Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros











Intervalo de ano de publicação
1.
Cell Commun Signal ; 22(1): 266, 2024 May 13.
Artigo em Inglês | MEDLINE | ID: mdl-38741139

RESUMO

Glioblastoma (GBM) is a type of brain cancer categorized as a high-grade glioma. GBM is characterized by limited treatment options, low patient survival rates, and abnormal serotonin metabolism. Previous studies have investigated the tumor suppressor function of aldolase C (ALDOC), a glycolytic enzyme in GBM. However, it is unclear how ALDOC regulates production of serotonin and its associated receptors, HTRs. In this study, we analyzed ALDOC mRNA levels and methylation status using sequencing data and in silico datasets. Furthermore, we investigated pathways, phenotypes, and drug effects using cell and mouse models. Our results suggest that loss of ALDOC function in GBM promotes tumor cell invasion and migration. We observed that hypermethylation, which results in loss of ALDOC expression, is associated with serotonin hypersecretion and the inhibition of PPAR-γ signaling. Using several omics datasets, we present evidence that ALDOC regulates serotonin levels and safeguards PPAR-γ against serotonin metabolism mediated by 5-HT, which leads to a reduction in PPAR-γ expression. PPAR-γ activation inhibits serotonin release by HTR and diminishes GBM tumor growth in our cellular and animal models. Importantly, research has demonstrated that PPAR-γ agonists prolong animal survival rates and increase the efficacy of temozolomide in an orthotopic brain model of GBM. The relationship and function of the ALDOC-PPAR-γ axis could serve as a potential prognostic indicator. Furthermore, PPAR-γ agonists offer a new treatment alternative for glioblastoma multiforme (GBM).


Assuntos
Glioblastoma , Agonistas PPAR-gama , Temozolomida , Animais , Humanos , Camundongos , Antineoplásicos Alquilantes/farmacologia , Antineoplásicos Alquilantes/uso terapêutico , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/patologia , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Linhagem Celular Tumoral , Progressão da Doença , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Glioblastoma/tratamento farmacológico , Glioblastoma/patologia , Glioblastoma/genética , Glioblastoma/metabolismo , PPAR gama/metabolismo , Agonistas PPAR-gama/farmacologia , Agonistas PPAR-gama/uso terapêutico , Serotonina/metabolismo , Transdução de Sinais/efeitos dos fármacos , Temozolomida/farmacologia , Temozolomida/uso terapêutico
2.
Adv Healthc Mater ; 12(29): e2301324, 2023 11.
Artigo em Inglês | MEDLINE | ID: mdl-37531231

RESUMO

Black phosphorus (BP) exhibits significant potential for clinical applications. However, further research is necessary to uncover the unknown biological functions of BP and broaden its applications across various fields. This study investigates the potential of BP as a targeting PPAR-γ agonist to overcome chemoresistance in the treatment of pancreatic adenocarcinoma (PAAD) using 2D and 3D cell lines, patient-derived organoids (PDOs), and mouse models. RNA-sequencing analysis shows that BP treatment enriches differentially expressed genes in the PPAR pathway, and molecular modeling predicts the potential docking site between BP and PPAR-γ. Transcriptional activity assays are further to verify the activation of PPAR-γ. BP-activated PPAR-γ inhibits cancer stem cell (CSC) properties and expression of biomarkers such as CD44 and c-Myc, which are involved in chemoresistance. Notably, CD44 overexpression in tumor cells renders them susceptible to BP while insensitive to gemcitabine. This indicates that BP preferentially targets stem-like cells, which exhibit heightened resistance to chemotherapeutic drugs. A combination treatment strategy involving BP and gemcitabine is developed, demonstrating enhanced treatment efficacy of PAAD in both in vitro and in vivo models. Thus, BP serves as a PPAR-γ agonist capable of reversing chemoresistance, establishing it as a potent anti-tumor approach for the treatment of PAAD.


Assuntos
Adenocarcinoma , Neoplasias Pancreáticas , Humanos , Camundongos , Animais , Neoplasias Pancreáticas/tratamento farmacológico , Neoplasias Pancreáticas/metabolismo , Agonistas PPAR-gama , Resistencia a Medicamentos Antineoplásicos , Adenocarcinoma/tratamento farmacológico , Adenocarcinoma/metabolismo , Gencitabina , PPAR gama/metabolismo , PPAR gama/uso terapêutico , Organoides/patologia , Linhagem Celular Tumoral , Neoplasias Pancreáticas
3.
Eur J Pharmacol ; 953: 175841, 2023 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-37329972

RESUMO

Pathological cardiac hypertrophy is associated with ventricular fibrosis leading to heart failure. The use of thiazolidinediones as Peroxisome Proliferator-Activated Receptor-gamma (PPARγ)-modulating anti-hypertrophic therapeutics has been restricted due to major side-effects. The present study aims to evaluate the anti-fibrotic potential of a novel PPARγ agonist, deoxyelephantopin (DEP) in cardiac hypertrophy. AngiotensinII treatment in vitro and renal artery ligation in vivo were performed to mimic pressure overload-induced cardiac hypertrophy. Myocardial fibrosis was evaluated by Masson's trichrome staining and hydroxyproline assay. Our results showed that DEP treatment significantly improves the echocardiographic parameters by ameliorating ventricular fibrosis without any bystander damage to other major organs. Following molecular docking, all-atomistic molecular dynamics simulation, reverse transcription-polymerase chain reaction and immunoblot analyses, we established DEP as a PPARγ agonist stably interacting with the ligand-binding domain of PPARγ. DEP specifically downregulated the Signal Transducer and Activator of Transcription (STAT)-3-mediated collagen gene expression in a PPARγ-dependent manner, as confirmed by PPARγ silencing and site-directed mutagenesis of DEP-interacting PPARγ residues. Although DEP impaired STAT-3 activation, it did not have any effect on the upstream Interleukin (IL)-6 level implying possible crosstalk of the IL-6/STAT-3 axis with other signaling mediators. Mechanistically, DEP increased the binding of PPARγ with Protein Kinase C-delta (PKCδ) which impeded the membrane translocation and activation of PKCδ, downregulating STAT-3 phosphorylation and resultant fibrosis. This study, therefore, for the first time demonstrates DEP as a novel cardioprotective PPARγ agonist. The therapeutic potential of DEP as an anti-fibrotic remedy can be exploited against hypertrophic heart failure in the future.


Assuntos
Insuficiência Cardíaca , PPAR gama , Humanos , PPAR gama/metabolismo , Interleucina-6 , Agonistas PPAR-gama , Simulação de Acoplamento Molecular , Cardiomegalia/patologia , Fibrose
4.
Molecules ; 28(9)2023 May 05.
Artigo em Inglês | MEDLINE | ID: mdl-37175309

RESUMO

The main objective of this research was to develop novel compounds from readily accessed natural products especially eugenol with potential biological activity. Eugenol, the principal chemical constituent of clove (Eugenia caryophyllata) from the family Myrtaceae is renowned for its pharmacological properties, which include analgesic, antidiabetic, antioxidant, anticancer, and anti-inflammatory effects. According to reports, PPARγ regulates inflammatory reactions. The synthesized compounds were structurally analyzed using FT-IR, 1HNMR, 13CNMR, and mass spectroscopy techniques. Molecular docking was performed to analyze binding free energy and important amino acids involved in the interaction between synthesized derivatives and the target protein. The development of the structure-activity relationship is based on computational studies. Additionally, the stability of the best-docked protein-ligand complexes was assessed using molecular dynamic modeling. The in-vitro PPARγ competitive binding Lanthascreen TR-FRET assay was used to confirm the affinity of compounds to the target protein. All the synthesized derivatives were evaluated for an in vitro anti-inflammatory activity using an albumin denaturation assay and HRBC membrane stabilization at varying concentrations from 6.25 to 400 µM. In this background, with the aid of computational research, we were able to design six novel derivatives of eugenol synthesized, analyzed, and utilized TR-FRET competitive binding assay to screen them for their ability to bind PPARγ. Anti-inflammatory activity evaluation through in vitro albumin denaturation and HRBC method revealed that 1f exhibits maximum inhibition of heat-induced albumin denaturation at 50% and 85% protection against HRBC lysis at 200 and 400 µM, respectively. Overall, we found novel derivatives of eugenol that could potentially reduce inflammation by PPARγ agonism.


Assuntos
Eugenol , PPAR gama , Humanos , Eugenol/farmacologia , PPAR gama/metabolismo , Simulação de Acoplamento Molecular , Agonistas PPAR-gama , Espectroscopia de Infravermelho com Transformada de Fourier , Anti-Inflamatórios/farmacologia , Inflamação , Relação Estrutura-Atividade , Simulação de Dinâmica Molecular , Albuminas
5.
Neurochem Res ; 48(9): 2870-2880, 2023 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-37204549

RESUMO

A growing body of evidence has shown that seizure can trigger inflammatory cascades through increasing the expression of several inflammatory cytokines. It has been proved that peroxisome proliferator-activated receptor-γ agonists have immunomodulatory, anti-inflammatory, and neuroprotective effects beyond the putative hypoglycemic effects. Thus, we investigated the inhibitory effect of rosiglitazone on the development of pentylenetetrazol (PTZ)-induced kindling via affecting the inflammatory pathway. Male C57BL/6 mice were randomly divided into vehicle group (0.1% DMSO), PTZ-group and rosiglitazone-PTZ-group. Kindling was induced by the administration of PTZ (40 mg/kg, i.p) every other day and mice were observed for 20 min after each PTZ injection. Twenty-four hours after the last dose, animals were euthanized and hippocampus was isolated. The level of Malondialdehyde (MDA), Superoxide Dismutase (SOD), and Catalase (CAT) activity were quantified in hippocampus by biochemical methods. The protein levels of IL-1ß, IL-6, IL-10, IFN-γ, TNF-α, caspase-3, iNOS, PPAR-γ, Bcl-2, or Bax factors were measured with western blotting. Also, the quantitative real-time PCR were used to evaluate the mRNA expression of those factors. Pretreatment with rosiglitazone significantly prevented the progression of kindling in comparison with control group. The rosiglitazone significantly decreased the MDA level and increased the CAT, and SOD levels in the rosiglitazone treated mice compared to those in the PTZ group (P < 0.01). Using real-time PCR and Western blotting assay, similar results were obtained. The expression levels of IL-1ß, IL-6, IL-10, IFN-γ, TNF-α, Bax or PPAR-γ were significantly changed in the brain. The results of this study suggest that effect of rosiglitazone may be crucial in its ability to protect against the neuronal damage caused by PTZ induced seizure.


Assuntos
Excitação Neurológica , Pentilenotetrazol , Animais , Masculino , Camundongos , Antioxidantes/farmacologia , Proteína X Associada a bcl-2/metabolismo , Citocinas/metabolismo , Interleucina-10/metabolismo , Interleucina-6/metabolismo , Camundongos Endogâmicos C57BL , Estresse Oxidativo , Pentilenotetrazol/toxicidade , PPAR gama/metabolismo , Agonistas PPAR-gama , Piroptose , Rosiglitazona/farmacologia , Convulsões/induzido quimicamente , Convulsões/tratamento farmacológico , Superóxido Dismutase/metabolismo , Fator de Necrose Tumoral alfa/metabolismo
6.
Pulm Pharmacol Ther ; 80: 102209, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-36907545

RESUMO

INTRODUCTION: Hyperoxia-induced lung injury is characterized by acute alveolar injury, disrupted epithelial-mesenchymal signaling, oxidative stress, and surfactant dysfunction, yet currently, there is no effective treatment. Although a combination of aerosolized pioglitazone (PGZ) and a synthetic lung surfactant (B-YL peptide, a surfactant protein B mimic) prevents hyperoxia-induced neonatal rat lung injury, whether it is also effective in preventing hyperoxia-induced adult lung injury is unknown. METHOD: Using adult mice lung explants, we characterize the effects of 24 and 72-h (h) exposure to hyperoxia on 1) perturbations in Wingless/Int (Wnt) and Transforming Growth Factor (TGF)-ß signaling pathways, which are critical mediators of lung injury, 2) aberrations of lung homeostasis and injury repair pathways, and 3) whether these hyperoxia-induced aberrations can be blocked by concomitant treatment with PGZ and B-YL combination. RESULTS: Our study reveals that hyperoxia exposure to adult mouse lung explants causes activation of Wnt (upregulation of key Wnt signaling intermediates ß-catenin and LEF-1) and TGF-ß (upregulation of key TGF-ß signaling intermediates TGF-ß type I receptor (ALK5) and SMAD 3) signaling pathways accompanied by an upregulation of myogenic proteins (calponin and fibronectin) and inflammatory cytokines (IL-6, IL-1ß, and TNFα), and alterations in key endothelial (VEGF-A and its receptor FLT-1, and PECAM-1) markers. All of these changes were largely mitigated by the PGZ + B-YL combination. CONCLUSION: The effectiveness of the PGZ + B-YL combination in blocking hyperoxia-induced adult mice lung injury ex-vivo is promising to be an effective therapeutic approach for adult lung injury in vivo.


Assuntos
Hiperóxia , Lesão Pulmonar , Animais , Camundongos , Hiperóxia/complicações , Hiperóxia/metabolismo , Pulmão , Lesão Pulmonar/etiologia , Lesão Pulmonar/prevenção & controle , Lesão Pulmonar/metabolismo , Pioglitazona/farmacologia , Pioglitazona/metabolismo , PPAR gama/agonistas , PPAR gama/metabolismo , Agonistas PPAR-gama , Tensoativos/metabolismo , Tensoativos/farmacologia , Fator de Crescimento Transformador beta/farmacologia
7.
EBioMedicine ; 90: 104506, 2023 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-36889064

RESUMO

BACKGROUND: Proteinuria is associated with many glomerular diseases and a risk factor for the progression to renal failure. We previously showed that heparanase (HPSE) is essential for the development of proteinuria, whereas peroxisome proliferator-activated receptor É£ (PPARÉ£) agonists can ameliorate proteinuria. Since a recent study showed that PPARÉ£ regulates HPSE expression in liver cancer cells, we hypothesized that PPARÉ£ agonists exert their reno-protective effect by inhibiting glomerular HPSE expression. METHODS: Regulation of HPSE by PPARÉ£ was assessed in the adriamycin nephropathy rat model, and cultured glomerular endothelial cells and podocytes. Analyses included immunofluorescence staining, real-time PCR, heparanase activity assay and transendothelial albumin passage assay. Direct binding of PPARÉ£ to the HPSE promoter was evaluated by the luciferase reporter assay and chromatin immunoprecipitation assay. Furthermore, HPSE activity was assessed in 38 type 2 diabetes mellitus (T2DM) patients before and after 16/24 weeks treatment with the PPARÉ£ agonist pioglitazone. FINDINGS: Adriamycin-exposed rats developed proteinuria, an increased cortical HPSE and decreased heparan sulfate (HS) expression, which was ameliorated by treatment with pioglitazone. In line, the PPARÉ£ antagonist GW9662 increased cortical HPSE and decreased HS expression, accompanied with proteinuria in healthy rats, as previously shown. In vitro, GW9662 induced HPSE expression in both endothelial cells and podocytes, and increased transendothelial albumin passage in a HPSE-dependent manner. Pioglitazone normalized HPSE expression in adriamycin-injured human endothelial cells and mouse podocytes, and adriamycin-induced transendothelial albumin passage was reduced as well. Importantly, we demonstrated a regulatory effect of PPARÉ£ on HPSE promoter activity and direct PPARy binding to the HPSE promoter region. Plasma HPSE activity of T2DM patients treated with pioglitazone for 16/24 weeks was related to their hemoglobin A1c and showed a moderate, near significant correlation with plasma creatinine levels. INTERPRETATION: PPARÉ£-mediated regulation of HPSE expression appears an additional mechanism explaining the anti-proteinuric and renoprotective effects of thiazolidinediones in clinical practice. FUNDING: This study was financially supported by the Dutch Kidney Foundation, by grants 15OI36, 13OKS023 and 15OP13. Consortium grant LSHM16058-SGF (GLYCOTREAT; a collaboration project financed by the PPP allowance made available by Top Sector Life Sciences & Health to the Dutch Kidney Foundation to stimulate public-private partnerships).


Assuntos
Diabetes Mellitus Tipo 2 , Nefropatias , Tiazolidinedionas , Ratos , Camundongos , Humanos , Animais , Pioglitazona/farmacologia , Pioglitazona/uso terapêutico , PPAR gama , Diabetes Mellitus Tipo 2/complicações , Agonistas PPAR-gama , Células Endoteliais/metabolismo , Tiazolidinedionas/farmacologia , Tiazolidinedionas/uso terapêutico , Proteinúria/tratamento farmacológico , Proteinúria/etiologia , Nefropatias/tratamento farmacológico , Doxorrubicina/efeitos adversos
8.
Bioorg Med Chem ; 78: 117130, 2023 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-36542958

RESUMO

PPAR gamma (PPARG) is a ligand activated transcription factor that regulates genes involved in inflammation, bone biology, lipid homeostasis, as well as a master regulator of adipogenesis and a potential lineage driver of luminal bladder cancer. While PPARG agonists lead to transcriptional activation of canonical target genes, inverse agonists have the opposite effect through inducing a transcriptionally repressive complex leading to repression of canonical target gene expression. While many agonists have been described and tested clinically, inverse agonists offer an underexplored avenue to modulate PPARG biology in vivo. Current inverse agonists lack favorable in vivo properties; herein we describe the discovery and characterization of a series of orally bioavailable 4-chloro-6-fluoroisophthalamides as covalent PPARG inverse-agonists, BAY-5516, BAY-5094, and BAY-9683. Structural studies of this series revealed distinct pre- and post-covalent binding positions, which led to the hypothesis that interactions in the pre-covalent conformation are primarily responsible for driving affinity, while interactions in the post-covalent conformation are more responsible for cellular functional effects by enhancing PPARG interactions with its corepressors. The need to simultaneously optimize for two distinct states may partially explain the steep SAR observed. Exquisite selectivity was achieved over related nuclear receptors in the subfamily due in part to a covalent warhead with low reactivity through an SNAr mechanism in addition to the specificity gained through covalent binding to a reactive cysteine uniquely positioned within the PPARG LBD. BAY-5516, BAY-5094, and BAY-9683 lead to pharmacodynamic regulation of PPARG target gene expression in vivo comparable to known inverse agonist SR10221 and represent new tools for future in vivo studies to explore their potential utility for treatment of disorders of hyperactivated PPARG including luminal bladder cancer and other disorders.


Assuntos
PPAR gama , Neoplasias da Bexiga Urinária , Humanos , PPAR gama/agonistas , Agonismo Inverso de Drogas , Agonistas PPAR-gama , Regulação da Expressão Gênica
9.
Araçatuba; s.n; 2020. 67 p. graf, tab, ilus.
Tese em Português | LILACS, BBO - Odontologia | ID: biblio-1438492

RESUMO

Os receptores ativados por proliferadores de peroxissoma de isoforma gama (PPARgama) são fatores de transcrição que se encontram no entrelaçamento de várias vias metabólicas e determinam a gênese da obesidade e doenças associadas. Estudos recentes têm relatado o potencial dos medicamentos agonistas PPARs, usados no tratamento de doenças cardiovasculares e diabetes tipo 2, de afetar a função das células ósseas e o risco de fratura. O objetivo deste estudo foi avaliar os efeitos da ativação do PPAR-gama no processo de reparo ósseo e osseointegração de implantes dentários. Foram utilizados 42 ratos Wistar alimentados com ração sólida padrão para roedores e água "ad Libitum". A administração do agonista PPAR-gama(durante 4 semanas) foi realizada incorporada à dieta perfazendo dois grupos experimentais: Grupo Controle (SC) e Grupo PPAR-gama (SC-gama). Os animais, em seguida, foram submetidos a procedimento cirúrgico para instalação de implantes dentários e confecção de defeitos ósseos nas tíbias direita e esquerda, respectivamente. Após períodos de 7, 15 e 45 dias os animais foram eutanasiados e as tíbias removidas seguiram para processamento e posterior análise histológica descritiva e histomorfometria. Em relação aos defeitos, no aspecto histológico descritivo, em todos os períodos analisados, há atraso no reparo ósseo do grupo SC-gama em relação ao SC. A histomorfometria apresenta diferença comparando os grupos teste e controle nos parâmetros ossoneoformado (ON) em 7 dias (P=0,0374) e 15 dias (P=0,0134) e osso maduro (OM) em 15 dias (P=0,0009). Em 45 dias, os valores tanto de OM quanto de ON não apresentam significância. Já as tíbias onde o implante foi instalado, nas áreas de roscas os dois grupos apresentam-se muito semelhantes tanto aos eventos celulares quanto à maturação óssea exceto no período de 15 dias onde o grupo teste apresentou atraso no reparo peri-implantar. Conclui-se que, em relação ao reparo ósseo o processo de diferenciação e remodelação é lento no grupo tratado; e em relação ao reparo peri-implantar o comportamento biológico demonstra atraso do grupo tratado apenas no período intermediário(AU)


The peroxisome proliferator-activated receptor-gamma isoform (PPAR­gamma) are transcription factors found in the interlacing of several metabolic pathways that determine the genesis of obesity and associated diseases. Recent studies have reported the potential of PPAR agonist drugs, used to treat cardiovascular disease and type 2 diabetes, to affect bone cell function and fracture risk. This study aimed to evaluate the effects of PPAR-gamma activation on the process of bone repair and osseointegration of dental implants. Thirty-six Wistar rats received standard rodent solid chow and "ad Libitum" water. The administration of the PPAR-gamma agonist (during four weeks) was performed incorporated into the diet, making two experimental groups: Control Group (SC) and PPAR gamma Group (SC-gama). The animals were then submitted to a surgical procedure to install dental implants and make bone defects in the right and left tibias, respectively. After periods of 7, 15, and 45 days the animals were euthanized, the tibiae were removed for histological protocols and subsequent descriptive histological analysis and histomorphometry. Concerning the defects, the descriptive histological aspect revealed a slight delay in bone repair in the SC-gamma group in comparison with the SC for all parameters. Conversely, the histomorphometry showed difference between the groups regarding neoformed bone (NB) in 7 days (P = 0.0374) and 15 days (P = 0.0134) and mature bone (MB) in 15 days (P = 0.0009). At 45 days, there were no differences between the groups regarding MB and NB. As for tibiae, where the implant was placed, the two groups were similar to both cellular events and bone maturation in the areas of threads, except at 15 days where the test group showed delay in peri-implant repair. In conclusion, regarding the bone defect, the process of differentiation and remodeling is slow in the treated group. Regarding the implant, a descriptive histological analysis showed biological behavior delay in the treated group only in the intermediate period(AU)


Assuntos
Animais , Ratos , Regeneração Óssea , Implantes Dentários , Osseointegração , PPAR gama , Células , Agonistas PPAR-gama , Obesidade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA