxicity testing using LA 2007.


Assuntos
Amiantos Anfibólicos , Amianto , Humanos , Ratos , Animais , Amiantos Anfibólicos/toxicidade , Fibras Minerais , Amianto/análise , Carcinógenos/toxicidade , Aerossóis
4.
Int J Mol Sci ; 21(8)2020 Apr 21.
Artigo em Inglês | MEDLINE | ID: mdl-32326149

RESUMO

Vasculogenesis and angiogenesis are physiological mechanisms occurring throughout the body. Any disruption to the precise balance of blood vessel growth necessary to support healthy tissue, and the inhibition of abnormal vessel sprouting has the potential to negatively impact stages of development and/or healing. Therefore, the identification of key regulators of these vascular processes is critical to identifying therapeutic means by which to target vascular-associated compromises and complications. Nuclear receptors are a family of transcription factors that have been shown to be involved in modulating different aspects of vascular biology in many tissues systems. Most recently, the role of nuclear receptors in ocular biology and vasculopathies has garnered interest. Herein, we review studies that have used in vitro assays and in vivo models to investigate nuclear receptor-driven pathways in two ocular vascular diseases associated with blindness, wet or exudative age-related macular degeneration, and proliferative diabetic retinopathy. The potential therapeutic targeting of nuclear receptors for ocular diseases is also discussed.


Assuntos
Suscetibilidade a Doenças , Neovascularização Patológica/metabolismo , Receptores Citoplasmáticos e Nucleares/metabolismo , Animais , Biomarcadores , Gerenciamento Clínico , Humanos , Imuno-Histoquímica , Degeneração Macular/etiologia , Degeneração Macular/metabolismo , Degeneração Macular/patologia , Terapia de Alvo Molecular , Receptores Citoplasmáticos e Nucleares/antagonistas & inibidores , Neovascularização Retiniana/etiologia , Neovascularização Retiniana/metabolismo , Neovascularização Retiniana/patologia , Transdução de Sinais
5.
J Biol Chem ; 291(48): 25255-25263, 2016 Nov 25.
Artigo em Inglês | MEDLINE | ID: mdl-27765815

RESUMO

Alcoholic liver disease is a pathological condition caused by overconsumption of alcohol. Because of the high morbidity and mortality associated with this disease, there remains a need to elucidate the molecular mechanisms underlying its etiology and to develop new treatments. Because peroxisome proliferator-activated receptor-ß/δ (PPARß/δ) modulates ethanol-induced hepatic effects, the present study examined alterations in gene expression that may contribute to this disease. Chronic ethanol treatment causes increased hepatic CYP2B10 expression inPparß/δ+/+ mice but not in Pparß/δ-/- mice. Nuclear and cytosolic localization of the constitutive androstane receptor (CAR), a transcription factor known to regulate Cyp2b10 expression, was not different between genotypes. PPARγ co-activator 1α, a co-activator of both CAR and PPARß/δ, was up-regulated in Pparß/δ+/+ liver following ethanol exposure, but not in Pparß/δ-/- liver. Functional mapping of the Cyp2b10 promoter and ChIP assays revealed that PPARß/δ-dependent modulation of SP1 promoter occupancy up-regulated Cyp2b10 expression in response to ethanol. These results suggest that PPARß/δ regulates Cyp2b10 expression indirectly by modulating SP1 and PPARγ co-activator 1α expression and/or activity independent of CAR activity. Ligand activation of PPARß/δ attenuates ethanol-induced Cyp2b10 expression in Pparß/δ+/+ liver but not in Pparß/δ-/- liver. Strikingly, Cyp2b10 suppression by ligand activation of PPARß/δ following ethanol treatment occurred in hepatocytes and was mediated by paracrine signaling from Kupffer cells. Combined, results from the present study demonstrate a novel regulatory role of PPARß/δ in modulating CYP2B10 that may contribute to the etiology of alcoholic liver disease.


Assuntos
Hidrocarboneto de Aril Hidroxilases/biossíntese , Família 2 do Citocromo P450/biossíntese , Regulação Enzimológica da Expressão Gênica , Hepatopatias Alcoólicas/metabolismo , Fígado/metabolismo , PPAR delta/metabolismo , PPAR beta/metabolismo , Regiões Promotoras Genéticas , Fator de Transcrição Sp1/metabolismo , Esteroide Hidroxilases/biossíntese , Animais , Hidrocarboneto de Aril Hidroxilases/genética , Família 2 do Citocromo P450/genética , Etanol/toxicidade , Hepatócitos/metabolismo , Hepatócitos/patologia , Células de Kupffer/metabolismo , Células de Kupffer/patologia , Fígado/patologia , Hepatopatias Alcoólicas/genética , Hepatopatias Alcoólicas/patologia , Camundongos , Camundongos Knockout , PPAR delta/genética , PPAR beta/genética , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Fator de Transcrição Sp1/genética , Esteroide Hidroxilases/genética
6.
Immunology ; 150(4): 456-467, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-27935639

