Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Heliyon ; 10(6): e27635, 2024 Mar 30.
Article in English | MEDLINE | ID: mdl-38509999

ABSTRACT

Seaweed has attracted attention as a bioactive source for preventing different chronic diseases, including liver injury and non-alcoholic fatty liver disease, the leading cause of liver-related mortality. Caulerpa lentillifera is characterized as tropical edible seaweed, currently being investigated for health benefits of its extracts and bioactive substances. This study examined the effects of C. lentillifera extract in ethyl acetate fraction (CLEA) on controlling lipid accumulation and lipid metabolism in HepG2 cells induced with oleic acid through the in vitro hepatic steatosis model. Gas chromatography-mass spectrometry (GC-MS) analysis indicated that CLEA contained diverse organic compounds, including hydrocarbons, amino acids, and carboxylic acids. Docked conformation of dl-2-phenyltryptophane and benzoic acid, two major bioactive CLEA components, showed high affinity binding to SIRT1 and AMPK as target molecules of lipid metabolism. CLEA reduced lipid accumulation and intracellular triglyceride levels in HepG2 cells stimulated with oleic acid. The effect of CLEA on regulating expression of lipid metabolism-related molecules was investigated by qPCR and immunoblotting. CLEA promoted expression of the SIRT1 gene in oleic acid-treated HepG2 cells. CLEA also reduced expression levels of SREBF1, FAS, and ACC genes, which might be related to activation of AMPK signaling in lipid-accumulated HepG2 cells. These findings suggest that CLEA contains bioactive compounds potentially reducing triglyceride accumulation in lipid-accumulated HepG2 hepatocytes by controlling lipid metabolism molecules.

2.
Mar Drugs ; 21(6)2023 Jun 03.
Article in English | MEDLINE | ID: mdl-37367670

ABSTRACT

Due to the challenge of prostate cancer (PCa) management, there has been a surge in efforts to identify more safe and effective compounds that can modulate the epithelial-mesenchymal transition (EMT) for driving metastasis. Holothurin A (HA), a triterpenoid saponin isolated from Holothuria scabra, has now been characterized for its diverse biological activities. However, the mechanisms of HA in EMT-driven metastasis of human PCa cell lines has not yet been investigated. Moreover, runt-related transcription factor 1 (RUNX1) acts as an oncogene in prostate cancer, but little is known about its role in the EMT. Thus, the purpose of this study was to determine how RUNX1 influences EMT-mediated metastasis, as well as the potential effect of HA on EMT-mediated metastasis in endogenous and exogenous RUNX1 expressions of PCa cell lines. The results demonstrated that RUNX1 overexpression could promote the EMT phenotype with increased EMT markers, consequently driving metastatic migration and invasion in PC3 cell line through the activation of Akt/MAPK signaling pathways. Intriguingly, HA treatment could antagonize the EMT program in endogenous and exogenous RUNX1-expressing PCa cell lines. A decreasing metastasis of both HA-treated cell lines was evidenced through a downregulation of MMP2 and MMP9 via the Akt/P38/JNK-MAPK signaling pathway. Overall, our approach first demonstrated that RUNX1 enhanced EMT-driven prostate cancer metastasis and that HA was capable of inhibiting the EMT and metastatic processes and should probably be considered as a candidate for metastasis PCa treatment.


Subject(s)
Epithelial-Mesenchymal Transition , Prostatic Neoplasms , Male , Humans , Proto-Oncogene Proteins c-akt/metabolism , Core Binding Factor Alpha 2 Subunit/genetics , Core Binding Factor Alpha 2 Subunit/pharmacology , Signal Transduction , Prostatic Neoplasms/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Cell Movement , Cell Line, Tumor , Neoplasm Metastasis , Neoplasm Invasiveness
3.
Int J Mol Sci ; 24(10)2023 May 10.
Article in English | MEDLINE | ID: mdl-37239873

ABSTRACT

Parkinson's disease (PD) is associated with dopaminergic neuron loss and alpha-synuclein aggregation caused by ROS overproduction, leading to mitochondrial dysfunction and autophagy impairment. Recently, andrographolide (Andro) has been extensively studied for various pharmacological properties, such as anti-diabetic, anti-cancer, anti-inflammatory, and anti-atherosclerosis. However, its potential neuroprotective effects on neurotoxin MPP+-induced SH-SY5Y cells, a cellular PD model, remain uninvestigated. In this study, we hypothesized that Andro has neuroprotective effects against MPP+-induced apoptosis, which may be mediated through the clearance of dysfunctional mitochondria by mitophagy and ROS by antioxidant activities. Herein, Andro pretreatment could attenuate MPP+-induced neuronal cell death that was reflected by reducing mitochondrial membrane potential (MMP) depolarization, alpha-synuclein, and pro-apoptotic proteins expressions. Concomitantly, Andro attenuated MPP+-induced oxidative stress through mitophagy, as indicated by increasing colocalization of MitoTracker Red with LC3, upregulations of the PINK1-Parkin pathway, and autophagy-related proteins. On the contrary, Andro-activated autophagy was compromised when pretreated with 3-MA. Furthermore, Andro activated the Nrf2/KEAP1 pathway, leading to increasing genes encoding antioxidant enzymes and activities. This study elucidated that Andro exhibited significant neuroprotective effects against MPP+-induced SH-SY5Y cell death in vitro by enhancing mitophagy and clearance of alpha-synuclein through autophagy, as well as increasing antioxidant capacity. Our results provide evidence that Andro could be considered a potential supplement for PD prevention.


Subject(s)
Neuroblastoma , Neuroprotective Agents , Parkinson Disease , Humans , Mitophagy , Antioxidants/pharmacology , Antioxidants/therapeutic use , Neurotoxins/pharmacology , alpha-Synuclein/metabolism , Neuroprotection , Kelch-Like ECH-Associated Protein 1/metabolism , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Reactive Oxygen Species/metabolism , NF-E2-Related Factor 2/metabolism , Autophagy , Apoptosis , Cell Line, Tumor , Parkinson Disease/drug therapy , Parkinson Disease/genetics , 1-Methyl-4-phenylpyridinium/toxicity
4.
BMC Complement Med Ther ; 22(1): 235, 2022 Sep 05.
Article in English | MEDLINE | ID: mdl-36064352

ABSTRACT

BACKGROUND: Lysiphyllum strychnifolium (Craib) A. Schmitz, a traditional Thai medicinal plant, is mainly composed of polyphenols and flavonoids and exhibits several pharmacological activities, including antioxidant, anticancer, antimicrobial, and antidiabetic activities. However, the mechanism by which pure compounds from L. strychnifolium inhibit glucose catalysis in the small intestine and their effect on the glucose transporter remain unknown. METHODS: The objectives of this research were to examine the effect of 3,5,7-trihydroxychromone-3-O-𝛼-L-rhamnopyranoside (compound 1) and 3,5,7,3',5'-pentahydroxy-flavanonol-3-O-𝛼-L-rhamnopyranoside (compound 2) on the inhibition of α-amylase and α-glucosidase, as well as glucose transporters, such as sodium-glucose cotransporter 1 (SGLT1), glucose transporter 2 (GLUT2), and glucose transporter 5 (GLUT5), using Caco-2 cells as a model of human intestinal epithelial cells. Additionally, the binding affinity and interaction patterns of compounds against two receptor proteins (SGLT1 and GLUT2) were determined for the first time utilizing a molecular docking approach. RESULTS: In the α-amylase inhibition assay, a concentration-dependent inhibitory response was observed against the enzyme. The results indicated that compound 1 inhibited α-amylase activity in a manner similar to that of acarbose (which exhibit IC50 values of 3.32 ± 0.30 µg/mL and 2.86 ± 0.10 µg/mL, respectively) in addition to a moderate inhibitory effect for compound 2 (IC50 = 10.15 ± 0.53 µg/mL). Interestingly, compounds 1 and 2 significantly inhibited α-glucosidase and exhibited better inhibition than that of acarbose, with IC50 values of 5.35 ± 1.66 µg/mL, 510.15 ± 1.46 µg/mL, and 736.93 ± 7.02 µg/mL, respectively. Additionally, α-glucosidase activity in the supernatant of the Caco-2 cell monolayer was observed. In comparison to acarbose, compounds 1 and 2 inhibited α-glucosidase activity more effectively in Caco-2 cells without cytotoxicity at a concentration of 62.5 µg/mL. Furthermore, the glucose uptake pathways mediated by SGLT1, GLUT2, and GLUT5- were downregulated in Caco-2 cells treated with compounds 1 and 2. Additionally, molecular modeling studies revealed that compounds 1 and 2 presented high binding activity with SGLT1 and GLUT2. CONCLUSION: In summary, our present study was the first to perform molecular docking with compounds present in L. strychnifolium extracts. Our findings indicated that compounds 1 and 2 reduced glucose uptake in Caco-2 cells by decreasing the expression of glucose transporter genes and inhibiting the binding sites of SGLT1 and GLUT2. Therefore, compounds 1 and 2 may be used as functional foods in dietary therapy for postprandial hyperglycemia modulation of type 2 diabetes.


Subject(s)
Diabetes Mellitus, Type 2 , Fabaceae , Acarbose , Caco-2 Cells , Diabetes Mellitus, Type 2/drug therapy , Glucose/metabolism , Humans , Hypoglycemic Agents/pharmacology , Molecular Docking Simulation , Polyphenols , alpha-Amylases/metabolism , alpha-Glucosidases/metabolism
5.
J Biomol Struct Dyn ; 40(23): 12674-12682, 2022.
Article in English | MEDLINE | ID: mdl-34514975

ABSTRACT

The androgen receptor (AR) plays a crucial role in the growth of prostate cancer, and has long been considered the cancer's primary strategic therapeutic target. However, despite the early susceptibility, patients receiving hormonal therapy targeting AR are likely to develops resistance to the treatment and progresses to the castration-resistant stage as a consequence of the mutation at the ligand binding pocket of AR. Interestingly, the surface pocket of the AR called binding function 3 (BF3) has been reported as a great benefit for treating a recurrent tumor. Herein, we investigate the potential of using a marine triterpenoid saponin, holothurin A, on targeting AR expression of prostate cancer using in vitro and in silico studies. Holothurin A reduced the PSA expression, leading to the growth inhibition of androgen sensitive prostate cancer cell line through a downregulation of AR activity. The molecular docking study demonstrated that holothurin A could bind strongly in the BF3 pocket by energetically favorable hydrogen acceptor and hydrophobic with a calculated binding affinity of -13.90 kcal/mol. Molecular dynamics simulations provided the additional evidence that holothurin A can form a stable complex with the BF3 pocket through the hydrophobic interactions with VAL676, ILE680, and ALA721. As a consequence, holothurin A modulates the activation function-2 (AF2) site of the AR through repositioning of the residues in the AF2 pocket. Targeting alternatives sites on the surface of AR via holothurin A will provide a potential candidate for future prostate cancer treatment.Communicated by Ramaswamy H. Sarma.


Subject(s)
Prostatic Neoplasms , Receptors, Androgen , Male , Humans , Receptors, Androgen/metabolism , Molecular Docking Simulation , Furylfuramide , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/genetics , Cell Line, Tumor
6.
J Microbiol Biotechnol ; 31(6): 775-783, 2021 Jun 28.
Article in English | MEDLINE | ID: mdl-33958506

ABSTRACT

Sea cucumber, Holothuria scabra, is a well-known traditional Asian medicine that has been used for suppressing inflammation, promoting wound healing, and improving immunity. Moreover, previous studies demonstrated that the extract from H. scabra contains many bioactive compounds with potent inhibitory effect on tumor cell survival and progression. However, the effect of the methanolic extract from the body wall of H. scabra (BWMT) on human prostate cancer cells has not yet been investigated. In this study, we aimed to investigate the effects and underlying mechanism of BWMT on prostate cancer cell viability and metastasis. BWMT was obtained by maceration with methanol. The effect of BWMT on cell viability was assessed by MTT and colony formation assays. The intracellular ROS accumulation was evaluated using a DCFH-DA fluorescence probe. Hoechst 33342 staining and Annexin V-FITC/PI staining were used to examine the apoptotic-inducing effect of the extract. A transwell migration assay was performed to determine the anti-metastasis effect. BWMT significantly reduced cell viability and triggered cellular apoptosis by accumulating intracellular ROS resulting in the upregulation of JNK and p38 signaling pathways. In addition, BWMT also inhibited the invasion of PC3 cells by downregulating MMP-2/-9 expression via the ERK pathway. Consequently, our study provides BWMT from H. scabra as a putative therapeutic agent that could be applicable against prostate cancer progression.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Holothuria/chemistry , MAP Kinase Signaling System/drug effects , Prostatic Neoplasms/pathology , Animals , Antineoplastic Agents/isolation & purification , Cell Cycle Checkpoints/drug effects , Cell Movement/drug effects , Cell Survival/drug effects , Humans , Male , Methanol/chemistry , PC-3 Cells , Reactive Oxygen Species/metabolism
7.
Nutr Cancer ; 73(10): 1964-1975, 2021.
Article in English | MEDLINE | ID: mdl-32878490

ABSTRACT

INTRODUCTION: Cancer cells utilize the modified glucose metabolism known as Warburg effect, with lactate production as the end product. In the search for alternative therapy, the body wall of sea cucumbers contains various substances with pharmacological activities. Herein, we investigate the effect of Holothuria scabra extract on the viability and Warburg effect of aggressive breast cancer cells. METHODS: Body wall of H. scabra was extracted using 95% ethanol. Triple-negative breast cancer cells, MDA-MB-231, were treated with the extract at various concentrations under normoglycemic and hyperglycemic conditions. Cytotoxicity test was performed using MTT assay. Apoptotic proteins were quantified using Western blot. Apoptotic cells were stained with Hoechst 33342. Lactate production was determined using L-lactate assay kit. RESULTS: By MTT assay, H. scabra extract suppressed the viability of breast cancer cells in a dose-dependent and time-dependent manner by enhancing apoptosis, indicated by a marked increase of proapoptotic Bax and pro-caspase three expressions, and decreased expression of anti-apoptotic Bcl-2. The extract could reduce hexokinase II expression, leading to reduced lactate production by blocking the Akt/mTOR/HIF-1 axis. DISCUSSION: Overall findings indicated that H. scabra extract could be a possible therapeutic against breast cancer progression in patients with hyperglycemia, for instance, diabetes mellitus.


Subject(s)
Breast Neoplasms , Holothuria , Triple Negative Breast Neoplasms , Animals , Apoptosis , Breast Neoplasms/drug therapy , Cell Line, Tumor , Cell Proliferation , Female , Humans , Plant Extracts/pharmacology , Proto-Oncogene Proteins c-akt/genetics , TOR Serine-Threonine Kinases , Triple Negative Breast Neoplasms/drug therapy
8.
Inflammopharmacology ; 28(4): 1027-1037, 2020 Aug.
Article in English | MEDLINE | ID: mdl-31813081

ABSTRACT

Sea cucumber, Holothuria scabra, is an echinoderm marine animal that has long been used as a traditional therapeutic in various diseases due to its chemical composition and protein enrichment. Many researchers have extensively studied the efficacy of sea cucumber extracts for many health benefits in recent years. Inflammation is a complex process involved in pro-/anti-inflammatory cytokine products. However, the role of the H. scabra extracts in anti-inflammation and its molecular regulations has not been apparently elucidated yet. In this study, we investigated the anti-inflammatory effect of H. scabra extracts by using lipopolysaccharide (LPS) from E. coli to induce an inflammatory response in RAW264.7 macrophage. It was found that ethyl acetate fraction of H. scabra extracts (EAHS) inhibited pro-inflammatory cytokines synthesis at both the transcriptional and translational levels, notably nitric oxide (NO), inducible nitric oxide synthase (iNOS), interleukin-1ß (IL-1ß), tumor necrosis factor-α (TNF-α), and prostaglandin E2 (PGE2). In addition, EAHS was able to downregulate IκB/NF-κB, and JNK expressions. These effects may be influenced by high contents of phenolic compound and triterpene glycosides in EAHS. Therefore, EAHS might have the potential to be developed as a natural anti-inflammatory agent.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Biological Products/pharmacology , Cytokines/metabolism , Holothuria/chemistry , Inflammation/drug therapy , Sea Cucumbers/chemistry , Signal Transduction/drug effects , Acetates/chemistry , Animals , Anti-Inflammatory Agents/chemistry , Biological Products/chemistry , Cell Line , Dinoprostone/metabolism , Disease Models, Animal , Escherichia coli/drug effects , Inflammation/chemically induced , Inflammation/metabolism , Interleukin-1beta/metabolism , Lipopolysaccharides/pharmacology , MAP Kinase Signaling System/drug effects , Macrophages/drug effects , Macrophages/metabolism , Mice , NF-kappa B/metabolism , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II/metabolism , RAW 264.7 Cells , Tumor Necrosis Factor-alpha/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...