Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 4.951
Filtrar
1.
Phytomedicine ; 134: 156017, 2024 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-39265443

RESUMO

BACKGROUND: Currently, there is a lack of validated pharmacological interventions for non-alcoholic fatty liver disease (NAFLD), which is characterized by the accumulation of hepatic triglyceride. Zhimu-Huangbai (ZH) herb-pair is a traditional Chinese medicine that regulates glucose and lipid metabolism disorders. However, the precise mechanisms underlying the preventive effects of hepatic triglyceride induced by high-fat diet (HFD) remain elusive. PURPOSE: The study aimed to examine the impact of ZH herb-pair on NAFLD in mice and explore the underlying mechanisms, particularly its effects on endoplasmic reticulum (ER) stress and lipid metabolism. METHODS: NAFLD was induced in mice using HFD, and the treated mice were orally administered ZH, metformin (Glucophage) or lovastatin. The lipid metabolism factors, ER stress markers, and the unfolded protein response (UPR) branch factors were measured using immunohistochemistry, western blotting or qRT-PCR. Co-Immunoprecipitation (CoIP) was performed to reveal the connection between SCAP and SREBP-1c. Tunicamycin (TM) and plasmid delivery were used to induce acute ER stress or crease XBP1 gain function models. The main compounds in ZH binding to IRE1α protein were studied by molecular docking and cellular thermal shift assay (CETSA). RESULTS: Treatment with ZH significantly ameliorated hepatic steatosis and reduced lipid synthesis process mainly inhibiting the expression of mature active form of SREBP-1c through relieving ER stress. The expression of IRE1α and XBP1s was inhibited after treatment with ZH. In addition, ZH improved the fatty liver phenotype caused by XBP1 overexpression via decreasing srebp1c transcription. In vitro experimental results suggested that the main compounds in ZH decreased cellular TG contents. Mechanistically, ZH targeted IRE1α and inhibited XBP1s mRNA expression to relieve ER stress and inhibit SREBP-1c production. CONCLUSIONS: ZH herb-pair can protect against NAFLD by reducing the expression of SREBP-1c, in part, via regulating IRE1α/XBP1s pathway.


Assuntos
Medicamentos de Ervas Chinesas , Estresse do Retículo Endoplasmático , Endorribonucleases , Camundongos Endogâmicos C57BL , Hepatopatia Gordurosa não Alcoólica , Proteínas Serina-Treonina Quinases , Proteína de Ligação a Elemento Regulador de Esterol 1 , Proteína 1 de Ligação a X-Box , Animais , Proteína de Ligação a Elemento Regulador de Esterol 1/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteína 1 de Ligação a X-Box/metabolismo , Medicamentos de Ervas Chinesas/farmacologia , Hepatopatia Gordurosa não Alcoólica/tratamento farmacológico , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Masculino , Endorribonucleases/metabolismo , Dieta Hiperlipídica/efeitos adversos , Metabolismo dos Lipídeos/efeitos dos fármacos , Camundongos , Humanos , Fígado/efeitos dos fármacos , Fígado/metabolismo , Triglicerídeos/metabolismo , Lovastatina/farmacologia , Resposta a Proteínas não Dobradas/efeitos dos fármacos , Metformina/farmacologia , Transdução de Sinais/efeitos dos fármacos
2.
Food Res Int ; 195: 114961, 2024 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-39277234

RESUMO

Monacolin K (MK), also known as lovastatin, is a polyketide compound with the ability to reduce plasma cholesterol levels and many other bio-activities. Red yeast rice (also named Hongqu) rich in MK derived from Monascus fermentation has attracted widespread attention due to its excellent performance in reducing blood lipids. However, industrial Monascus fermentation suffers from the limitations such as low yield of MK, long fermentation period, and susceptibility to contamination. In this study, we firstly blocked the competitive pathway of MK biosynthesis to create polyketide synthase gene pigA (the key gene responsible for the biosynthesis of Monascus azaphilone pigments) deficient strain A1. Then, based on the strategies to increase precursor supply for MK biosynthesis, acetyl-CoA carboxylase gene acc overexpression strains C1 and C2 were constructed with WT and A1 as the parent, respectively. Finally, histone deacetylase gene hos2 overexpression strain H1 was constructed by perturbation of histone acetylation modification. HPLC detection revealed all these four strains significantly increased their abilities to produce MK. After 14 days of solid-state fermentation, the MK yields of strains A1, C1, C2, and H1 reached 2.03 g/100 g, 1.81 g/100 g, 2.45 g/100 g and 2.52 g/100 g, which increased by 28.5 %, 14.7 %, 43.9 % and 36.1 % compared to WT, respectively. RT-qPCR results showed that overexpression of hos2 significantly increased the expression level of almost all genes responsible for MK biosynthesis after 5-day growth. Overall, the abilities of these strains to produce MK has been greatly improved, and MK production period has been shortened to 14 days from 20 days, providing new approaches for efficient production of Hongqu rich in MK.


Assuntos
Fermentação , Histonas , Lovastatina , Monascus , Monascus/metabolismo , Monascus/genética , Acetilação , Histonas/metabolismo , Acetil-CoA Carboxilase/metabolismo , Acetil-CoA Carboxilase/genética , Policetídeo Sintases/genética , Policetídeo Sintases/metabolismo , Hipolipemiantes/farmacologia , Produtos Biológicos/metabolismo , Histona Desacetilases/metabolismo , Histona Desacetilases/genética
3.
Nutrients ; 16(17)2024 Sep 04.
Artigo em Inglês | MEDLINE | ID: mdl-39275298

RESUMO

The risk of cardiovascular disease (CVD) is approximately doubled in subjects with hypercholesterolemia compared to those with normal blood cholesterol levels. Monacolin K (MK), the main active substance in rice fermented by the Monascus purpureus, acts on cholesterol metabolism. Rice also contains other bioactive compounds such as γ-oryzanol (OZ) and γ-aminobutyric acid (GABA). In a randomized, placebo-controlled, double-blind trial, the efficacy and tolerability of a food supplement (FS) based on an ingredient standardized to contain monacolins (4.5%), OZ, and GABA were evaluated in subjects with mild dyslipidemia. For the duration of the trial, enrolled subjects (n = 44, each group) received the FS or placebo and were instructed to use an isocaloric diet. Compared to the placebo group, after a 3 months of the FS, the mean low-density lipoprotein cholesterol and mean TC values were reduced by 19.3 and 8.3%, respectively, while the mean high-density lipoprotein cholesterol value increased by 29.3%. On average, the subjects shifted from very high to moderate CVD risk. Glucose metabolism and hepatic and renal parameters did not change after the treatment and no adverse events were reported. Guidelines to handle hypercholesterolemia with food supplements in specific clinical settings are needed to better manage mild dyslipidemia.


Assuntos
Suplementos Nutricionais , Dislipidemias , Fenilpropionatos , Ácido gama-Aminobutírico , Humanos , Método Duplo-Cego , Masculino , Feminino , Pessoa de Meia-Idade , Dislipidemias/tratamento farmacológico , Dislipidemias/sangue , Fenilpropionatos/uso terapêutico , Adulto , Lovastatina/uso terapêutico , LDL-Colesterol/sangue , Resultado do Tratamento , HDL-Colesterol/sangue
4.
Int J Surg ; 110(8): 4716-4726, 2024 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-39143707

RESUMO

BACKGROUND: In recent years, the widespread use of lipid-lowering drugs, especially statins, has attracted people's attention. Statin use may be potentially associated with a reduced risk of breast cancer. OBJECTIVE: To explore the relationship between statin use and cancer risk. And further explore the potential role of statins in the adjuvant treatment of breast cancer. METHODS: Data for the Mendelian randomization portion of the study were obtained from genome-wide association studies of common cancers in the UK Biobank and FinnGen studies and from the Global Lipid Genetics Consortium's low density lipoprotein (LDL). In addition, the impacts of statins and chemotherapy drugs on breast cancer were examined using both in vitro and in vivo models, with particular attention to the expression levels of the immune checkpoint protein PD-L1 and its potential to suppress tumor growth. RESULTS: Data from about 3.8 million cancer patients and ~1.3 million LDL-measuring individuals were analyzed. Genetically proxied HMGCR inhibition (statins) was associated with breast cancer risk reduction (P=0.0005). In vitro experiments showed that lovastatin significantly inhibited paclitaxel-induced PD-L1 expression and assisted paclitaxel in suppressing tumor cell growth. Furthermore, the combination therapy involving lovastatin and paclitaxel amplified CD8+ T-cell infiltration, bolstering their tumor-killing capacity and enhancing in vivo efficacy. CONCLUSION: The utilization of statins is correlated with improved prognoses for breast cancer patients and may play a role in facilitating the transition from cold to hot tumors. Combination therapy with lovastatin and paclitaxel enhances CD8+ T-cell activity and leads to better prognostic characteristics.


Assuntos
Antígeno B7-H1 , Neoplasias da Mama , Linfócitos T CD8-Positivos , Inibidores de Hidroximetilglutaril-CoA Redutases , Paclitaxel , Neoplasias da Mama/tratamento farmacológico , Antígeno B7-H1/antagonistas & inibidores , Antígeno B7-H1/metabolismo , Feminino , Paclitaxel/farmacologia , Paclitaxel/administração & dosagem , Humanos , Inibidores de Hidroximetilglutaril-CoA Redutases/farmacologia , Prognóstico , Linfócitos T CD8-Positivos/efeitos dos fármacos , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Camundongos , Animais , Lovastatina/farmacologia , Lovastatina/administração & dosagem , Estudo de Associação Genômica Ampla
5.
J Gene Med ; 26(8): e3722, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-39117601

RESUMO

BACKGROUND: Lovastatin, a type of statin usually considered as a lipid-lowering drug that lowers blood cholesterol and low-density lipoprotein cholesterol levels, has been rediscovered to have anticancer activity. Fewer studies exist regarding the effect of lovastatin on esophageal squamous cell carcinoma (ESCC). METHODS: Here, we report that lovastatin shows anticancer effect on ESCC By affecting the mitochondrial autophagy pathway. Moreover, based on proteomics and computer molecular simulations found that RAB38 and RAB27A may be a target of lovastatin. RESULTS: We observed that autophagy of mitochondria is inhibited by lovastatin, affecting esophageal squamous cell proliferation. There is a possible link between the expression of RAB38, RAB27A and immune cell invasion in esophageal cancer. CONCLUSIONS: These results demonstrate the huge potential of lovastatin as an RAB38, RAB27A inhibitor in esophageal cancer chemotherapy and chemoprevention.


Assuntos
Autofagia , Proliferação de Células , Neoplasias Esofágicas , Carcinoma de Células Escamosas do Esôfago , Lovastatina , Proteômica , Lovastatina/farmacologia , Humanos , Carcinoma de Células Escamosas do Esôfago/tratamento farmacológico , Carcinoma de Células Escamosas do Esôfago/metabolismo , Carcinoma de Células Escamosas do Esôfago/patologia , Proliferação de Células/efeitos dos fármacos , Proteômica/métodos , Neoplasias Esofágicas/tratamento farmacológico , Neoplasias Esofágicas/metabolismo , Neoplasias Esofágicas/patologia , Linhagem Celular Tumoral , Autofagia/efeitos dos fármacos , Proteínas rab de Ligação ao GTP/metabolismo , Mitocôndrias/metabolismo , Mitocôndrias/efeitos dos fármacos , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Simulação de Acoplamento Molecular
6.
J Nat Med ; 78(4): 845-848, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-38834898

RESUMO

Recently, health hazards, such as kidney damage, have been reported owing to the ingestion of a health food product, so-called "foods with functional claims (FFC)'', containing beni-koji (red yeast rice). Although not an expected compound in the FFC, the detection of puberulic acid has also been reported. Further investigations of these health food products, such as the identification of other unintended compounds and clarifying the health impacts of puberulic acid, are required. To clarify the causes of these health issues, we investigated the presence of unintended compounds in the FFC containing beni-koji using comprehensive instrumental analyses. Using differential analysis, novel compounds 1 and 2 were detected as unexpected components between the samples with and without adverse event reports. Although limited to the samples available for analyses in this study, both compounds 1 and 2 were detected in all the samples that also contained puberulic acid. Compounds 1 and 2, with molecular formulas of C23H34O7 and C28H42O8, respectively, may be lovastatin derivatives. Their structures were confirmed using NMR analyses and are novel natural compounds. For definitive confirmation, we are in the process of synthesizing compounds 1 and 2 from lovastatin. The route of contamination of these compounds are currently under investigation. The findings of this study could be used to address the growing health hazards associated with health food products.


Assuntos
Produtos Biológicos , Produtos Biológicos/química , Humanos , Alimento Funcional , Estrutura Molecular , Lovastatina , Espectroscopia de Ressonância Magnética
7.
Expert Opin Drug Deliv ; 21(5): 779-796, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38795359

RESUMO

BACKGROUND: Controlled and targeted drug delivery to treat nonalcoholic fatty liver disease (NAFLD) can benefit from additive attributes of natural formulation ingredients incorporated into the drug delivery vehicles. METHODS: Lovastatin (LVN) loaded, bile acid (BA) and fatty acid (FA) integrated nanoemulsomes (NES) were formulated by thin layer hydration technique for synergistic and targeted delivery of LVN to treat NAFLD. Organic phase NES was comprised of stearic acid with garlic (GL) and ginger (GR) oils, separately. Ursodeoxycholic acid and linoleic acid were individually incorporated as targeting moieties. RESULTS: Stability studies over 90 days showed average NES particle size, surface charge, polydispersity index, and entrapment efficiency values of 270 ± 27.4 nm, -23.8 ± 3.5 mV, 0.2 ± 0.04 and 81.36 ± 3.4%, respectively. Spherical NES were observed under a transmission electron microscope. In-vitro LVN release depicted non-fickian release mechanisms from GL and GR oils-based NES. Ex-vivo permeation of BA/FA integrated NES through isolated rat intestines showed greater flux than non-integrated ones. CONCLUSION: Liver histopathology of experimental rats together with in-vivo lipid profiles and liver function tests illustrated that these NES possess the clinical potential to be promising drug carriers for NAFLD.


Assuntos
Ácidos e Sais Biliares , Sistemas de Liberação de Medicamentos , Estabilidade de Medicamentos , Emulsões , Ácidos Graxos , Lovastatina , Hepatopatia Gordurosa não Alcoólica , Tamanho da Partícula , Animais , Hepatopatia Gordurosa não Alcoólica/tratamento farmacológico , Ratos , Ácidos e Sais Biliares/química , Masculino , Lovastatina/administração & dosagem , Lovastatina/farmacocinética , Lovastatina/química , Ácidos Graxos/química , Ácidos Graxos/administração & dosagem , Nanopartículas/química , Ratos Sprague-Dawley , Portadores de Fármacos/química
8.
Atherosclerosis ; 395: 117573, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38796407

RESUMO

BACKGROUND AND AIMS: Activation of vascular smooth muscle cell inflammation is recognised as an important early driver of vascular disease. We have previously identified the let-7 miRNA family as important regulators of inflammation in in vitro and in vivo models of atherosclerosis. Here we investigated a dual statin/let-7d-5p miRNA combination therapy approach to target human aortic SMC (HAoSMC) activation and inflammation. METHODS: In vitro studies using primary HAoSMCs were performed to investigate the effects of let-7d-5p miRNA overexpression and inhibition. HAoSMCs were treated with combinations of the inflammatory cytokine tumor necrosis factor-α (TNF-α), and atorvastatin or lovastatin. HAoSMC Bulk RNA-seq transcriptomics of HAoSMCs revealed downstream regulatory networks modulated by let-7d-5p miRNA overexpression and statins. Proteome profiler cytokine array, Western blotting and quantitative PCR analyses were performed on HAoSMCs to validate key findings. RESULTS: Let-7d-5p overexpression significantly attenuated TNF-α-induced upregulation of IL-6, ICAM1, VCAM1, CCL2, CD68, MYOCD gene expression in HAoSMCs (p<0.05). Statins (atorvastatin, lovastatin) significantly attenuated inflammatory gene expression and upregulated Let-7d levels in HAoSMCs (p<0.05). Bulk RNA-seq analysis of a dual Let-7d-5p overexpression/statin therapy in HAoSMCs revealed that let-7d-5p activation and statins converge on key inflammatory pathways (IL-6, IL-1ß, TNF-α, IFN-γ). Let-7d-5p overexpression led to reduced expression of the ox-LDL receptor OLR1, and this was associated with lower ox-LDL uptake in HAoSMCs. In silico analysis of smooth muscle cell phenotypic switching shows that overexpression of let-7d-5p in HAoSMCs maintains a contractile phenotype. CONCLUSIONS: Targeting the Let-7 network alongside statins can modulate HAoSMC activation and attenuate key inflammatory pathway signals.


Assuntos
Aorta , Atorvastatina , Inibidores de Hidroximetilglutaril-CoA Redutases , MicroRNAs , Músculo Liso Vascular , Miócitos de Músculo Liso , Fenótipo , Transdução de Sinais , MicroRNAs/metabolismo , MicroRNAs/genética , Humanos , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/efeitos dos fármacos , Músculo Liso Vascular/patologia , Miócitos de Músculo Liso/metabolismo , Miócitos de Músculo Liso/efeitos dos fármacos , Aorta/metabolismo , Aorta/patologia , Atorvastatina/farmacologia , Inibidores de Hidroximetilglutaril-CoA Redutases/farmacologia , Células Cultivadas , Fator de Necrose Tumoral alfa/metabolismo , Inflamação/metabolismo , Inflamação/genética , Lovastatina/farmacologia , Mediadores da Inflamação/metabolismo , Regulação da Expressão Gênica , Citocinas/metabolismo
9.
Microb Cell Fact ; 23(1): 134, 2024 May 09.
Artigo em Inglês | MEDLINE | ID: mdl-38724934

RESUMO

BACKGROUND: Lovastatin has widespread applications thanks to its multiple pharmacological effects. Fermentation by filamentous fungi represents the major way of lovastatin production. However, the current lovastatin productivity by fungal fermentation is limited and needs to be improved. RESULTS: In this study, the lovastatin-producing strains of Aspergillus terreus from marine environment were screened, and their lovastatin productions were further improved by genetic engineering. Five strains of A. terreus were isolated from various marine environments. Their secondary metabolites were profiled by metabolomics analysis using Ultra Performance Liquid Chromatography-Mass spectrometry (UPLC-MS) with Global Natural Products Social Molecular Networking (GNPS), revealing that the production of secondary metabolites was variable among different strains. Remarkably, the strain of A. terreus MJ106 could principally biosynthesize the target drug lovastatin, which was confirmed by High Performance Liquid Chromatography (HPLC) and gene expression analysis. By one-factor experiment, lactose was found to be the best carbon source for A. terreus MJ106 to produce lovastatin. To improve the lovastatin titer in A. terreus MJ106, genetic engineering was applied to this strain. Firstly, a series of strong promoters was identified by transcriptomic and green fluorescent protein reporter analysis. Then, three selected strong promoters were used to overexpress the transcription factor gene lovE encoding the major transactivator for lov gene cluster expression. The results revealed that compared to A. terreus MJ106, all lovE over-expression mutants exhibited significantly more production of lovastatin and higher gene expression. One of them, LovE-b19, showed the highest lovastatin productivity at a titer of 1512 mg/L, which represents the highest production level reported in A. terreus. CONCLUSION: Our data suggested that combination of strain screen and genetic engineering represents a powerful tool for improving the productivity of fungal secondary metabolites, which could be adopted for large-scale production of lovastatin in marine-derived A. terreus.


Assuntos
Aspergillus , Fermentação , Engenharia Genética , Lovastatina , Lovastatina/biossíntese , Lovastatina/metabolismo , Aspergillus/metabolismo , Aspergillus/genética , Organismos Aquáticos/metabolismo , Organismos Aquáticos/genética
10.
Int J Biol Sci ; 20(6): 2130-2148, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38617541

RESUMO

Triple-negative breast cancer (TNBC) is the most aggressive subtype of breast cancer with limited effective therapeutic options readily available. We have previously demonstrated that lovastatin, an FDA-approved lipid-lowering drug, selectively inhibits the stemness properties of TNBC. However, the intracellular targets of lovastatin in TNBC remain largely unknown. Here, we unexpectedly uncovered ribosome biogenesis as the predominant pathway targeted by lovastatin in TNBC. Lovastatin induced the translocation of ribosome biogenesis-related proteins including nucleophosmin (NPM), nucleolar and coiled-body phosphoprotein 1 (NOLC1), and the ribosomal protein RPL3. Lovastatin also suppressed the transcript levels of rRNAs and increased the nuclear protein level and transcriptional activity of p53, a master mediator of nucleolar stress. A prognostic model generated from 10 ribosome biogenesis-related genes showed outstanding performance in predicting the survival of TNBC patients. Mitochondrial ribosomal protein S27 (MRPS27), the top-ranked risky model gene, was highly expressed and correlated with tumor stage and lymph node involvement in TNBC. Mechanistically, MRPS27 knockdown inhibited the stemness properties and the malignant phenotypes of TNBC. Overexpression of MRPS27 attenuated the stemness-inhibitory effect of lovastatin in TNBC cells. Our findings reveal that dysregulated ribosome biogenesis is a targetable vulnerability and targeting MRPS27 could be a novel therapeutic strategy for TNBC patients.


Assuntos
Neoplasias de Mama Triplo Negativas , Humanos , Neoplasias de Mama Triplo Negativas/tratamento farmacológico , Neoplasias de Mama Triplo Negativas/genética , Lovastatina/farmacologia , Lovastatina/uso terapêutico , Proteínas Ribossômicas/genética , Proteínas Nucleares , Ribossomos/genética , Proteínas Mitocondriais
11.
Appl Microbiol Biotechnol ; 108(1): 255, 2024 Mar 06.
Artigo em Inglês | MEDLINE | ID: mdl-38446219

RESUMO

Monascus spp. are commercially important fungi due to their ability to produce beneficial secondary metabolites such as the cholesterol-lowering agent lovastatin and natural food colorants azaphilone pigments. Although hyphal branching intensively influenced the production of these secondary metabolites, the pivotal regulators of hyphal development in Monascus spp. remain unclear. To identify these important regulators, we developed an artificial intelligence (AI)-assisted image analysis tool for quantification of hyphae-branching and constructed a random T-DNA insertion library. High-throughput screening revealed that a STE kinase, MpSTE1, was considered as a key regulator of hyphal branching based on the hyphal phenotype. To further validate the role of MpSTE1, we generated an mpSTE1 gene knockout mutant, a complemented mutant, and an overexpression mutant (OE::mpSTE1). Microscopic observations revealed that overexpression of mpSTE1 led to a 63% increase in branch number while deletion of mpSTE1 reduced the hyphal branching by 68% compared to the wild-type strain. In flask cultures, the strain OE::mpSTE1 showed accelerated growth and glucose consumption. More importantly, the strain OE::mpSTE1 produced 9.2 mg/L lovastatin and 17.0 mg/L azaphilone pigments, respectively, 47.0% and 30.1% higher than those of the wild-type strain. Phosphoproteomic analysis revealed that MpSTE1 directly phosphorylated 7 downstream signal proteins involved in cell division, cytoskeletal organization, and signal transduction. To our best knowledge, MpSTE1 is reported as the first characterized regulator for tightly regulating the hyphal branching in Monascus spp. These findings significantly expanded current understanding of the signaling pathway governing the hyphal branching and development in Monascus spp. Furthermore, MpSTE1 and its analogs were demonstrated as promising targets for improving production of valuable secondary metabolites. KEY POINTS: • MpSTE1 is the first characterized regulator for tightly regulating hyphal branching • Overexpression of mpSTE1 significantly improves secondary metabolite production • A high-throughput image analysis tool was developed for counting hyphal branching.


Assuntos
Hifas , Monascus , Monascus/genética , Inteligência Artificial , Proteínas Serina-Treonina Quinases , Lovastatina , Treonina , Serina
12.
Toxicol In Vitro ; 97: 105806, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38432573

RESUMO

INTRODUCTION: Statins have demonstrated chondroprotective effects by reducing inflammation and mitigating extracellular matrix degradation. However, statins are also reported to be cytotoxic to several types of cells. Early-onset osteoarthritis (OA) is characterized by synovial inflammation, which adversely affects hyaluronan (HA) production in fibroblast-like synoviocytes (FLSs). Nevertheless, the precise effects of statins on the synovium remain unclear. METHODS: This study investigated the impact of lovastatin on human FLSs, and HA secretion-related genes, signaling pathways, and production were evaluated. RESULTS: The findings revealed that high doses of lovastatin (20 or 40 µM) decreased FLS viability and increased cell death. FLS proliferation ceased when cultured in a medium containing 5 or 10 µM lovastatin. mRNA expression analysis demonstrated that lovastatin (5 and 10 µM) upregulated the gene level of hyaluronan synthase 1 (HAS1), HAS2, and proteoglycan 4 (PRG4), but not HAS3. While the expression of multidrug resistance-associated protein 5 transporter gene remained unaffected, both inward-rectifying potassium channel and acid-sensing ion channel 3 were upregulated. Western blot further confirmed that lovastatin increased the production of HAS1 and PRG4, and activated the PKC-α, ERK1/2, and p38-MAPK signaling pathways. Additionally, lovastatin elevated intracellular cAMP levels and HA production in FLSs. CONCLUSION: Lovastatin impairs cellular proliferation but enhances HA production in human FLSs.


Assuntos
Inibidores de Hidroximetilglutaril-CoA Redutases , Sinoviócitos , Humanos , Sinoviócitos/metabolismo , Ácido Hialurônico/metabolismo , Lovastatina/farmacologia , Lovastatina/metabolismo , Inibidores de Hidroximetilglutaril-CoA Redutases/farmacologia , Fibroblastos/metabolismo , Proliferação de Células , Inflamação/metabolismo , Células Cultivadas
13.
Semergen ; 50(4): 102156, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38310834

RESUMO

Monacolin K is the major active component in red yeast rice (RYR) which is structurally identical to lovastatin and has the most powerful effect, in terms of reducing blood cholesterol levels. This review aimed to examine the effect and safety of different doses of monacolin K on blood cholesterol levels. PubMed and Cochrane were searched for articles published between 2012 and 2023 for clinical-trials and randomized-controlled-trials. Eligible studies included participants>18-years-old, of any gender and ethnicity. The intervention/exposure of interest was monacolin K. Hypercholesterolemia was considered the outcome of interest defined as the elevated total or low-density-lipoprotein (LDL) cholesterol levels. 12 randomized-controlled-trials were eligible for inclusion in the analysis including 769 participants>18-years-old. 11 out of 12 studies were assessed with high methodological quality and one study with low methodological quality. Monacolin K supplementation varied between 2mg and 10mg per day and the maximum period of supplementation was 12 weeks. All studies indicated a beneficial effect of monacolin supplementation on LDL and total cholesterol levels (p<0.05) regardless the dose and period of supplementation. Also, 3 of the included studies reported adverse side effects after treatment with monacolin K. Low doses of monacolin K equal to 3mg/day exert potential cholesterol-lowering effects although the number of relative studies is limited. Regarding the safety of monacolin K supplementation, findings seem to be more controversial and therefore, it is suggested for all patients treated with monacolin K to be routinely monitored regardless the dose of supplementation.


Assuntos
Anticolesterolemiantes , Produtos Biológicos , LDL-Colesterol , Ácidos Dicarboxílicos , Suplementos Nutricionais , Ácidos Graxos , Hipercolesterolemia , Lovastatina , Ensaios Clínicos Controlados Aleatórios como Assunto , Humanos , Hipercolesterolemia/tratamento farmacológico , Lovastatina/administração & dosagem , Lovastatina/efeitos adversos , Anticolesterolemiantes/administração & dosagem , Anticolesterolemiantes/efeitos adversos , LDL-Colesterol/sangue , Relação Dose-Resposta a Droga , Colesterol/sangue
14.
J Nat Prod ; 87(4): 753-763, 2024 04 26.
Artigo em Inglês | MEDLINE | ID: mdl-38372239

RESUMO

In this study, we isolated lovastatin derivatives, including aculeatiols A-G (1-7) and three known compounds (8-10), from Aspergillus aculeatus. Their structures and absolute configurations were experimentally determined by high-resolution electrospray ionization mass spectrometry, nuclear magnetic resonance spectroscopy, and X-ray diffraction analyses, and the results were corroborated by quantum-chemical calculations. As members of the lovastatin derivatives, aculeatiols A-C (1-3) possess a γ-lactone functional group in the side chain. Compound 6 represents the first example that features an undescribed aromatized heterotetracyclic 6/6/6/6 ring system. Biologically, the lipid-lowering effects of all of these compounds were evaluated by analyzing the free fatty acid-induced intracellular lipid accumulation. In addition, compound 5, which regulated the transcription of genes associated with lipid uptake and synthesis, inhibited the accumulation of lipids.


Assuntos
Aspergillus , Lovastatina , Aspergillus/química , Lovastatina/farmacologia , Lovastatina/química , Estrutura Molecular , Humanos
15.
Nutrients ; 16(3)2024 Feb 02.
Artigo em Inglês | MEDLINE | ID: mdl-38337728

RESUMO

Red yeast rice (RYR) has a cholesterol-lowering effect due to the presence of bioactive components (monacolins, mainly monacolin K) that act by inhibiting the activity of 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) reductase. The European Food Safety Authority (EFSA) assessed the use of RYR and, while pointing out several uncertainties regarding the available data, raised a warning related to the safety of RYR when used as a food supplement at a dose of monacolin as low as 3 mg/day. In their decision in June 2023, EFSA approved the use of monacolins from RYR at doses less than 3 mg/day. We therefore decided to interrogate the different adverse event reporting systems (FAERS and CAERS) and analyse the characteristics of the cases reported to be associated with RYR supplements, and we reviewed the most recent meta-analyses with a focus on the occurrence of muscle symptoms and liver dysfunction. In terms of all musculoskeletal disorders from September 2013 (when the first case related to RYR consumption was recorded) to 30 September 2023, 363,879 cases were reported in the FAERS, with the number of cases related to RYR consumption being very small and accounting for 0.008% of cases. In the same time frame, 27,032 cases of hepatobiliary disorders were reported, and the cases attributable to RYR ingestion accounted for 0.01% of all cases. A low rate of muscle symptoms and liver dysfunction attributed to RYR ingestion was also observed in the CAERS database, where only 34 cases of adverse muscle events and 10 cases of adverse liver events reported RYR as the suspect product, while 19 cases of both muscle events and 10 cases of adverse liver events reported it as a concomitant product. This profile mirrors that of meta-analyses of randomised clinical trials of RYR, in which RYR use was not associated with either liver dysfunction or muscular adverse symptoms.


Assuntos
Produtos Biológicos , Hepatopatias , Humanos , Lovastatina , Suplementos Nutricionais/efeitos adversos , Suplementos Nutricionais/análise , Produtos Biológicos/efeitos adversos , Músculos/química , Hepatopatias/epidemiologia , Hepatopatias/etiologia , Hepatopatias/tratamento farmacológico , Extratos Vegetais
16.
Cancer Lett ; 588: 216765, 2024 Apr 28.
Artigo em Inglês | MEDLINE | ID: mdl-38408604

RESUMO

Current immune checkpoint blockade (ICB) immunotherapeutics have revolutionized cancer treatment. However, many cancers especially the "immunologically cold" tumors, do not respond to ICB, prompting the search for additional strategies to achieve durable responses. The cGAS-STING pathway, as an essential immune response pathway, has been demonstrated for a potent target to sensitize ICB immunotherapy. However, the low efficiency of conventional STING agonists limits their clinical application. Recent studies have shown that DNA topoisomerase I (TOPI) inhibitor chemodrug SN38 can activate the cGAS-STING pathway and induce an immune response through DNA damage, while the traditional statins medication lovastatin was found to inhibit DNA damage repair, which may in turn upregulate the damaged DNA level. Herein, we have developed a liposomal carrier co-loaded with SN38 and lovastatin (SL@Lip), which can be accumulated in tumors and efficiently released SN38 and lovastatin, addressing the problem of weak solubility of these two drugs. Importantly, lovastatin can increase DNA damage and enhance the activation of cGAS-STING pathway, coordinating with SN38 chemotherapy and exhibiting the enhanced combinational immunotherapy of PD-1 antibody by remodeling the tumor microenvironment in mouse colorectal cancer of both subcutaneous and orthotopic xenograft models. Overall, this study demonstrates that lovastatin-assisted cGAS-STING stimulation mediated by liposomal delivery system significantly strengthened both chemotherapy and immunotherapy of colorectal cancer, providing a clinically translational strategy for combinational ICB therapy in the "immunologically cold" tumors.


Assuntos
Neoplasias do Colo , Inibidores de Hidroximetilglutaril-CoA Redutases , Neoplasias , Humanos , Animais , Camundongos , Lovastatina/farmacologia , Inibidores de Checkpoint Imunológico , Lipossomos , Neoplasias do Colo/tratamento farmacológico , Imunoterapia , Microambiente Tumoral
17.
Appl Microbiol Biotechnol ; 108(1): 154, 2024 Jan 19.
Artigo em Inglês | MEDLINE | ID: mdl-38240803

RESUMO

Monascus pilosus has been used to produce lipid-lowering drugs rich in monacolin K (MK) for a long period. Genome mining reveals there are still many potential genes worth to be explored in this fungus. Thereby, efficient genetic manipulation tools will greatly accelerate this progress. In this study, we firstly developed the protocol to prepare protoplasts for recipient of CRISPR/Cas9 system. Subsequently, the vector and donor DNA were co-transformed into recipients (106 protoplasts/mL) to produce 60-80 transformants for one test. Three genes (mpclr4, mpdot1, and mplig4) related to DNA damage response (DDR) were selected to compare the gene replacement frequencies (GRFs) of Agrobacterium tumefaciens-mediated transformation (ATMT) and CRISPR/Cas9 gene editing system (CGES) in M. pilosus MS-1. The results revealed that GRF of CGES was approximately five times greater than that of ATMT, suggesting that CGES was superior to ATMT as a targeting gene editing tool in M. pilosus MS-1. The inactivation of mpclr4 promoted DDR via the non-homologous end-joining (NHEJ) and increased the tolerances to DNA damaging agents. The inactivation of mpdot1 blocked DDR and led to the reduced tolerances to DNA damaging agents. The inactivation of mplig4 mainly blocked the NHEJ pathway and led to obviously reduced tolerances to DNA damaging agents. The submerged fermentation showed that the ability to produce MK in strain Δmpclr4 was improved by 52.6% compared to the wild type. This study provides an idea for more effective exploration of gene functions in Monascus strains. KEY POINTS: • A protocol of high-quality protoplasts for CGES has been developed in M. pilosus. • The GRF of CGES was about five times that of ATMT in M. pilosus. • The yield of MK for Δmpclr4 was enhanced by 52.6% compared with the wild type.


Assuntos
Edição de Genes , Monascus , Monascus/genética , Monascus/metabolismo , Sistemas CRISPR-Cas , Marcação de Genes/métodos , Lovastatina/metabolismo , Agrobacterium tumefaciens/genética , DNA/metabolismo
18.
Expert Rev Clin Pharmacol ; 17(3): 255-261, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38275183

RESUMO

BACKGROUND: Concerns regarding statin-related neurocognitive disorders have emerged in recent years. However, previous studies have reported inconsistent results. We evaluated the association between statins and neurocognitive disorders using the FDA Adverse Event Reporting System (FAERS). RESEARCH DESIGN AND METHODS: Data from 2004 to 2022 were obtained from the FAERS database. After deduplication and standardization of drug names, we extracted neurocognitive disorder event (NCDE) cases reported with statins as the suspected drugs. The significant association between statins and NCDE was evaluated using the reporting odds ratio (ROR) and information component. RESULTS: In total, 6,959 NCDE cases with statins as the primary suspected drugs were identified. Signals were detected in pravastatin (ROR, 1.49; 95% CI: 1.32-1.67), atorvastatin (ROR, 1.39; 95% CI: 1.34-1.44), and simvastatin (ROR, 1.31; 95% CI: 1.25-1.38). Age-stratified analysis showed that (1) in the population aged 65 years and older, signals were detected for atorvastatin, simvastatin, rosuvastatin, pravastatin, lovastatin, fluvastatin, and pitavastatin; and (2) in populations under 65 years of age, signals were detected for atorvastatin, simvastatin, rosuvastatin, pravastatin, and lovastatin. CONCLUSIONS: This study suggests a significant association between the NCDE and statins, including atorvastatin, simvastatin, and pravastatin. The intensity of the association increased with age.


With the extensive use of statins worldwide in recent years, some patients have reported that statins lead to cognitive impairment. Researchers have conducted studies on this issue; however, the results have been inconsistent. Some believe that statins have no impact on cognitive function, while others believe they are beneficial, and others believe they have negative effects.To further investigate this issue, we analyzed data from the FDA adverse event reporting system, which collects adverse drug reactions reported by people worldwide, to evaluate the association between statins and cognitive impairment. Our findings suggest that some statins are associated with cognitive impairment. Therefore, when cognitive changes occur in patients taking statins, they should be taken seriously.


Assuntos
Inibidores de Hidroximetilglutaril-CoA Redutases , Humanos , Inibidores de Hidroximetilglutaril-CoA Redutases/efeitos adversos , Rosuvastatina Cálcica/efeitos adversos , Atorvastatina/efeitos adversos , Pravastatina/efeitos adversos , Farmacovigilância , Sistemas de Notificação de Reações Adversas a Medicamentos , Sinvastatina/efeitos adversos , Lovastatina , Transtornos Neurocognitivos/induzido quimicamente
19.
Pediatr Dermatol ; 41(2): 296-297, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-37726979

RESUMO

A 2-year-old boy presented with an extensive, asymptomatic, photosensitive eruption refractory to topical steroids and tretinoin; examination and biopsies were consistent with generalized linear porokeratosis involving the face, limbs, and trunk. Treatment with topical cholesterol-lovastatin was initiated, and it successfully improved early erythematous lesions. Whole exome sequencing that targeted mevalonate pathway genes crucial in cholesterol synthesis later revealed a pathogenic, paternally inherited, porokeratosis-associated MVD, c.70+5 G>A, mutation. Topical cholesterol-lovastatin is a safe and effective empiric treatment for porokeratosis when used in the early, erythematous phase, and its success is likely mediated through its role in targeting mevalonate pathway mutations.


Assuntos
Lovastatina , Poroceratose , Pré-Escolar , Humanos , Masculino , Colesterol , Lovastatina/uso terapêutico , Ácido Mevalônico/metabolismo , Poroceratose/tratamento farmacológico , Poroceratose/diagnóstico , Resultado do Tratamento
20.
Int J Biol Macromol ; 255: 128208, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-37979745

RESUMO

Increasing data suggested that histone methylation modification plays an important role in regulating biosynthesis of secondary metabolites (SMs). Monascus spp. have been applied to produce hypolipidemic drug lovastatin (also called monacolin K, MK) and edible Monascus-type azaphilone pigments (MonAzPs). However, little is known about how histone methylation regulates MK and MonAzPs. In this study, we constructed H3K9 methyltransferase deletion strain ΔMpDot1 and H4K20 methyltransferase deletion strain ΔMpSet9 using Monascus pilosus MS-1 as the parent. The result showed that deletion of MpDot1 reduced the production of MK and MonAzPs, and deletion of MpSet9 increased MonAzPs production. Real-time quantitative PCR (RT-qPCR) showed inactivation of mpdot1 and mpset9 disturbed the expression of genes responsible for the biosynthesis of MK and MonAzPs. Western blot suggested that deletion of MpDot1 reduced H3K79me and H4K16ac, and deletion of MpSet9 decreased H4K20me3 and increased H4pan acetylation. Chromatin immunoprecipitation coupled with quantitative PCR (ChIP-qPCR) showed ΔMpDot1 strain and ΔMpSet9 strain reduced the enrichment of H3K79me2 and H4K20me3 in the promoter regions of key genes for MK and MonAzPs biosynthesis, respectively. These results suggested that MpDot1 and MpSet9 affected the synthesis of SMs by regulating gene transcription and histone crosstalk, providing alternative approach for regulation of lovastatin and MonAzPs.


Assuntos
Lovastatina , Monascus , Lovastatina/farmacologia , Histonas/metabolismo , Histona-Lisina N-Metiltransferase/genética , Histona-Lisina N-Metiltransferase/metabolismo , Monascus/genética , Monascus/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA