Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
J Ethnopharmacol ; 330: 118226, 2024 Aug 10.
Article in English | MEDLINE | ID: mdl-38670401

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Idiopathic pulmonary fibrosis (IPF) is a progressive fibrosing pulmonary disorder that has a poor prognosis and high mortality. Although there has been extensive effort to introduce several new anti-fibrotic agents in the past decade, IPF remains an incurable disease. Mimosa pudica L., an indigenous Vietnamese plant, has been empirically used to treat respiratory disorders. Nevertheless, the therapeutic effects of M. pudica (MP) on lung fibrosis and the mechanisms underlying those effects remain unclear. AIM OF THE STUDY: This study investigated the protective effect of a crude ethanol extract of the above-ground parts of MP against pulmonary fibrogenesis. MATERIALS AND METHODS: Inflammatory responses triggered by TNFα in structural lung cells were examined in normal human lung fibroblasts and A549 alveolar epithelial cells using Western blot analysis, reverse transcription-quantitative polymerase chain reaction assays, and immunocytochemistry. The epithelial-to-mesenchymal transition (EMT) was examined via cell morphology observations, F-actin fluorescent staining, gene and protein expression measurements, and a wound-healing assay. Anti-fibrotic assays including collagen release, differentiation, and measurements of fibrosis-related gene and protein expression levels were performed on TGFß-stimulated human lung fibroblasts and lung fibroblasts derived from mice with fibrotic lungs. Finally, in vitro anti-fibrotic activities were validated using a mouse model of bleomycin-induced pulmonary fibrosis. RESULTS: MP alleviated the inflammatory responses of A549 alveolar epithelial cells and lung fibroblasts, as revealed by inhibition of TNFα-induced chemotactic cytokine and chemokine expression, along with inactivation of the MAPK and NFκB signalling pathways. MP also partially reversed the TGFß-promoted EMT via downregulation of mesenchymal markers in A549 cells. Importantly, MP decreased the expression levels of fibrosis-related genes/proteins including collagen I, fibronectin, and αSMA; moreover, it suppressed collagen secretion and prevented myofibroblast differentiation in lung fibroblasts. These effects were mediated by FOXO3 stabilization through suppression of TGFß-induced ERK1/2 phosphorylation. MP consistently protected mice from the onset and progression of bleomycin-induced pulmonary fibrosis. CONCLUSION: This study explored the multifaceted roles of MP in counteracting the pathobiological processes of lung fibrosis. The results suggest that further evaluation of MP could yield candidate therapies for IPF.


Subject(s)
Epithelial-Mesenchymal Transition , Forkhead Box Protein O3 , MAP Kinase Signaling System , Mice, Inbred C57BL , Plant Extracts , Pulmonary Fibrosis , Animals , Humans , Plant Extracts/pharmacology , Plant Extracts/therapeutic use , A549 Cells , Mice , MAP Kinase Signaling System/drug effects , Epithelial-Mesenchymal Transition/drug effects , Pulmonary Fibrosis/drug therapy , Pulmonary Fibrosis/chemically induced , Forkhead Box Protein O3/metabolism , Fibroblasts/drug effects , Fibroblasts/metabolism , Male , Bleomycin , Antifibrotic Agents/pharmacology , Lung/drug effects , Lung/pathology , Lung/metabolism , Idiopathic Pulmonary Fibrosis/drug therapy , Idiopathic Pulmonary Fibrosis/chemically induced , Idiopathic Pulmonary Fibrosis/pathology
2.
Acta Pharm Sin B ; 14(1): 273-291, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38261813

ABSTRACT

Obesity has been known to negatively modulate the life-span and immunosuppressive potential of mesenchymal stromal cells (MSC). However, it remains unclear what drives the compromised potency of obese MSC. In this study, we examined the involvement of adiponectin, an adipose tissue-derived hormone, in obesity-induced impaired therapeutic function of MSC. Diet-induced obesity leads to a decrease in serum adiponectin, accompanied by impairment of survival and immunomodulatory effects of adipose-derived MSC (ADSC). Interestingly, priming with globular adiponectin (gAcrp) improved the immunomodulatory potential of obese ADSC. Similar effects were also observed in lean ADSC. In addition, gAcrp potentiated the therapeutic effectiveness of ADSC in a mouse model of DSS-induced colitis. Mechanistically, while obesity inhibited the glycolytic capacity of MSC, gAcrp treatment induced a metabolic shift toward glycolysis through activation of adiponectin receptor type 1/p38 MAPK/hypoxia inducible factor-1α axis. These findings suggest that activation of adiponectin signaling is a promising strategy for enhancing the therapeutic efficacy of MSC against immune-mediated disorders.

3.
J Control Release ; 361: 443-454, 2023 09.
Article in English | MEDLINE | ID: mdl-37558053

ABSTRACT

Triple-negative breast cancer (TNBC) is highly aggressive and has no standard treatment. Although being considered as an alternative to conventional treatments for TNBC, immunotherapy has to deal with many challenges that hinder its efficacy, particularly the poor immunogenic condition of the tumor microenvironment (TME). Herein, we designed a liposomal nanoparticle (LN) platform that delivers simultaneously toll-like receptor 7 (imiquimod, IQ) and toll-like receptor 3 (poly(I:C), IC) agonists to take advantage of the different toll-like receptor (TLR) signaling pathways, which enhances the condition of TME from a "cold" to a "hot" immunogenic state. The optimized IQ/IC-loaded LN (IQ/IC-LN) was effectively internalized by cancer cells, macrophages, and dendritic cells, followed by the release of the delivered drugs and subsequent stimulation of the TLR3 and TLR7 signaling pathways. This stimulation encouraged the secretion of type I interferon (IFN-α, IFN-ß) and CXCLl0, a T-cell and antigen-presenting cells (APCs) recruitment chemokine, from both cancer cells and macrophages and polarized macrophages to the M1 subtype in in vitro studies. Notably, systemic administration of IQ/IC-LN allowed for the high accumulation of drug content in the tumor, followed by the effective uptake by immune cells in the TME. IQ/IC-LN treatment comprehensively enhanced the immunogenic condition in the TME, which robustly inhibited tumor growth in tumor-bearing mice. Furthermore, synergistic antitumor efficacy was obtained when the IQ/IC-LN-induced immunogenic state in TME was combined with anti-PD1 antibody therapy. Thus, our results suggest the potential of combining 2 TLR agonists to reform the TME from a "cold" to a "hot" state, supporting the therapeutic efficacy of immune checkpoint inhibitors.


Subject(s)
Toll-Like Receptor 3 , Triple Negative Breast Neoplasms , Humans , Animals , Mice , Triple Negative Breast Neoplasms/drug therapy , Adjuvants, Immunologic , Liposomes , Poly I-C/therapeutic use , Immunotherapy/methods , Tumor Microenvironment
4.
Exp Mol Med ; 55(2): 313-324, 2023 02.
Article in English | MEDLINE | ID: mdl-36750692

ABSTRACT

Mesenchymal stem cell (MSC) therapy is an emerging treatment strategy to counteract metabolic syndromes, including obesity and its comorbid disorders. However, its effectiveness is challenged by various factors in the obese environment that negatively impact MSC survival and function. The identification of these detrimental factors will provide opportunities to optimize MSC therapy for the treatment of obesity and its comorbidities. Dysregulated production of adipokines, a group of cytokines and hormones derived from adipose tissue, has been postulated to play a pivotal role in the development of obesity-associated complications. Intriguingly, adipokines have also been implicated in the modulation of viability, self-renewal, proliferation, and other properties of MSC. However, the involvement of adipokine imbalance in impaired MSC functionality has not been completely understood. On the other hand, treatment of obese individuals with MSC can restore the serum adipokine profile, suggesting the bidirectionality of the adipokine-MSC relationship. In this review, we aim to discuss the current knowledge on the central role of adipokines in the crosstalk between obesity and MSC dysfunction. We also summarize recent advances in the use of MSC for the treatment of obesity-associated diseases to support the hypothesis that adipokines modulate the benefits of MSC therapy in obese patients.


Subject(s)
Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells , Metabolic Syndrome , Humans , Adipokines/metabolism , Obesity/therapy , Obesity/complications , Metabolic Syndrome/metabolism , Adipose Tissue/metabolism , Mesenchymal Stem Cells/metabolism
5.
Mol Ther ; 31(3): 890-908, 2023 03 01.
Article in English | MEDLINE | ID: mdl-36566348

ABSTRACT

Mesenchymal stem cells (MSCs) are ubiquitous multipotent cells that exhibit significant therapeutic potentials in a variety of disorders. Nevertheless, their clinical efficacy is limited owing to poor survival, low rate of engraftment, and impaired potency upon transplantation. Spheroidal three-dimensional (3D) culture of MSCs (MSC3D) has been proven to better preserve their in vivo functional properties. However, the molecular mechanisms underlying the improvement in MSC function by spheroid formation are not clearly understood. NLRP3 inflammasomes, a key component of the innate immune system, have recently been shown to play a role in cell fate decision of MSCs. The present study examined the role of NLRP3 inflammasomes in the survival and potency of MSC spheroids. We found that MSC3D led to decreased activation of NLRP3 inflammasomes through alleviation of ER stress in an autophagy-dependent manner. Importantly, downregulation of NLRP3 inflammasomes signaling critically contributes to the enhanced survival rate in MSC3D through modulation of pyroptosis and apoptosis. The critical role of NLRP3 inflammasome suppression in the enhanced therapeutic efficacy of MSC spheroids was further confirmed in an in vivo mouse model of DSS-induced colitis. These findings suggest that 3D culture confers survival and functional advantages to MSCs by suppressing NLRP3 inflammasome activation.


Subject(s)
Colitis , Inflammasomes , Mesenchymal Stem Cells , Animals , Mice , Colitis/chemically induced , Colitis/genetics , Colitis/immunology , Inflammasomes/genetics , Inflammasomes/immunology , Mesenchymal Stem Cells/immunology , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Signal Transduction , Cell Culture Techniques, Three Dimensional
6.
Sci Adv ; 8(34): eabn8614, 2022 08 26.
Article in English | MEDLINE | ID: mdl-36001671

ABSTRACT

Immunomodulation is an essential consideration for cell replacement procedures. Unfortunately, lifelong exposure to nonspecific systemic immunosuppression results in immunodeficiency and has toxic effects on nonimmune cells. Here, we engineered hybrid spheroids of mesenchymal stem cells (MSCs) with rapamycin-releasing poly(lactic-co-glycolic acid) microparticles (RAP-MPs) to prevent immune rejection of islet xenografts in diabetic C57BL/6 mice. Hybrid spheroids were rapidly formed by incubating cell-particle mixture in methylcellulose solution while maintaining high cell viability. RAP-MPs were uniformly distributed in hybrid spheroids and sustainably released RAP for ~3 weeks. Locoregional transplantation of hybrid spheroids containing low doses of RAP-MPs (200- to 4000-ng RAP per recipient) significantly prolonged islet survival times and promoted the generation of regional regulatory T cells. Enhanced programmed death-ligand 1 expression by MSCs was found to be responsible for the immunomodulatory performance of hybrid spheroids. Our results suggest that these hybrid spheroids offer a promising platform for the efficient use of MSCs in the transplantation field.


Subject(s)
Mesenchymal Stem Cells , Spheroids, Cellular , Animals , Humans , Immunomodulation , Mesenchymal Stem Cells/metabolism , Mice , Mice, Inbred C57BL , Transplantation, Heterologous
7.
Antioxid Redox Signal ; 37(13-15): 1030-1050, 2022 11.
Article in English | MEDLINE | ID: mdl-35286219

ABSTRACT

Aims: Raloxifene, a selective estrogen receptor (ER) modulator, has been reported to exert the tumor-suppressive effects in both ER-positive and ER-negative cancer cells; however, the mechanisms underlying its ER-independent anti-cancer effects are poorly understood. The NLRP3 inflammasome, a critical component of the innate immune system, has recently received growing attention owing to its multifaceted roles in various aspects of cancer development. The present study aimed at examining the involvement of NLRP3 inflammasomes in the anti-breast cancer effects of raloxifene and its underlying mechanisms. Results: Raloxifene significantly inhibited the activation of NLRP3 inflammasomes in various breast cancer cell lines. Importantly, forced expression of a gain-of-function variant of NLRP3 rescued breast cancer cells from growth arrest by raloxifene, suggesting that the suppression of NLRP3 inflammasomes activation mediates the raloxifene-induced inhibition of breast cancer growth. Mechanistically, raloxifene suppressed NLRP3 inflammasomes activation by lowering the cellular levels of reactive oxygen species (ROS) through the modulation of redox signaling mediated via aryl hydrocarbon receptor (AhR)-nuclear factor erythroid 2-related factor 2 (Nrf2)-heme oxygenase-1 (HO-1) axis or the impaired generation of mitochondrial ROS in a mitophagy-dependent manner. Further, the blockage of AhR signaling or inhibition of mitophagy abolished the tumor-suppressive effect of raloxifene in a human breast tumor xenograft model. Innovation: We elucidate a novel molecular mechanism underlying the breast tumor suppressing effect of raloxifene. Conclusion: The results observed in this study suggest that the modulation of NLRP3 inflammasomes activation is a critical event in the inhibition of breast tumor growth by raloxifene. Antioxid. Redox Signal. 37, 1030-1050.


Subject(s)
Breast Neoplasms , Inflammasomes , Humans , Female , Inflammasomes/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Mitophagy , Receptors, Aryl Hydrocarbon , Reactive Oxygen Species/metabolism , Raloxifene Hydrochloride/pharmacology , Oxidation-Reduction , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism
8.
Heliyon ; 8(3): e09070, 2022 Mar.
Article in English | MEDLINE | ID: mdl-35287327

ABSTRACT

Balanophora laxiflora, a medicinal plant traditionally used to treat fever, pain, and inflammation in Vietnam, has been reported to possess prominent anti-inflammatory activity. This study examined the active constituents and molecular mechanisms underlying these anti-inflammatory effects using bioactivity-guided isolation in combination with cell-based assays and animal models of inflammation. Among the isolated compounds, the triterpenoid (21α)-22-hydroxyhopan-3-one (1) showed the most potent inhibitory effect on COX-2 expression in LPS-stimulated Raw 264.7 macrophages. Furthermore, 1 suppressed the expression of the inflammatory mediators iNOS, IL-1ß, INFß, and TNFα in activated Raw 264.7 macrophages and alleviated the inflammatory response in carrageenan-induced paw oedema and a cotton pellet-induced granuloma model. Mechanistically, the anti-inflammatory effects of 1 were mediated via decreasing cellular reactive oxygen species (ROS) levels by inhibiting NADPH oxidases (NOXs) and free radical scavenging activities. By downregulating ROS signalling, 1 reduced the activation of MAPK signalling pathways, leading to decreased AP-1-dependent transcription of inflammatory mediators. These findings shed light on the chemical constituents that contribute to the anti-inflammatory actions of B. laxiflora and suggest that 1 is a promising candidate for treating inflammation-related diseases.

9.
J Exp Clin Cancer Res ; 41(1): 9, 2022 Jan 05.
Article in English | MEDLINE | ID: mdl-34986886

ABSTRACT

BACKGROUND: Adiponectin, the most abundant adipokine derived from adipose tissue, exhibits a potent suppressive effect on the growth of breast cancer cells; however, the underlying molecular mechanisms for this effect are not completely understood. Fatty acid metabolic reprogramming has recently been recognized as a crucial driver of cancer progression. Adiponectin demonstrates a wide range of metabolic activities for the modulation of lipid metabolism under physiological conditions. However, the biological actions of adiponectin in cancer-specific lipid metabolism and its role in the regulation of cancer cell growth remain elusive. METHODS: The effects of adiponectin on fatty acid metabolism were evaluated by measuring the cellular neutral lipid pool, free fatty acid level, and fatty acid oxidation (FAO). Colocalization between fluorescent-labeled lipid droplets and LC3/lysosomes was employed to detect lipophagy activation. Cell viability and apoptosis were examined by MTS assay, caspase-3/7 activity measurement, TUNEL assay, and Annexin V binding assay. Gene expression was determined by real time-quantitative polymerase chain reaction (RT-qPCR) and western blot analysis. The transcriptional activity of SREBP-1 was examined by a specific dsDNA binding assay. The modulatory roles of SIRT-1 and adiponectin-activated mediators were confirmed by gene silencing and/or using their pharmacological inhibitors. Observations from in vitro assays were further validated in an MDA-MB-231 orthotopic breast tumor model. RESULTS: Globular adiponectin (gAcrp) prominently decreased the cellular lipid pool in different breast cancer cells. The cellular lipid deficiency promoted apoptosis by causing disruption of lipid rafts and blocking raft-associated signal transduction. Mechanistically, dysregulated cellular lipid homeostasis by adiponectin was induced by two concerted actions: 1) suppression of fatty acid synthesis (FAS) through downregulation of SREBP-1 and FAS-related enzymes, and 2) stimulation of lipophagy-mediated lipolysis and FAO. Notably, SIRT-1 induction critically contributed to the adiponectin-induced metabolic alterations. Finally, fatty acid metabolic remodeling by adiponectin and the key role of SIRT-1 were confirmed in nude mice bearing breast tumor xenografts. CONCLUSION: This study elucidates the multifaceted role of adiponectin in tumor fatty acid metabolic reprogramming and provides evidence for the connection between its metabolic actions and suppression of breast cancer.


Subject(s)
Adiponectin/metabolism , Breast Neoplasms/genetics , Cell Death/physiology , Fatty Acids/metabolism , Animals , Breast Neoplasms/pathology , Female , Humans , Mice , Mice, Nude , Transfection
10.
Arch Pharm Res ; 45(1): 38-50, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34797495

ABSTRACT

Adiponectin, an adipose tissue-derived hormone, exhibits a modulatory effect on cell death/survival and possesses potent anti-inflammatory properties. However, the underlying molecular mechanisms remain elusive. Sestrin2, a stress-inducible metabolic protein, has shown cytoprotective and inflammation-modulatory effects under stressful conditions. In this study, we examined the role of sestrin2 signaling in the modulation of cell survival and inflammatory responses by globular adiponectin (gAcrp) in macrophages. We observed that gAcrp induced a significant increase in sestrin2 expression in both RAW 264.7 murine macrophages and primary murine macrophages. Notably, gAcrp treatment markedly increased expression of hypoxia inducible factor-1 α (HIF-1α) and gene silencing of HIF-1α blocked sestrin2 induction by gAcrp. In addition, pretreatment with a pharmacological inhibitor of ERK or PI3K abrogated both sestrin2 and HIF-1α expression by gAcrp, indicating that ERK/PI3K-mediated HIF-1α signaling pathway plays a critical role in sestrin2 induction by gAcrp. Furthermore, sestrin2 induction is implicated in autophagy activation, and knockdown of sestrin2 prevented enhanced cell viability by gAcrp. Moreover, gene silencing of sestrin2 caused restoration of gAcrp-induced expression of anti-inflammatory genes in a gene-selective manner. Taken together, these results indicate that sestrin2 induction critically contributes to cell survival and anti-inflammatory responses by gAcrp in macrophages.


Subject(s)
Adiponectin/pharmacology , Anti-Inflammatory Agents/pharmacology , Cell Survival/drug effects , Macrophages, Peritoneal/drug effects , Peroxidases/metabolism , RAW 264.7 Cells/drug effects , Animals , Autophagy/drug effects , Blotting, Western , Gene Silencing , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , MAP Kinase Signaling System/drug effects , Macrophages, Peritoneal/metabolism , Mice , RAW 264.7 Cells/metabolism , Reactive Oxygen Species/metabolism , Real-Time Polymerase Chain Reaction
11.
Int J Mol Sci ; 22(3)2021 Feb 01.
Article in English | MEDLINE | ID: mdl-33535537

ABSTRACT

Adiposity is associated with an increased risk of various types of carcinoma. One of the plausible mechanisms underlying the tumor-promoting role of obesity is an aberrant secretion of adipokines, a group of hormones secreted from adipose tissue, which have exhibited both oncogenic and tumor-suppressing properties in an adipokine type- and context-dependent manner. Increasing evidence has indicated that these adipose tissue-derived hormones differentially modulate cancer cell-specific metabolism. Some adipokines, such as leptin, resistin, and visfatin, which are overproduced in obesity and widely implicated in different stages of cancer, promote cellular glucose and lipid metabolism. Conversely, adiponectin, an adipokine possessing potent anti-tumor activities, is linked to a more favorable metabolic phenotype. Adipokines may also play a pivotal role under the reciprocal regulation of metabolic rewiring of cancer cells in tumor microenvironment. Given the fact that metabolic reprogramming is one of the major hallmarks of cancer, understanding the modulatory effects of adipokines on alterations in cancer cell metabolism would provide insight into the crosstalk between obesity, adipokines, and tumorigenesis. In this review, we summarize recent insights into putative roles of adipokines as mediators of cellular metabolic rewiring in obesity-associated tumors, which plays a crucial role in determining the fate of tumor cells.


Subject(s)
Adipokines/metabolism , Disease Progression , Neoplasms/complications , Neoplasms/metabolism , Obesity/complications , Obesity/metabolism , Adipose Tissue/metabolism , Adiposity , Animals , Glucose/metabolism , Glycolysis , Humans , Inflammation/pathology , Leptin/metabolism , Lipid Metabolism , Mice , Mitochondria/metabolism , Nicotinamide Phosphoribosyltransferase/metabolism , Oxidation-Reduction , Phenotype , Reactive Oxygen Species , Resistin/metabolism , Tumor Microenvironment
12.
Mol Oncol ; 15(2): 657-678, 2021 02.
Article in English | MEDLINE | ID: mdl-33226729

ABSTRACT

Leptin, a hormone predominantly derived from adipose tissue, is well known to induce growth of breast cancer cells. However, its underlying mechanisms remain unclear. In this study, we examined the role of reprogramming of lipid metabolism and autophagy in leptin-induced growth of breast cancer cells. Herein, leptin induced significant increase in fatty acid oxidation-dependent ATP production in estrogen receptor-positive breast cancer cells. Furthermore, leptin induced both free fatty acid release and intracellular lipid accumulation, indicating a multifaceted effect of leptin in fatty acid metabolism. These findings were further validated in an MCF-7 tumor xenograft mouse model. Importantly, all the aforementioned metabolic effects of leptin were mediated via autophagy activation. In addition, SREBP-1 induction driven by autophagy and fatty acid synthase induction, which is mediated by SREBP-1, plays crucial roles in leptin-stimulated metabolic reprogramming and are required for growth of breast cancer cell, suggesting a pivotal contribution of fatty acid metabolic reprogramming to tumor growth by leptin. Taken together, these results highlighted a crucial role of autophagy in leptin-induced cancer cell-specific metabolism, which is mediated, at least in part, via SREBP-1 induction.


Subject(s)
Autophagy , Breast Neoplasms/metabolism , Cellular Reprogramming , Fatty Acids/metabolism , Leptin/metabolism , Neoplasm Proteins/metabolism , Sterol Regulatory Element Binding Protein 1/metabolism , Animals , Breast Neoplasms/genetics , Fatty Acids/genetics , Female , Humans , Leptin/genetics , MCF-7 Cells , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Proteins/genetics , Sterol Regulatory Element Binding Protein 1/genetics
13.
Article in English | WPRIM (Western Pacific) | ID: wpr-895081

ABSTRACT

Balanophora laxiflora Hemsl. (Balanophoraceae) is a traditional medicinal plant with a diverse array of biological activities. In our exploration of new bioactive constituents from B. laxiflora, we isolated five compounds, including a new lignan, balanophorone (5), and four known phenolic compounds (1–4). The chemical structures of these compounds were determined by extensive spectroscopic analyses, including 1D and 2D NMR, HR-ESI-MS, and CD. In addition, we evaluated the effects of each of the isolates (1–5) on the messenger RNA expression levels of tumor necrosis factor (TNF)-α and cyclooxygenase (COX)-2 in lipopolysaccharide (LPS)-stimulated RAW 264.7 macrophages and cytotoxicity against MCF-7 and MDA-MB-231 breast cancer cells. Compound 2 showed significant inhibition of LPS-induced COX-2 and TNF-α expression in RAW 264.7 macrophages, while compound 4 showed moderate cytotoxicity against MCF-7 and MDA-MB-231 breast cancer cells, with IC 50 values of 18.3 and 30.7 μM, respectively. No significant effects on the viability of normal mammary epithelial cells were observed.

14.
Article in English | WPRIM (Western Pacific) | ID: wpr-902785

ABSTRACT

Balanophora laxiflora Hemsl. (Balanophoraceae) is a traditional medicinal plant with a diverse array of biological activities. In our exploration of new bioactive constituents from B. laxiflora, we isolated five compounds, including a new lignan, balanophorone (5), and four known phenolic compounds (1–4). The chemical structures of these compounds were determined by extensive spectroscopic analyses, including 1D and 2D NMR, HR-ESI-MS, and CD. In addition, we evaluated the effects of each of the isolates (1–5) on the messenger RNA expression levels of tumor necrosis factor (TNF)-α and cyclooxygenase (COX)-2 in lipopolysaccharide (LPS)-stimulated RAW 264.7 macrophages and cytotoxicity against MCF-7 and MDA-MB-231 breast cancer cells. Compound 2 showed significant inhibition of LPS-induced COX-2 and TNF-α expression in RAW 264.7 macrophages, while compound 4 showed moderate cytotoxicity against MCF-7 and MDA-MB-231 breast cancer cells, with IC 50 values of 18.3 and 30.7 μM, respectively. No significant effects on the viability of normal mammary epithelial cells were observed.

15.
Arch Pharm Res ; 43(10): 997-1016, 2020 Oct.
Article in English | MEDLINE | ID: mdl-33078304

ABSTRACT

Aberrant production of adipokines, a group of adipocytes-derived hormones, is considered one of the most important pathological characteristics of obesity. In individuals with obesity, beneficial adipokines, such as adiponectin are downregulated, whereas leptin and other pro-inflammatory adipokines are highly upregulated. Hence, the imbalance in levels of these adipokines is thought to promote the development of obesity-linked complications. However, the mechanisms by which adipokines contribute to the pathogenesis of various diseases have not been clearly understood. Inflammasomes represent key signaling platform that triggers the inflammatory and immune responses through the processing of the interleukin family of pro-inflammatory cytokines in a caspase-1-dependent manner. Beyond their traditional function as a component of the innate immune system, inflammasomes have been recently integrated into the pathological process of multiple metabolism- and obesity-related disorders such as cardiovascular diseases, diabetes, fatty liver disease, and cancer. Interestingly, emerging evidence also highlights the role of adipokines in the modulation of inflammasomes activation, making it a promising mechanism underlying distinct biological actions of adipokines in diseases driven by inflammation and metabolic disorders. In this review, we summarize the effects of adipokines, in particular adiponectin, leptin, visfatin and apelin, on inflammasomes activation and their implications in the pathophysiology of obesity-linked complications.


Subject(s)
Adipokines/metabolism , Inflammasomes/immunology , Metabolic Diseases/complications , Obesity/complications , Adipocytes/cytology , Adipocytes/immunology , Adipocytes/metabolism , Animals , Cardiovascular Diseases/immunology , Cardiovascular Diseases/metabolism , Diabetes Mellitus/immunology , Diabetes Mellitus/metabolism , Disease Models, Animal , Down-Regulation/immunology , Fatty Liver/immunology , Fatty Liver/metabolism , Humans , Inflammasomes/metabolism , Metabolic Diseases/immunology , Metabolic Diseases/metabolism , Neoplasms/immunology , Neoplasms/metabolism , Obesity/immunology , Obesity/metabolism , Up-Regulation/immunology
16.
Cancers (Basel) ; 12(3)2020 Mar 06.
Article in English | MEDLINE | ID: mdl-32155890

ABSTRACT

Adiponectin, an adipokine predominantly derived from adipose tissue, exhibits potent antitumor properties in breast cancer cells. However, its mechanisms of action remain elusive. Inflammasomes-intracellular multimeric protein complexes-modulate cancer cell growth in a complicated manner, as well as playing a role in the innate immune system. Herein, we examined the potential role of inflammasomes in the antitumor activity of adiponectin and found that globular adiponectin (gAcrp) significantly suppressed inflammasomes activation in breast cancer cells both in vitro and in vivo conditions, as determined by decreased expression of inflammasomes components, including NOD-like receptor pyrin domain-containing protein 3 (NLRP3) and the apoptosis-associated speck-like protein containing a CARD (ASC), and inhibition of interleukin-1ß and caspase-1 activation. Treatment with pharmacological inhibitors of inflammasomes caused decrease in cell viability, apoptosis induction, and G0/G1 cell cycle arrest, suggesting that inflammasomes activation is implicated in the growth of breast cancer cells. In addition, treatment with gAcrp generated essentially similar results to those of inflammasomes inhibitors, further indicating that suppression of breast cancer cell growth by gAcrp is mediated via modulation of inflammasomes. Mechanistically, gAcrp suppressed inflammasomes activation through sestrin2 (SESN2) induction, liver kinase B1 (LKB-1)-dependent AMP-activated protein kinase (AMPK) phosphorylation, and alleviation of endoplasmic reticulum (ER) stress. Taken together, these results demonstrate that gAcrp inhibits growth of breast cancer cells by suppressing inflammasomes activation, at least in part, via SESN2 induction and AMPK activation-dependent mechanisms.

17.
Int J Pharm ; 555: 63-76, 2019 Jan 30.
Article in English | MEDLINE | ID: mdl-30448315

ABSTRACT

The first objective of this study was to optimize a supersaturatable self-nanoemulsifying drug delivery system (S-SNEDDS) containing silymarin through the investigation of the single and synergistic effect of either SNEDDS or a precipitation inhibitor on dissolution efficiency (DE) of silymarin. The bioavailability and hepatoprotective activity of S-SNEDDS were then compared to those of a branded product (Legalon®, Meda). SNEDDS containing silymarin was developed by titration technique, and Poloxamer 407 was selected as the optimal precipitation inhibitor by using casting film and solvent-shift method. The interaction of silybin (the major active constituent of silymarin) and the polymer was then determined by differential scanning calorimetry, powder X-ray diffractometry (PXRD), Fourier transforms infrared spectroscopy and 1H NMR analysis. The combination of two techniques including SNEDDS and addition of 10% of Poloxamer 407 remarkably increased DE4h (88.28%) compared to the reference product (6.41%). The relative bioavailability of S-SNEDDS versus Legalon® was about 760%. The hepatoprotective activity of S-SNEDDS in CCl4-induced mice was also superior to the commercial product in declining both the levels of serum transaminases (ALT, AST) and lipid peroxidation as well as glutathione and superoxide dismutase (SOD) activities under tested doses calculated as silybin (10, 25 and 50 mg/kg). These biopharmaceutical and pharmacological advantages of S-SNEDDS indicated prospects in the development of a novel product that offers lower strength of silymarin while enhancing therapeutic outcomes.


Subject(s)
Drug Delivery Systems , Liver Diseases/prevention & control , Nanoparticles , Silymarin/administration & dosage , Animals , Biological Availability , Carbon Tetrachloride/toxicity , Chemistry, Pharmaceutical/methods , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Carriers/chemistry , Drug Liberation , Emulsions , Male , Mice , Poloxamer/chemistry , Protective Agents/administration & dosage , Protective Agents/pharmacokinetics , Protective Agents/pharmacology , Rabbits , Silybin/administration & dosage , Silybin/chemistry , Silymarin/pharmacokinetics , Silymarin/pharmacology , Solubility
18.
Article in English | WPRIM (Western Pacific) | ID: wpr-820701

ABSTRACT

OBJECTIVE@#To screen Vietnamese medicinal plants for xanthine oxidase (XO) inhibitory activity and to isolate XO inhibitor(s) from the most active plant.@*METHODS@#The plants materials were extracted by methanol. The active plant materials were fractionated using different organic solvents, including n-hexane, ethyl acetate, and n-butanol. Bioassay-guided fractionation and column chromatography were used to isolate compounds. The compounds structures were elucidated by analysis of spectroscopic data, including IR, MS, and NMR.@*RESULTS@#Three hundreds and eleven methanol extracts (CME) belonging to 301 Vietnamese herbs were screened for XO inhibitory activity. Among these plants, 57 extracts displayed XO inhibitory activity at 100 μg/mL with inhibition rates of over 50%. The extracts of Archidendron clypearia (A. clypearia), Smilax poilanei, Linociera ramiflora and Passiflora foetida exhibited the greatest potency with IC values below 30 μg/mL. Chemical study performed on the extract of A. clypearia resulted in the isolation of six compounds, including 1-octacosanol, docosenoic acid, daucosterol, methyl gallate, quercitrin and (-)-7-O-galloyltricetiflavan. The compound (-)-7-O-galloyltricetiflavan showed the most potent XO inhibitory activity with an IC value of 25.5 μmol/L.@*CONCLUSIONS@#From this investigation, four Vietnamese medicinal plants were identified to have XO inhibitory effects with IC values of the methanol extracts below 30 μg/mL. Compound (-)-7-O- galloyltricetiflavan was identified as an XO inhibitor from A. clypearia with IC value of 25.5 μmol/L.

19.
Article in Chinese | WPRIM (Western Pacific) | ID: wpr-972610

ABSTRACT

Objective To screen Vietnamese medicinal plants for xanthine oxidase (XO) inhibitory activity and to isolate XO inhibitor(s) from the most active plant. Methods The plants materials were extracted by methanol. The active plant materials were fractionated using different organic solvents, including n-hexane, ethyl acetate, and n-butanol. Bioassay-guided fractionation and column chromatography were used to isolate compounds. The compounds structures were elucidated by analysis of spectroscopic data, including IR, MS, and NMR. Results Three hundreds and eleven methanol extracts (CME) belonging to 301 Vietnamese herbs were screened for XO inhibitory activity. Among these plants, 57 extracts displayed XO inhibitory activity at 100 μg/mL with inhibition rates of over 50%. The extracts of Archidendron clypearia (A. clypearia), Smilax poilanei, Linociera ramiflora and Passiflora foetida exhibited the greatest potency with IC

SELECTION OF CITATIONS
SEARCH DETAIL
...