RESUMO

The peroxisome proliferator-activated receptor-ß/δ (PPARß/δ) is known to have multiple anti-inflammatory effects, typically observed in endothelial cells, macrophages, T cells and B cells. Despite the fact that mast cells are important mediators of inflammation, to date, the role of PPARß/δ in mast cells has not been examined. Hence, the present study examined the hypothesis that PPARß/δ modulates mast cell phenotype. Bone-marrow-derived mast cells (BMMCs) and peritoneal mast cells from Pparß/δ+/+ mice expressed higher levels of high-affinity IgE receptor (FcεRI) compared with Pparß/δ-/- mice. BMMCs from Pparß/δ+/+ mice also exhibited dense granules, associated with higher expression of enzymes and proteases compared with Pparß/δ-/- mice. Resting BMMCs from Pparß/δ+/+ mice secreted lower levels of inflammatory cytokines, associated with the altered activation of phospholipase Cγ1 and extracellular signal-regulated kinases compared with Pparß/δ-/- mice. Moreover, the production of cytokines by mast cells induced by various stimuli was highly dependent on PPARß/δ expression. This study demonstrates that PPARß/δ is an important regulator of mast cell phenotype.


Assuntos
Células da Medula Óssea/fisiologia , Diferenciação Celular , Mastócitos/fisiologia , PPAR delta/metabolismo , PPAR beta/metabolismo , Animais , Células Cultivadas , Citocinas/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Feminino , Mediadores da Inflamação/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , PPAR delta/genética , PPAR beta/genética , Fenótipo , Receptores de IgE/genética , Receptores de IgE/metabolismo , Transdução de Sinais/genética
7.
Mol Carcinog ; 56(5): 1472-1483, 2017 05.
Artigo em Inglês | MEDLINE | ID: mdl-27996177

RESUMO

Neuroblastoma is a common childhood cancer typically treated by inducing differentiation with retinoic acid (RA). Peroxisome proliferator-activated receptor-ß/δ, (PPARß/δ) is known to promote terminal differentiation of many cell types. In the present study, PPARß/δ was over-expressed in three human neuroblastoma cell lines, NGP, SK-N-BE(2), and IMR-32, that exhibit high, medium, and low sensitivity, respectively, to retinoic acid-induced differentiation to determine if PPARß/δ and retinoic acid receptors (RARs) could be jointly targeted to increase the efficacy of treatment. All-trans-RA (atRA) decreased expression of SRY (sex determining region Y)-box 2 (SOX2), a stem cell regulator and marker of de-differentiation, in NGP and SK-N-BE(2) cells with inactive or mutant tumor suppressor p53, respectively. However, atRA did not suppress SOX2 expression in IMR-32 cells carrying wild-type p53. Over-expression and/or ligand activation of PPARß/δ reduced the average volume and weight of ectopic tumor xenografts from NGP, SK-N-BE(2), or IMR-32 cells compared to controls. Compared with that found with atRA, PPARß/δ suppressed SOX2 expression in NGP and SK-N-BE(2) cells and ectopic xenografts, and was also effective in suppressing SOX2 expression in IMR-32 cells that exhibit higher p53 expression compared to the former cell lines. Combined, these observations demonstrate that activating or over-expressing PPARß/δ induces cell differentiation through p53- and SOX2-dependent signaling pathways in neuroblastoma cells and tumors. This suggests that combinatorial activation of both RARα and PPARß/δ may be suitable as an alternative therapeutic approach for RA-resistant neuroblastoma patients.


Assuntos
Neuroblastoma/patologia , PPAR delta/metabolismo , PPAR beta/metabolismo , Fatores de Transcrição SOXB1/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Animais , Diferenciação Celular/fisiologia , Linhagem Celular Tumoral , Resistencia a Medicamentos Antineoplásicos , Feminino , Humanos , Camundongos Nus , Neuroblastoma/tratamento farmacológico , Neuroblastoma/metabolismo , PPAR delta/genética , PPAR beta/genética , PTEN Fosfo-Hidrolase/metabolismo , Receptor alfa de Ácido Retinoico/metabolismo , Tretinoína/farmacologia , Ensaios Antitumorais Modelo de Xenoenxerto
8.
J Biol Chem ; 290(38): 23416-31, 2015 Sep 18.
Artigo em Inglês | MEDLINE | ID: mdl-26242735

RESUMO

Ppard(-/-) mice exhibit smaller litter size compared with Ppard(+/+) mice. To determine whether peroxisome proliferator-activated receptor-D (PPARD) could possibly influence this phenotype, the role of PPARD in testicular biology was examined. Atrophic testes and testicular degeneration were observed in Ppard(-/-) mice compared with Ppard(+/+) mice, indicating that PPARD modulates spermatogenesis. Higher expression of p27 and decreased expression of proliferating cellular nuclear antigen in Sertoli cells were observed in Ppard(+/+) mice as compared with Ppard(-/-) mice, and these were associated with decreased Sertoli cell number in Ppard(+/+) mice. Cyclin D1 and cyclin D2 expression was lower in Ppard(+/+) as compared with Ppard(-/-) mice. Ligand activation of PPARD inhibited proliferation of a mouse Sertoli cell line, TM4, and an inverse agonist of PPARD (DG172) rescued this effect. Temporal inhibition of extracellular signal-regulated kinase (ERK) activation by PPARD in the testis was observed in Ppard(+/+) mice and was associated with decreased serum follicle-stimulating hormone and higher claudin-11 expression along the blood-testis barrier. PPARD-dependent ERK activation also altered expression of claudin-11, p27, cyclin D1, and cyclin D2 in TM4 cells, causing inhibition of cell proliferation, maturation, and formation of tight junctions in Sertoli cells, thus confirming a requirement for PPARD in accurate Sertoli cell function. Combined, these results reveal for the first time that PPARD regulates spermatogenesis by modulating the function of Sertoli cells during early testis development.


Assuntos
Proliferação de Células/fisiologia , Sistema de Sinalização das MAP Quinases/fisiologia , Receptores Citoplasmáticos e Nucleares/metabolismo , Células de Sertoli/metabolismo , Espermatogênese/fisiologia , Animais , Linhagem Celular , Claudinas/biossíntese , Claudinas/genética , Ciclina D1/biossíntese , Ciclina D1/genética , Ciclina D2/biossíntese , Ciclina D2/genética , MAP Quinases Reguladas por Sinal Extracelular/biossíntese , MAP Quinases Reguladas por Sinal Extracelular/genética , Masculino , Camundongos , Receptores Citoplasmáticos e Nucleares/genética , Células de Sertoli/citologia
9.
Biol Reprod ; 86(5): 160, 1-10, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22321834

RESUMO

Testicular dysgenesis syndrome refers to a collection of diseases in men, including testicular cancer, that arise as a result of abnormal testicular development. Phthalates are a class of chemicals used widely in the production of plastic products and other consumer goods. Unfortunately, phthalate exposure has been linked to reproductive dysfunction and has been shown to adversely affect normal germ cell development. In this study, we show that mono-(2-ethylhexyl) phthalate (MEHP) induces matrix metalloproteinase 2 (MMP2) expression in testicular embryonal carcinoma NT2/D1 cells but has no significant effect on MMP9 expression. NT2/D1 cells also have higher levels of MYC expression following MEHP treatment. It is widely recognized that activation of MMP2 and MYC is tightly associated with tumor metastasis and tumor progression. Gelatin zymographic analysis indicates that MEHP strongly activates MMP2 in NT2/D1 cells. Addition of the MMP2-specific inhibitor SB-3CT inhibited MEHP-enhanced cell invasion and migration, demonstrating that MMP2 plays a functional role in promoting testicular embryonal carcinoma progression in response to MEHP exposure. Furthermore, we investigated genome-wide gene expression profiles of NT2/D1 cells following MEHP exposure at 0, 3, and 24 h. Microarray analysis and semiquantitative RT-PCR revealed that MEHP exposure primarily influenced genes in cell adhesion and transcription in NT2/D1 cells. Gap junction protein-alpha 1, vinculin, and inhibitor of DNA-binding protein-1 were significantly down-regulated by MEHP treatment, while claudin-6 and beta 1-catenin expression levels were up-regulated. This study provides insight into mechanisms that may account for modulating testicular cancer progression following phthalate exposure.


Assuntos
Movimento Celular/fisiologia , Dietilexilftalato/análogos & derivados , Células-Tronco de Carcinoma Embrionário/patologia , Neoplasias Testiculares/patologia , Cateninas/biossíntese , Adesão Celular/efeitos dos fármacos , Adesão Celular/fisiologia , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Claudinas/biossíntese , Conexina 43/biossíntese , Dietilexilftalato/efeitos adversos , Células-Tronco de Carcinoma Embrionário/efeitos dos fármacos , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/fisiologia , Compostos Heterocíclicos com 1 Anel/farmacologia , Humanos , Proteína 1 Inibidora de Diferenciação/biossíntese , Masculino , Metaloproteinase 2 da Matriz/biossíntese , Metaloproteinase 9 da Matriz/biossíntese , Invasividade Neoplásica , Sulfonas/farmacologia , Neoplasias Testiculares/tratamento farmacológico , Transcrição Gênica/efeitos dos fármacos , Transcrição Gênica/fisiologia , Vinculina/biossíntese , delta Catenina
10.
Biol Reprod ; 85(6): 1203-15, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21832167

RESUMO

Our previous studies showed that the prototypical testicular toxic phthalate monoester, mono-(2-ethylhexyl) phthalate (MEHP), suppresses Sertoli cell TIMP2 levels and allows for the activation of MMP2 in seminiferous epithelium. Activation of MMP2 is important for triggering germ cell apoptosis and instigating germ cell detachment from Sertoli cells. These novel findings led us to examine the transcriptional regulation of the Timp2 gene that accounts for the decrease in Sertoli cell TIMP2 levels following MEHP exposure. Sequential deletion of the Timp2 5'-upstream activating sequence (1200 bp) was used to survey transcriptional activation in the Timp2 promoter region in response to MEHP. Results indicate that under control conditions in rat Sertoli cells, CCAAT enhancer-binding protein alpha (CEBPA) acts as a transactivator to initiate Timp2 gene transcription, and its action is deactivated by exposure to MEHP. By contrast, MYC protein acts as an inhibitor of Timp2 gene transcription, and its activity is increased after MEHP treatment. Addition of follicle-stimulating hormone (FSH) to cells causes translocation of CEBPA into the Sertoli cell nucleus and rescues MEHP-suppressed TIMP2 levels. Down-regulation of TIMP2 expression by MEHP exposure is blocked by forskolin (a cAMP-elevating agent), suggesting that the decrease in Sertoli cell TIMP2 expression following MEHP exposure is cAMP-dependent. Taken together, these data indicate that MEHP both disrupts the FSH-stimulated cAMP signaling pathway and activates the inhibitory signaling mediated by MYC protein, to ultimately account for the cellular mechanism underlying the decreased expression of TIMP2 in Sertoli cells.


Assuntos
Proteínas Estimuladoras de Ligação a CCAAT/metabolismo , Dietilexilftalato/análogos & derivados , Proteínas Proto-Oncogênicas c-myc/metabolismo , Células de Sertoli/metabolismo , Inibidor Tecidual de Metaloproteinase-2/metabolismo , Animais , Técnicas de Cocultura , Dietilexilftalato/toxicidade , Hormônio Foliculoestimulante/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Regiões Promotoras Genéticas , RNA Mensageiro/metabolismo , Sistemas do Segundo Mensageiro , Células de Sertoli/efeitos dos fármacos , Inibidor Tecidual de Metaloproteinase-2/genética
11.
Biol Reprod ; 82(3): 516-27, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19828778

RESUMO

Tight junctions between Sertoli cells of the testicular seminiferous epithelium establishes the blood-testis barrier (BTB) and creates a specialized adluminal microenvironment above the BTB that is required for the development of the germ cells that reside there. Actin filament-based anchoring junctions between Sertoli cells and germ cells are important for maintaining close physical contact between these cells as well as regulating the release of mature spermatids into the lumen. Previously, we reported that Sertoli cell injury in rodents after mono-(2-ethylhexyl) phthalate (MEHP) exposure results in the activation of matrix metalloproteinase 2 (MMP2) and increases the sensitivity of germ cells to undergo apoptosis. A disruption in the physical association between Sertoli cells and germ cells and premature loss of germ cells from the seminiferous epithelium has been widely described after phthalate treatment. In this study, we investigate the functional participation of MMP2 in the mechanism underlying MEHP-induced disruption of junction complexes and the resultant loss of germ cells. Exposure of C57BL/6J mice to MEHP (1 g/kg, oral gavage) decreased the expression of occludin in the tight junctions between Sertoli cells and caused gaps between adjacent Sertoli cells as observed by transmission electron microscopy. A reduced expression of laminin-gamma3 and beta1-integrin in apical ectoplasmic specializations between Sertoli cells and germ cells in a time-dependent manner was also observed. Treatment with specific MMP2 inhibitors (TIMP2 and SB-3CT) both in vitro and in vivo significantly suppressed MEHP-induced germ cell sloughing and changes in the expression of these junctional proteins, indicating that MMP-2 plays a primary role in this process. Furthermore, the detachment of germ cells from Sertoli cells appears to be independent of the apoptotic signaling process since MEHP-induced germ cell detachment from Sertoli cells could not be prevented by the addition of a pan-caspase inhibitor (z-VAD-FMK).


Assuntos
Junções Aderentes/efeitos dos fármacos , Dietilexilftalato/análogos & derivados , Metaloproteinase 2 da Matriz/fisiologia , Epitélio Seminífero/efeitos dos fármacos , Testículo/efeitos dos fármacos , Junções Aderentes/metabolismo , Animais , Barreira Hematotesticular/efeitos dos fármacos , Barreira Hematotesticular/metabolismo , Adesão Celular/efeitos dos fármacos , Células Cultivadas , Dietilexilftalato/farmacologia , Ativação Enzimática/efeitos dos fármacos , Masculino , Metaloproteinase 2 da Matriz/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Complexos Multiproteicos/efeitos dos fármacos , Complexos Multiproteicos/metabolismo , Ratos , Ratos Endogâmicos F344 , Epitélio Seminífero/metabolismo , Doenças Testiculares/induzido quimicamente , Doenças Testiculares/metabolismo , Testículo/metabolismo , Testículo/ultraestrutura
12.
JCI Insight ; 5(1)2020 01 16.
Artigo em Inglês | MEDLINE | ID: mdl-31829999

RESUMO

Effective treatments and animal models for the most prevalent neurodegenerative form of blindness in elderly people, called age-related macular degeneration (AMD), are lacking. Genome-wide association studies have identified lipid metabolism and inflammation as AMD-associated pathogenic pathways. Given liver X receptors (LXRs), encoded by the nuclear receptor subfamily 1 group H members 2 and 3 (NR1H3 and NR1H2), are master regulators of these pathways, herein we investigated the role of LXR in human and mouse eyes as a function of age and disease and tested the therapeutic potential of targeting LXR. We identified immunopositive LXR fragments in human extracellular early dry AMD lesions and a decrease in LXR expression within the retinal pigment epithelium (RPE) as a function of age. Aged mice lacking LXR presented with isoform-dependent ocular pathologies. Specifically, loss of the Nr1h3 isoform resulted in pathobiologies aligned with AMD, supported by compromised visual function, accumulation of native and oxidized lipids in the outer retina, and upregulation of ocular inflammatory cytokines, while absence of Nr1h2 was associated with ocular lipoidal degeneration. LXR activation not only ameliorated lipid accumulation and oxidant-induced injury in RPE cells but also decreased ocular inflammatory markers and lipid deposition in a mouse model, thereby providing translational support for pursuing LXR-active pharmaceuticals as potential therapies for dry AMD.


Assuntos
Receptores X do Fígado/genética , Receptores X do Fígado/metabolismo , Degeneração Macular/genética , Degeneração Macular/metabolismo , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Envelhecimento/patologia , Animais , Modelos Animais de Doenças , Células Endoteliais , Feminino , Estudo de Associação Genômica Ampla , Humanos , Inflamação/metabolismo , Degeneração Macular/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Pessoa de Meia-Idade , Fenótipo , Retina/metabolismo , Retina/patologia , Epitélio Pigmentado da Retina , Transcriptoma , Adulto Jovem
13.
Toxicol Sci ; 159(2): 436-448, 2017 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-28962521

RESUMO

Skin tumorigenesis results from DNA damage, increased inflammation, and evasion of apoptosis. The peroxisome proliferator-activated receptors (PPARs) can modulate these mechanisms in non-melanoma skin cancer. However, limited data exists regarding the role of PPARs in melanoma. This study examined the effect of proliferator-activated receptor-ß/δ (PPARß/δ) and PPARγ on cell proliferation, anchorage-dependent clonogenicity, and ectopic xenografts in the UACC903 human melanoma cell line. Stable overexpression of either PPARß/δ or PPARγ enhanced ligand-induced expression of a PPARß/δ/PPARγ target gene in UACC903 cell lines as compared with controls. The induction of target gene expression by ligand activation of PPARγ was not altered by overexpression of PPARß/δ, or vice versa. Stable overexpression of either PPARß/δ or PPARγ reduced the percentage of cells in the G1 and S phase of the cell cycle, and increased the percentage of cells in the G2/M phase of the cell cycle in UACC903 cell lines as compared with controls. Ligand activation of PPARß/δ did not further alter the distribution of cells within each phase of the cell cycle. By contrast, ligand activation of PPARγ enhanced these changes in stable UACC903 cells overexpressing PPARγ compared with controls. Stable overexpression of either PPARß/δ or PPARγ and/or ligand activation of either PPARß/δ or PPARγ inhibited cell proliferation, and anchorage-dependent clonogenicity of UACC903 cell lines as compared with controls. Further, overexpression of either PPARß/δ or PPARγ and/or ligand activation of either PPARß/δ or PPARγ inhibited ectopic xenograft tumorigenicity derived from UACC903 melanoma cells as compared with controls, and this was likely due in part to induction of apoptosis. Results from these studies demonstrate the antitumorigenic effects of both PPARß/δ and PPARγ and suggest that targeting these receptors may be useful for primary or secondary melanoma chemoprevention.


Assuntos
Apoptose/fisiologia , Inflamação/fisiopatologia , Melanoma/patologia , Receptores Ativados por Proliferador de Peroxissomo/fisiologia , Neoplasias Cutâneas/patologia , Animais , Adesão Celular/fisiologia , Ciclo Celular/fisiologia , Linhagem Celular Tumoral , Proliferação de Células/fisiologia , Xenoenxertos , Humanos , Ligantes , Camundongos , Camundongos Nus , Receptores Ativados por Proliferador de Peroxissomo/genética
14.
J Clin Oncol ; 23(5): 953-64, 2005 Feb 10.
Artigo em Inglês | MEDLINE | ID: mdl-15598976

RESUMO

PURPOSE: Inflammation plays a critical role in cancer progression. In this study we investigate the pro-tumorigenic activities and gene expression profiles of lung cancer cells after interaction with macrophages. MATERIALS AND METHODS: We measured intratumoral microvessel counts and macrophage density in 41 lung cancer tumor specimens and correlated these with the patients' clinical outcome. The interaction between macrophages and cancer cell lines was assessed using a transwell coculture system. The invasive potential was evaluated by in vitro invasion assay. The matrix-degrading activity was assayed by gelatin zymography. The microarray was applied to a large-scale analysis of the genes involved in the interaction, as well as to monitor the gene expression profiles of lung cancer cells responding to anti-inflammatory drugs in cocultures. RESULTS: The macrophage density positively correlated with microvessel counts and negatively correlated with patient relapse-free survival (P < .05). After coculture with macrophages, lung cancer cell lines exhibited higher invasive potentials and matrix-degrading activities. We identified 50 genes by microarray that were upregulated more than two-fold in cancer cells after coculture. Northern blot analyses confirmed some gene expression such as interleukin-6, interleukin-8, and matrix metalloproteinase 9. The two-dimensional hierarchical clustering also demonstrated that the gene expression profiles of lung cancer cells responding to various anti-inflammatory drugs in cocultures are distinct. CONCLUSION: The interaction of lung cancer cells and macrophages can promote the invasiveness and matrix-degrading activity of cancer cells. Our results also suggest that a great diversity of gene expression occurs in this interaction, which may assist us in understanding the process of cancer metastasis.


Assuntos
Neoplasias Pulmonares/patologia , Macrófagos Alveolares/patologia , Adenocarcinoma/irrigação sanguínea , Adenocarcinoma/genética , Adenocarcinoma/patologia , Anti-Inflamatórios/uso terapêutico , Carcinoma Pulmonar de Células não Pequenas/irrigação sanguínea , Carcinoma Pulmonar de Células não Pequenas/genética , Carcinoma Pulmonar de Células não Pequenas/patologia , Carcinoma de Células Escamosas/irrigação sanguínea , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/patologia , Contagem de Células , Linhagem Celular Tumoral , Técnicas de Cocultura , Progressão da Doença , Intervalo Livre de Doença , Feminino , Gelatinases/genética , Regulação Neoplásica da Expressão Gênica , Humanos , Interleucina-6/genética , Interleucina-8/genética , Neoplasias Pulmonares/irrigação sanguínea , Neoplasias Pulmonares/genética , Macrófagos Alveolares/efeitos dos fármacos , Masculino , Metaloproteinase 9 da Matriz/genética , Microcirculação/patologia , Pessoa de Meia-Idade , Invasividade Neoplásica , Regulação para Cima/genética
15.
Aging (Albany NY) ; 8(9): 1952-1978, 2016 09 08.
Artigo em Inglês | MEDLINE | ID: mdl-27622388

RESUMO

Peroxisome proliferator-activated receptor-ß/δ (PPARß/δ) is a nuclear receptor that regulates differentiation, inflammation, lipid metabolism, extracellular matrix remodeling, and angiogenesis in multiple tissues. These pathways are also central to the pathogenesis of age-related macular degeneration (AMD), the leading cause of vision loss globally. With the goal of identifying signaling pathways that may be important in the development of AMD, we investigated the impact of PPARß/δ activation on ocular tissues affected in the disease. PPARß/δ is expressed and can be activated in AMD vulnerable cells, including retinal pigment epithelial (RPE) and choroidal endothelial cells. Further, PPARß/δ knockdown modulates AMD-related pathways selectively. Specifically, genetic ablation of Pparß/δ in aged mice resulted in exacerbation of several phenotypic features of early dry AMD, but attenuation of experimentally induced choroidal neovascular (CNV) lesions. Antagonizing PPARß/δ in both in vitro angiogenesis assays and in the in vivo experimentally induced CNV model, inhibited angiogenesis and angiogenic pathways, while ligand activation of PPARß/δ, in vitro, decreased RPE lipid accumulation, characteristic of dry AMD. This study demonstrates for the first time, selective regulation of a nuclear receptor in the eye and establishes that selective targeting of PPARß/δ may be a suitable strategy for treatment of different clinical sub-types of AMD.


Assuntos
Degeneração Macular/metabolismo , Neovascularização Patológica/metabolismo , PPAR beta/metabolismo , Epitélio Pigmentado da Retina/metabolismo , Idoso , Animais , Linhagem Celular , Movimento Celular/efeitos dos fármacos , Movimento Celular/fisiologia , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Eletrorretinografia , Feminino , Humanos , Macaca mulatta , Degeneração Macular/genética , Masculino , Camundongos , Camundongos Transgênicos , Pessoa de Meia-Idade , Neovascularização Patológica/genética , PPAR beta/agonistas , PPAR beta/antagonistas & inibidores , PPAR beta/genética , Fenótipo , Epitélio Pigmentado da Retina/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Sulfonas/farmacologia , Tiazóis/farmacologia , Tiofenos/farmacologia , Adulto Jovem
16.
Front Biosci ; 10: 853-65, 2005 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-15569594

RESUMO

Abstract Interleukin (IL)-8, a cytokine of the CXC chemokine family that was originally classified as a neutrophil chemoattractant, is now reported to play an important role in tumor progression and metastasis in a variety of human cancers, including lung cancers. IL-8 biologic activity in tumors and the tumor microenvironment may contribute to tumor progression through its potential function in the regulation of angiogenesis, cancer cell growth and survival, tumor cell motion, leukocyte infiltration and modification of immune responses. Recently, infiltrating macrophages in tumor stroma have been considered to be able to stimulate cancer growth, enhance angiogenesis and promote metastasis, and has prognostic significance in several human cancers. Accumulating evidence also shows that cancer cells and stromal cell interaction can stimulate cancer cells, as well as stromal cells in the expression of IL-8 and other growth factors. Here, we summarize current information about IL-8 biology in human lung cancers and focus on its effect on tumor angiogenesis, regulation of IL-8 expression in tumors, its prognostic significances, the role of tumor infiltrating macrophages in the production of IL-8 in cancer cells and the tumor microenvironment, gene expression profiles after cancer cell-stromal cell interaction, and the effect of a variety anti- inflammatory drugs on the modification of IL-8 and other gene expressions in cancer cells and the tumor microenvironment in lung cancers.


Assuntos
Regulação Neoplásica da Expressão Gênica , Interleucina-8/fisiologia , Microcirculação , Neovascularização Patológica , Anti-Inflamatórios/farmacologia , Humanos , Interleucina-8/metabolismo , Leucócitos/metabolismo , Macrófagos/metabolismo , Metástase Neoplásica , Neoplasias/metabolismo
17.
Respir Res ; 6: 89, 2005 Aug 08.
Artigo em Inglês | MEDLINE | ID: mdl-16083514

RESUMO

BACKGROUND: Theophylline has been used widely as a bronchodilator for the treatment of bronchial asthma and has been suggested to modulate immune response. While the importance of macrophages in asthma has been reappraised and emphasized, their significance has not been well investigated. We conducted a genome-wide profiling of the gene expressions of macrophages in response to theophylline. METHODS: Microarray technology was used to profile the gene expression patterns of macrophages modulated by theophylline. Northern blot and real-time quantitative RT-PCR were also used to validate the microarray data, while Western blot and ELISA were used to measure the levels of IL-13 and LTC4. RESULTS: We identified dozens of genes in macrophages that were dose-dependently down- or up-regulated by theophylline. These included genes related to inflammation, cytokines, signaling transduction, cell adhesion and motility, cell cycle regulators, and metabolism. We observed that IL-13, a central mediator of airway inflammation, was dramatically suppressed by theophylline. Real-time quantitative RT-PCR and ELISA analyses also confirmed these results, without respect to PMA-treated THP-1 cells or isolated human alveolar macrophages. Theophylline, rolipram, etazolate, db-cAMP and forskolin suppressed both IL-13 mRNA expression (~25%, 2.73%, 8.12%, 5.28%, and 18.41%, respectively) and protein secretion (<10% production) in macrophages. These agents also effectively suppressed LTC4 expression. CONCLUSION: Our results suggest that the suppression of IL-13 by theophylline may be through cAMP mediation and may decrease LTC4 production. This study supports the role of theophylline as a signal regulator of inflammation, and that down regulation of IL-13 by theophylline may have beneficial effects in inflammatory airway diseases.


Assuntos
Bronquite/metabolismo , Bronquite/patologia , Fatores Imunológicos/metabolismo , Macrófagos/metabolismo , Alvéolos Pulmonares/metabolismo , Teofilina/administração & dosagem , Anti-Inflamatórios/administração & dosagem , Linhagem Celular , Relação Dose-Resposta a Droga , Perfilação da Expressão Gênica , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Interleucina-13/metabolismo , Leucotrieno C4/metabolismo , Ativação de Macrófagos/efeitos dos fármacos , Macrófagos/efeitos dos fármacos , Proteoma/metabolismo , Alvéolos Pulmonares/patologia
18.
Clin Cancer Res ; 9(2): 729-37, 2003 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-12576442

RESUMO

PURPOSE: To evaluate the interaction between tumor-infiltrating macrophages and cancer cells and its effect on the expression of a potent angiogenic factor, interleukin-8 (IL-8), tumor angiogenesis, and patient outcome in non-small cell lung cancer (NSCLC). EXPERIMENTAL DESIGN: We measured tumor IL-8 mRNA expression (by real-time quantitative reverse transcription-PCR), intratumor microvessel counts, and tumor-infiltrating macrophage density (by immunohistochemical staining) in 35 NSCLC surgical specimens and correlated with the patient's clinical outcome. We then investigated the interaction between macrophages (cell line THP-1) and six different human cancer cell lines (four NSCLCs, one osteosarcoma, and one hepatoma) and its effect on IL-8 mRNA expression using a macrophage/cancer cell coculture system, IL-8 mRNA expression in lung cancer cells, and macrophages being measured separately after coculture in the presence or absence of six anti-inflammatory agents, i.e., pentoxifylline, aspirin, indomethacin, dexamethasone, celecoxib (a selective cyclooxygenase-2 inhibitor), and pyrrolidine dithiocarbamate, a specific nuclear factor kappaB (NF-kappaB) inhibitor. NF-kappaB transcriptional activity and protein levels were measured by reporter gene assay and Western blot. RESULTS: The tumor-infiltrating macrophage density correlated significantly and positively with tumor IL-8 mRNA expression and intratumor microvessel counts and significantly and negatively with patient survival. In addition, after cell-cell interaction in cancer cell:macrophage cocultures, marked IL-8 mRNA expression was induced in lung cancer cells (approximately 270-fold) and, to a lesser degree, in macrophages (4.5-fold). The increase in IL-8 mRNA expression correlated with the in vitro metastatic potential of the cancer cells. All six anti-inflammatory agents suppressed induction of IL-8 mRNA expression in lung cancer cells by >90%, four (pentoxifylline, celecoxib, pyrrolidine dithiocarbamate, and dexamethasone) having a dose-dependent effect. NF-kappaB transcriptional regulation and protein levels were simultaneously increased in the nuclei of cancer cells in macrophage/cancer cell cocultures, this effect also being suppressed by all six anti-inflammatory agents. CONCLUSIONS: The interaction between infiltrating macrophages and cancer cells up-regulates IL-8 mRNA expression, especially in the cancer cells; this may contribute greatly to the increased tumor angiogenesis and adverse outcome in NSCLC patients with a high density of tumor-infiltrating macrophages. Anti-inflammatory agents can suppress the induction of IL-8 mRNA expression seen in lung cancer cells after coculture with macrophages, and this suppression is mediated, in part, through the NF-kappaB pathway.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/imunologia , Regulação Neoplásica da Expressão Gênica , Interleucina-8/genética , Neoplasias Pulmonares/imunologia , Macrófagos/imunologia , Neovascularização Patológica/imunologia , Transcrição Gênica , Carcinoma Pulmonar de Células não Pequenas/irrigação sanguínea , Carcinoma Pulmonar de Células não Pequenas/genética , Carcinoma Pulmonar de Células não Pequenas/mortalidade , Linhagem Celular , Técnicas de Cocultura , Feminino , Regulação Neoplásica da Expressão Gênica/imunologia , Humanos , Imuno-Histoquímica , Neoplasias Pulmonares/irrigação sanguínea , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/mortalidade , Macrófagos/patologia , Masculino , Microcirculação/imunologia , Microcirculação/patologia , Pessoa de Meia-Idade , Neovascularização Patológica/genética , Neovascularização Patológica/patologia , RNA Mensageiro/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Análise de Sobrevida , Células Tumorais Cultivadas
19.
Curr Pharmacol Rep ; 1(2): 121-128, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26000234

RESUMO

The role of peroxisome proliferator-activated receptor-ß/δ (PPARß/δ) in cancer remains contentious due in large part to divergent publications indicating opposing effects in different rodent and human cell culture models. During the past 10 years, some facts regarding PPARß/δ in cancer have become clearer, while others remain uncertain. For example, it is now well accepted that (1) expression of PPARß/δ is relatively lower in most human tumors as compared to the corresponding non-transformed tissue, (2) PPARß/δ promotes terminal differentiation, and (3) PPARß/δ inhibits pro-inflammatory signaling in multiple in vivo models. However, whether PPARß/δ is suitable to target with natural and/or synthetic agonists or antagonists for cancer chemoprevention is hindered because of the uncertainty in the mechanism of action and role in carcinogenesis. Recent findings that shed new insight into the possibility of targeting this nuclear receptor to improve human health will be discussed.

20.
Oncotarget ; 6(34): 36319-37, 2015 Nov 03.
Artigo em Inglês | MEDLINE | ID: mdl-26431381

RESUMO

Peroxisome proliferator-activated receptor-ß/δ (PPARß/δ) has important physiological functions in control of cell growth, lipid and glucose homeostasis, differentiation and inflammation. To investigate the role of PPARß/δ in cancer, stable human testicular embryonal carcinoma cell lines were developed that constitutively express PPARß/δ. Expression of PPARß/δ caused enhanced activation of the receptor, and this significantly decreased proliferation, migration, invasion, anchorage-independent growth, and also reduced tumor mass and volume of ectopic xenografts derived from NT2/D1 cells compared to controls. The changes observed in xenografts were associated with decreased PPARß/δ-dependent expression of proliferating cell nuclear antigen and octamer-binding transcription factor-3/4, suggesting suppressed tumor proliferation and induction of differentiation. Inhibition of migration and invasion was mediated by PPARß/δ competing with formation of the retinoic acid receptor (RAR)/retinoid X receptor (RXR) complex, resulting in attenuation of RARα-dependent matrix metalloproteinase-2 expression and activity. These results demonstrate that PPARß/δ mediates attenuation of human testicular embryonal carcinoma cell progression through a novel RAR-dependent mechanism and suggest that activation of PPARß/δ inhibits RAR/RXR dimerization and represents a new therapeutic strategy.


Assuntos
PPAR delta/metabolismo , Receptores do Ácido Retinoico/metabolismo , Neoplasias Testiculares/metabolismo , Neoplasias Testiculares/patologia , Adolescente , Adulto , Carcinogênese , Diferenciação Celular , Linhagem Celular Tumoral , Proliferação de Células/fisiologia , Humanos , Masculino , Adulto Jovem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA