Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 356
Filtrar
1.
Int J Biol Sci ; 20(11): 4297-4313, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39247816

RESUMO

The heart relies on various defense mechanisms, including metabolic plasticity, to maintain its normal structure and function under high-altitude hypoxia. Pioglitazone, a peroxisome proliferator-activated receptor γ (PPARγ), sensitizes insulin, which in turn regulates blood glucose levels. However, its preventive effects against hypoxia-induced cardiac dysfunction at high altitudes have not been reported. In this study, pioglitazone effectively prevented cardiac dysfunction in hypoxic mice for 4 weeks, independent of its effects on insulin sensitivity. In vitro experiments demonstrated that pioglitazone enhanced the contractility of primary cardiomyocytes and reduced the risk of QT interval prolongation under hypoxic conditions. Additionally, pioglitazone promoted cardiac glucose metabolic reprogramming by increasing glycolytic capacity; enhancing glucose oxidation, electron transfer, and oxidative phosphorylation processes; and reducing mitochondrial reactive ROS production, which ultimately maintained mitochondrial membrane potential and ATP production in cardiomyocytes under hypoxic conditions. Notably, as a PPARγ agonist, pioglitazone promoted hypoxia-inducible factor 1α (HIF-1α) expression in hypoxic myocardium. Moreover, KC7F2, a HIF-1α inhibitor, disrupted the reprogramming of cardiac glucose metabolism and reduced cardiac function in pioglitazone-treated mice under hypoxic conditions. In conclusion, pioglitazone effectively prevented high-altitude hypoxia-induced cardiac dysfunction by reprogramming cardiac glucose metabolism.


Assuntos
Glucose , Hipóxia , Miócitos Cardíacos , PPAR gama , Pioglitazona , Pioglitazona/farmacologia , Pioglitazona/uso terapêutico , Animais , PPAR gama/metabolismo , PPAR gama/agonistas , Camundongos , Glucose/metabolismo , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/efeitos dos fármacos , Hipóxia/complicações , Hipóxia/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Tiazolidinedionas/farmacologia , Tiazolidinedionas/uso terapêutico , Espécies Reativas de Oxigênio/metabolismo
2.
Cells ; 13(17)2024 Sep 09.
Artigo em Inglês | MEDLINE | ID: mdl-39273076

RESUMO

Epithelial-to-mesenchymal transition (EMT) plays a major role in breast cancer progression and the development of drug resistance. We have previously demonstrated a trans-differentiation therapeutic approach targeting invasive dedifferentiated cancer cells. Using a combination of PPARγ agonists and MEK inhibitors, we forced the differentiation of disseminating breast cancer cells into post-mitotic adipocytes. Utilizing murine breast cancer cells, we demonstrated a broad class effect of PPARγ agonists and MEK inhibitors in inducing cancer cell trans-differentiation into adipocytes. Both Rosiglitazone and Pioglitazone effectively induced adipogenesis in cancer cells, marked by PPARγ and C/EBPα upregulation, cytoskeleton rearrangement, and lipid droplet accumulation. All tested MEK inhibitors promoted adipogenesis in the presence of TGFß, with Cobimetinib showing the most prominent effects. A metastasis ex vivo culture from a patient diagnosed with triple-negative breast cancer demonstrated a synergistic upregulation of PPARγ with the combination of Pioglitazone and Cobimetinib. Our results highlight the potential for new therapeutic strategies targeting cancer cell plasticity and the dedifferentiation phenotype in aggressive breast cancer subtypes. Combining differentiation treatments with standard therapeutic approaches may offer a strategy to overcome drug resistance.


Assuntos
Diferenciação Celular , PPAR gama , Pioglitazona , PPAR gama/metabolismo , PPAR gama/agonistas , Humanos , Animais , Camundongos , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Feminino , Pioglitazona/farmacologia , Inibidores de Proteínas Quinases/farmacologia , Adipócitos/efeitos dos fármacos , Adipócitos/metabolismo , Adipogenia/efeitos dos fármacos , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Rosiglitazona/farmacologia , Azetidinas/farmacologia , Neoplasias da Mama/patologia , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/metabolismo , Piperidinas/farmacologia
3.
Wiad Lek ; 77(7): 1325-1330, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39241129

RESUMO

OBJECTIVE: Aim: The aim of this study was to determine the effect of application of drug with circadian activity (pioglitazone) for treatment of patients with periodontist. PATIENTS AND METHODS: Materials and Methods: Group I - 18 individuals with healthy periodontium. Group II - 12 participants with stage II, grade B periodontitis treated with a standard treatment protocol. Group III - 12 participants with stage II, grade B periodontitis, treated with a regimen that included the standard protocol along with the administration of pioglitazone prescribed at recommended times of intake. Group IV - 12 participants with stage II, grade B periodontitis treated with the standard protocol and pioglitazone against the recommended intake hours. RESULTS: Results: The Simplified Oral Hygiene Index in Group I was 0.406±0.034. In Group II, it was 2.5±0.06. In Group III, the hygiene index was 2.633±0.056 and in Group IV it was 2.5±0.059. The Papillary-Marginal-Alveolar Index in Group I was 0.033±0.004. In Group II, it was 0.366±0.011. For Group III, the PMA index was 0.38±0.012 and for Group IV it was 0.378±0.01. The Russell's Periodontal Index in Group I was 0.111±0.008. In Group II, it was 4.668±0.155. For Group III - 4.708±0.132 and for Group IV it was 4.575±0.089. The Gingival Bleeding Index (GBI) in Group I was 0.031±0.003, while in Group II, it was 0.266±0.009, in Group III, it was 0.273±0.007 and in Group IV it was 0.278±0.006. CONCLUSION: Conclusions: The administration of pioglitazone (the drug with circadian activity) according to its circadian stage indeed has a positive effect on the periodontal status changes of patients with stage II, grade B periodontitis.


