Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.641
Filtrar
Mais filtros

Intervalo de ano de publicação
1.
J Cell Mol Med ; 28(13): e18529, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38984945

RESUMO

In this in vitro study, for the first time, we evaluate the effects of simvastatin-loaded liposome nanoparticles (SIM-LipoNPs) treatment on fibrosis-induced liver microtissues, as simvastatin (SIM) has shown potential benefits in the non-alcoholic fatty liver disease process. We developed multicellular liver microtissues composed of hepatic stellate cells, hepatoblastoma cells and human umbilical vein endothelial cells. The microtissues were supplemented with a combination of palmitic acid and oleic acid to develop fibrosis models. Subsequently, various groups of microtissues were exposed to SIM and SIM-LipoNPs at doses of 5 and 10 mg/mL. The effectiveness of the treatments was evaluated by analysing cell viability, production of reactive oxygen species (ROS) and nitric oxide (NO), the expression of Kruppel-like factor (KLF) 2, and pro-inflammatory cytokines (interleukin(IL)-1 α, IL-1 ß, IL-6 and tumour necrosis factor-α), and the expression of collagen I. Our results indicated that SIM-LipoNPs application showed promising results. SIM-LipoNPs effectively amplified the SIM-klf2-NO pathway at a lower dosage compatible with a high dosage of free SIM, which also led to reduced oxidative stress by decreasing ROS levels. SIM-LipoNPs administration also resulted in a significant reduction in pro-inflammatory cytokines and Collagen I mRNA levels, as a marker of fibrosis. In conclusion, our study highlights the considerable therapeutic potential of using SIM-LipoNPs to prevent liver fibrosis progress, underscoring the remarkable properties of SIM-LipoNPs in activating the KLF2-NO pathway and anti-oxidative and anti-inflammatory response.


Assuntos
Células Estreladas do Fígado , Fatores de Transcrição Kruppel-Like , Lipossomos , Cirrose Hepática , Nanopartículas , Espécies Reativas de Oxigênio , Sinvastatina , Humanos , Sinvastatina/farmacologia , Cirrose Hepática/tratamento farmacológico , Cirrose Hepática/patologia , Cirrose Hepática/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Nanopartículas/química , Fatores de Transcrição Kruppel-Like/metabolismo , Fatores de Transcrição Kruppel-Like/genética , Células Estreladas do Fígado/efeitos dos fármacos , Células Estreladas do Fígado/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Citocinas/metabolismo , Células Endoteliais da Veia Umbilical Humana/metabolismo , Óxido Nítrico/metabolismo
2.
Biochem Biophys Res Commun ; 734: 150591, 2024 Nov 19.
Artigo em Inglês | MEDLINE | ID: mdl-39255745

RESUMO

Breast cancer is a prominent cause of death among women and is distinguished by a high occurrence of metastasis. From this perspective, apart from conventional therapies, several alternative approaches have been researched and explored in recent years, including the utilization of nano-albumin and statin medications like simvastatin. The objective of this study was to prepare albumin nanoparticles incorporating simvastatin by the self-assembly method and evaluate their impact on breast cancer metastasis and apoptosis. The data showed the prepared nanoparticles have a diameter of 185 ± 24nm and a drug loading capacity of 8.85 %. The findings exhibit improved release in a lysosomal-like environment and under acidic pH conditions. MTT data showed that nanoparticles do not exhibit a dose-dependent effect on cells. Additionally, the results from MTT, flow cytometry, and qPCR analyses demonstrated that nanoparticles have a greater inhibitory and lethal effect on MDA-MB-231 cells compared to normal simvastatin. And cause cells to accumulate in the G0/G1 phase, initiating apoptotic pathways by inhibiting cell cycle progression. Nanoparticles containing simvastatin can prevent cell invasion and migration in both monolayer and spheroid models, as compared to simvastatin alone, at microscopic levels and in gene expression. The obtained data clearly showed that, compared to simvastatin, nanoparticles containing simvastatin demonstrated significant efficacy in suppressing the growth, proliferation, invasion, and migration of cancer cells in monolayer (2D) and spheroid (3D) models.


Assuntos
Apoptose , Neoplasias da Mama , Nanopartículas , Sinvastatina , Esferoides Celulares , Sinvastatina/farmacologia , Sinvastatina/química , Sinvastatina/administração & dosagem , Humanos , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/patologia , Neoplasias da Mama/metabolismo , Nanopartículas/química , Feminino , Linhagem Celular Tumoral , Esferoides Celulares/efeitos dos fármacos , Esferoides Celulares/patologia , Apoptose/efeitos dos fármacos , Antineoplásicos/farmacologia , Antineoplásicos/química , Albuminas/química , Albuminas/farmacologia , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos
3.
Mol Genet Metab ; 143(1-2): 108570, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39244853

RESUMO

Smith-Lemli-Opitz syndrome (SLOS) is a rare, multiple malformation/intellectual disability disorder caused by pathogenic variants of DHCR7. DHCR7 catalyzes the reduction of 7-dehydrocholesterol (7DHC) to cholesterol in the final step of cholesterol biosynthesis. This results in accumulation of 7DHC and a cholesterol deficiency. Although the biochemical defect is well delineated and multiple mechanisms underlying developmental defects have been explored, the post developmental neuropathological consequences of altered central nervous system sterol composition have not been studied. Preclinical studies suggest that astroglial activation may occur in SLOS. To determine if astroglial activation is present in individuals with SLOS, we quantified cerebrospinal fluid (CSF) glial fibrillary acidic protein using a Quanterix Simoa® GFAP Discovery Kit for SR-X™. Relative to an age-appropriate comparison group, we found that CSF GFAP levels were elevated 3.9-fold in SLOS (3980 ± 3732 versus 1010 ± 577 pg/ml, p = 0.0184). Simvastatin, a 3-hydroxy-3-methylglutaryl-coenzyme A reductase inhibitor, has previously been shown to increase expression of hypomorphic DHCR7 alleles and in a placebo-controlled trial improved serum sterol levels and decreased irritability. Using archived CSF samples from that prior study, we observed a significant decrease (p = 0.0119) in CSF GFAP levels in response to treatment with simvastatin. Although further work needs to be done to understand the potential contribution of neuroinflammation to SLOS neuropathology and cognitive dysfunction, these data confirm astroglial activation in SLOS and suggest that CSF GFAP may be a useful biomarker to monitor therapeutic responses.


Assuntos
Proteína Glial Fibrilar Ácida , Oxirredutases atuantes sobre Doadores de Grupo CH-CH , Síndrome de Smith-Lemli-Opitz , Síndrome de Smith-Lemli-Opitz/líquido cefalorraquidiano , Síndrome de Smith-Lemli-Opitz/genética , Humanos , Proteína Glial Fibrilar Ácida/líquido cefalorraquidiano , Proteína Glial Fibrilar Ácida/genética , Masculino , Feminino , Criança , Pré-Escolar , Oxirredutases atuantes sobre Doadores de Grupo CH-CH/genética , Adolescente , Lactente , Colesterol/líquido cefalorraquidiano , Astrócitos/metabolismo , Astrócitos/patologia , Desidrocolesteróis/líquido cefalorraquidiano , Adulto , Sinvastatina/farmacologia , Adulto Jovem
4.
BMC Cancer ; 24(1): 1167, 2024 Sep 19.
Artigo em Inglês | MEDLINE | ID: mdl-39300376

RESUMO

BACKGROUND: Metastatic pancreatic ductal adenocarcinoma (mPDAC) patients have very poor prognosis highlighting the urgent need of novel treatments. In this regard, repurposing non-oncology already-approved drugs might be an attractive strategy to offer more-effective treatment easily tested in clinical trials. Accumulating evidence suggests that epigenetic deregulation is a hallmark of cancer contributing to treatment resistance in several solid tumors, including PDAC. Histone deacetylase inhibitors (HDACi) are epigenetic drugs we have investigated preclinically and clinically as anticancer agents. Valproic acid (VPA) is a generic low-cost anticonvulsant and mood stabilizer with HDAC inhibitory activity, and anticancer properties also demonstrated in PDAC models. Statins use was reported to be associated with lower mortality risk in patients with pancreatic cancer and statins have been shown to have a direct antitumor effect when used alone or in combination therapy. We recently showed capability of VPA/Simvastatin (SIM) combination to potentiate the antitumor activity of gemcitabine/nab-paclitaxel in vitro and in vivo PDAC preclinical models. METHODS/DESIGN: VESPA is a patient-centric open label randomized multicenter phase-II investigator-initiated trial, evaluating the feasibility, safety, and efficacy of VPA/SIM plus first line gemcitabine/nab-paclitaxel-based regimens (AG or PAXG) (experimental arm) versus chemotherapy alone (standard arm) in mPDAC patients. The study involves Italian and Spanish oncology centers and includes an initial 6-patients safety run-in-phase. A sample size of 240 patients (120 for each arm) was calculated under the hypothesis that the addition of VPA/SIM to gemcitabine and nab-paclitaxel-based regimens may extend progression free survival from 6 to 9 months in the experimental arm. Secondary endpoints are overall survival, response rate, disease control rate, duration of response, CA 19.9 reduction, toxicity, and quality of life. The study includes a patient engagement plan and complementary biomarkers studies on tumor and blood samples. CONCLUSIONS: VESPA is the first trial evaluating efficacy and safety of two repurposed drugs in oncology such as VPA and SIM, in combination with standard chemotherapy, with the aim of improving mPDAC survival. The study is ongoing. Enrollment started in June 2023 and a total of 63 patients have been enrolled as of June 2024. TRIAL REGISTRATION: EudraCT number: 2022-004154-63; ClinicalTrials.gov identifier NCT05821556, posted 2023/04/20.


Assuntos
Albuminas , Protocolos de Quimioterapia Combinada Antineoplásica , Desoxicitidina , Gencitabina , Paclitaxel , Neoplasias Pancreáticas , Sinvastatina , Ácido Valproico , Adulto , Idoso , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Albuminas/administração & dosagem , Albuminas/uso terapêutico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Carcinoma Ductal Pancreático/tratamento farmacológico , Carcinoma Ductal Pancreático/patologia , Desoxicitidina/análogos & derivados , Desoxicitidina/administração & dosagem , Desoxicitidina/uso terapêutico , Reposicionamento de Medicamentos/métodos , Paclitaxel/administração & dosagem , Paclitaxel/uso terapêutico , Neoplasias Pancreáticas/tratamento farmacológico , Neoplasias Pancreáticas/patologia , Sinvastatina/administração & dosagem , Sinvastatina/uso terapêutico , Ácido Valproico/uso terapêutico , Ácido Valproico/administração & dosagem , Ensaios Clínicos Controlados Aleatórios como Assunto , Estudos Multicêntricos como Assunto , Ensaios Clínicos Fase II como Assunto
5.
Neurochem Res ; 49(3): 732-743, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38063948

