Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
Invest Ophthalmol Vis Sci ; 64(13): 36, 2023 10 03.
Artículo en Inglés | MEDLINE | ID: mdl-37870848

RESUMEN

Purpose: The purpose of this study was to critically test the hypothesis that mitochondrial pyruvate carrier (MPC) function is essential for maintenance of the corneal myofibroblast phenotype in vitro and in vivo. Methods: Protein and mRNA for canonical profibrotic markers were assessed in cultured cat corneal myofibroblasts generated via transforming growth factor (TGF)-ß1 stimulation and treated with either the thiazolidinedione (TZD) troglitazone or the MPC inhibitor alpha-cyano-beta-(1-phenylindol-3-yl) acrylate (UK-5099). RNA sequencing was used to gain insight into signaling modules related to instructive, permissive, or corollary changes in gene expression following treatment. A feline photorefractive keratectomy (PRK) model of corneal wounding was used to test the efficacy of topical troglitazone at reducing α-smooth muscle actin (SMA)-positive staining when applied 2 to 4 weeks postoperatively, during peak fibrosis. Results: Troglitazone caused cultured myofibroblasts to adopt a fibroblast-like phenotype through a noncanonical, peroxisome proliferator-activated receptor (PPAR)-γ-independent mechanism. Direct MPC inhibition using UK-5099 recapitulated this effect, but classic inhibitors of oxidative phosphorylation (OXPHOS) did not. Gene Set Enrichment Analysis (GSEA) of RNA sequencing data converged on energy substrate utilization and the Mitochondrial Permeability Transition pore as key players in myofibroblast maintenance. Finally, troglitazone applied onto an established zone of active fibrosis post-PRK significantly reduced stromal α-SMA expression. Conclusions: Our results provide empirical evidence that metabolic remodeling in myofibroblasts creates selective vulnerabilities beyond simply mitochondrial energy production, and that these are critical for maintenance of the myofibroblast phenotype. For the first time, we provide proof-of-concept data showing that this remodeling can be exploited to treat existing corneal fibrosis via inhibition of the MPC.


Asunto(s)
Fibroblastos , Miofibroblastos , Animales , Gatos , Miofibroblastos/patología , Troglitazona/farmacología , Fibroblastos/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Fibrosis , PPAR gamma/genética , PPAR gamma/metabolismo , Fenotipo , Piruvatos/metabolismo , Actinas/metabolismo , Células Cultivadas
2.
Toxicol Lett ; 375: 8-20, 2023 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-36596350

RESUMEN

The role of peroxisome proliferator activated receptor gamma (PPARγ) in the regulation of adipocyte differentiation has been well characterized. Besides adipose tissue, PPARγ is also highly expressed in the intestine. However, the functional role of PPARγ in the regulation of intestinal function still remains poorly understood. In the present study, we sought to understand the role of PPARγ activation on regulation of intestinal barrier function in intestinal porcine epithelial cells (IPEC-J2) and weaned piglets exposed to the mycotoxin, deoxynivalenol (DON). PPARγ activation by rosiglitazone and troglitazone, two pharmacological PPARγ ligands, increased the protein expression of tight junction proteins (TJP), claudin-3 and 4. PPARγ inhibition increased endocytosis of claudin-4 which was reversed by its activation with troglitazone. DON exposure decreased the protein expression of TJP, and also significantly suppressed PPARγ transcriptional activity. Interestingly, PPARγ activation reversed the reduction of claudin-3 and 4 caused by DON in vitro and in vivo. PPARγ activation also partially restored the transepithelial electrical resistance (TEER) and reduced the permeability of fluorescein isothiocyanate-dextran (FITC-dextran) that have been negatively impacted by DON. These effects were lost in the presence of a specific PPARγ antagonist or in PPARγ knockout cells, confirming the importance of PPARγ in the regulation of intestinal barrier function and integrity. Likewise, in weaned pigs exposed to DON, the PPARγ agonist pioglitazone mitigated the impaired villus-crypt morphology caused by DON. Therefore, pharmacological and natural bioactive compounds with PPARγ stimulatory activities could be effective in preventing DON-induced gut barrier dysfunction.


Asunto(s)
Enfermedades Intestinales , PPAR gamma , Porcinos , Animales , PPAR gamma/genética , PPAR gamma/metabolismo , Claudina-4/genética , Claudina-4/metabolismo , Claudina-3/metabolismo , Troglitazona/farmacología , Uniones Estrechas , Células Epiteliales , Mucosa Intestinal/metabolismo , Proteínas de Uniones Estrechas/metabolismo , Endocitosis
3.
Biol Pharm Bull ; 45(3): 276-283, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35228393

RESUMEN

Lung cancer is the leading cause of cancer-related deaths worldwide. Troglitazone (TGZ), a peroxisome proliferator-activated receptor gamma (PPARγ) ligand, is a potential antitumor agent. However, the action mechanism of TGZ in lung adenocarcinoma cells has not been completely elucidated. To assess this mechanism and the anticancer effects of TGZ in human lung adenocarcinoma cell lines (A549 and H1975), we investigated the involvement of PPARγ, apoptosis, the mitogen-activated protein kinase (MAPK) pathway, protein kinase B (Akt)/mammalian target of rapamycin (mTOR) pathway, and autophagy. Cell viability was measured using fluorescence-based assays. Apoptotic cells were detected by Hoechst 33342 and Annexin V-fluorescein isothiocyanate (FITC)/propidium iodide (PI) double staining; protein expression was detected by Western blotting. TGZ inhibited cell proliferation in a dose-dependent manner in both cell lines, and the effect was not suppressed by a PPARγ inhibitor. Additionally, TGZ increased apoptotic cell number and upregulated p38 and c-Jun N-terminal kinase (JNK) phosphorylation; however, p38 and JNK inhibitors did not block TGZ-mediated inhibition of cell proliferation in either cell line. TGZ also upregulated extracellular signal-regulated kinase 1/2 (ERK1/2) phosphorylation, whereas an ERK1/2 inhibitor enhanced TGZ-mediated cytotoxicity in A549 cells. Additionally, TGZ increased LC3-II expression, and chloroquine (an autophagy inhibitor) attenuated TGZ-mediated inhibition of cell proliferation. These findings suggest that TGZ-induced inhibition of cell proliferation is PPARγ independent. TGZ-mediated inhibition of cell proliferation was accompanied by apoptosis and independent of the MAPK signaling pathway. These results suggest that TGZ inhibits cell proliferation through autophagy-induced cytotoxicity. This study demonstrated that chemotherapy using TGZ may be effective for lung adenocarcinoma.


Asunto(s)
Adenocarcinoma del Pulmón , Tiazolidinedionas , Adenocarcinoma del Pulmón/tratamiento farmacológico , Apoptosis , Autofagia , Línea Celular , Línea Celular Tumoral , Proliferación Celular , Cromanos/farmacología , Humanos , Tiazolidinedionas/farmacología , Troglitazona/farmacología
4.
J Virol ; 95(24): e0093821, 2021 11 23.
Artículo en Inglés | MEDLINE | ID: mdl-34613794

RESUMEN

Sodium taurocholate cotransporting polypeptide (NTCP) is a receptor that is essential for hepatitis B virus (HBV) entry into the host cell. A number of HBV entry inhibitors targeting NTCP have been reported to date; these inhibitors have facilitated a mechanistic analysis of the viral entry process. However, the mechanism of HBV internalization into host cells after interaction of virus with NTCP remains largely unknown. Recently, we reported that troglitazone, a thiazolidinedione derivative, specifically inhibits both HBV internalization and NTCP oligomerization, resulting in inhibition of HBV infection. Here, using troglitazone as a chemical probe to investigate entry process, the contribution of NTCP oligomerization to HBV internalization was evaluated. Using surface plasmon resonance and transporter kinetics, we found that troglitazone directly interacts with NTCP and noncompetitively interferes with NTCP-mediated bile acid uptake, suggesting that troglitazone allosterically binds to NTCP, rather than to the bile acid-binding pocket. Additionally, alanine scanning mutagenesis showed that a mutation at phenylalanine 274 of NTCP (F274A) caused a loss of HBV susceptibility and disrupted both the oligomerization of NTCP and HBV internalization without affecting viral attachment to the cell surface. An inhibitor of the interaction between NTCP and epidermal growth factor receptor (EGFR), another host cofactor essential for HBV internalization, impeded NTCP oligomerization. Meanwhile, coimmunoprecipitation analysis revealed that neither troglitazone nor the F274A mutation in NTCP affects the NTCP-EGFR interaction. These findings suggest that NTCP oligomerization is initiated downstream of the NTCP-EGFR interaction and then triggers HBV internalization. This study provides significant insight into the HBV entry mechanisms. IMPORTANCE Hepatitis B virus (HBV) infection is mediated by a specific interaction with sodium taurocholate cotransporting polypeptide (NTCP), a viral entry receptor. Although the virus-receptor interactions are believed to trigger viral internalization into host cells, the exact molecular mechanisms of HBV internalization are not understood. In this study, we revealed the mode of action whereby troglitazone, a specific inhibitor of HBV internalization, impedes NTCP oligomerization and identified NTCP phenylalanine 274 as a residue essential for this oligomerization. We further analyzed the association between NTCP oligomerization and HBV internalization, a process that is mediated by epidermal growth factor receptor (EGFR), another essential host cofactor for HBV internalization. Our study provides critical information on the mechanism of HBV entry and suggests that oligomerization of the viral receptor serves as an attractive target for drug discovery.


Asunto(s)
Virus de la Hepatitis B/fisiología , Transportadores de Anión Orgánico Sodio-Dependiente/metabolismo , Multimerización de Proteína , Receptores Virales/metabolismo , Simportadores/metabolismo , Internalización del Virus/efectos de los fármacos , Transporte Biológico , Receptores ErbB/genética , Receptores ErbB/metabolismo , Células Hep G2 , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Hepatocitos/virología , Humanos , Transportadores de Anión Orgánico Sodio-Dependiente/genética , Simportadores/genética , Troglitazona/farmacología , Acoplamiento Viral/efectos de los fármacos
5.
Cancer Lett ; 519: 289-303, 2021 10 28.
Artículo en Inglés | MEDLINE | ID: mdl-34302921

RESUMEN

CD73, a cell surface-localized ecto-5'-nucleotidase, is the major enzymatic source of extracellular adenosine. Canonically, it plays multiple roles in cancer-related processes via its metabolite. As a druggable target, clinical trials targeting CD73 in various malignant diseases are currently ongoing. Here, we report the ecto-5'-nucleotidase-independent functions of CD73 in pancreatic ductal adenocarcinoma (PDAC). Our findings support that the elevated expression of CD73 in PDAC cells promotes gemcitabine (GEM) resistance by activating AKT. We discovered that a large amount of intracellular CD73 are localized in the endoplasmic reticulum membrane. Intracellular CD73 physically interacts with major vault protein to activate the SRC-AKT circuit. Troglitazone (TGZ) is a peroxisome proliferator-activated receptor gamma agonist that could inhibit the expression of CD73. The administration of TGZ markedly enhances sensitivity to GEM via downregulating CD73 in PDAC. Our findings support that CD73 could be targeted to overcome chemoresistance in PDAC.


Asunto(s)
5'-Nucleotidasa/genética , Carcinoma Ductal Pancreático/tratamiento farmacológico , Carcinoma Ductal Pancreático/genética , Desoxicitidina/análogos & derivados , Resistencia a Antineoplásicos/genética , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/genética , Animales , Línea Celular Tumoral , Desoxicitidina/farmacología , Regulación hacia Abajo/genética , Retículo Endoplásmico/genética , Femenino , Proteínas Ligadas a GPI/genética , Regulación Neoplásica de la Expresión Génica/genética , Células HEK293 , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Troglitazona/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto/métodos , Gemcitabina
6.
Mol Med Rep ; 23(6)2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33846779

RESUMEN

Prion diseases, which involve the alteration of cellular prion protein into a misfolded isoform, disrupt the central nervous systems of humans and animals alike. Prior research has suggested that peroxisome proliferator­activator receptor (PPAR)γ and autophagy provide some protection against neurodegeneration. PPARs are critical to lipid metabolism regulation and autophagy is one of the main cellular mechanisms by which cell function and homeostasis is maintained. The present study examined the effect of troglitazone, a PPARγ agonist, on autophagy flux in a prion peptide (PrP) (106­126)­mediated neurodegeneration model. Western blot analysis confirmed that treatment with troglitazone increased LC3­II and p62 protein expression, whereas an excessive increase in autophagosomes was verified by transmission electron microscopy. Troglitazone weakened PrP (106­126)­mediated neurotoxicity via PPARγ activation and autophagy flux inhibition. A PPARγ antagonist blocked PPARγ activation as well as the neuroprotective effects induced by troglitazone treatment, indicating that PPARγ deactivation impaired troglitazone­mediated protective effects. In conclusion, the present study demonstrated that troglitazone protected primary neuronal cells against PrP (106­126)­induced neuronal cell death by inhibiting autophagic flux and activating PPARγ signals. These results suggested that troglitazone may be a useful therapeutic agent for the treatment of neurodegenerative disorders and prion diseases.


Asunto(s)
Autofagia/efectos de los fármacos , Hipoglucemiantes/farmacología , Neuronas/metabolismo , PPAR gamma/metabolismo , Fragmentos de Péptidos/efectos adversos , Priones/efectos adversos , Troglitazona/farmacología , Animales , Proteína 5 Relacionada con la Autofagia/genética , Línea Celular , Humanos , Ratones , Ratones Endogámicos ICR , Neuronas/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , PPAR gamma/agonistas , Proteínas Priónicas
7.
Biol Pharm Bull ; 43(9): 1375-1381, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32879212

RESUMEN

Adipogenic differentiation is a complex process by which fibroblast-like undifferentiated cells are converted into cells that accumulate lipid droplets. We here investigated the effect of gene deletion of calcium-independent phospholipase A2γ (iPLA2γ), a membrane-bound PLA2 enzyme, on adipogenic differentiation in mice. Since iPLA2γ knockout (KO) mice showed reduced fat volume and weight, we prepared mouse embryonic fibroblasts (MEF) from wild-type (WT) and iPLA2γ KO mice and examined the effect of iPLA2γ deletion on in vitro adipogenic differentiation. iPLA2γ increased during adipogenic differentiation in WT mouse-derived MEFs, and the differentiation was partially abolished in iPLA2γ KO-derived MEFs. In KO-derived MEFs, the inductions of peroxisome proliferator activator receptor γ (PPARγ) and CAAT/enhancer-binding protein α (C/EBPα) were also reduced during adipogenic differentiation, and the reductions in PPARγ and C/EBPα expressions and the defect in adipogenesis were restored by treatment with troglitazone, a PPARγ ligand. These results indicate that iPLA2γ might play a critical role in adipogenic differentiation by regulating PPARγ expression.


Asunto(s)
Adipogénesis/fisiología , Fibroblastos/metabolismo , Fosfolipasas A2 Grupo VI/metabolismo , Lisofosfolipasa/metabolismo , PPAR gamma/metabolismo , Adipogénesis/efectos de los fármacos , Animales , Proteínas Potenciadoras de Unión a CCAAT , Diferenciación Celular/efectos de los fármacos , Fibroblastos/efectos de los fármacos , Fosfolipasas A2 Grupo VI/genética , Lisofosfolipasa/genética , Ratones , Ratones Noqueados , Cultivo Primario de Células , Troglitazona/farmacología
8.
PLoS One ; 15(8): e0237976, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32822399

RESUMEN

Environmental exposure to arsenite (As3+) has a strong association with the development of human urothelial cancer (UC) and is the 5th most common cancer in men and the 12th most common cancer in women. Muscle invasive urothelial cancer (MIUC) are grouped into basal or luminal molecular subtypes based on their gene expression profile. The basal subtype is more aggressive and can be associated with squamous differentiation, characterized by high expression of keratins (KRT1, 5, 6, 14, and 16) and epidermal growth factor receptor (EGFR) within the tumors. The luminal subtype is less aggressive and is predominately characterized by elevated gene expression of peroxisome proliferator-activated receptor- gamma (PPARγ) and forkhead box protein A1 (FOXA1). We have previously shown that As3+-transformed urothelial cells (As-T) exhibit a basal subtype of UC expressing genes associated with squamous differentiation. We hypothesized that the molecular subtype of the As-T cells could be altered by inducing the expression of PPARγ and/or inhibiting the proliferation of the cells. Non-transformed and As-T cells were treated with Troglitazone (TG, PPARG agonist, 10 µM), PD153035 (PD, an EGFR inhibitor, 1 µM) or a combination of TG and PD for 3 days. The results obtained demonstrate that treatment of the As-T cells with TG upregulated the expression of PPARγ and FOXA1 whereas treatment with PD decreased the expression of some of the basal keratins. However, a combined treatment of TG and PD resulted in a consistent decrease of several proteins associated with the basal subtype of bladder cancers (KRT1, KRT14, KRT16, P63, and TFAP2A). Our data suggests that activation of PPARγ while inhibiting cell proliferation facilitates the regulation of genes involved in maintaining the luminal subtype of UC. In vivo animal studies are needed to address the efficacy of using PPARγ agonists and/or proliferation inhibitors to reduce tumor grade/stage of MIUC.


Asunto(s)
Arsenitos/farmacología , Proliferación Celular/efectos de los fármacos , PPAR gamma/metabolismo , Troglitazona/farmacología , Animales , Biomarcadores de Tumor/metabolismo , Línea Celular Tumoral , Regulación hacia Abajo/efectos de los fármacos , Receptores ErbB/antagonistas & inhibidores , Receptores ErbB/metabolismo , Factor Nuclear 3-alfa del Hepatocito/genética , Factor Nuclear 3-alfa del Hepatocito/metabolismo , Humanos , Queratinas/genética , Queratinas/metabolismo , Ratones , Ratones Desnudos , PPAR gamma/agonistas , Quinazolinas/farmacología , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Transcriptoma/efectos de los fármacos , Trasplante Heterólogo , Regulación hacia Arriba/efectos de los fármacos , Neoplasias de la Vejiga Urinaria/metabolismo , Neoplasias de la Vejiga Urinaria/patología
9.
Anticancer Res ; 40(3): 1247-1254, 2020 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-32132021

RESUMEN

AIM: We attempted to clarify the role of Peroxisome proliferator-activated receptor γ (PPARγ) and its ligand, troglitazone (TRO) on oral squamous cell carcinoma (SCC). MATERIALS AND METHODS: The expression of PPARγ gene was examined in 47 human oral SCC tissues and two human oral SCC cell lines, CA9-22 and HSC-4. The effects of TRO on the growth and cell-cycle progression of human oral SCC cells were examined. RESULTS: PPARγ mRNA was detected in 20 of 47 oral SCC tissues and two human oral SCC cells. TRO significantly suppressed the growth of the cells, but did not induce apoptosis. CA9-22 cells treated with TRO showed an increased fraction in the G1 phase and decreased fractions in the S and G2-M phases. CONCLUSION: TRO did not induce apoptosis in oral SCC cells, but did inhibit the growth of the cells by arresting the cell cycle at G1 phase.


Asunto(s)
Hipoglucemiantes/uso terapéutico , Neoplasias de la Boca/tratamiento farmacológico , PPAR gamma/metabolismo , Carcinoma de Células Escamosas de Cabeza y Cuello/tratamiento farmacológico , Troglitazona/farmacología , Puntos de Control del Ciclo Celular/efectos de los fármacos , Procesos de Crecimiento Celular/efectos de los fármacos , Humanos , Hipoglucemiantes/farmacología , Ligandos , Neoplasias de la Boca/metabolismo , Neoplasias de la Boca/patología , PPAR gamma/biosíntesis , PPAR gamma/genética , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Carcinoma de Células Escamosas de Cabeza y Cuello/genética , Carcinoma de Células Escamosas de Cabeza y Cuello/metabolismo , Carcinoma de Células Escamosas de Cabeza y Cuello/patología
10.
Mol Cell Probes ; 51: 101543, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32105703

RESUMEN

Deformities in human soft tissue caused by trauma or burn present a difficult problem in plastic surgery. In this study, we encapsulated troglitazone and angiotensin 1-7 mimetic AVE0991 in gelation microspheres with the goal of inducing epithelial transformation for potential applications in tissue reconstruction. After troglitazone or AVE0991 were encapsulated to gelation microspheres, their release kinetics and bioactivity were examined. Surface morphology and diameter of the gelation microspheres were evaluated using light microscopy. The release of the drugs was assessed in the presence of human adipose-derived stem cells (ADSCs). Treatment with troglitazone microspheres increased cell viability and activated the ß-catenin in ADSCs. Moreover, the AVE0991 microspheres also increased cell viability and C-myc expression of ADSCs. These results showed that troglitazone and AVE0991 microspheres promoted the activity of ADSCs. Furthermore, ADSCs were co-treated with troglitazone and AVE0991 microspheres. Western blot and immunofluorescent staining showed that co-treatment with troglitazone and AVE0991 microspheres elevated the expression of epithelialization associated protein CK14 in ADSCs. In conclusion, our findings indicate that microspheres with troglitazone and AVE0991 can significantly improve the viability and epithelialization of ADSCs, which provides a new approach for the construction of tissue-engineered skin.


Asunto(s)
Gelatina/química , Imidazoles/farmacocinética , Células Madre Mesenquimatosas/efectos de los fármacos , Ingeniería de Tejidos/métodos , Troglitazona/farmacocinética , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Liberación de Fármacos , Humanos , Hipoglucemiantes/farmacología , Imidazoles/farmacología , Células Madre Mesenquimatosas/metabolismo , Microesferas , Tamaño de la Partícula , Proteínas Proto-Oncogénicas c-myc/genética , Proteínas Proto-Oncogénicas c-myc/metabolismo , Repitelización , Reacción en Cadena en Tiempo Real de la Polimerasa , Troglitazona/farmacología , beta Catenina/metabolismo
11.
FASEB J ; 34(3): 4283-4292, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31970838

RESUMEN

The environmental hypoxia of high altitude (HA) increases the incidence of intrauterine growth restriction (IUGR) approximately threefold. The peroxisome proliferator-activated receptor γ (PPAR-γ), a ligand-activated nuclear receptor that promotes vasorelaxation by increasing nitric oxide and downregulating endothelin-1 (ET-1) production, has been implicated in IUGR. Based on our prior work indicating that pharmacologic activation of the PPARγ pathway protects against hypoxia-associated IUGR, we used an experimental murine model to determine whether such effects may be attributed to vasodilatory effects in the uteroplacental circulation. Using wire myography, ex vivo vasoreactivity studies were conducted in uterine arteries (UtA) isolated from pregnant mice exposed to hypoxia or normoxia from gestational day 14.5 to 18.5. Exposure to troglitazone, a high-affinity PPARγ agonist-induced vasorelaxation in UtA preconstricted with phenylephrine, with HA-UtA showing increased sensitivity. Troglitazone blunted ET-1-induced contraction of UtA in hypoxic and normoxic dams equivalently. Immunohistological analysis revealed enhanced staining for ET-1 receptors in the placental labyrinthine zone in hypoxic compared to normoxic dams. Our results suggest that pharmacologic PPAR-γ activation, via its vasoactive properties, may protect the fetal growth under hypoxic conditions by improving uteroplacental perfusion and thereby justify further investigation into PPARγ as a therapeutic target for IUGR in pregnancies complicated by hypoxia.


Asunto(s)
Endotelina-1/metabolismo , PPAR gamma/metabolismo , Placenta/metabolismo , Arteria Uterina/metabolismo , Animales , Modelos Animales de Enfermedad , Femenino , Retardo del Crecimiento Fetal/metabolismo , Hipoxia/metabolismo , Inmunohistoquímica , Ratones , Fenilefrina/farmacología , Placenta/efectos de los fármacos , Embarazo , Tiazolidinedionas/farmacología , Troglitazona/farmacología , Arteria Uterina/efectos de los fármacos
12.
Oncol Rep ; 43(2): 711-717, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31894283

RESUMEN

The aim of the present study was to investigate the metabolic and anticancer effects of troglitazone (TGZ) with a focus on the potential role of mitochondrial pyruvate utilization. 2­Deoxyglucose (2­DG) was more cytotoxic in CT26 cancer cells compared with T47D cells, despite a smaller suppression of glucose uptake. On the other hand, TGZ caused a more prominent shift to glycolytic metabolism and was more cytotoxic in T47D cells. Both effects of TGZ on T47D cells were dose­dependently reversed by addition of methyl pyruvate (mPyr), indicating suppression of mitochondrial pyruvate availability. Furthermore, UK5099, a specific mitochondrial pyruvate carrier inhibitor, closely simulated the metabolic and antitumor effects of TGZ and their reversal by mPyr. This was accompanied by a substantial reduction of activated p70S6K. In CT26 cells, UK5099 did not reduce activated p70S6K and only modestly decreased cell proliferation. In these cells, combining glutamine restriction with UK5099 further increased glucose uptake and completely suppressed cell proliferation. Thus, TGZ­mediated inhibition of mitochondrial pyruvate utilization is an effective treatment for cancer cells that are more dependent on mitochondrial glucose metabolism. By contrast, cancer cells that are more glycolysis­dependent may require suppression of glutamine utilization in addition to blocking mitochondrial pyruvate availability for a full antitumor effect.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias de la Mama/metabolismo , Mitocondrias/metabolismo , Ácido Pirúvico/metabolismo , Troglitazona/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Desoxiglucosa/farmacología , Relación Dosis-Respuesta a Droga , Femenino , Glucólisis/efectos de los fármacos , Humanos , Mitocondrias/efectos de los fármacos , Proteínas Quinasas S6 Ribosómicas 70-kDa/metabolismo
13.
BMC Genomics ; 21(1): 64, 2020 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-31959126

RESUMEN

BACKGROUND: The advent of Next Generation Sequencing has allowed transcriptomes to be profiled with unprecedented accuracy, but the high costs of full-length mRNA sequencing have posed a limit on the accessibility and scalability of the technology. To address this, we developed 3'Pool-seq: a simple, cost-effective, and scalable RNA-seq method that focuses sequencing to the 3'-end of mRNA. We drew from aspects of SMART-seq, Drop-seq, and TruSeq to implement an easy workflow, and optimized parameters such as input RNA concentrations, tagmentation conditions, and read depth specifically for bulk-RNA. RESULTS: Thorough optimization resulted in a protocol that takes less than 12 h to perform, does not require custom sequencing primers or instrumentation, and cuts over 90% of the costs associated with TruSeq, while still achieving accurate gene expression quantification (Pearson's correlation coefficient with ERCC theoretical concentration r = 0.96) and differential gene detection (ROC analysis of 3'Pool-seq compared to TruSeq AUC = 0.921). The 3'Pool-seq dual indexing scheme was further adapted for a 96-well plate format, and ERCC spike-ins were used to correct for potential row or column pooling effects. Transcriptional profiling of troglitazone and pioglitazone treatments at multiple doses and time points in HepG2 cells was then used to show how 3'Pool-seq could distinguish the two molecules based on their molecular signatures. CONCLUSIONS: 3'Pool-seq can accurately detect gene expression at a level that is on par with TruSeq, at one tenth of the total cost. Furthermore, its unprecedented TruSeq/Nextera hybrid indexing scheme and streamlined workflow can be applied in several different formats, including 96-well plates, which allows users to thoroughly evaluate biological systems under several conditions and timepoints. Care must be taken regarding experimental design and plate layout such that potential pooling effects can be accounted for and corrected. Lastly, further studies using multiple sets of ERCC spike-ins may be used to simulate differential gene expression in a system with known ground-state values.


Asunto(s)
RNA-Seq/métodos , Animales , Análisis Costo-Beneficio , Células Hep G2 , Humanos , Ratones , Pioglitazona/farmacología , RNA-Seq/economía , Transcriptoma/efectos de los fármacos , Troglitazona/farmacología
14.
Eur J Med Chem ; 187: 111939, 2020 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-31838327

RESUMEN

Breast cancer is a major medical threat which cannot be sufficiently addressed by current therapies because of spontaneous or acquired treatment resistance. Besides, triple-negative breast cancer (TNBC) tumors do not respond to targeted therapies, thus new therapeutic strategies are needed. In this context, we designed and prepared new desulfured troglitazone (TGZ)-derived molecules and evaluated them in vitro for their anti-proliferative activity, with a special focus on triple-negative breast cancer cell lines. Optimization of the synthetic strategies and deracemization of the lead compound were performed to give highly active compound 10 with low-micromolar potency. Further studies revealed that this compound triggers apoptosis rather than cell cycle arrest as observed with TGZ.


Asunto(s)
Antineoplásicos/farmacología , Troglitazona/farmacología , Antineoplásicos/síntesis química , Antineoplásicos/química , Apoptosis/efectos de los fármacos , Puntos de Control del Ciclo Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Estructura Molecular , Relación Estructura-Actividad , Troglitazona/síntesis química , Troglitazona/química
15.
Diabetes ; 69(2): 146-157, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31757794

RESUMEN

Incretin hormone dysregulation contributes to reduced insulin secretion and hyperglycemia in patients with type 2 diabetes mellitus (T2DM). Resistance to glucose-dependent insulinotropic polypeptide (GIP) action may occur through desensitization or downregulation of ß-cell GIP receptors (GIP-R). Studies in rodents and cell lines show GIP-R expression can be regulated through peroxisome proliferator-activated receptor γ (PPARγ) response elements (PPREs). Whether this occurs in humans is unknown. To test this, we conducted a randomized, double-blind, placebo-controlled trial of pioglitazone therapy on GIP-mediated insulin secretion and adipocyte GIP-R expression in subjects with well-controlled T2DM. Insulin sensitivity improved, but the insulinotropic effect of infused GIP was unchanged following 12 weeks of pioglitazone treatment. In parallel, we observed increased GIP-R mRNA expression in subcutaneous abdominal adipocytes from subjects treated with pioglitazone. Treatment of cultured human adipocytes with troglitazone increased PPARγ binding to GIP-R PPREs. These results show PPARγ agonists regulate GIP-R expression through PPREs in human adipocytes, but suggest this mechanism is not important for regulation of the insulinotropic effect of GIP in subjects with T2DM. Because GIP has antilipolytic and lipogenic effects in adipocytes, the increased GIP-R expression may mediate accretion of fat in patients with T2DM treated with PPARγ agonists.


Asunto(s)
Diabetes Mellitus Tipo 2/tratamiento farmacológico , Polipéptido Inhibidor Gástrico/metabolismo , Glucosa/metabolismo , Pioglitazona/farmacología , Pioglitazona/uso terapéutico , Receptores de Superficie Celular/metabolismo , Adipocitos/efectos de los fármacos , Diabetes Mellitus Tipo 2/metabolismo , Método Doble Ciego , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Hipoglucemiantes/farmacología , Hipoglucemiantes/uso terapéutico , Secreción de Insulina , PPAR gamma/metabolismo , Receptores de Superficie Celular/genética , Troglitazona/farmacología
16.
Theriogenology ; 142: 276-283, 2020 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-31708195

RESUMEN

The peroxisome proliferator-activated receptor gamma (PPARG, also called NR1C3) is a nuclear receptor of the peroxisome proliferator-activated receptor family (PPAR). PPARs are involved in the regulation of apoptosis, cell cycle, estradiol and progesterone synthesis, and metabolism. However, the role of PPARs and their regulation during follicular development and ovulation in monovular species remain poorly understood. In this study, a well-established intrafollicular injection model was used to investigate if the PPARG participates in the regulation of dominant follicle development and ovulation in cattle. Findings from this study revealed that the relative mRNA abundance of PPARG was similar between dominant and subordinate follicles around follicle deviation, decreased after the LH surge, and increased before ovulation. In addition, a quadratic correlation was found between PPARG mRNA levels in granulosa cells and progesterone concentration in the follicular fluid. Intrafollicular injection of 50 µM Troglitazone (TGZ; a PPARG agonist) inhibited follicular growth and decreased CYP19A1 mRNA abundance in granulosa cells. These findings indicate that PPARG is involved in the regulation of steroidogenesis, follicle growth and ovulation in cattle.


Asunto(s)
Folículo Ovárico/efectos de los fármacos , Folículo Ovárico/fisiología , PPAR gamma/agonistas , Troglitazona/farmacología , Animales , Bovinos , Células Cultivadas , Regulación hacia Abajo/efectos de los fármacos , Femenino , Expresión Génica/efectos de los fármacos , Células de la Granulosa/efectos de los fármacos , Células de la Granulosa/fisiología , Oogénesis/efectos de los fármacos , Oogénesis/genética , Ovulación/efectos de los fármacos , Ovulación/genética , PPAR gamma/genética , PPAR gamma/metabolismo
17.
J Toxicol Sci ; 44(11): 811-820, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31708537

RESUMEN

Troglitazone, the first peroxisome proliferator-associated receptor γ agonist developed as an antidiabetic drug, was withdrawn from the market due to idiosyncratic severe liver toxicity. One proposed mechanism by which troglitazone causes liver injury is induction of mitochondrial membrane permeability transition (MPT), which occurs in a calcium-independent phospholipase A2 (iPLA2)-dependent manner at a concentration of 10 µM. MPT, induced by opening of the MPT pore, leads to the release of cytochrome c and consequent apoptosis or necrosis. In the present study, we aimed to clarify the mechanism of troglitazone-induced MPT in more detail using isolated rat liver mitochondria. We focused on extra-mitochondrial Ca2+ and membrane potential as triggers of iPLA2 activation or MPT induction. As a link between iPLA2 and MPT, we focused on cardiolipin (CL), a unique, mitochondria-specific phospholipid with four acyl chains that affects respiration, the morphology, and other mitochondrial functions. We found that (1) Ca2+ release from the mitochondrial matrix was induced prior to troglitazone-induced onset of MPT, (2) released Ca2+ was involved in troglitazone-induced MPT, (3) mild depolarization (approximately 10%) may be a trigger of troglitazone-induced MPT and (4) enhanced decomposition of CL following mitochondrial iPLA2 activation might mediate troglitazone-induced MPT.


Asunto(s)
Calcio/metabolismo , Hipoglucemiantes/farmacología , Mitocondrias Hepáticas/efectos de los fármacos , Proteínas de Transporte de Membrana Mitocondrial/metabolismo , Fosfolipasas A2 Calcio-Independiente/metabolismo , Troglitazona/farmacología , Animales , Masculino , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Mitocondrias Hepáticas/metabolismo , Mitocondrias Hepáticas/fisiología , Poro de Transición de la Permeabilidad Mitocondrial , Ratas Wistar
18.
BMC Vet Res ; 15(1): 324, 2019 Sep 06.
Artículo en Inglés | MEDLINE | ID: mdl-31492181

RESUMEN

BACKGROUND: Obesity in cats has been associated with alterations in adipokines including: adiponectin, interleukin-6 (IL6), and tumor necrosis factor-α (TNFα). Omega-3 polyunsaturated fatty acids have multiple beneficial effects on obesity-associated disorders, and therefore may alleviate these alterations. This study aimed to determine the effects of body condition, fat depot, troglitazone, and different fatty acids on secretion of adiponectin, IL6 and TNFα from adipose tissue of healthy cats. Subcutaneous and visceral adipose tissue samples were collected from 18 healthy intact female cats, and body condition score (Range 3-7/9) was determined. Concentrations of adiponectin were measured in mature adipocytes cultures and concentrations of IL6 and TNFα were measured in stromovascular cells cultures following treatment with control medium, troglitazone at 10 µM, eicosapentaenoic acid, arachidonic acid, or palmitic acid, at 25, 50, or 100 µM. RESULTS: Stromovascular cells of visceral origin secreted higher concentrations of IL6 than corresponding cells of subcutaneous origin (P = 0.003). Arachidonic acid treatment at 25, 50, and 100 µM increased IL6 secretion in subcutaneous (P = 0.045, P = 0.002, and P < 0.001, respectively) and visceral (P = 0.034, P = 0.001, and P < 0.001, respectively) stromovascular cells. Eicosapentaenoic acid treatment increased TNFα secretion in subcutaneous stromovascular cells at 25, 50, and 100 µM (P = 0.002, P = 0.001, and P = 0.015, respectively) and in visceral stromovascular cells at 50 µM (P < 0.001). No significant effect on medium adiponectin concentration was observed following troglitazone treatment (P = 0.4) or fatty acids treatments at 25 (P = 0.2), 50 (P = 0.8), or 100 (P = 0.7) µM. Body condition score did not have significant effects on medium concentrations of adiponectin (P = 0.4), IL6 (P = 0.1), or TNFα (P = 0.8). CONCLUSIONS: This study demonstrated higher basal secretion of IL6 from visceral compared to subcutaneous adipose tissue, a stimulatory effect of arachidonic acid on secretion of IL6 and a stimulatory effect of eicosapentaenoic acid on TNFα from feline adipose tissue.


Asunto(s)
Adipoquinas/metabolismo , Tejido Adiposo/efectos de los fármacos , Grasas de la Dieta/farmacología , Ácidos Grasos/farmacología , Tejido Adiposo/metabolismo , Animales , Ácido Araquidónico/metabolismo , Constitución Corporal , Gatos , Células Cultivadas , Ácido Eicosapentaenoico/análogos & derivados , Ácido Eicosapentaenoico/metabolismo , Femenino , Interleucina-6/metabolismo , Ácido Palmítico/metabolismo , Troglitazona/farmacología , Factor de Necrosis Tumoral alfa/metabolismo
19.
Mol Pharm ; 16(10): 4230-4240, 2019 10 07.
Artículo en Inglés | MEDLINE | ID: mdl-31509422

RESUMEN

Interruption of bile acid (BA) homeostasis has been hypothesized for a variety of liver diseases and for drug-induced liver injury (DILI). Consequently, BA is gaining increasing prominence as a potential biomarker. The objective of this work was to evaluate the effect of troglitazone (TZN, associated with severe DILI), pioglitazone (PZN, rarely associated with DILI), and acetylsalicylic acid (ASA, or aspirin, not associated with DILI) on the in vitro BA homeostasis in hepatocytes co-cultured with nonparenchymal cells by monitoring the disposition of 36 BAs. The cells were supplemented with 2.5 µM d4-cholic acid, d4-chenodeoxycholic acid, d4-lithocholic acid, d4-deoxycholic acid, d4-ursodeoxycholic acid, and hyodeoxycholic acid. Concentration-time profiles of BAs were used to determine the area under the curve from the supernatant, lysate, or bile compartments, in the presence or absence of TZN, PZN, or ASA. When applicable, IC50 describing depletion of individual BAs was calculated, or accumulation greater than 200% of dimethyl sulfoxide control was noted. Thiazolidinediones significantly altered the concentration of glycine and sulfate conjugates; however, more BAs were impacted by TZN than with PZN. For commonly shared BAs, TZN exhibited 3- to 13-fold stronger inhibition than PZN. In contrast, no changes were observed with ASA. Modulation of BA disposition by thiazolidinediones and ASA was appropriately differentiated. Particularly for thiazolidinediones, TZN was more potent in interrupting BA homeostasis, and, when also considering its higher dose, may explain differences in their clinical instances of DILI. This is one of the first works which comprehensively evaluated the disposition of primary and secondary BAs along with their metabolites in an in vitro system. Differing degrees of BA homeostasis modulation was observed with various perpetrators associated with varying clinical instances of DILI. These data indicate that in vitro systems such as hepatocyte co-cultures may be a promising tool to gain a detailed insight into how drugs affect BA handling to further probe into the mechanism of DILI related to BA homeostasis.


Asunto(s)
Aspirina/farmacología , Ácidos y Sales Biliares/metabolismo , Enfermedad Hepática Inducida por Sustancias y Drogas/tratamiento farmacológico , Hepatocitos/fisiología , Homeostasis , Pioglitazona/farmacología , Troglitazona/farmacología , Antiinflamatorios no Esteroideos/química , Antiinflamatorios no Esteroideos/farmacología , Aspirina/química , Células Cultivadas , Enfermedad Hepática Inducida por Sustancias y Drogas/metabolismo , Enfermedad Hepática Inducida por Sustancias y Drogas/patología , Técnicas de Cocultivo , Hepatocitos/citología , Hepatocitos/efectos de los fármacos , Humanos , Hipoglucemiantes/química , Hipoglucemiantes/farmacología , Pioglitazona/química , Troglitazona/química
20.
Sci Rep ; 9(1): 10506, 2019 07 19.
Artículo en Inglés | MEDLINE | ID: mdl-31324820

RESUMEN

For augmentation or reconstruction of urinary bladder after cystectomy, bladder urothelium derived from human induced pluripotent stem cells (hiPSCs) has recently received focus. However, previous studies have only shown the emergence of cells expressing some urothelial markers among derivatives of hiPSCs, and no report has demonstrated the stratified structure, which is a particularly important attribute of the barrier function of mature bladder urothelium. In present study, we developed a method for the directed differentiation of hiPSCs into mature stratified bladder urothelium. The caudal hindgut, from which the bladder urothelium develops, was predominantly induced via the high-dose administration of CHIR99021 during definitive endoderm induction, and this treatment subsequently increased the expressions of uroplakins. Terminal differentiation, characterized by the increased expression of uroplakins, CK13, and CK20, was induced with the combination of Troglitazone + PD153035. FGF10 enhanced the expression of uroplakins and the stratification of the epithelium, and the transwell culture system further enhanced such stratification. Furthermore, the barrier function of our urothelium was demonstrated by a permeability assay using FITC-dextran. According to an immunohistological analysis, the stratified uroplakin II-positive epithelium was observed in the transwells. This method might be useful in the field of regenerative medicine of the bladder.


Asunto(s)
Células Madre Pluripotentes Inducidas/citología , Vejiga Urinaria/citología , Urotelio/citología , Factor de Transcripción CDX2/biosíntesis , Factor de Transcripción CDX2/genética , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Dextranos/metabolismo , Dextranos/farmacocinética , Endodermo/citología , Receptores ErbB/antagonistas & inhibidores , Factor 10 de Crecimiento de Fibroblastos/farmacología , Fluoresceína-5-Isotiocianato/análogos & derivados , Fluoresceína-5-Isotiocianato/metabolismo , Fluoresceína-5-Isotiocianato/farmacocinética , Glucógeno Sintasa Quinasa 3 beta/antagonistas & inhibidores , Humanos , Células Madre Pluripotentes Inducidas/efectos de los fármacos , PPAR gamma/agonistas , Permeabilidad , Piridinas/farmacología , Pirimidinas/farmacología , Quinazolinas/farmacología , Proteínas Recombinantes/farmacología , Medicina Regenerativa/métodos , Factores de Transcripción SOXF/biosíntesis , Factores de Transcripción SOXF/genética , Troglitazona/farmacología , Uroplaquinas/biosíntesis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...