Assuntos
Índice Periodontal , Periodontite , Pioglitazona , Humanos , Masculino , Feminino , Periodontite/tratamento farmacológico , Pioglitazona/uso terapêutico , Pioglitazona/administração & dosagem , Pioglitazona/farmacologia , Adulto , Pessoa de Meia-Idade , Ritmo Circadiano/efeitos dos fármacos , Tiazolidinedionas/uso terapêutico , Tiazolidinedionas/administração & dosagem , Índice de Higiene Oral , Hipoglicemiantes/uso terapêutico , Hipoglicemiantes/administração & dosagem
4.
Acta Neurobiol Exp (Wars) ; 84(2): 153-164, 2024 Jun 26.
Artigo em Inglês | MEDLINE | ID: mdl-39087839

RESUMO

The aim of the present study was to evaluate the effect of rosiglitazone (RSG) or pioglitazone (POG) on the synaptic plasticity, neuronal apoptosis, brain-derived neurotrophic factor (BDNF), and nitric oxide (NO) metabolites in the hippocampus of juvenile hypothyroid rats. The animals were divided into four groups: control; propylthiouracil (PTU), 0.05% dose in drinking water for 42 days; PTU-POG; and PTU-RSG. The POG (20 mg/kg) and the RSG (4 mg/kg) were administered by IP injection. We conducted long­term potentiation (LTP) in the cornu ammonis 1 area of the hippocampus using high­frequency stimulation of the Schaffer collateral pathway. Then, the hippocampal tissues were collected to determine BDNF and NO levels and the degree of apoptosis. PTU administration decreased the slope (10-90%) and amplitude of the fEPSPs compared to control. Injection of RSG or POG increased the slope, slope (10-90%), and amplitude of the fEPSP in the PTU­POG or PTU­RSG groups compared to the PTU group. TUNEL­positive neurons and NO metabolites in the hippocampus of the PTU group were higher than those of the control group. RSG or POG increased BDNF content in PTU-POG or PTU-RSG groups. Treatment of the rats with POG or RSG decreased apoptotic neurons and NO metabolites in the hippocampus of PTU-POG or PTU-RSG groups, respectively, compared to the PTU group. This study's results revealed that POG or RSG normalized LTP impairment, neuronal apoptosis, and improved BDNF content in the hippocampal tissue of juvenile hypothyroid rats.


Assuntos
Apoptose , Fator Neurotrófico Derivado do Encéfalo , Hipocampo , Hipotireoidismo , Potenciação de Longa Duração , PPAR gama , Ratos Wistar , Rosiglitazona , Animais , Apoptose/efeitos dos fármacos , Hipotireoidismo/tratamento farmacológico , Hipotireoidismo/induzido quimicamente , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Masculino , Rosiglitazona/farmacologia , Potenciação de Longa Duração/efeitos dos fármacos , PPAR gama/agonistas , PPAR gama/metabolismo , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Tiazolidinedionas/farmacologia , Pioglitazona/farmacologia , Ratos , Propiltiouracila/farmacologia , Modelos Animais de Doenças , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Óxido Nítrico/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/metabolismo
5.
Immun Inflamm Dis ; 12(8): e70001, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-39172009

RESUMO

BACKGROUND: The inhalation of paraquat (PQ), one of the most widely used herbicides in the world, can result in lung injury. Curcuma longa (Cl) has long history in traditional and folk medicine for the treatment of a wide range of disorders including respiratory diseases. AIM: The aim of the present work was to evaluate the preventive effect of Cl on inhaled PQ-induced lung injury in rats. METHODS: Male Wistar rats were divided into 8 groups (n = 7), one group exposed to saline (control) and other groups exposed to PQ aerosol. Saline (PQ), Cl extract, (two doses), curcumin (Cu), pioglitazone (Pio), and the combination of Cl-L + Pio and dexamethasone (Dex) were administered during the exposure period to PQ. Total and differential white blood cell (WBC) counts, oxidant and antioxidant indicators in the bronchoalveolar lavage (BALF), interleukin (IL)-10, and tumor necrosis alpha (TNF-α) levels in the lung tissues, lung histologic lesions score, and air way responsiveness to methacholine were evaluated. RESULTS: WBC counts (Total and differential), malondialdehyde level, tracheal responsiveness (TR), IL-10, TNF-α and histopathological changes of the lung were markedly elevated but total thiol content and the activities of catalase and superoxide dismutase were decreased in the BALF in the PQ group. Both doses of Cl, Cu, Pio, Cl-L + Pio, and Dex markedly improved all measured variables in comparison with the PQ group. CONCLUSION: CI, Pio, and Cl-L + Pio improved PQ-induced lung inflammation and oxidative damage comparable with the effects of Dex.


Assuntos
Curcuma , PPAR gama , Paraquat , Pioglitazona , Extratos Vegetais , Ratos Wistar , Animais , Pioglitazona/farmacologia , Pioglitazona/uso terapêutico , Paraquat/toxicidade , Masculino , Ratos , Curcuma/química , PPAR gama/agonistas , PPAR gama/metabolismo , Extratos Vegetais/farmacologia , Extratos Vegetais/uso terapêutico , Pulmão/patologia , Pulmão/efeitos dos fármacos , Pulmão/metabolismo , Lesão Pulmonar/induzido quimicamente , Lesão Pulmonar/prevenção & controle , Lesão Pulmonar/tratamento farmacológico , Lesão Pulmonar/patologia , Lesão Pulmonar/metabolismo , Dexametasona/farmacologia , Líquido da Lavagem Broncoalveolar/citologia , Estresse Oxidativo/efeitos dos fármacos , Tiazolidinedionas/farmacologia , Tiazolidinedionas/uso terapêutico , Antioxidantes/farmacologia , Curcumina/farmacologia , Curcumina/uso terapêutico
6.
J Neuroinflammation ; 21(1): 194, 2024 Aug 03.
Artigo em Inglês | MEDLINE | ID: mdl-39097742

RESUMO

Chronic neuroinflammation and microglial activation are key mediators of the secondary injury cascades and cognitive impairment that follow exposure to repetitive mild traumatic brain injury (r-mTBI). Peroxisome proliferator-activated receptor-γ (PPARγ) is expressed on microglia and brain resident myeloid cell types and their signaling plays a major anti-inflammatory role in modulating microglial responses. At chronic timepoints following injury, constitutive PPARγ signaling is thought to be dysregulated, thus releasing the inhibitory brakes on chronically activated microglia. Increasing evidence suggests that thiazolidinediones (TZDs), a class of compounds approved from the treatment of diabetes mellitus, effectively reduce neuroinflammation and chronic microglial activation by activating the peroxisome proliferator-activated receptor-γ (PPARγ). The present study used a closed-head r-mTBI model to investigate the influence of the TZD Pioglitazone on cognitive function and neuroinflammation in the aftermath of r-mTBI exposure. We revealed that Pioglitazone treatment attenuated spatial learning and memory impairments at 6 months post-injury and reduced the expression of reactive microglia and astrocyte markers in the cortex, hippocampus, and corpus callosum. We then examined whether Pioglitazone treatment altered inflammatory signaling mechanisms in isolated microglia and confirmed downregulation of proinflammatory transcription factors and cytokine levels. To further investigate microglial-specific mechanisms underlying PPARγ-mediated neuroprotection, we generated a novel tamoxifen-inducible microglial-specific PPARγ overexpression mouse line and examined its influence on microglial phenotype following injury. Using RNA sequencing, we revealed that PPARγ overexpression ameliorates microglial activation, promotes the activation of pathways associated with wound healing and tissue repair (such as: IL10, IL4 and NGF pathways), and inhibits the adoption of a disease-associated microglia-like (DAM-like) phenotype. This study provides insight into the role of PPARγ as a critical regulator of the neuroinflammatory cascade that follows r-mTBI in mice and demonstrates that the use of PPARγ agonists such as Pioglitazone and newer generation TZDs hold strong therapeutic potential to prevent the chronic neurodegenerative sequelae of r-mTBI.


Assuntos
Disfunção Cognitiva , Microglia , PPAR gama , Pioglitazona , Animais , Masculino , Camundongos , Concussão Encefálica/metabolismo , Concussão Encefálica/tratamento farmacológico , Disfunção Cognitiva/metabolismo , Disfunção Cognitiva/tratamento farmacológico , Camundongos Endogâmicos C57BL , Microglia/efeitos dos fármacos , Microglia/metabolismo , Pioglitazona/farmacologia , Pioglitazona/uso terapêutico , PPAR gama/metabolismo
7.
Gene ; 931: 148892, 2024 Dec 30.
Artigo em Inglês | MEDLINE | ID: mdl-39187138

RESUMO

Sepsis-related brain injury (SRBI) refers to brain dysfunction and structural damage caused by sepsis, which is characterized by inflammation, oxidative stress, and destruction of the blood-brain barrier. Pioglitazone is a PPAR-γ agonist in which PPAR-γ acts as an inflammatory modulator, determining the relationship between PPAR-γ and SRBI and inflammatory state is critical for the disease. This study aimed to construct a drug-target-disease network for SRBI and Pioglitazone based on network pharmacology, and to investigate the therapeutic effect and potential mechanism of Pioglitazone in SRBI induced by lipopolysaccharide (LPS) in rats through transcriptomics. To establish a rat Model of SRBI by intraperitoneal injection of LPS (10 mg/kg): SD rats were divided into Control, Model (LPS), Pioglitazone, (LPS + Pioglitazone) and GW9662 group (LPS+GW9662). The effects and potential mechanisms of Pioglitazone in the treatment of SRBI were studied using biochemical indexes, pathological changes and transcriptome-sequencing (RNA-seq). RNA-seq results showed 620 DEGs between the Model and the Pioglitazone groups. Enrichment analysis involved multiple inflammatory response processes and chemokine receptor binding functions. TLR4 and CXCL10 in the Toll signaling pathway may play an important role in SRBI as important targets. Pioglitazone may ameliorate SRBI through the PPAR-γ/TLR4/CXCL10 pathway.


Assuntos
Lipopolissacarídeos , PPAR gama , Pioglitazona , Ratos Sprague-Dawley , Sepse , Transcriptoma , Pioglitazona/farmacologia , Animais , Ratos , PPAR gama/metabolismo , PPAR gama/genética , Masculino , Transcriptoma/efeitos dos fármacos , Sepse/tratamento farmacológico , Sepse/genética , Sepse/complicações , Sepse/metabolismo , Receptor 4 Toll-Like/metabolismo , Receptor 4 Toll-Like/genética , Quimiocina CXCL10/genética , Quimiocina CXCL10/metabolismo , Modelos Animais de Doenças , Transdução de Sinais/efeitos dos fármacos , Anilidas/farmacologia , Lesões Encefálicas/tratamento farmacológico , Lesões Encefálicas/metabolismo , Lesões Encefálicas/genética , Lesões Encefálicas/etiologia , Perfilação da Expressão Gênica , Encefalopatia Associada a Sepse/tratamento farmacológico , Encefalopatia Associada a Sepse/genética , Encefalopatia Associada a Sepse/metabolismo
8.
Artigo em Inglês | MEDLINE | ID: mdl-39179196

RESUMO

BACKGROUND: The clinical manifestation of autism spectrum disorder (ASD) is linked to the disruption of fundamental neurodevelopmental pathways. Emerging evidences claim to have an upregulation of canonical Wnt/ß-catenin pathway while downregulation of PPARγ pathway in ASD. This study aims to investigate the therapeutic potential of pioglitazone, a PPARγ agonist, in rat model of ASD. The study further explores the possible role of PPARγ and Wnt/ß-catenin pathway and their interaction in ASD by using their modulators. MATERIAL AND METHODS: Pregnant female Wistar rats received 600 mg/kg of valproic acid (VPA) to induce autistic symptoms in pups. Pioglitazone (10 mg/kg) was used to evaluate neurobehaviors, relative mRNA expression of inflammatory (IL-1ß, IL-6, IL-10, TNF-α), apoptotic markers (Bcl-2, Bax, & Caspase-3) and histopathology (H&E, Nissl stain, Immunohistochemistry). Effect of pioglitazone was evaluated on Wnt pathway and 4 µg/kg dose of 6-BIO (Wnt modulator) was used to study the PPARγ pathway. RESULTS: ASD model was established in pups as indicated by core autistic symptoms, increased neuroinflammation, apoptosis and histopathological neurodegeneration in cerebellum, hippocampus and amygdala. Pioglitazone significantly attenuated these alterations in VPA-exposed rats. The expression study results indicated an increase in key transcription factor, ß-catenin in VPA-rats suggesting an upregulation of canonical Wnt pathway in them. Pioglitazone significantly downregulated the Wnt signaling by suppressing the expression of Wnt signaling-associated proteins. The inhibiting effect of Wnt pathway on PPARγ activity was indicated by downregulation of PPARγ-associated protein in VPA-exposed rats and those administered with 6-BIO. CONCLUSION: In the present study, upregulation of canonical Wnt/ß-catenin pathway was demonstrated in ASD rat model. Pioglitazone administration significantly ameliorated these symptoms potentially through its neuroprotective effect and its ability to downregulate the Wnt/ß-catenin pathway. The antagonism between the PPARγ and Wnt pathway offers a promising therapeutic approach for addressing ASD.


Assuntos
Transtorno do Espectro Autista , Modelos Animais de Doenças , Fármacos Neuroprotetores , PPAR gama , Pioglitazona , Ratos Wistar , Via de Sinalização Wnt , Animais , Transtorno do Espectro Autista/tratamento farmacológico , Transtorno do Espectro Autista/metabolismo , Transtorno do Espectro Autista/induzido quimicamente , PPAR gama/agonistas , PPAR gama/metabolismo , Pioglitazona/farmacologia , Feminino , Via de Sinalização Wnt/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Ratos , Gravidez , Ácido Valproico/farmacologia , beta Catenina/metabolismo , Agonistas PPAR-gama
9.
J Dermatolog Treat ; 35(1): 2381763, 2024 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-39034037

RESUMO

BACKGROUND: Psoriasis is a common autoimmune disease in clinical practice, and previous observational studies have suggested that PPARG agonists such as Pioglitazone may be potential therapeutic agents. However, due to interference from various confounding factors, different observational studies have not reached a unified conclusion. We aim to evaluate the potential use of PPARG agonists for treating psoriasis from a new perspective through drug-targeted Mendelian randomization (MR) analysis. MATERIALS AND METHODS: This study includes data on 8,876 individuals for acute myocardial infarction from GWAS, and LDL cholesterol data from 343,621 Europeans. FinnGen contributed psoriasis vulgaris data for 403,972 individuals. The DrugBank10 databases function to identify genes encoding protein products targeted by active constituents of lipid-modifying targets. A two-sample MR analysis and summary-data-based MR (SMR) analysis estimated the associations between expressions of drug target genes and symptoms of psoriasis vulgaris. A multivariable MR study was further conducted to examine if the observed association was direct association. RESULTS: SMR analysis revealed that enhanced PPARG gene expression in the blood (equivalent to a one standard deviation increase) was a protective factor for psoriasis vulgaris (beta = -0.2017, se = 0.0723, p = 0.0053). Besides, there exists an MR association between LDL mediated by PPARG and psoriasis vulgaris outcomes (beta = -3.9169, se = 0.5676, p = 5.17E-12). These results indicate that PPARG is a therapeutic target for psoriasis, suggesting that psoriasis may be a potential indication for PPARG agonists. CONCLUSION: This study confirms that therapeutic activation of PPARG helps suppress the development of psoriasis. Psoriasis may be a new indication for PPARG agonists, such as Pioglitazone. In the future, new anti-psoriatic drugs could be developed targeting PPARG.


Assuntos
Análise da Randomização Mendeliana , PPAR gama , Psoríase , Humanos , Psoríase/tratamento farmacológico , Psoríase/genética , Psoríase/patologia , PPAR gama/genética , PPAR gama/agonistas , Estudo de Associação Genômica Ampla , LDL-Colesterol/sangue , Pioglitazona/farmacologia , Polimorfismo de Nucleotídeo Único
10.
Int Immunopharmacol ; 139: 112757, 2024 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-39067401

RESUMO

Sepsis is a severe immune response to an infection. It is associated with multiple organ dysfunction syndrome (MODs) along with systemic and neuronal inflammatory response. This study focused on the acute neurologic dysfunction associated with sepsis by exploring the role of PPARγ/SIRT1 pathway against sepsis. We studied the role of this axis in ameliorating sepsis-associated encephalopathy (SAE) and its linked neurobehavioral disorders by using pioglitazone (PIO). This PPARγ agonist showed neuroprotective actions in neuroinflammatory disorders. Sepsis was induced in mice by LPS (10 mg/kg). Survival rate and MODs were assessed. Furthermore, behavioral deficits, cerebral oxidative, inflammatory, and apoptotic markers, and the cerebral expression level of SIRT1 were determined. In this study, we observed that PIO attenuated sepsis-induced cerebral injury. PIO significantly enhanced survival rate, attenuated MODs, and systemic inflammatory response in septic mice. PIO also promoted cerebral SIRT1 expression and reduced cerebral activation of microglia, oxidative stress, HMGB, iNOS, NLRP3 and caspase-3 along with an obvious improvement in behavioral deficits and cerebral pathological damage induced by LPS. Most of the neuroprotective effects of PIO were abolished by EX-527, a SIRT1 inhibitor. These results highlight that the neuroprotective effect of PIO in SAE is mainly SIRT1-dependent.


Assuntos
Lipopolissacarídeos , Fármacos Neuroprotetores , Pioglitazona , Encefalopatia Associada a Sepse , Transdução de Sinais , Sirtuína 1 , Animais , Sirtuína 1/metabolismo , Pioglitazona/uso terapêutico , Pioglitazona/farmacologia , Encefalopatia Associada a Sepse/tratamento farmacológico , Transdução de Sinais/efeitos dos fármacos , Masculino , Camundongos , Fármacos Neuroprotetores/uso terapêutico , Fármacos Neuroprotetores/farmacologia , Sepse/tratamento farmacológico , Sepse/complicações , PPAR gama/metabolismo , PPAR gama/agonistas , Camundongos Endogâmicos C57BL , Estresse Oxidativo/efeitos dos fármacos , Encéfalo/efeitos dos fármacos , Encéfalo/patologia , Encéfalo/metabolismo , Modelos Animais de Doenças , Microglia/efeitos dos fármacos
11.
Theranostics ; 14(8): 3221-3245, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38855177

RESUMO

The availability of non-invasive drug delivery systems capable of efficiently transporting bioactive molecules across the blood-brain barrier to specific cells at the injury site in the brain is currently limited. Delivering drugs to neurons presents an even more formidable challenge due to their lower numbers and less phagocytic nature compared to other brain cells. Additionally, the diverse types of neurons, each performing specific functions, necessitate precise targeting of those implicated in the disease. Moreover, the complex synthetic design of drug delivery systems often hinders their clinical translation. The production of nanomaterials at an industrial scale with high reproducibility and purity is particularly challenging. However, overcoming this challenge is possible by designing nanomaterials through a straightforward, facile, and easily reproducible synthetic process. Methods: In this study, we have developed a third-generation 2-deoxy-glucose functionalized mixed layer dendrimer (2DG-D) utilizing biocompatible and cost-effective materials via a highly facile convergent approach, employing copper-catalyzed click chemistry. We further evaluated the systemic neuronal targeting and biodistribution of 2DG-D, and brain delivery of a neuroprotective agent pioglitazone (Pio) in a pediatric traumatic brain injury (TBI) model. Results: The 2DG-D exhibits favorable characteristics including high water solubility, biocompatibility, biological stability, nanoscale size, and a substantial number of end groups suitable for drug conjugation. Upon systemic administration in a pediatric mouse model of traumatic brain injury (TBI), the 2DG-D localizes in neurons at the injured brain site, clears rapidly from off-target locations, effectively delivers Pio, ameliorates neuroinflammation, and improves behavioral outcomes. Conclusions: The promising in vivo results coupled with a convenient synthetic approach for the construction of 2DG-D makes it a potential nanoplatform for addressing brain diseases.


Assuntos
Dendrímeros , Desoxiglucose , Sistemas de Liberação de Medicamentos , Neurônios , Animais , Dendrímeros/química , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Sistemas de Liberação de Medicamentos/métodos , Desoxiglucose/farmacologia , Desoxiglucose/farmacocinética , Fármacos Neuroprotetores/farmacocinética , Fármacos Neuroprotetores/administração & dosagem , Fármacos Neuroprotetores/farmacologia , Camundongos , Pioglitazona/farmacologia , Pioglitazona/administração & dosagem , Pioglitazona/farmacocinética , Barreira Hematoencefálica/metabolismo , Barreira Hematoencefálica/efeitos dos fármacos , Lesões Encefálicas Traumáticas/tratamento farmacológico , Lesões Encefálicas Traumáticas/metabolismo , Encéfalo/metabolismo , Encéfalo/efeitos dos fármacos , Encefalopatias/tratamento farmacológico , Humanos , Modelos Animais de Doenças , Distribuição Tecidual , Masculino
12.
Ulus Travma Acil Cerrahi Derg ; 30(6): 406-414, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38863295

RESUMO

BACKGROUND: This study evaluated the use of metformin or pioglitazone in preventing or reducing the development of post-operative intra-abdominal adhesion (PIAA) by employing histopathological, immunohistochemical, and biochemical analyses in an experimental adhesion model. METHODS: Fifty Wistar-Albino rats were divided into five groups: Group I (Control), Group II (Sham Treatment), Group III (Hy-aluronic Acid), Group IV (Metformin), and Group V (Pioglitazone). Adhesions were induced in the experimental groups, except for the sham group, using the scraping method. After 10 days, rats were euthanized for evaluation. Macroscopic adhesion degrees were assessed using Nair's scoring system. Immunohistochemical and enzyme-linked immunosorbent assay (ELISA) methods were utilized to assess serum, peritoneal lavage, and intestinal tissue samples. Fructosamine, interleukin-6 (IL-6), transforming growth factor-beta (TGF-ß), and fibronectin levels were measured in serum and peritoneal lavage samples. RESULTS: The groups exhibited similar Nair scores and Type I or Type III Collagen staining scores (all, p>0.05). Pioglitazone significantly reduced serum IL-6 and TGF-ß levels compared to controls (p=0.002 and p=0.008, respectively). Both metformin and pioglitazone groups showed elevated IL-6 in peritoneal lavage relative to controls, while fibronectin levels in the lavage were lower in pioglitazone-treated rats compared to the sham group (all, p<0.005). CONCLUSION: Pioglitazone, but not metformin, demonstrated a positive biochemical impact on preventing PIAA formation in an experimental rat model, although histological impacts were not observed. Further experimental studies employing different dose/duration regimens of pioglitazone are needed to enhance our understanding of its effect on PIAA formation.


Assuntos
Modelos Animais de Doenças , Metformina , Pioglitazona , Ratos Wistar , Animais , Pioglitazona/farmacologia , Metformina/farmacologia , Aderências Teciduais/prevenção & controle , Aderências Teciduais/tratamento farmacológico , Ratos , Hipoglicemiantes/farmacologia , Masculino , Tiazolidinedionas/farmacologia , Complicações Pós-Operatórias/prevenção & controle
13.
Clin Transl Sci ; 17(5): e13834, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38771175

RESUMO

Pioglitazone is class of thiazolidinediones that activates peroxisome proliferator-activated receptors (PPARs) in adipocytes to improve glucose metabolism and insulin sensitivity and has been used as a treatment for type 2 diabetes. However, the underlying mechanisms of associated pioglitazone-induced effects remain unclear. Our study aimed to investigate endogenous metabolite alterations associated with pioglitazone administration in healthy male subjects using an untargeted metabolomics approach. All subjects received 30 mg of pioglitazone once daily in the assigned sequence and period. Urine samples were collected before pioglitazone administration and for 24 h after 7 days of administration. A total of 1465 compounds were detected and filtered using a coefficient of variance below 30% and 108 metabolites were significantly altered upon pioglitazone administration via multivariate statistical analysis. Fourteen significant metabolites were identified using authentic standards and public libraries. Additionally, pathway analysis revealed that metabolites from purine and beta-alanine metabolisms were significantly altered after pioglitazone administration. Further analysis of quantification of metabolites from purine metabolism, revealed that the xanthine/hypoxanthine and uric acid/xanthine ratios were significantly decreased at post-dose. Pioglitazone-dependent endogenous metabolites and metabolic ratio indicated the potential effect of pioglitazone on the activation of PPAR and fatty acid synthesis. Additional studies involving patients are required to validate these findings.


Assuntos
Voluntários Saudáveis , Pioglitazona , Purinas , Tiazolidinedionas , Humanos , Masculino , Pioglitazona/farmacologia , Pioglitazona/administração & dosagem , Purinas/administração & dosagem , Purinas/metabolismo , Adulto , Tiazolidinedionas/administração & dosagem , Tiazolidinedionas/farmacologia , Tiazolidinedionas/efeitos adversos , Metabolômica/métodos , Adulto Jovem , Hipoglicemiantes/farmacologia , Hipoglicemiantes/administração & dosagem
14.
Molecules ; 29(9)2024 May 04.
Artigo em Inglês | MEDLINE | ID: mdl-38731628

RESUMO

Fluorescence lifetime imaging microscopy (FLIM) has proven to be a useful method for analyzing various aspects of material science and biology, like the supramolecular organization of (slightly) fluorescent compounds or the metabolic activity in non-labeled cells; in particular, FLIM phasor analysis (phasor-FLIM) has the potential for an intuitive representation of complex fluorescence decays and therefore of the analyzed properties. Here we present and make available tools to fully exploit this potential, in particular by coding via hue, saturation, and intensity the phasor positions and their weights both in the phasor plot and in the microscope image. We apply these tools to analyze FLIM data acquired via two-photon microscopy to visualize: (i) different phases of the drug pioglitazone (PGZ) in solutions and/or crystals, (ii) the position in the phasor plot of non-labelled poly(lactic-co-glycolic acid) (PLGA) nanoparticles (NPs), and (iii) the effect of PGZ or PGZ-containing NPs on the metabolism of insulinoma (INS-1 E) model cells. PGZ is recognized for its efficacy in addressing insulin resistance and hyperglycemia in type 2 diabetes mellitus, and polymeric nanoparticles offer versatile platforms for drug delivery due to their biocompatibility and controlled release kinetics. This study lays the foundation for a better understanding via phasor-FLIM of the organization and effects of drugs, in particular, PGZ, within NPs, aiming at better control of encapsulation and pharmacokinetics, and potentially at novel anti-diabetics theragnostic nanotools.


Assuntos
Nanopartículas , Pioglitazona , Pioglitazona/farmacologia , Pioglitazona/química , Nanopartículas/química , Animais , Linhagem Celular Tumoral , Humanos , Microscopia de Fluorescência/métodos , Ratos , Copolímero de Ácido Poliláctico e Ácido Poliglicólico/química , Hipoglicemiantes/farmacologia , Hipoglicemiantes/química
15.
J Clin Pharmacol ; 64(8): 984-992, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38563070

RESUMO

As a selective and potent inhibitor targeting the isocitrate dehydrogenase-2 (IDH2) mutant protein, enasidenib obtained approval from the US Food and Drug Administration (FDA) in 2017 for adult patients with acute myeloid leukemia (AML) with an IDH2 mutation. In vitro investigations demonstrated that enasidenib affects various drug metabolic enzymes and transporters. This current investigation aimed to assess enasidenib on the pharmacokinetics (PKs) of CYP substrates, including dextromethorphan (CYP2D6 probe drug), flurbiprofen (CYP2C9 probe drug), midazolam (CYP3A4 probe drug), omeprazole (CYP2C19 probe drug), and pioglitazone (CYP2C8 probe drug), in patients with AML or myelodysplastic syndrome. Results showed that following the co-administration of enasidenib (100 mg, once daily) for 28 days, the PK parameters AUC(0-∞) and Cmax of dextromethorphan increased by 1.37 (90% confidence interval (CI): 0.96, 1.96) and 1.24 (90% CI: 0.94, 1.65)-fold, respectively, compared to dextromethorphan alone. For flurbiprofen, these parameters increased by 1.14 (90%CI: 1.01, 1.29) and 0.97 (90% CI 0.86, 1.08)-fold, respectively, when compared to flurbiprofen alone. Conversely, midazolam exhibited decreases to 0.57 (90% CI 0.34, 0.97) and 0.77 (90% CI 0.39, 1.53)-fold, respectively, in comparison to midazolam alone. The parameters for omeprazole increased by 1.86 (90% CI: 1.33, 2.60) and 1.47 (0.93, 2.31)-fold, respectively, compared to omeprazole alone, while those for pioglitazone decreased to 0.80 (90% CI: 0.62, 1.03) and 0.87 (90% CI: 0.65, 1.16)-fold, respectively, in comparison to pioglitazone alone. These findings provide valuable insights into dose recommendations concerning drugs acting as substrates of CYP2D6, CYP2C9, CYP3A4, CYP2C19, and CYP2C8 when administered concurrently with enasidenib.


Assuntos
Dextrometorfano , Interações Medicamentosas , Leucemia Mieloide Aguda , Midazolam , Síndromes Mielodisplásicas , Pioglitazona , Piridinas , Humanos , Masculino , Pessoa de Meia-Idade , Leucemia Mieloide Aguda/tratamento farmacológico , Feminino , Idoso , Síndromes Mielodisplásicas/tratamento farmacológico , Midazolam/farmacocinética , Pioglitazona/farmacocinética , Pioglitazona/farmacologia , Dextrometorfano/farmacocinética , Dextrometorfano/administração & dosagem , Piridinas/farmacocinética , Piridinas/administração & dosagem , Flurbiprofeno/farmacocinética , Flurbiprofeno/administração & dosagem , Flurbiprofeno/análogos & derivados , Triazinas/farmacocinética , Triazinas/uso terapêutico , Triazinas/administração & dosagem , Adulto , Omeprazol/farmacocinética , Omeprazol/administração & dosagem , Sistema Enzimático do Citocromo P-450/metabolismo , Antineoplásicos/farmacocinética , Antineoplásicos/uso terapêutico , Antineoplásicos/administração & dosagem , Recidiva , Idoso de 80 Anos ou mais , Aminopiridinas
16.
Hum Cell ; 37(4): 959-971, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38607518

RESUMO

Acute kidney injury (AKI) induced by renal ischemia/reperfusion injury (IRI) is a severe clinical condition. ROS accumulation, antioxidant pathways deficiency, and inflammation are involved in IRI. Pioglitazone (Pio) exerts anti-inflammatory and antioxidant effects. The aim of this study was to explore the protective effects of pioglitazone against IRI-induced AKI. Pathogen-free Sprague-Dawley (SD) rats were arbitrarily divided into four groups: Sham operation group Control (CON) group, CON + Pio group, I/R + Saline group, and I/R + Pio group. In addition, HK-2 cells were subjected to hypoxia and reoxygenation to develop an H/R model for investigation of the protective mechanism of Pio. Pretreatment with pioglitazone in the model rats reduced urea nitrogen and creatinine levels, histopathological scores, and cytotoxicity after IRI. Pioglitazone treatment significantly attenuated renal cell apoptosis, decreased cytotoxicity, increased Bcl-2 expression, and downregulated Bax expression. Besides, the levels of ROS and inflammatory factors, including NLRP3, ASC, pro-IL-1ß, pro-caspase-1, cleaved-caspase-1, TNF-α, IL-6, and IL-1ß, in I/R rats and H/R cells were normalized by the pioglitazone treatment. Pioglitazone improved IRI-induced AKI by attenuating oxidative stress and NLRP3 inflammasome activation. Therefore, pioglitazone has the potential to serve as a novel agent for renal IRI treatment and prevention.


Assuntos
Injúria Renal Aguda , Inflamassomos , Proteína 3 que Contém Domínio de Pirina da Família NLR , Estresse Oxidativo , Pioglitazona , Ratos Sprague-Dawley , Traumatismo por Reperfusão , Pioglitazona/farmacologia , Traumatismo por Reperfusão/prevenção & controle , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Injúria Renal Aguda/etiologia , Injúria Renal Aguda/prevenção & controle , Injúria Renal Aguda/tratamento farmacológico , Injúria Renal Aguda/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Inflamassomos/metabolismo , Animais , Humanos , Masculino , Modelos Animais de Doenças , Ratos , Apoptose/efeitos dos fármacos , Antioxidantes/farmacologia , Linhagem Celular
17.
Am J Physiol Regul Integr Comp Physiol ; 327(1): R25-R34, 2024 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-38682243

RESUMO

Insulin insensitivity decreases exogenous glucose oxidation and metabolic clearance rate (MCR) during aerobic exercise in unacclimatized lowlanders at high altitude (HA). Whether use of an oral insulin sensitizer before acute HA exposure enhances exogenous glucose oxidation is unclear. This study investigated the impact of pioglitazone (PIO) on exogenous glucose oxidation and glucose turnover compared with placebo (PLA) during aerobic exercise at HA. With the use of a randomized crossover design, native lowlanders (n = 7 males, means ± SD, age: 23 ± 6 yr, body mass: 84 ± 11 kg) consumed 145 g (1.8 g/min) of glucose while performing 80 min of steady-state (1.43 ± 0.16 V̇o2 L/min) treadmill exercise at HA (460 mmHg; [Formula: see text] 96.6 mmHg) following short-term (5 days) use of PIO (15 mg oral dose per day) or PLA (microcrystalline cellulose pill). Substrate oxidation and glucose turnover were determined using indirect calorimetry and stable isotopes ([13C]glucose and 6,6-[2H2]glucose). Exogenous glucose oxidation was not different between PIO (0.31 ± 0.03 g/min) and PLA (0.32 ± 0.09 g/min). Total carbohydrate oxidation (PIO: 1.65 ± 0.22 g/min, PLA: 1.68 ± 0.32 g/min) or fat oxidation (PIO: 0.10 ± 0.0.08 g/min, PLA: 0.09 ± 0.07 g/min) was not different between treatments. There was no treatment effect on glucose rate of appearance (PIO: 2.46 ± 0.27, PLA: 2.43 ± 0.27 mg/kg/min), disappearance (PIO: 2.19 ± 0.17, PLA: 2.20 ± 0.22 mg/kg/min), or MCR (PIO: 1.63 ± 0.37, PLA: 1.73 ± 0.40 mL/kg/min). Results from this study indicate that PIO is not an effective intervention to enhance exogenous glucose oxidation or MCR during acute HA exposure. Lack of effect with PIO suggests that the etiology of glucose metabolism dysregulation during acute HA exposure may not result from insulin resistance in peripheral tissues.NEW & NOTEWORTHY Short-term (5 days) use of the oral insulin sensitizer pioglitazone does not alter circulating glucose or insulin responses to enhance exogenous glucose oxidation during steady-state aerobic exercise in young healthy men under simulated acute (8 h) high-altitude (460 mmHg) conditions. These results indicate that dysregulations in glucose metabolism in native lowlanders sojourning at high altitude may not be due to insulin resistance at peripheral tissue.


Assuntos
Altitude , Estudos Cross-Over , Exercício Físico , Glucose , Hipoglicemiantes , Oxirredução , Pioglitazona , Humanos , Pioglitazona/administração & dosagem , Pioglitazona/farmacologia , Masculino , Adulto Jovem , Exercício Físico/fisiologia , Adulto , Glucose/metabolismo , Hipoglicemiantes/administração & dosagem , Hipoglicemiantes/farmacologia , Hipoglicemiantes/farmacocinética , Taxa de Depuração Metabólica , Glicemia/metabolismo , Glicemia/efeitos dos fármacos , Insulina/sangue , Insulina/metabolismo
18.
Nano Lett ; 24(15): 4354-4361, 2024 Apr 17.
Artigo em Inglês | MEDLINE | ID: mdl-38563599

RESUMO

The recent focus of cancer therapeutics research revolves around modulating the immunosuppressive tumor microenvironment (TME) to enhance efficacy. The tumor stroma, primarily composed of cancer-associated fibroblasts (CAFs), poses significant obstacles to therapeutic penetration, influencing resistance and tumor progression. Reprogramming CAFs into an inactivated state has emerged as a promising strategy, necessitating innovative approaches. This study pioneers the design of a nanoformulation using pioglitazone, a Food and Drug Administration-approved anti-diabetic drug, to reprogram CAFs in the breast cancer TME. Glutathione (GSH)-responsive dendritic mesoporous organosilica nanoparticles loaded with pioglitazone (DMON-P) are designed for the delivery of cargo to the GSH-rich cytosol of CAFs. DMON-P facilitates pioglitazone-mediated CAF reprogramming, enhancing the penetration of doxorubicin (Dox), a therapeutic drug. Treatment with DMON-P results in the downregulation of CAF biomarkers and inhibits tumor growth through the effective delivery of Dox. This innovative approach holds promise as an alternative strategy for enhancing therapeutic outcomes in CAF-abundant tumors, particularly in breast cancer.


Assuntos
Neoplasias da Mama , Fibroblastos Associados a Câncer , Nanopartículas , Humanos , Feminino , Pioglitazona/farmacologia , Pioglitazona/uso terapêutico , Doxorrubicina/farmacologia , Doxorrubicina/uso terapêutico , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/patologia , Microambiente Tumoral
19.
Naunyn Schmiedebergs Arch Pharmacol ; 397(8): 6177-6195, 2024 08.
Artigo em Inglês | MEDLINE | ID: mdl-38441571

RESUMO

Gastric ulcer is a disturbing disease that impacts many people worldwide. Pioglitazone (Piog), a thiazolidinedione, and ligustrazine (Ligu), a natural component of Ligusticum chuanxiong possess gastroprotective properties. However, the underlying mechanism is not well elucidated. The present study aimed to investigate the gastroprotective effects of Piog (15 mg/kg, p.o.), Ligu (15 mg/kg, p.o.), and their combination against ethanol-induced gastric ulcer in rats. Omeprazole (10 mg/kg) was used as a standard. Pre-treatment for 7 days with Piog, Ligu, and (Piog+Ligu) effectively alleviated ethanol-predisposed oxidative stress and inflammation through restoring HO-1, GSH, and SOD tissue levels and decreasing elevated MDA, TNF-α, ICAM, I-NOS, and IL-1ß contents. Moreover, Piog, Ligu, and (Piog+Ligu) markedly inhibited the ethanol-induced increase of gastric NF-KB and BAX. In contrast, this pre-treatment regimen significantly accelerated protein expression of SIRT1, Nrf2, and Bcl-2, along with autophagic proteins, ATG5 and Beclin. Interestingly, macroscopic, histopathological examination and mucin content were in harmony with previous results, where pre-treatment with Piog, Ligu, and (Piog+Ligu) showed a declined mucosal injury as evidenced by the remarkable decrease of the ulcer area percentage by 62.3%, 38.7%, and 91.2%, respectively, compared to the ethanol-ulcerated group. In conclusion, Piog and Ligu exhibited remarkable gastroprotective properties. Our study was the first to show that Piog, Ligu, and (Piog+Ligu) ameliorated oxidative stress, inflammation, and apoptosis and accelerated the autophagic process via the upregulation of the upstream SIRT1 protein. It is worth mentioning that future studies are needed to pave the way for the clinical use of Piog and Ligu as gastro-protective agents.


Assuntos
Etanol , Estresse Oxidativo , Pioglitazona , Pirazinas , Sirtuína 1 , Úlcera Gástrica , Regulação para Cima , Animais , Úlcera Gástrica/induzido quimicamente , Úlcera Gástrica/prevenção & controle , Úlcera Gástrica/patologia , Úlcera Gástrica/metabolismo , Etanol/toxicidade , Sirtuína 1/metabolismo , Masculino , Pirazinas/farmacologia , Pirazinas/uso terapêutico , Pioglitazona/farmacologia , Pioglitazona/uso terapêutico , Estresse Oxidativo/efeitos dos fármacos , Regulação para Cima/efeitos dos fármacos , Ratos , Antiulcerosos/farmacologia , Antiulcerosos/uso terapêutico , Ratos Sprague-Dawley , Mucosa Gástrica/efeitos dos fármacos , Mucosa Gástrica/patologia , Mucosa Gástrica/metabolismo
20.
Toxicon ; 241: 107687, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38484848

RESUMO

The effects of safranal and pioglitazone alone and their combination on inhaled paraquat (PQ)-induced systemic oxidative stress and inflammation as well as behavioral changes were examined in rats. In this study, animals were exposed to saline (Ctrl) or PQ (PQ groups) aerosols. PQ exposed animals were treated with dexamethasone, 0.8 and 3.2 mg/kg/day safranal (Saf-L and Saf-H), 5 mg/kg/day pioglitazone (Pio), and Saf-L + Pio for 16 days during PQ exposure period. PQ group showed increased numbers of total and differential WBCs in blood and bronchoalveolar lavage fluid (BALF), increased malondialdehyde (MDA), in the serum BALF and brain reduced thiol, catalase (CAT), and superoxide dismutase (SOD) levels compared to the control group (for all, p < 0.001). The escape latency and traveled distance were enhanced, but the time spent in the target quadrant in the probe day and the latency to enter the dark room 3, 24, 48, and 72 h after receiving an electrical shock, (in the shuttle box test) were decreased in the PQ group (p < 0.05 to P < 0.001). In all treated groups, all measure values were improved compared to PQ group (p < 0.05 to p < 0.001). In combination treated group of Saf-L + Pio, most measured values were more improved than the Saf-L and Pio groups (p < 0.05 to p < 0.001). Saf and Pio improved PQ-induced changes similar to dexamethasone but the effects produced by combination treatments of Saf-L + Pio were more prominent than Pio and Saf-L alone, suggesting a potentiating effect for the combination of the two agents.


Assuntos
Lesão Pulmonar Aguda , Cicloexenos , Paraquat , Edema Pulmonar , Terpenos , Ratos , Animais , Paraquat/toxicidade , Pulmão , Pioglitazona/farmacologia , Estresse Oxidativo , Inflamação/induzido quimicamente , Inflamação/tratamento farmacológico , Dexametasona/farmacologia , Dexametasona/uso terapêutico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...