RESUMO

Astrocytes have key regulatory roles in central nervous system (CNS), integrating metabolic, inflammatory and synaptic responses. In this regard, type I interferon (IFN) receptor signaling in astrocytes can regulate synaptic plasticity. Simvastatin is a cholesterol-lowering drug that has shown anti-inflammatory properties, but its effects on astrocytes, a main source of cholesterol for neurons, remain to be elucidated. Herein, we investigated the effects of simvastatin in inflammatory and functional parameters of primary cortical and hypothalamic astrocyte cultures obtained from IFNα/ß receptor knockout (IFNα/ßR-/-) mice. Overall, simvastatin decreased extracellular levels of tumor necrosis factor-α (TNF-α) and interleukin-1ß (IL-1ß), which were related to a downregulation in gene expression in hypothalamic, but not in cortical astrocytes. Moreover, there was an increase in anti-inflammatory interleukin-10 (IL-10) in both structures. Effects of simvastatin in inflammatory signaling also involved a downregulation of cyclooxygenase 2 (COX-2) gene expression as well as an upregulation of nuclear factor κB subunit p65 (NFκB p65). The expression of cytoprotective genes sirtuin 1 (SIRT1) and nuclear factor erythroid derived 2 like 2 (Nrf2) was also increased by simvastatin. In addition, simvastatin increased glutamine synthetase (GS) activity and glutathione (GSH) levels only in cortical astrocytes. Our findings provide evidence that astrocytes from different regions are important cellular targets of simvastatin in the CNS, even in the absence of IFNα/ßR, which was showed by the modulation of cytokine production and release, as well as the expression of cytoprotective genes and functional parameters.


Assuntos
Astrócitos , Sinvastatina , Camundongos , Animais , Astrócitos/metabolismo , Sinvastatina/farmacologia , Camundongos Knockout , Fator de Necrose Tumoral alfa/metabolismo , Interferon-alfa/metabolismo , Interferon-alfa/farmacologia , Anti-Inflamatórios/farmacologia , Colesterol/metabolismo , Células Cultivadas
6.
Mol Pharm ; 21(2): 873-882, 2024 Feb 05.
Artigo em Inglês | MEDLINE | ID: mdl-38229228

RESUMO

Prostate cancer (PC), particularly its metastatic castration-resistant form (mCRPC), is a leading cause of cancer-related deaths among men in the Western world. Traditional systemic treatments, including hormonal therapy and chemotherapy, offer limited effectiveness due to tumors' inherent resistance to these therapies. Moreover, they often come with significant side effects. We have developed a delivery method using a tumor-cell-specific heptamethine carbocyanine dye (DZ) designed to transport therapeutic agents directly to tumor cells. This research evaluated simvastatin (SIM) as the antitumor payload because of its demonstrated chemopreventive effects on human cancers and its well-documented safety profile. We designed and synthesized a DZ-SIM conjugate for tumor cell targeting. PC cell lines and xenograft tumor models were used to assess tumor-cell targeting specificity and killing activity and to investigate the corresponding mechanisms. DZ-SIM treatment effectively killed PC cells regardless of their androgen receptor status or inherent therapeutic resistance. The conjugate targeted and suppressed xenograft tumor formation without harming normal cells of the host. In cancer cells, DZ-SIM was enriched in subcellular organelles, including mitochondria, where the conjugate formed adducts with multiple proteins and caused the loss of transmembrane potential, promoting cell death. The DZ-SIM specifically targets PC cells and their mitochondria, resulting in a loss of mitochondrial function and cell death. With a unique subcellular targeting strategy, the conjugate holds the potential to outperform existing chemotherapeutic drugs. It presents a novel strategy to circumvent therapeutic resistance, offering a more potent treatment for mCRPC.


Assuntos
Neoplasias de Próstata Resistentes à Castração , Sinvastatina , Masculino , Humanos , Sinvastatina/farmacologia , Sinvastatina/uso terapêutico , Neoplasias de Próstata Resistentes à Castração/metabolismo , Próstata/metabolismo , Carbocianinas , Linhagem Celular Tumoral
7.
Neuroendocrinology ; 114(10): 934-949, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38964285

RESUMO

INTRODUCTION: The aim of this study was to investigate the autoinflammatory effect and biological behaviour of simvastatin (SIM) on adamantinomatous craniopharyngioma (ACP) cells. METHODS: Craniopharyngiomas imaging, intraoperative observations, and tumour histopathology were employed to investigate the correlation between esters and craniopharyngiomas. Filipin III fluorescent probe verified the validity of SIM on the alternations of synthesized cholesterol in craniopharyngioma cells. The cell counting kit-8 (CCK8) assay detected the impacts of SIM on cell proliferation and determined the IC50 value of tumour cells. Reverse transcription polymerase chain reaction (RT-PCR) measured the expression of inflammatory factors. Flow cytometry technique detected the cell cycle and apoptosis, and cell scratch assay judged the cell migration. Meanwhile, Western blot was adopted to determine the expression of proteins related to inflammation, proliferation, and apoptosis signalling pathways. RESULTS: In the ACP tumour parenchyma, many cholesterol crystalline clefts were observed, and the deposition of esters was closely associated with craniopharyngioma inflammation. After SIM intervention, a reduction in cholesterol synthesis within ACP was noted. RT-PCR analysis revealed SIM inhibited the transcription of inflammatory factors in ACP cells. Western blot analysis demonstrated SIM inhibited nuclear factor-kappa B p65 activation expression while promoted the expressions of Cl-caspase-3 and P38 MAPK. CCK8 assay indicated a decrease in ACP cell activity upon SIM treatment. Scratch assay signified that SIM hindered ACP cell migration. Flow cytometry results suggested that the drug promoted ACP cell apoptosis. CONCLUSION: SIM suppressed the inflammatory response to craniopharyngiomas by inhibiting craniopharyngioma cholesterol synthesis, inhibited proliferation of ACP cells, and promoted their apoptosis.


Assuntos
Apoptose , Proliferação de Células , Craniofaringioma , Neoplasias Hipofisárias , Sinvastatina , Humanos , Craniofaringioma/metabolismo , Craniofaringioma/tratamento farmacológico , Craniofaringioma/patologia , Neoplasias Hipofisárias/metabolismo , Neoplasias Hipofisárias/tratamento farmacológico , Neoplasias Hipofisárias/patologia , Sinvastatina/farmacologia , Apoptose/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Feminino , Masculino , Adulto , Inflamação/metabolismo , Inflamação/tratamento farmacológico , Inflamação/patologia , Linhagem Celular Tumoral , Pessoa de Meia-Idade , Adolescente , Movimento Celular/efeitos dos fármacos , Criança , Inibidores de Hidroximetilglutaril-CoA Redutases/farmacologia , Adulto Jovem
8.
Pharmacol Res ; 200: 107075, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38228255

RESUMO

Claudin-5 (CLDN5) is an essential component of tight junctions (TJs) and is critical for the integrity of the blood-brain barrier (BBB), ensuring homeostasis and protection from damage to the central nervous system (CNS). Currently, many researchers have summarized the role and mechanisms of CLDN5 in CNS diseases. However, it is noteworthy that CLDN5 also plays a significant role in tumor growth and metastasis. In addition, abnormal CLDN5 expression is involved in the development of respiratory diseases, intestinal diseases, cardiac diseases, and diabetic ocular complications. This paper aims to review the structure, expression, and regulation of CLDN5, focusing on its role in tumors, including its expression and regulation, effects on malignant phenotypes, and clinical significance. Furthermore, this paper will provide an overview of the role and mechanisms of CLDN5 in respiratory diseases, intestinal diseases, cardiac diseases, and diabetic ocular complications.


Assuntos
Doenças do Sistema Nervoso Central , Diabetes Mellitus , Cardiopatias , Enteropatias , Neoplasias , Humanos , Claudina-5/genética , Claudina-5/metabolismo , Neoplasias/genética
9.
Pharmacol Res ; : 107466, 2024 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-39419133

RESUMO

Myocardial ischemia causes extensive damage, further exacerbated by reperfusion, a phenomenon called myocardial ischemia/reperfusion injury (MIRI). Nowadays, the pathological mechanisms of MIRI have received extensive attention. Oxidative stress, multiple programmed cell deaths, inflammation and others are all essential pathological mechanisms contributing to MIRI. Mitochondria are the energy supply centers of cells. Numerous studies have found that abnormal mitochondrial function is an essential "culprit" of MIRI, and mitophagy mediated by the phosphatase and tensin homolog (PTEN)-induced kinase 1 (PINK1)/Parkin signaling pathway is an integral part of maintaining mitochondrial function. Therefore, exploring the association between the PINK1/Parkin signaling pathway-mediated mitophagy and MIRI is crucial. This review will mainly summarize the crucial role of the PINK1/Parkin signaling pathway-mediated mitophagy in MIR-induced several pathological mechanisms and various potential interventions that affect the PINK1/Parkin signaling pathway-mediated mitophagy, thus ameliorating MIRI.

10.
Mol Biol Rep ; 51(1): 824, 2024 Jul 18.
Artigo em Inglês | MEDLINE | ID: mdl-39023688

RESUMO

BACKGROUND: Hepatocellular carcinoma (HCC) is a global life-threatening problem and therapeutic interventions are still encountered. IQGAP genes are involved in HCC oncogenesis. The modulatory effect of statins on the expression of IQGAP genes is still unclear. This study aims to study the effect of free SV and chitosan (CS) decorated simvastatin (SV) loaded solid lipid nanoparticles (C-SV-SLNs) on HCC mortality. METHODS AND RESULTS: Plain, SV-SLN, and C-SV- SLN were prepared and characterized in terms of particle size (PS), zeta potential (ZP), and polydispersity index (PDI). The biosafety of different SLN was investigated using fresh erythrocytes, moreover, cytotoxicity was investigated using HepG2 cell lines. The effect of SLNs on IQGAPs gene expression as well as JNK, HDAC6, and HDAC8 activity was investigated using PCR and MOE-docking. The current results displayed that SV-SLNs have nanosized, negative ZP and are homogenous, CS decoration shifts the ZP of SLN into cationic ZP. Furthermore, all SLNs exhibited desirable biosafety in terms of no deleterious effect on erythrocyte integrity. SV solution and SV-SLN significantly increase the mortality of HepG2 compared to undertreated cells, however, the effect of SV-SLN is more pronounced compared to free SV. Remarkably, C-SV-SLN elicits high HepG2 cell mortality compared to free SV and SV-SLN. The treatment of HepG2 cells with SV solution, SV-SLN, or C-SV-SLN significantly upregulates the IQGAP2 gene with repression of IQGAP1 and IQGAP3 genes. MOE-docking studies revealed both SV and tenivastatin exhibit interactions with the active sites of JNK, HDAC6, and HDAC8. Moreover, tenivastatin exhibited greater interactions with magnesium and zinc compared to SV. CONCLUSIONS: This research provides novel insights into the therapeutic potential of SV, SV-SLN and C-SV-SLNs in HCC treatment, modulating critical signaling cascades involving IQGAPs, JNK, and HDAC. The development of C-SV-SLNs presents a promising strategy for effective HCC therapy.


Assuntos
Carcinoma Hepatocelular , Quitosana , Histona Desacetilases , Neoplasias Hepáticas , Nanopartículas , Proteínas Ativadoras de ras GTPase , Humanos , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/tratamento farmacológico , Carcinoma Hepatocelular/patologia , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/tratamento farmacológico , Neoplasias Hepáticas/patologia , Células Hep G2 , Histona Desacetilases/metabolismo , Histona Desacetilases/genética , Quitosana/farmacologia , Proteínas Ativadoras de ras GTPase/genética , Proteínas Ativadoras de ras GTPase/metabolismo , Nanopartículas/química , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Tamanho da Partícula , Lipossomos , Proteínas Repressoras
11.
BMC Pulm Med ; 24(1): 224, 2024 May 08.
Artigo em Inglês | MEDLINE | ID: mdl-38720270

RESUMO

BACKGROUND: Simvastatin (Sim), a hydroxy-methylglutaryl coenzyme A (HMG-CoA) reductase inhibitor, has been widely used in prevention and treatment of cardiovascular diseases. Studies have suggested that Sim exerts anti-fibrotic effects by interfering fibroblast proliferation and collagen synthesis. This study was to determine whether Sim could alleviate silica-induced pulmonary fibrosis and explore the underlying mechanisms. METHODS: The rat model of silicosis was established by the tracheal perfusion method and treated with Sim (5 or 10 mg/kg), AICAR (an AMPK agonist), and apocynin (a NOX inhibitor) for 28 days. Lung tissues were collected for further analyses including pathological histology, inflammatory response, oxidative stress, epithelial mesenchymal transformation (EMT), and the AMPK-NOX pathway. RESULTS: Sim significantly reduced silica-induced pulmonary inflammation and fibrosis at 28 days after administration. Sim could reduce the levels of interleukin (IL)-1ß, IL-6, tumor necrosis factor-α and transforming growth factor-ß1 in lung tissues. The expressions of hydroxyproline, α-SMA and vimentin were down-regulated, while E-cad was increased in Sim-treated rats. In addition, NOX4, p22pox, p40phox, p-p47phox/p47phox expressions and ROS levels were all increased, whereas p-AMPK/AMPK was decreased in silica-induced rats. Sim or AICAR treatment could notably reverse the decrease of AMPK activity and increase of NOX activity induced by silica. Apocynin treatment exhibited similar protective effects to Sim, including down-regulating of oxidative stress and inhibition of the EMT process and inflammatory reactions. CONCLUSIONS: Sim attenuates silica-induced pulmonary inflammation and fibrosis by downregulating EMT and oxidative stress through the AMPK-NOX pathway.


Assuntos
Proteínas Quinases Ativadas por AMP , Fibrose Pulmonar , Dióxido de Silício , Sinvastatina , Animais , Masculino , Ratos , Acetofenonas/farmacologia , Aminoimidazol Carboxamida/análogos & derivados , Aminoimidazol Carboxamida/farmacologia , Proteínas Quinases Ativadas por AMP/metabolismo , Modelos Animais de Doenças , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Inibidores de Hidroximetilglutaril-CoA Redutases/farmacologia , Pulmão/patologia , Pulmão/efeitos dos fármacos , Pulmão/metabolismo , NADPH Oxidase 4/metabolismo , NADPH Oxidases/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Pneumonia/induzido quimicamente , Pneumonia/prevenção & controle , Pneumonia/tratamento farmacológico , Pneumonia/metabolismo , Pneumonia/patologia , Fibrose Pulmonar/induzido quimicamente , Fibrose Pulmonar/tratamento farmacológico , Ribonucleotídeos/farmacologia , Transdução de Sinais/efeitos dos fármacos , Silicose/tratamento farmacológico , Silicose/patologia , Silicose/metabolismo , Sinvastatina/farmacologia , Fator de Crescimento Transformador beta1/metabolismo
12.
Pharmacology ; 109(1): 43-51, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38016432

RESUMO

INTRODUCTION: There is still no effective treatment for heart failure with preserved left ventricular ejection fraction (HFpEF), and therapies to improve prognosis are urgently needed. Clinical studies in patients with HFpEF have shown that statins and HMG-CoA reductase inhibitors may reduce their mortality rate. However, the mechanisms underlying the effects of statins on HFpEF remain unknown. In the present study, we examined whether simvastatin administration inhibits the development of cardiac fibrosis in HFpEF model mice. We further examined the contribution of the Smad and mitogen-activated protein (MAP) kinase pathways to the transforming growth factor-ß (TGF-ß) signaling pathway in the development of HFpEF. METHODS: HFpEF animals were prepared by feeding C57BL/6 N mice a high-fat diet and providing water containing N[w]-nitro-l-arginine methyl ester hydrochloride (l-NAME) for 15 weeks. Simvastatin (30 mg/kg/day) or vehicle was administered orally daily during the experimental period. Cardiac function was measured by echocardiography, and cardiac fibrosis was evaluated by Masson's trichrome staining. Changes in the TGF-ß signaling proteins in myocardial tissue were examined by Western blotting. RESULTS: A high-fat diet and l-NAME solution load induced cardiac diastolic dysfunction with cardiac fibrosis. Simvastatin treatment markedly attenuated cardiac fibrosis and reduced cardiac diastolic dysfunction. In addition, simvastatin prevented the increase in phosphorylation levels of Smad (Smad2 and Smad3) and MAPK (c-Raf, Erk1/2) pathway proteins downstream of the TGF-ß receptor in cardiac tissue. CONCLUSIONS: Our present study demonstrated that simvastatin attenuated diastolic dysfunction by reducing cardiac fibrosis in HFpEF hearts. Furthermore, our findings suggest that the mechanisms by which simvastatin attenuates HFpEF development involve, at least in part, inhibition of the TGF-ß signaling pathway, which is activated in the HFpEF heart.


Assuntos
Cardiopatias , Insuficiência Cardíaca , Inibidores de Hidroximetilglutaril-CoA Redutases , Humanos , Camundongos , Animais , Insuficiência Cardíaca/tratamento farmacológico , Insuficiência Cardíaca/metabolismo , Sinvastatina/farmacologia , Sinvastatina/uso terapêutico , Volume Sistólico , Fator de Crescimento Transformador beta/metabolismo , Inibidores de Hidroximetilglutaril-CoA Redutases/farmacologia , Inibidores de Hidroximetilglutaril-CoA Redutases/uso terapêutico , Função Ventricular Esquerda , Camundongos Endogâmicos C57BL , Cardiopatias/tratamento farmacológico , Transdução de Sinais , Fibrose
13.
Pediatr Dermatol ; 2024 Aug 21.
Artigo em Inglês | MEDLINE | ID: mdl-39168486

RESUMO

Conradi-Hünermann-Happle syndrome is a rare genodermatosis affecting cholesterol metabolism caused by pathogenic variants in the emopamil binding protein (EBP) gene. It presents with skin, skeletal, and ophthalmological alterations. Cutaneous findings include hyperkeratotic lesions following Blaschko lines that subsequently improve leaving scarring alopecia and patches of atrophy. The purpose of this case report is to present a case of a patient treated with simvastatin-cholesterol ointment.

14.
BMC Musculoskelet Disord ; 25(1): 748, 2024 Sep 18.
Artigo em Inglês | MEDLINE | ID: mdl-39294613

RESUMO

BACKGROUND: Osteoporosis is a major health concern for postmenopausal women, and the effect of simvastatin (Sim) on bone metabolism is controversial. This study aimed to investigate the effect of simvastatin on the bone microstructure and bone mechanical properties in ovariectomized (OVX) mice. METHODS: 24 female C57BL/6J mice (8-week-old) were randomly allocated into three groups including the OVX + Sim group, the OVX group and the control group. At 8 weeks after operation, the L4 vertebral bones were dissected completely for micro-Computed Tomography (micro-CT) scanning and micro-finite element analysis (µFEA). The differences between three groups were compared using ANOVA with a LSD correction, and the relationship between bone microstructure and mechanical properties was analyzed using linear regression. RESULTS: Bone volume fraction, trabecular number, connectivity density and trabecular tissue mineral density in the OVX + Sim group were significantly higher than those in the OVX group (P < 0.05). For the mechanical properties detected via µFEA, the OVX + Sim group had lower total deformation, equivalent elastic strain and equivalent stress compared to the OVX group (P < 0.05). In the three groups, the mechanical parameters were significantly correlated with bone volume fraction and trabecular bone mineral density. CONCLUSIONS: The findings suggested that simvastatin had a potential role in the treatment of osteoporosis. The results of this study could guide future research on simvastatin and support the development of simvastatin-based treatments to improve bone health.


Assuntos
Densidade Óssea , Análise de Elementos Finitos , Camundongos Endogâmicos C57BL , Ovariectomia , Sinvastatina , Microtomografia por Raio-X , Sinvastatina/farmacologia , Animais , Feminino , Camundongos , Densidade Óssea/efeitos dos fármacos , Osteoporose/diagnóstico por imagem , Osteoporose/tratamento farmacológico , Osteoporose/prevenção & controle , Inibidores de Hidroximetilglutaril-CoA Redutases/farmacologia , Modelos Animais de Doenças , Vértebras Lombares/efeitos dos fármacos , Vértebras Lombares/diagnóstico por imagem , Fenômenos Biomecânicos/efeitos dos fármacos
15.
Allergol Immunopathol (Madr) ; 52(5): 36-43, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39278849

RESUMO

The aim of this study was to elucidate the therapeutic effect of simvastatin on experimental autoimmune encephalomyelitis (EAE) by regulating the balance between Th17 and Treg cells in mice. C57BL/6 mice were randomly divided into four groups: normal group, EAE group, simvastatin (2 and 10 mg/kg) group, and AG490 group (with AG490 serving as the positive control). Neurological function scores of mice were assessed daily. The four groups received treatments of normal saline, normal saline, and simvastatin (2 and 10 mg/kg), respectively. In the AG490 group, mice were injected intraperitoneally with AG490 (1 mg) every other day, and treatment was halted after 3 weeks. The spinal cord was stained with hematoxylin and eosin (H&E), and immunohistochemical staining for retinoic acid receptor-related orphan receptor γ(RORγ) and Foxp3 (Foxp3) was performed. Spleen samples were taken for Th17 and Treg analysis using flow cytometry. The levels of interleukin-17 and transforming growth factor-ß (TGF-ß) were detected using enzyme-linked immunosorbent assay (ELISA). In the simvastatin and AG490 groups, recovery from neurological impairment was earlier compared to the EAE group, and the symptoms were notably improved. Both simvastatin and AG490 reduced focal inflammation, decreased RORγ-positive cell infiltration, and significantly increased the number of FOXP3-positive cells. The number of Th17 cells and the level of IL-17 in the spleen were decreased in the simvastatin and AG490 treatment groups, while the number of Treg cells and TGF-ß levels were significantly increased across all treatment groups. Simvastatin exhibits anti-inflammatory and immunomodulatory effects, potentially alleviating symptoms of neurological dysfunction of EAE. Regulating the balance between Th17 and Treg may represent a therapeutic mechanism for simvastatin in treating EAE.


Assuntos
Encefalomielite Autoimune Experimental , Camundongos Endogâmicos C57BL , Sinvastatina , Linfócitos T Reguladores , Células Th17 , Animais , Encefalomielite Autoimune Experimental/tratamento farmacológico , Encefalomielite Autoimune Experimental/imunologia , Células Th17/imunologia , Células Th17/efeitos dos fármacos , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/efeitos dos fármacos , Sinvastatina/farmacologia , Sinvastatina/administração & dosagem , Camundongos , Feminino , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares/metabolismo , Interleucina-17/metabolismo , Fatores de Transcrição Forkhead/metabolismo , Medula Espinal/imunologia , Medula Espinal/efeitos dos fármacos , Medula Espinal/patologia , Humanos , Fator de Crescimento Transformador beta/metabolismo , Modelos Animais de Doenças
16.
Clin Oral Investig ; 28(1): 71, 2024 Jan 04.
Artigo em Inglês | MEDLINE | ID: mdl-38172458

RESUMO

OBJECTIVES: The present study goal was to assess clinically and radiographically using simvastatin (SMV) loaded xenograft for guided bone regeneration (GBR) around simultaneously placed implants with alveolar ridge splitting in patients with horizontally atrophic jaw defect. MATERIALS AND METHODS: Randomized distribution of the twenty-two patients into two groups (11 patients each) was performed. Group I participants received alveolar ride splitting (ARS) with GBR using SMV gel mixed bone graft and a barrier membrane with simultaneous implant placement. Group II received the same treatment protocol without SMV gel. At the baseline, 6- and 9-months post-surgery, clinical and radiological alterations were assessed. RESULTS: Six months after therapy, PES records of group I were statistically significantly improved than those of group II (P < .001). Group I exhibited statistically significant expansion of the alveolar ridge over group II after 6 and 9 months (P < .001). When compared to group II over the evaluation interval between 6 and 9 months, group I demonstrated statistically substantially minimal loss of the mean marginal bone level (P < .001). At the 6- and 9-month observation periods, bone density gain was considerably higher in group I than that in group II (P < .001). CONCLUSION: Alveolar ridge splitting along with GBR-augmented SMV improve the clinical and radiographical outcomes around dental implant over GBR alone. CLINICAL RELEVANCE: Augmenting GBR with SMV in alveolar ridge splitting could boost implant osseointegration and enhance peri-implant tissue changes. CLINICAL TRIAL REGISTRATION: NCT05020405.


Assuntos
Aumento do Rebordo Alveolar , Implantes Dentários , Humanos , Implantação Dentária Endóssea/métodos , Xenoenxertos , Aumento do Rebordo Alveolar/métodos , Processo Alveolar/cirurgia , Regeneração Óssea
17.
Clin Oral Investig ; 28(1): 86, 2024 Jan 10.
Artigo em Inglês | MEDLINE | ID: mdl-38195898

RESUMO

OBJECTIVES: Conducting a scoping review (SR) to assess scientific evidence for topical simvastatin's impact on alveolar bone regeneration and determine its level of support for clinical applications. MATERIALS AND METHODS: This SR followed the PRISMA-ScR and OSF registries protocol; systematic searching was conducted on MEDLINE/PubMed, Cochrane, Embase, Scopus, Web of Science, and LILACS, to identify relevant articles until June 2023. Inclusion criteria covered clinical trials, case series, prospective and retrospective studies, along with in vivo investigations, involving participants of any sex and age. RESULTS: Out of 1312 identified studies, 20 (9 in vivo, 11 RCTs) met inclusion criteria. RCTs focused on third molar extraction, in vivo on mandibular incisor surgery. The majority of RCTs employed a collagen sponge and a simvastatin concentration of 10mg; conversely, most in vivo studies favored polylactide-co-glycolide and a 2 mg simvastatin concentration. RCTs had 3-month follow-ups; in vivo, studies extended to 8 weeks. Seven RCTs assessed pain outcomes, simvastatin did not significantly affect pain in six studies. Among four RCTs on postoperative swelling, only two observed a significant increase in the simvastatin group. In general, positive bone formation and the absence of adverse effects directly linked to topical simvastatin were observed across the study models. CONCLUSIONS: Intra-alveolar simvastatin post-tooth extraction has been to be shown to be effective and safe for preserving alveolar bone, with varied concentrations and carriers, with no significant adverse effects. CLINICAL RELEVANCE: This review provides critical insights into the effects of simvastatin on alveolar bone regeneration, informing potential benefits and possible challenges associated with its post-extraction application. OSF REGISTRY PROTOCOL: osf.io/q3bnf.


Assuntos
Incisivo , Extração Dentária , Humanos , Estudos Prospectivos , Estudos Retrospectivos , Dor
18.
Clin Oral Investig ; 28(4): 210, 2024 Mar 12.
Artigo em Inglês | MEDLINE | ID: mdl-38467945

RESUMO

OBJECTIVES: The present study aimed to assess clinically and radiographically the usage of autogenous tooth bone graft (ATBG) combined with and without Simvastatin (SMV) around immediately placed dental implants in periodontally compromised sites. METHODS: Thirty-nine patients required a single extraction of periodontally compromised tooth were divided into three groups (13 patients each). Group I received immediate implant placement (IIP) without grafting. Group II received IIP with ATBG filling the gap around IIP. Group III received SMV gel mixed with ATBG around IIP. Radiographic changes were reported at the baseline, 6-, and 12-months post-surgery. RESULTS: All implants achieved the success criteria with no complications. At 6- and 12-months post-surgery, group III showed a statistically lower mean ridge width loss compared to Group I and Group II (P < .001). Group II revealed less reduction in the mean alveolar ridge width compared to group I (P < .001). Group III showed a statistically significantly less MBL loss than group I and group II (P < .001). All groups showed a statistically significant increase in BD gain compared to baseline (P < .001). Group III showed statistically significant high BD compared to group II (P < .001). Group II showed statistically significantly higher mean BD gain than that of group I (P < .001). CONCLUSION: SMV combined with ATBG boosts the hard tissue parameters around dental implants over ATBG alone. Clinical trial registration was on August 1, 2021 (NCT04992416). CLINICAL RELEVANCE: ATBG with SMV in periodontally compromised sites could improve implant osseointegration and promote favorable changes in peri-implant tissues.


Assuntos
Perda do Osso Alveolar , Implantes Dentários , Humanos , Processo Alveolar/cirurgia , Osseointegração , Extração Dentária , Transplante Ósseo , Alvéolo Dental/cirurgia , Implantação Dentária Endóssea , Perda do Osso Alveolar/diagnóstico por imagem , Perda do Osso Alveolar/cirurgia , Seguimentos
19.
J Biol Phys ; 50(3-4): 255-269, 2024 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-38935192

RESUMO

Bone is a complex tissue that fulfills the role of a resistance structure. This quality is most commonly assessed by bone densitometry, but bone strength may not only be related to bone mineral density but also to the preservation of bone cytoarchitectonics. The study included two groups of rats, ovariectomized and non-ovariectomized. Each group was divided into three batches: control, simvastatin-treated, and fenofibrate-treated. In the ovariectomized group, hypolipidemic treatment was instituted at 12 weeks post ovariectomy. One rat from each of the 6 batches was sacrificed 8 weeks after the start of treatment in the group. The experimental study was performed using a Bruker Minispec mq 20 spectrometer operating at a frequency of 20 MHz, subsequently also performed by 1H T2-T2 molecular exchange maps. The results were represented by T2-T2 molecular exchange maps that showed, comparatively, both pore size and their interconnectivity at the level of the femoral epiphysis, being able to evaluate both the effect of estrogen on bone tissue biology and the effect of the lipid-lowering medication, simvastatin, and fenofibrate, in both the presence and absence of estrogen. T2-T2 molecular exchange maps showed that the absence of estrogen results in an increase in bone tissue pore size and interconnectivity. In the presence of estrogen, lipid-lowering medication, both simvastatin and fenofibrate alter bone tissue cytoarchitectonics by reducing pore interconnectivity. In the absence of estrogen, fenofibrate improves bone tissue cytoarchitectonics, the T2-T2 molecular exchange map being similar to that of non-osteoporotic bone tissue.


Assuntos
Sinvastatina , Animais , Ratos , Feminino , Sinvastatina/farmacologia , Osso e Ossos , Fenofibrato/farmacologia , Ovariectomia , Espectroscopia de Prótons por Ressonância Magnética , Ratos Wistar , Fêmur
20.
Int J Mol Sci ; 25(4)2024 Feb 17.
Artigo em Inglês | MEDLINE | ID: mdl-38397055

RESUMO

Statins, widely prescribed for lipid disorders, primarily target 3-hydroxy-3-methylglutaryl-coenzyme A (HMG-CoA) reductase competitively and reversibly, resulting in reduced low-density lipoprotein cholesterol (LDL-C). This mechanism proves effective in lowering the risk of lipid-related diseases such as ischemic cerebrovascular and coronary artery diseases. Beyond their established use, statins are under scrutiny for potential applications in treating bone diseases. The focus of research centers mainly on simvastatin, a lipophilic statin demonstrating efficacy in preventing osteoporosis and aiding in fracture and bone defect healing. Notably, these effects manifest at elevated doses (20 mg/kg/day) of statins, posing challenges for systematic administration due to their limited bone affinity. Current investigations explore intraosseous statin delivery facilitated by specialized carriers. This paper outlines various carrier types, characterizing their structures and underscoring various statins' potential as local treatments for bone diseases.


Assuntos
Inibidores de Hidroximetilglutaril-CoA Redutases , Osteoporose , Humanos , Inibidores de Hidroximetilglutaril-CoA Redutases/farmacologia , Inibidores de Hidroximetilglutaril-CoA Redutases/uso terapêutico , Sinvastatina/farmacologia , Sinvastatina/uso terapêutico , LDL-Colesterol , Osteoporose/tratamento farmacológico , Osso e Ossos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA