Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 78
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Exp Cell Res ; 417(1): 113210, 2022 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-35597298

RESUMEN

Cancer cells acquire immunoediting ability to evade immune surveillance and thus escape eradication. It is widely known that mutant proteins encoded from tumor suppressor TP53 exhibit gain-of-function in cancer cells, thereby promoting progression; however, how mutant p53 contributes to the sheltering of cancer cells from host anticancer immunity remains unclear. Herein, we report that murine p53 missense mutation G242A (corresponding to human G245A) suppresses the activation of host natural killer (NK) cells, thereby enabling breast cancer cells to avoid immune assault. We found that serial injection of EMT6 breast cancer cells that carry wild-type (wt) Trp53, like normal fibroblasts, promoted NK activity in mice, while SVTneg2 cells carrying Trp53 G242A+/+ mutation decreased NK cell numbers and increased CD8+ T lymphocyte numbers in spleen. Innate immunity based on NK cells and CD8 T cells was reduced in p53 mutant-carrying transgenic mice (Trp53 R172H/+, corresponding to human R175H/+). Further, upon co-culture with isolated NK cells, EMT6 cells substantively activated NK cells and proliferation thereof, increasing interferon-gamma (IFN-γ) production; however, SVTneg2 cells suppressed NK cell activation. Further mechanistic study elucidated that p53 can modulate expression by cancer cells of Mult-1 and H60a, which are activating and inhibitory ligands for NKG2D receptors of NK cells, respectively, to enhance immune surveillance against cancer. Our findings demonstrate that wt p53 is requisite for NK cell-based immune recognition and elimination of cancerous cells, and perhaps more importantly, that p53 missense mutant presence in cancer cells impairs NK cell-attributable responses, thus veiling cancerous cells from host immunity and enabling cancer progression.


Asunto(s)
Neoplasias de la Mama , Células Asesinas Naturales , Proteína p53 Supresora de Tumor , Animales , Neoplasias de la Mama/genética , Neoplasias de la Mama/inmunología , Femenino , Células Asesinas Naturales/metabolismo , Ratones , Ratones Transgénicos , Subfamilia K de Receptores Similares a Lectina de Células NK/genética , Subfamilia K de Receptores Similares a Lectina de Células NK/metabolismo , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo
2.
Prostate ; 77(8): 888-899, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28240383

RESUMEN

BACKGROUND: Chronic inflammation has been associated with the development and progression of human cancers including prostate cancer. The exact role of the inflammatory Th17-IL-17 pathway in prostate cancer remains unknown. In this study, we aimed to determine the importance of Th17 cells and IL-17 in a Pten-null prostate cancer mouse model. METHODS: The Pten-null mice were treated by Th17 inhibitor SR1001 or anti-mouse IL-17 monoclonal antibody from 6 weeks of age up to 12 weeks of age. For SR1001 treatment, the mice were injected intraperitoneally (i.p.) twice a day with vehicle or SR1001, which was dissolved in a dimethylsulfoxide (DMSO) solution. All mice were euthanized for necropsy at 12 weeks of age. For IL-17 antibody treatment, the mice were injected intravenously (i.v.) once every two weeks with control IgG or rat anti-mouse IL-17 monoclonal antibody, which was dissolved in PBS. The injection time points were at 6, 8, and 10 weeks old. All mice were analyzed for the prostate phenotypes at 12 weeks of age. RESULTS: We found that either SR1001 or anti-IL-17 antibody treatment decreased the formation of micro-invasive prostate cancer in Pten-null mice. The SR1001 or anti-IL-17 antibody treated mouse prostates had reduced proliferation, increased apoptosis, and reduced angiogenesis, as well as reduced inflammatory cell infiltration. By assessing the epithelial-to-mesenchymal transition (EMT) markers, we found that SR1001 or anti-IL-17 antibody treated prostate tissues had weaker EMT phenotype compared to the control treated prostates. CONCLUSIONS: These results demonstrated that Th17-IL-17 pathway plays a key role in prostate cancer progression in Pten-null mice. Targeting Th17-IL-17 pathway could prevent micro-invasive prostate cancer formation in mice. Prostate 77:888-899, 2017. © 2017 Wiley Periodicals, Inc.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Inflamación/inmunología , Interleucina-17 , Invasividad Neoplásica , Neoplasias de la Próstata , Sulfonamidas/farmacología , Células Th17/inmunología , Tiazoles/farmacología , Animales , Modelos Animales de Enfermedad , Monitoreo de Drogas/métodos , Factores Inmunológicos/farmacología , Inyecciones Intraperitoneales , Interleucina-17/antagonistas & inhibidores , Interleucina-17/inmunología , Masculino , Ratones , Terapia Molecular Dirigida/métodos , Invasividad Neoplásica/patología , Invasividad Neoplásica/prevención & control , Próstata/inmunología , Próstata/patología , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/inmunología , Neoplasias de la Próstata/patología , Neoplasias de la Próstata/fisiopatología , Resultado del Tratamiento
3.
J Immunol ; 194(8): 4019-28, 2015 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-25769926

RESUMEN

The symptoms of vaginal candidiasis exacerbate in the second half of the menstrual cycle in premenopausal women when the serum estradiol level is elevated. Estradiol has been shown to inhibit Th17 differentiation and production of antifungal IL-17 cytokines. However, little is known about the mechanisms. In the present study, we used mouse splenocytes and found that estradiol inhibited Th17 differentiation through downregulation of Rorγt mRNA and protein expression. Estradiol activated estrogen receptor (ER)α to recruit repressor of estrogen receptor activity (REA) and form the ERα/REA complex. This complex bound to three estrogen response element (ERE) half-sites on the Rorγt promoter region to suppress Rorγt expression. Estradiol induced Rea mRNA and protein expression in mouse splenocytes. Using Rea small interfering RNA to knock down Rea expression enhanced Rorγt expression and Th17 differentiation. Alternatively, histone deacetylase 1 and 2 bound to the three ERE half-sites, independent of estradiol. Histone deacetylase inhibitor MS-275 dose- and time-dependently increased Rorγt expression and subsequently enhanced Th17 differentiation. In 15 healthy premenopausal women, high serum estradiol levels are correlated with low RORγT mRNA levels and high REA mRNA levels in the vaginal lavage. These results demonstrate that estradiol upregulates REA expression and recruits REA via ERα to the EREs on the RORγT promoter region, thus inhibiting RORγT expression and Th17 differentiation. This study suggests that the estradiol/ERα/REA axis may be a feasible target in the management of recurrent vaginal candidiasis.


Asunto(s)
Diferenciación Celular/inmunología , Estradiol/inmunología , Receptor alfa de Estrógeno/inmunología , Complejos Multiproteicos/inmunología , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/inmunología , Proteínas Represoras/inmunología , Elementos de Respuesta/inmunología , Células Th17/inmunología , Transcripción Genética/inmunología , Adulto , Animales , Benzamidas/farmacología , Candidiasis/inmunología , Candidiasis/patología , Relación Dosis-Respuesta a Droga , Femenino , Inhibidores de Histona Desacetilasas/farmacología , Humanos , Ratones , Prohibitinas , Piridinas/farmacología , Células Th17/patología , Transcripción Genética/efectos de los fármacos , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/inmunología , Vagina/inmunología , Vagina/patología
4.
Int J Gynecol Cancer ; 27(2): 196-205, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27870715

RESUMEN

OBJECTIVE: Estrogen is a well-known oncogenic driver in endometrial (ECs) and breast cancers (BCs). Programmed cell death protein 1 (PD-1) and its ligands PD-1 Ligand 1 (PD-L1) and PD-L2 have been shown to mediate immune evasion of the tumor cells. The purpose of the present study was to assess the effects of estrogen on PD-L1 and PD-L2 expression in EC and BC cell lines. METHODS: 17ß-Estradiol (E2)-induced expression of PD-L1 and PD-L2 and possible signaling pathway were investigated in EC and BC cells. Coculture of T cells and cancer cells with E2 stimulation was performed to assess the functions of T cells. RESULTS: We found that E2 increased expression of PD-L1, but not PD-L2, protein via activation of phosphoinositide 3-kinase (PI3K)/Akt pathway in Ishikawa and Michigan Cancer Foundation-7 (MCF-7) cells. Phosphoinositide 3-kinase and Akt inhibitors could block E2's effects. 17ß-Estradiol did not increase PD-L1 mRNA transcription, but stabilized PD-L1 mRNA. 17ß-Estradiol's effects were only observed in estrogen receptor α (ERα)-positive Ishikawa and MCF-7 cells, but not in ERα-negative MDA-MB-231 cells. Coculture of Ishikawa or MCF-7 cells with T cells inhibited expression of interferon-γ and interleukin-2 and increased BCL-2-interacting mediator of cell death expression in the presence of E2. CONCLUSIONS: This study provides the first evidence that estrogen upregulates PD-L1 protein expression in ERα-positive EC and BC cells to suppress immune functions of T cells in the tumor microenvironment, demonstrating a new mechanism of how estrogen drives cancer progression.


Asunto(s)
Antígeno B7-H1/biosíntesis , Neoplasias de la Mama/metabolismo , Neoplasias Endometriales/metabolismo , Estradiol/farmacología , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Procesamiento Postranscripcional del ARN/efectos de los fármacos , Antígeno B7-H1/genética , Neoplasias de la Mama/enzimología , Neoplasias de la Mama/genética , Neoplasias de la Mama/inmunología , Línea Celular Tumoral , Neoplasias Endometriales/enzimología , Neoplasias Endometriales/genética , Neoplasias Endometriales/inmunología , Receptor alfa de Estrógeno/biosíntesis , Femenino , Humanos , Células Jurkat , Células MCF-7 , ARN Mensajero/genética , ARN Mensajero/metabolismo , Transducción de Señal/efectos de los fármacos , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología
5.
Prostate ; 76(15): 1420-30, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27325602

RESUMEN

BACKGROUND: Bone metastasis from primary prostate cancer leads to markedly diminished quality of life with poor long-term survival. Bone seeking treatment options are limited with adverse consequences on rapidly proliferating tissues such as bone marrow. In the present study, we seek to determine the bone-enriching capabilities of monomethyl auristatin E phosphate (MMAEp), a derivative of the potent antimitotic monomethyl auristatin E (MMAE). METHODS: The in vitro actions and mechanisms of cytotoxicity were assessed using cell viability, immunofluorescence, flow cytometry, and Western blot analysis. In vivo efficacy was determined using an intratibial xenograft mouse model of human prostate cancer and live animal imaging. RESULTS: The half maximal inhibitory concentration (IC50) of MMAE and MMAEp was determined to be approximately 2 and 48 nM, respectively, in PC-3 and C4-2B cell lines. MMAEp retained the mechanism of action of MMAE in reducing microtubule polymerization and stalling cell cycle progression at the G2/M transition. MMAEp was able to bind hydroxyapatite in in vitro assays. MMAEp significantly reduced intratibial tumor growth compared to the vehicle control treatment. CONCLUSIONS: MMAEp is an antimitotic compound that binds to calcium ions in the bone and inhibits prostate tumor growth in the bone. Prostate 76:1420-1430, 2016. © 2016 Wiley Periodicals, Inc.


Asunto(s)
Antimitóticos/farmacología , Neoplasias Óseas/tratamiento farmacológico , Proliferación Celular/efectos de los fármacos , Oligopéptidos/farmacología , Neoplasias de la Próstata/tratamiento farmacológico , Animales , Apoptosis/efectos de los fármacos , Neoplasias Óseas/secundario , Calcio/metabolismo , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Durapatita/metabolismo , Humanos , Iones/metabolismo , Masculino , Ratones , Microtúbulos/efectos de los fármacos , Fosfatos/farmacología , Neoplasias de la Próstata/patología , Tibia/patología , Moduladores de Tubulina/farmacología , Carga Tumoral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
6.
Int J Mol Sci ; 17(4): 522, 2016 Apr 07.
Artículo en Inglés | MEDLINE | ID: mdl-27070576

RESUMEN

The functions of interleukin-17A (IL-17A) in adipose tissues and adipocytes have not been well understood. In the present study, male mice were fed with a regular diet (n = 6, lean mice) or a high-fat diet (n = 6, obese mice) for 30 weeks. Subcutaneous adipose tissue (SAT) and visceral adipose tissue (VAT) were analyzed for IL-17A levels. SAT and VAT were treated with IL-17A and analyzed for inflammatory and metabolic gene expression. Mouse 3T3-L1 pre-adipocytes were differentiated into adipocytes, followed with IL-17A treatment and analysis for inflammatory and metabolic gene expression. We found that IL-17A levels were higher in obese SAT than lean SAT; the basal expression of inflammatory and metabolic genes was different between SAT and VAT and between lean and obese adipose tissues. IL-17A differentially induced expression of inflammatory and metabolic genes, such as tumor necrosis factor α, Il-6, Il-1ß, leptin, and glucose transporter 4, in adipose tissues of lean and obese mice. IL-17A also differentially induced expression of inflammatory and metabolic genes in pre-adipocytes and adipocytes, and IL-17A selectively activated signaling pathways in adipose tissues and adipocytes. These findings suggest that IL-17A differentially induces inflammatory and metabolic gene expression in the adipose tissues of lean and obese mice.


Asunto(s)
Tejido Adiposo/inmunología , Regulación de la Expresión Génica , Interleucina-17/inmunología , Obesidad/inmunología , Adipocitos/inmunología , Adipocitos/metabolismo , Adipocitos/patología , Adipoquinas/genética , Adipoquinas/inmunología , Adipoquinas/metabolismo , Tejido Adiposo/metabolismo , Tejido Adiposo/patología , Animales , Línea Celular , Dieta Alta en Grasa/efectos adversos , Interleucina-17/genética , Interleucina-17/metabolismo , Masculino , Ratones , Ratones Obesos , Obesidad/genética , Obesidad/metabolismo , Obesidad/patología , Transducción de Señal
7.
Prostate ; 75(8): 883-95, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25683512

RESUMEN

BACKGROUND: Extravasation is a critical step in cancer metastasis, in which adhesion of intravascular cancer cells to the vascular endothelial cells is controlled by cell surface adhesion molecules. The role of interleukin-17 (IL-17), insulin, and insulin-like growth factor 1 (IGF1) in adhesion of prostate cancer cells to the vascular endothelial cells is unknown, which is the subject of the present study. METHODS: Human umbilical vein endothelial cells (HUVECs) and human prostate cancer cell lines (PC-3, DU-145, LNCaP, and C4-2B) were analyzed for expression of vascular cell adhesion molecule 1 (VCAM-1), integrins, and cluster of differentiation 44 (CD44) using flow cytometry and Western blot analysis. The effects of IL-17, insulin, and IGF1 on VCAM-1 expression and adhesion of prostate cancer cells to HUVECs were examined. The interaction of VCAM-1 and CD44 was assessed using immunoprecipitation assays. RESULTS: Insulin and IGF1 acted with IL-17 to increase VCAM-1 expression in HUVECs. PC-3, DU-145, LNCaP, and C4-2B cells expressed ß1 integrin but not α4 integrin. CD44 was expressed by PC-3 and DU-145 cells but not by LNCaP or C4-2B cells. When HUVECs were treated with IL-17, insulin or IGF1, particularly with a combination of IL-17 and insulin (or IGF1), adhesion of PC-3 and DU-145 cells to HUVECs was significantly increased. In contrast, adhesion of LNCaP and C4-2B cells to HUVECs was not affected by treatment of HUVECs with IL-17 and/or insulin/IGF1. CD44 expressed in PC-3 cells physically bound to VCAM-1 expressed in HUVECs. CONCLUSIONS: CD44-VCAM-1 interaction mediates the adhesion between prostate cancer cells and HUVECs. IL-17 and insulin/IGF1 enhance adhesion of prostate cancer cells to vascular endothelial cells through increasing VCAM-1 expression in the vascular endothelial cells. These findings suggest that IL-17 may act with insulin/IGF1 to promote prostate cancer metastasis.


Asunto(s)
Células Endoteliales/metabolismo , Receptores de Hialuranos/metabolismo , Factor I del Crecimiento Similar a la Insulina/farmacología , Insulina/farmacología , Interleucina-17/farmacología , Neoplasias de la Próstata/metabolismo , Molécula 1 de Adhesión Celular Vascular/metabolismo , Adhesión Celular/efectos de los fármacos , Adhesión Celular/fisiología , Línea Celular Tumoral , Células Endoteliales de la Vena Umbilical Humana , Humanos , Masculino
8.
Int J Mol Sci ; 16(5): 11750-65, 2015 May 22.
Artículo en Inglés | MEDLINE | ID: mdl-26006246

RESUMEN

Aminomethylphosphonic acid (AMPA) and its parent compound herbicide glyphosate are analogs to glycine, which have been reported to inhibit proliferation and promote apoptosis of cancer cells, but not normal cells. Methoxyacetic acid (MAA) is the active metabolite of ester phthalates widely used in industry as gelling, viscosity and stabilizer; its exposure is associated with developmental and reproductive toxicities in both rodents and humans. MAA has been reported to suppress prostate cancer cell growth by inducing growth arrest and apoptosis. However, it is unknown whether AMPA and MAA can inhibit cancer cell growth. In this study, we found that AMPA and MAA inhibited cell growth in prostate cancer cell lines (LNCaP, C4-2B, PC-3 and DU-145) through induction of apoptosis and cell cycle arrest at the G1 phase. Importantly, the AMPA-induced apoptosis was potentiated with the addition of MAA, which was due to downregulation of the anti-apoptotic gene baculoviral inhibitor of apoptosis protein repeat containing 2 (BIRC2), leading to activation of caspases 7 and 3. These results demonstrate that the combination of AMPA and MAA can promote the apoptosis of prostate cancer cells, suggesting that they can be used as potential therapeutic drugs in the treatment of prostate cancer.


Asunto(s)
Acetatos/farmacología , Antineoplásicos/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Apoptosis/efectos de los fármacos , Organofosfonatos/farmacología , Próstata/efectos de los fármacos , Neoplasias de la Próstata/tratamiento farmacológico , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Fase G1/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Isoxazoles , Masculino , Próstata/metabolismo , Próstata/patología , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/patología , Tetrazoles
9.
Am J Physiol Lung Cell Mol Physiol ; 307(6): L497-508, 2014 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-25038189

RESUMEN

Recent findings demonstrate that inhaled cigarette smoke, the predominant lung carcinogen, elicits a T helper 17 (Th17) inflammatory phenotype. Interleukin-17A (IL-17), the hallmark cytokine of Th17 inflammation, displays pro- and antitumorigenic properties in a manner that varies according to tumor type and assay system. To investigate the role of IL-17 in lung tumor growth, we used an autochthonous tumor model (K-Ras(LA1) mice) with lung delivery of a recombinant adenovirus that expresses IL-17A. Virus-mediated expression of IL-17A in K-Ras(LA1) mice at 8-10 wk of age doubled lung tumor growth in 3 wk relative to littermates that received a green fluorescent protein-expressing control adenovirus. IL-17 induced matrix metalloproteinase-9 (MMP-9) expression in vivo and in vitro. In accord with this finding, selective and specific inhibitors of MMP-9 repressed the increased motility and invasiveness of IL-17-treated lung tumor cells in culture. Knockdown or mutation of p53 promoted the motility of murine lung tumor cells and abrogated the promigratory role of IL-17. Coexpression of siRNA-resistant wild-type, but not mutant, human p53 rescued both IL-17-mediated migration and MMP-9 mRNA induction in p53 knockdown lung tumor cells. IL-17 increased MMP-9 mRNA stability by reducing interaction with the mRNA destabilizing serine/arginine-rich splicing factor 1 (SRSF1). Taken together, our results indicate that IL-17 stimulates lung tumor growth and regulates MMP-9 mRNA levels in a p53- and SRSF1-dependent manner.


Asunto(s)
Movimiento Celular , Interleucina-17/biosíntesis , Neoplasias Pulmonares/metabolismo , Animales , Estabilidad de Enzimas/genética , Técnicas de Silenciamiento del Gen , Humanos , Interleucina-17/genética , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Metaloproteinasa 9 de la Matriz/biosíntesis , Metaloproteinasa 9 de la Matriz/genética , Ratones , Ratones Transgénicos , Invasividad Neoplásica , Proteínas Nucleares/biosíntesis , Proteínas Nucleares/genética , Proteínas Proto-Oncogénicas p21(ras)/genética , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Factores de Empalme de ARN , Proteínas de Unión al ARN/biosíntesis , Proteínas de Unión al ARN/genética , Factores de Empalme Serina-Arginina , Proteína p53 Supresora de Tumor/biosíntesis , Proteína p53 Supresora de Tumor/genética
10.
Prostate ; 74(8): 869-79, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24691769

RESUMEN

BACKGROUND: Interleukin-17 (IL-17) has been demonstrated to promote formation and growth of hormone-naïve prostate adenocarcinoma in mice. IL-17's role in development of castration-resistant prostate cancer is unknown. In the present study, we investigated IL-17's role in castration-resistant prostate cancer in a mouse model. METHODS: IL-17 receptor C (IL-17RC) deficient mice were interbred with Pten conditional mutant mice to produce RC(+) mice that maintained IL-17RC expression and RC(-) mice that were IL-17RC deficient. Male RC(+) and RC(-) mice were Pten-null and were castrated at 16 weeks of age when invasive prostate cancer had already formed. At 30 weeks of age, all male mice were analyzed for the prostate phenotypes. RESULTS: RC(-) mice displayed prostates that were smaller than RC(+) mice. Approximately 23% of prostatic glands in RC(-) mice, in contrast to 65% of prostatic glands in RC(+) mice, developed invasive adenocarcinomas. Compared to castrate RC(+) mice, castrate RC(-) mouse prostate had lower rates of cellular proliferation and higher rates of apoptosis as well as lower levels of MMP7, YBX1, MTA1, and UBE2C proteins. In addition, castrate RC(-) mouse prostate had less angiogenesis, which was associated with decreased levels of COX-2 and VEGF. Moreover, castrate RC(-) mouse prostate had fewer inflammatory cells including lymphocytes, myeloid-derived suppressor cells, and macrophages. CONCLUSIONS: Taken together, our findings suggest that IL-17 promotes development of invasive prostate adenocarcinomas under castrate conditions, potentially through creating an immunotolerant and pro-angiogenic tumor microenvironment.


Asunto(s)
Tolerancia Inmunológica , Interleucina-17/fisiología , Neovascularización Patológica/metabolismo , Neoplasias de la Próstata Resistentes a la Castración/metabolismo , Receptores de Interleucina-17/fisiología , Microambiente Tumoral/genética , Animales , Tolerancia Inmunológica/genética , Interleucina-17/deficiencia , Masculino , Ratones , Ratones Noqueados , Neovascularización Patológica/genética , Neovascularización Patológica/inmunología , Orquiectomía , Neoplasias de la Próstata Resistentes a la Castración/genética , Neoplasias de la Próstata Resistentes a la Castración/inmunología , Receptores de Interleucina-17/deficiencia
11.
Int J Mol Sci ; 15(4): 6941-60, 2014 Apr 22.
Artículo en Inglés | MEDLINE | ID: mdl-24758934

RESUMEN

Embryonic development of articular cartilage has not been well understood and the role of doublecortin (DCX) in determination of chondrocyte phenotype is unknown. Here, we use a DCX promoter-driven eGFP reporter mouse model to study the dynamic gene expression profiles in mouse embryonic handplates at E12.5 to E13.5 when the condensed mesenchymal cells differentiate into either endochondral chondrocytes or joint interzone cells. Illumina microarray analysis identified a variety of genes that were expressed differentially in the different regions of mouse handplate. The unique expression patterns of many genes were revealed. Cytl1 and 3110032G18RIK were highly expressed in the proximal region of E12.5 handplate and the carpal region of E13.5 handplate, whereas Olfr538, Kctd15, and Cited1 were highly expressed in the distal region of E12.5 and the metacarpal region of E13.5 handplates. There was an increasing gradient of Hrc expression in the proximal to distal direction in E13.5 handplate. Furthermore, when human DCX protein was expressed in human adipose stem cells, collagen II was decreased while aggrecan, matrilin 2, and GDF5 were increased during the 14-day pellet culture. These findings suggest that DCX may play a role in defining chondrocyte phenotype.


Asunto(s)
Condrocitos/metabolismo , Proteínas Asociadas a Microtúbulos/metabolismo , Neuropéptidos/metabolismo , Animales , Diferenciación Celular , Células Cultivadas , Condrocitos/citología , Proteínas de Dominio Doblecortina , Proteína Doblecortina , Embrión de Mamíferos/metabolismo , Desarrollo Embrionario , Genes Reporteros , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Células Madre Mesenquimatosas/citología , Ratones , Proteínas Asociadas a Microtúbulos/genética , Neuropéptidos/genética , Fenotipo , Transcriptoma
12.
Biomedicines ; 12(4)2024 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-38672111

RESUMEN

Interleukin-17 (IL-17) is a pro-inflammatory cytokine that participates in innate and adaptive immune responses and plays an important role in host defense, autoimmune diseases, tissue regeneration, metabolic regulation, and tumor progression. Post-translational modifications (PTMs) are crucial for protein function, stability, cellular localization, cellular transduction, and cell death. However, PTMs of IL-17 receptor A (IL-17RA) have not been investigated. Here, we show that human IL-17RA was targeted by F-box and WD repeat domain-containing 11 (FBXW11) for ubiquitination, followed by proteasome-mediated degradation. We used bioinformatics tools and biochemical techniques to determine that FBXW11 ubiquitinated IL-17RA through a lysine 27-linked polyubiquitin chain, targeting IL-17RA for proteasomal degradation. Domain 665-804 of IL-17RA was critical for interaction with FBXW11 and subsequent ubiquitination. Our study demonstrates that FBXW11 regulates IL-17 signaling pathways at the IL-17RA level.

13.
J Natl Cancer Inst ; 116(3): 421-433, 2024 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-37847647

RESUMEN

BACKGROUND: Although the fusion of the transmembrane serine protease 2 gene (TMPRSS2) with the erythroblast transformation-specific-related gene (ERG), or TMPRSS2-ERG, occurs frequently in prostate cancer, its impact on clinical outcomes remains controversial. Roughly half of TMPRSS2-ERG fusions occur through intrachromosomal deletion of interstitial genes and the remainder via insertional chromosomal rearrangements. Because prostate cancers with deletion-derived TMPRSS2-ERG fusions are more aggressive than those with insertional fusions, we investigated the impact of interstitial gene loss on prostate cancer progression. METHODS: We conducted an unbiased analysis of transcriptome data from large collections of prostate cancer samples and employed diverse in vitro and in vivo models combined with genetic approaches to characterize the interstitial gene loss that imposes the most important impact on clinical outcome. RESULTS: This analysis identified FAM3B as the top-ranked interstitial gene whose loss is associated with a poor prognosis. The association between FAM3B loss and poor clinical outcome extended to fusion-negative prostate cancers where FAM3B downregulation occurred through epigenetic imprinting. Importantly, FAM3B loss drives disease progression in prostate cancer. FAM3B acts as an intermediator of a self-governing androgen receptor feedback loop. Specifically, androgen receptor upregulates FAM3B expression by binding to an intronic enhancer to induce an enhancer RNA and facilitate enhancer-promoter looping. FAM3B, in turn, attenuates androgen receptor signaling. CONCLUSION: Loss of FAM3B in prostate cancer, whether through the TMPRSS2-ERG translocation or epigenetic imprinting, causes an exit from this autoregulatory loop to unleash androgen receptor activity and prostate cancer progression. These findings establish FAM3B loss as a new driver of prostate cancer progression and support the utility of FAM3B loss as a biomarker to better define aggressive prostate cancer.


Asunto(s)
Neoplasias de la Próstata , Receptores Androgénicos , Masculino , Humanos , Receptores Androgénicos/genética , Retroalimentación , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/metabolismo , Transcriptoma , Proteínas de Fusión Oncogénica/genética , Regulador Transcripcional ERG/genética , Regulador Transcripcional ERG/metabolismo , Proteínas de Neoplasias/genética , Citocinas/genética
14.
Cancer Cell Int ; 13(1): 16, 2013 Feb 14.
Artículo en Inglés | MEDLINE | ID: mdl-23409704

RESUMEN

A fribotic tumor microenvironment promotes progression of cancer. In this study, we utilize a reconstituted basement membrane mimics Matrigel based three-dimensional organotypic culture (rBM 3-D) to investigate the mechanisms that mediate the tumor promoting effects of the fibrogenic mediators TGF-ß1 and type I collagen (Col-1) on lung adenocarcinoma cells. Similar to normal alveolar epithelial cells, the well-differentiated lung adenocarcinoma cells in rBM 3-D culture undergo acinar morphogeneis that features polarized epithelial cell spheres with a single central lumen. Either TGF-ß1 or Col-1 modestly distorts acinar morphogenesis. On the other hand, TGF-ß1 and Col-1 synergistically induce a transition from acinar morphology into stellate morphology that is characteristic of invasive and metastatic cancer cells. Inhibition of the Src kinase activity abrogates induction of stellate morphology, activation of Akt and mTOR, and the expression of tumor promoting genes by TGF-ß1 and Col-1. To a similar extent, pharmacological inhibition of mTOR abrogates the cellular responses to TGF-ß1 and Col-1. In summary, we demonstrate that TGF-ß1 and Col-1 promote stellate morphogenesis of lung cancer cells. Our findings further suggest that the Src-Akt-mTOR axis mediates stellate morphogenesis. These findings also indicate that rBM 3-D culture can serve as an ideal platform for swift and cost-effective screening of therapeutic candidates at the interface of the tumor and its microenvironment.

15.
J Pineal Res ; 55(4): 377-87, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24033914

RESUMEN

Obesity is a chronic inflammation with increased serum levels of insulin, insulin-like growth factor 1 (IGF1), and interleukin-17 (IL-17). The objective of this study was to test a hypothesis that insulin and IGF1 enhance IL-17-induced expression of inflammatory chemokines/cytokines through a glycogen synthase kinase 3ß (GSK3B)-dependent mechanism, which can be inhibited by melatonin. We found that insulin/IGF1 and lithium chloride enhanced IL-17-induced expression of C-X-C motif ligand 1 (Cxcl1) and C-C motif ligand 20 (Ccl20) in the Gsk3b(+/+) , but not in Gsk3b(-/-) mouse embryonic fibroblast (MEF) cells. IL-17 induced higher levels of Cxcl1 and Ccl20 in the Gsk3b(-/-) MEF cells, compared with the Gsk3b(+/+) MEF cells. Insulin and IGF1 activated Akt to phosphorylate GSK3B at serine 9, thus inhibiting GSK3B activity. Melatonin inhibited Akt activation, thus decreasing P-GSK3B at serine 9 (i.e., increasing GSK3B activity) and subsequently inhibiting expression of Cxcl1 and Ccl20 that was induced either by IL-17 alone or by a combination of insulin and IL-17. Melatonin's inhibitory effects were only observed in the Gsk3b(+/+) , but in not Gsk3b(-/-) MEF cells. Melatonin also inhibited expression of Cxcl1, Ccl20, and Il-6 that was induced by a combination of insulin and IL-17 in the mouse prostatic tissues. Further, nighttime human blood, which contained high physiologic levels of melatonin, decreased expression of Cxcl1, Ccl20, and Il-6 in the PC3 human prostate cancer xenograft tumors. Our data support our hypothesis and suggest that melatonin may be used to dampen IL-17-mediated inflammation that is enhanced by the increased levels of insulin and IGF1 in obesity.


Asunto(s)
Glucógeno Sintasa Quinasa 3/metabolismo , Factor I del Crecimiento Similar a la Insulina/farmacología , Insulina/farmacología , Interleucina-17/metabolismo , Animales , Western Blotting , Línea Celular Tumoral , Glucógeno Sintasa Quinasa 3/genética , Glucógeno Sintasa Quinasa 3 beta , Humanos , Insulina/metabolismo , Factor I del Crecimiento Similar a la Insulina/metabolismo , Cloruro de Litio/farmacología , Melatonina/metabolismo , Melatonina/farmacología , Ratones , Ratones Noqueados , Fosforilación/efectos de los fármacos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal
16.
Arthroscopy ; 29(9): 1559-65, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23910000

RESUMEN

PURPOSE: Our goals were (1) to characterize metal microparticles created by standard arthroscopic instruments and (2) to examine the in vitro cellular responses induced by those particles, including possible synergistic effects with local anesthetic. METHODS: We applied standard surgical tools to 16 foam bone blocks immersed in saline solution (plus 3 non-instrumented controls). Eight specimens underwent 4 minutes of exposure to a 4.0-mm full-radius shaver rotating forward at 6,000 rpm. In the other blocks, 4 holes were created with a 3.0-mm drill through a sleeve. Particles were isolated onto silicon wafers by density gradient ultra-centrifugation, and scanning electron microscopy was used to analyze a minimum of 1,000 particles per wafer. Metal particles were then isolated and purified. Aliquots of sterilized micro-particles were applied to cultured bovine chondrocytes (with or without local anesthetic) and to cultured human or bovine synoviocytes. Chondrocyte viability was assessed with live/dead cell assay by flow cytometry. Synoviocyte responses were assessed with quantitative polymerase chain reaction. RESULTS: Stainless steel or aluminum particles were found in each sample (the same composition as surgical instruments). The mean particle size was 1 to 2 µm (range, 50 nm to 20 µm). Chondrocyte exposure (1 hour) to metal debris induced a small but statistically significant increase in cell death, without any synergistic effect of local anesthetic. Proinflammatory chemokines were consistently upregulated in both human and bovine synoviocytes exposed to metallic microparticles for 3, 24, and 48 hours. CONCLUSIONS: This study shows that metallic microdebris is liberated by common arthroscopic instruments, at scales much smaller than previously recognized. These particles are bioactive, as shown by the in vitro synoviocyte responses initiated by metallic microparticles. CLINICAL RELEVANCE: Our findings suggest that metallic microparticles could induce intra-articular damage through a synoviocyte-mediated cytokine response if their concentrations reach clinically significant levels.


Asunto(s)
Artroscopía/instrumentación , Condrocitos , Cuerpos Extraños/etiología , Articulaciones/lesiones , Metales , Tamaño de la Partícula , Membrana Sinovial/citología , Anestésicos Locales/farmacología , Animales , Bovinos , Muerte Celular , Supervivencia Celular , Centrifugación por Gradiente de Densidad/métodos , Condrocitos/efectos de los fármacos , Humanos , Microscopía Electrónica de Rastreo
17.
Lupus Sci Med ; 10(1)2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-37041033

RESUMEN

OBJECTIVE: SLE is an autoimmune disease characterised by persistent inflammation and autoantibody production. Genetic predisposition and environmental factors such as a high-fat diet (HFD) may contribute to lupus development. However, the immune cell profile and gender difference in response to HFD in lupus have not been reported. Here we investigated the impact of HFD on lupus pathogenesis and autoimmunity using lupus-prone mice. METHODS: Thirty male and 30 female MRL/lymphoproliferation (lpr) mice were fed with regular diet (RD) or HFD. Body weights were recorded weekly. SLE progression was monitored by skin lesion, urine protein, titres of antidouble-strand DNA (dsDNA) and ANA. At week 14, kidney and skin tissue sections were stained with H&E and periodic acid-Schiff to detect histological kidney index and skin score. Splenocytes were identified by immunofluorescence staining and flow cytometry. RESULTS: HFD significantly increased body weight and lipid levels compared with RD (p<0.01). Skin lesions were observed in 55.6% of the HFD group compared with 11.1% of the RD group, with greater histopathological skin scores in the female HFD group (p<0.01). Although both male and female mice had higher serum IgG in the HFD group than in the RD group, only the male HFD group showed an increased trend in anti-dsDNA Ab and ANA titres. Kidney pathological changes in the HFD group were more severe in male mice than in female mice (p<0.05), detected by proteinuria, kidney index and glomerular cell proliferation. Significant increases of germinal centre B cells and T follicular helper cells were observed in the spleens of HFD mice (p<0.05). CONCLUSION: HFD induced an accelerated and exacerbated lupus development and autoimmunity in MRL/lpr mice. Our results parallel many known clinical lupus phenotypes and sexual dimorphism in which male patients are likelier to have a severe disease (nephritis) than female lupus patients who may have a broader range of lupus symptoms.


Asunto(s)
Lupus Eritematoso Sistémico , Nefritis Lúpica , Masculino , Femenino , Humanos , Animales , Ratones , Autoinmunidad , Dieta Alta en Grasa , Ratones Endogámicos MRL lpr , Obesidad
18.
Prostate ; 72(12): 1306-16, 2012 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-22213096

RESUMEN

BACKGROUND: Neuroendocrine differentiation (NED) is one of the mechanisms underlying development of castration-resistant prostate cancer (CRPC). In this study, we investigated IL-6-induced NED in two LNCaP sublines. METHODS: LNCaP-S17, an LNCaP subline that secretes IL-6, and LNCaP-C3, a control subline that does not express IL-6, were analyzed for IL-6-induced NED, activation of JAK2 and STAT3 pathways, and expression of IL-6/IL-6R signaling proteins and downstream target genes. RESULTS: IL-6 did not induce NED in LNCaP-S17 cells, even though IL-6 induced NED in LNCaP-C3 cells. IL-6 activated JAK2 and STAT3 pathways in LNCaP-C3 cells but not in LNCaP-S17 cells. IL-6 did not activate ERK1/2, AKT, or NF-κB pathways in either cell line. Both LNCaP-C3 and LNCaP-S17 cell lines expressed IL-6R, gp130, and TYK2 at almost the same levels and did not express JAK1 or JAK3. The basal level of JAK2 expression was slightly higher in LNCaP-C3 cells than in LNCaP-S17 cells. Two suppressors of cytokine signaling, SOCS7 and cytokine-inducible SH2 protein (CIS), were expressed constitutively at higher levels in LNCaP-S17 cells than in LNCaP-C3 cells, while SOCS1 to SOCS6 were expressed at approximately the same levels. Using siRNA to knockdown SOCS7 and CIS expression in LNCaP-S17 cells led to increased phosphorylation of STAT3 upon IL-6 stimulation. CONCLUSIONS: LNCaP-S17 cells are resistant to exogenous IL-6-induced NED due to increased levels of CIS/SOCS7 that block activation of JAK2-STAT3 pathways.


Asunto(s)
Comunicación Autocrina/fisiología , Diferenciación Celular/fisiología , Interleucina-6/biosíntesis , Células Neuroendocrinas/metabolismo , Células Neuroendocrinas/patología , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Comunicación Autocrina/genética , Diferenciación Celular/genética , Línea Celular Tumoral , Técnicas de Cocultivo , Regulación Neoplásica de la Expresión Génica , Humanos , Interleucina-6/genética , Janus Quinasa 2/antagonistas & inhibidores , Janus Quinasa 2/biosíntesis , Masculino , Proteínas Nucleares/biosíntesis , Factor de Transcripción STAT3/antagonistas & inhibidores , Factor de Transcripción STAT3/biosíntesis , Proteínas Supresoras de la Señalización de Citocinas/biosíntesis , Regulación hacia Arriba/genética , Regulación hacia Arriba/fisiología
19.
Arthroscopy ; 28(3): 397-404, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22169763

RESUMEN

PURPOSE: The purpose of this study was to determine the effects of temperature or 0.25% bupivacaine treatment in combination with supraphysiologic temperatures on chondrocyte viability. METHODS: Bovine articular chondrocytes in suspension culture were treated with phosphate-buffered saline solution at 20°C, 37°C, 40°C, 42°C, 45°C, 47°C, and 50°C for 15, 30, and 60 minutes or with phosphate-buffered saline solution at 37°C, 45°C, and 50°C for 30 and 60 minutes followed by 0.25% bupivacaine at 20°C for 60 minutes. Chondrocyte viability was analyzed by flow cytometry with the LIVE/DEAD Viability/Cytotoxicity Kit (Molecular Probes, Eugene, OR). Annexin V and ethidium double staining determined whether apoptosis or necrosis occurred. RESULTS: Temperatures from 20°C to 42°C did not cause chondrocyte death. Temperatures at or above 45°C caused significant chondrocyte death, particularly at 50°C for 60 minutes, compared with 37°C at 60 minutes (P < .01). When the chondrocytes were incubated at 50°C, subsequent exposure to bupivacaine significantly increased chondrocyte death compared with the saline solution-treated control group (P < .001). There were additive cytotoxic effects when bupivacaine was combined with supraphysiologic temperatures. It was also found that bupivacaine at supraphysiologic temperatures caused necrosis of articular chondrocytes. CONCLUSIONS: Temperatures at or above 45°C caused significant chondrocyte death. Bupivacaine treatment in the presence of 45°C and 50°C temperatures significantly increased necrosis of bovine articular chondrocytes in this in vitro study. CLINICAL RELEVANCE: Immediate intra-articular injection of bupivacaine after heat-generating procedures may cause damage to the cartilage because of the additive cytotoxic effects of bupivacaine and elevated temperature.


Asunto(s)
Anestésicos Locales/efectos adversos , Bupivacaína/efectos adversos , Cartílago Articular/efectos de los fármacos , Condrocitos/efectos de los fármacos , Calor/efectos adversos , Anestésicos Locales/administración & dosificación , Animales , Apoptosis/efectos de los fármacos , Bupivacaína/administración & dosificación , Cartílago Articular/citología , Bovinos , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Citometría de Flujo , Necrosis/etiología
20.
J Shoulder Elbow Surg ; 21(12): 1651-5, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22743071

RESUMEN

PURPOSE: Lateral elbow pain has multiple etiologies; most common is lateral epicondylitis. Radio-capitellar arthritis, posterolateral rotatory instability (PLRI), plica and radial tunnel syndromes may produce similar pain. The purpose of this study is to report on a rare subset of patients who had an acute injury during treatment for chronic lateral epicondylitis, exacerbating symptoms and lessening function. Indications for surgery were a failure of another round of nonoperative management and diagnosis of a new injury to the lateral ligaments in addition to the lateral epicondylitis. Surgical intervention revealed the acute injury to the radial ulno-humeral ligament (RUHL) complex, superimposed on chronic lateral epicondylitis, which we believe caused worsening of symptoms. Surgical repair of both lesions provided satisfactory results. MATERIALS AND METHODS: Seven patients (range, 29-46 years; mean, 40.7) being treated for chronic lateral epicondylitis each sustained an acute elbow injury resulting in PLRI. Study data, including Andrews-Carson Elbow Score (ACES) and Mayo Elbow Performance Score (MEPS), were collected in the initial evaluation and at regular postoperative intervals, with a follow-up period of 12-24 months. Indications for surgery were pain, functional impairment, and failure of other treatments. All surgeries were performed on an outpatient basis under general anesthesia in the prone position. RESULTS: All patients showed arthroscopic evidence of chronic lateral epicondylitis and acute RUHL injury. All showed significant improvement in total ACES and MEPS after repair of both lesions, full range of motion, and objective improvement in strength and function, with no adverse effects or complications. CONCLUSION: Patients with chronic lateral epicondylitis who sustain an acute injury may damage the RUHL complex. Early recognition of this additional injury may allow surgical repair of both injuries with satisfactory results.


Asunto(s)
Artroscopía/métodos , Ligamentos Colaterales/lesiones , Lesiones de Codo , Traumatismos de la Mano/complicaciones , Codo de Tenista/complicaciones , Enfermedad Aguda , Adulto , Enfermedad Crónica , Ligamentos Colaterales/cirugía , Articulación del Codo/cirugía , Femenino , Estudios de Seguimiento , Traumatismos de la Mano/diagnóstico , Traumatismos de la Mano/cirugía , Humanos , Masculino , Persona de Mediana Edad , Radio (Anatomía) , Estudios Retrospectivos , Codo de Tenista/diagnóstico , Resultado del Tratamiento , Cúbito
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA