Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
Phytomedicine ; 123: 155188, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38056146

ABSTRACT

BACKGROUND: Non-alcoholic steatohepatitis (NASH) is a global health problem with no effective treatment. Isoquercitrin (IQ) alters hepatic lipid metabolism and inhibits adipocyte differentiation. The underlying regulatory mechanisms of IQ in regulating insulin resistance (IR) and lipid metabolism remain unclear. PURPOSE: This study was aimed at investigating the effects of IQ on NASH and deciphering whether the underlying mechanisms are via modulation of galectin-3 mediated IR and lipid metabolism. METHODS: IR-HepG2 cell lines were used to demonstrate the ability of IQ to modulate galectin-3-mediated glucose disposal and lipid metabolism. A 20-week high-fat diet (HFD)-induced NASH model was established in C57BL/6J mice, and the protective effect of IQ on lipid disposal in the liver was verified. Further, the mRNA and protein levels of glucose and lipid metabolism were investigated, and lysophosphatidylcholine (LPC) and acylcarnitine (AC) profiling were performed to characterize the changes in endogenous substances associated with mitochondrial function and lipid metabolism in serum and cells. Furthermore, the pharmacokinetic features of IQ were explored in a rat model of NASH. RESULTS: IQ restored liver function and ameliorated inflammation and lipid accumulationin NASH model mice. Notably, significant regulation of the proteins included fatty acid-generating and transporting, cholesterol metabolism enzymes, nuclear transcription factors, mitochondrial metabolism, and IR-related enzymes was noted to be responsible for the therapeutic mechanisms of IQ against experimental NASH. Serum lipid metabolism-related metabolomic assay confirmed that LPC and AC biosynthesis mostly accounted for the therapeutic effect of IQ in mice with NASH and that IQ maintained the homeostasis of LPC and AC levels. CONCLUSION: This is the first study showing that IQ protects against of NASH by modulating galectin-3-mediated IR and lipid metabolism. The mechanisms responsible for liver protection and improved lipid metabolic disorder by IQ may be related to the suppression of IR and regulation of mitochondrial function and lipid metabolism. Galectin-3 down-regulation represents a potentially novel approach for the treatment and prevention of NASH.


Subject(s)
Insulin Resistance , Non-alcoholic Fatty Liver Disease , Quercetin/analogs & derivatives , Mice , Animals , Rats , Non-alcoholic Fatty Liver Disease/drug therapy , Non-alcoholic Fatty Liver Disease/metabolism , Galectin 3/genetics , Galectin 3/metabolism , Galectin 3/pharmacology , Lipid Metabolism , Mice, Inbred C57BL , Liver , Diet, High-Fat/adverse effects , Glucose/metabolism , Lipids
2.
Immunopharmacol Immunotoxicol ; 45(6): 692-700, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37358143

ABSTRACT

OBJECTIVE: Treatment with TNF-α inhibitors improve psoriasis with minimize/minor neutrophils infiltration and CXCL-1/8 expression in psoriatic lesions. However, the fine mechanism of TNF-α initiating psoriatic inflammation by tuning keratinocytes is unclear. Our previous research identified the deficiency of intracellular galectin-3 was sufficient to promote psoriasis inflammation characterized by neutrophil accumulation. This study aims to investigate whether TNF-α participated in psoriasis development through dysregulating galectin-3 expression. METHODS: mRNA levels were assessed through quantitative real-time PCR. Flow cytometry was used to detect cell cycle/apoptosis. Western blot was used to evaluate the activation of the NF-κB signaling pathway. HE staining and immunochemistry were used to detect epidermal thickness and MPO expression, respectively. Specific small interfering RNA (siRNA) was used to knock down hsa-miR-27a-3p while plasmids transfection was used to overexpress galectin-3. Further, the multiMiR R package was utilized to predict microRNA-target interaction. RESULTS AND DISCUSSION: We found that TNF-α stimulation altered cell proliferation and differentiation and promoted the production of psoriasis-related inflammatory mediators along with the inhibition of galectin-3 expression in keratinocytes. Supplement of galectin-3 could counteract the rise of CXCL-1/8 but not the other phenotypes of keratinocytes induced by TNF-α. Mechanistically, inhibition of the NF-κB signaling pathway could counteract the decrease of galectin-3 and the increase of hsa-miR-27a-3p expression whereas silence of hsa-miR-27a-3p could counteract the decrease of galectin-3 expression induced by TNF-α treatment in keratinocytes. Intradermal injection of murine anti-CXCL-2 antibody greatly alleviated imiquimod-induced psoriasis-like dermatitis. CONCLUSION: TNF-α initiates psoriatic inflammation by increasing CXCL-1/8 in keratinocytes mediated by the axis of NF-κB-hsa-miR-27a-3p-galectin-3 pathway.


Subject(s)
Galectin 3 , Keratinocytes , MicroRNAs , Psoriasis , Tumor Necrosis Factor-alpha , Tumor Necrosis Factor-alpha/pharmacology , Keratinocytes/metabolism , HaCaT Cells , Humans , MicroRNAs/genetics , Chemokine CXCL1/metabolism , Interleukin-8/metabolism , Galectin 3/genetics , Psoriasis/genetics , Psoriasis/pathology , NF-kappa B/metabolism , Signal Transduction , Female , Animals , Mice , Mice, Inbred C57BL
3.
Bioengineered ; 13(2): 4260-4270, 2022 02.
Article in English | MEDLINE | ID: mdl-35191812

ABSTRACT

Cardiac hypertrophy results from the adaptive response of the myocardium to pressure overload on the heart. Tanshinone IIA (Tan IIA) is the major active compound extracted from Salvia miltiorrhiza Bunge, which possesses various pharmacological benefits. In the present study, the effect and mechanism of action of Tan IIA on cardiac hypertrophy were studied. Ang II-induced and transverse aortic constriction (TAC)-induced cardiomyocyte hypertrophy models were used to evaluate the effect of Tan IIA. An adenoviral vector system was utilized to overexpress galectin-3. The results revealed that Tan IIA significantly inhibited Ang II-induced hypertrophy in vitro and TAC-induced cardiac hypertrophy in vivo. Furthermore, Tan IIA notably inhibited the expression of galectin-3. Rescue experiments indicated that galectin-3 overexpression reversed the effects of Tan IIA, which further validated the interaction between Tan IIA and galectin-3. Additionally, Tan IIA suppressed alkB homolog 5, RNA demethylase (ALKBH5)-mediated N6-methyladenosine (m6A) modification of galectin-3. In summary, the results of the present study indicated that Tan IIA attenuates cardiac hypertrophy by targeting galectin-3, suggesting that galectin-3 plays a critical role in cardiac hypertrophy and represents a new therapeutic target.


Subject(s)
Abietanes/administration & dosage , Cardiomegaly/drug therapy , Drugs, Chinese Herbal/administration & dosage , Galectin 3/metabolism , Animals , Cardiomegaly/genetics , Cardiomegaly/metabolism , Galectin 3/genetics , Humans , Male , Methylation , Mice , Mice, Inbred C57BL , Salvia miltiorrhiza/chemistry , Signal Transduction
4.
Biochem Pharmacol ; 178: 114113, 2020 08.
Article in English | MEDLINE | ID: mdl-32579956

ABSTRACT

Tumor-associated macrophages (TAMs) have been shown to be associated with poor prognosis of cancer and are predominately localized in the hypoxia regions of tumor. We demonstrated in this study that hypoxia increases the synthesis and secretion of galectin-3 by TAMs. The increased expression of galectin-3 in TAMs was seen to be associated with nucleation of transcription factor NF-κB through generation and activation of ROS and promoted tumor growth and metastasis in vitro and in mice through multiple molecular mechanisms. It was found that the TAMs-mediated promotion of tumor growth and metastasis in hypoxia was inhibited by administration of macrophage-depletion agent clodronate liposomal (CL) or galectin-3 inhibitor modified citric pectin (MCP) in orthotopic syngeneic mammary adenocarcinoma model and metastasis model. Co-administration of anti-angiogenesis agent sorafenib or bevacizumab with CL and MCP showed to cause stronger inhibition of tumor growth and metastasis than administration of each agent alone. These results indicate that hypoxia-induced galectin-3 expression and secretion from TAMs promotes tumor growth and metastasis. Targeting the actions of galectin-3 in hypoxia may be a potential therapeutic strategy for cancer treatment.


Subject(s)
Adenocarcinoma/drug therapy , Bevacizumab/pharmacology , Breast Neoplasms/drug therapy , Galectin 3/antagonists & inhibitors , Gene Expression Regulation, Neoplastic , Hypoxia/drug therapy , Mammary Neoplasms, Experimental/drug therapy , Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Animals , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Clodronic Acid/pharmacology , Coculture Techniques , Disease Progression , Female , Galectin 3/genetics , Galectin 3/metabolism , Humans , Hypoxia/genetics , Hypoxia/metabolism , Hypoxia/pathology , Lymphatic Metastasis , Macrophages/drug effects , Macrophages/metabolism , Macrophages/pathology , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/metabolism , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Inbred BALB C , Mice, Nude , NF-kappa B/genetics , NF-kappa B/metabolism , Neovascularization, Pathologic , Pectins/pharmacology , Signal Transduction , Sorafenib/pharmacology
5.
Biomed Pharmacother ; 126: 110071, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32172066

ABSTRACT

Myocardial fibrosis (MF) plays a key role in the development and progression of heart failure (HF) with limited effective therapies. Galectin-3 (Gal-3) is a biomarker associated with fibrosis and inflammation in patients with HF. The Gal-3 inhibitor modified citrus pectin (MCP) protects against cardiac dysfunction, though the underlying mechanism remains unclear. The aim of this study was to investigate the effect and mechanism of MCP on MF using an isoproterenol (ISO)-induced rat model of HF. Cardiac function was analyzed by echocardiography and electrocardiography. Histopathological changes in the heart tissue were assessed by hematoxylin-eosin and Masson trichrome staining. The mRNA and protein expression levels of signaling molecules and pro-inflammatory cytokines were monitored by immunohistochemistry, western blot, qRT-PCR and ELISA analyses. The results demonstrated that MCP ameliorated cardiac dysfunction, decreased myocardial injury and reduced collagen deposition. Furthermore, MCP downregulated the expression of Gal-3, TLR4 and MyD88, thereby inhibiting NF-κB-p65 activation. MCP also decreased the expression of IL-1ß, IL-18 and TNF-α, which have been implicated in the pathogenesis of HF. These inhibitory effects were observed on day 15 and continued until day 22. Taken together, these results suggest that MCP ameliorates cardiac dysfunction through inhibiting inflammation and MF. These effects may be through downregulating Gal-3 expression and suppressing activation of the TLR4/MyD88/NF-κB signaling pathway. The present study supports the use of Gal-3 as a therapeutic target for the treatment of MF after myocardial infarction.


Subject(s)
Cardiomyopathies/etiology , Cardiomyopathies/metabolism , Galectin 3/metabolism , Myeloid Differentiation Factor 88/metabolism , NF-kappa B/metabolism , Pectins/pharmacology , Signal Transduction/drug effects , Toll-Like Receptor 4/metabolism , Animals , Biomarkers , Biopsy , Cardiomyopathies/diagnosis , Cardiomyopathies/drug therapy , Cytokines/metabolism , Disease Susceptibility , Echocardiography , Electrocardiography , Fibrosis , Galectin 3/genetics , Heart Function Tests , Immunohistochemistry , Inflammation , Inflammation Mediators/metabolism , Male , Models, Biological , Rats
6.
Carbohydr Res ; 490: 107903, 2020 Apr.
Article in English | MEDLINE | ID: mdl-32171073

ABSTRACT

This study intends to investigate the inhibitory potential of different plant derived saccharides on cell migration and adhesion of pancreatic ductal adenocarcinoma (PDAC) cells to microvascular liver endothelium, particularly considering the role of transmembranous galectin-3. PDAC cell lines PancTu1 and Panc1 were characterized by considerable (transmembranous) galectin-3 (Gal3) expression. SiRNA mediated Gal3 knockdown as well as treatment with differentially processed pectins and arabinogalactan-proteins (AGPs) did not impact on cell migration of either PDAC cell line. In contrast, Gal3 knockdown reduced adhesion of PDAC cells to the liver endothelial cell line TMNK-1 being more pronounced in Panc1 cells. Similarly, plant derived substances did not impact cell adhesion of PancTu1 cells while partially hydrolyzed citrus pectin (MCP), pectinase-treated MCP (MCPPec) and partially hydrolized AGP (AGPTFA) clearly diminished adhesive properties of Panc1 cells. MCPPec or AGPTFA could not further intensify the adhesion reducing effect of galectin-3 knockdown, indicating that these plant derived polysaccharides are able to inhibit PDAC cell adhesion to liver endothelial cells in a galectin-3 dependent manner. Overall, these data suggest an inhibitory potential of plant derived processed saccharides which have undergone chemical modification in impairing PDAC cell adhesion to liver endothelium.


Subject(s)
Carcinoma, Pancreatic Ductal/metabolism , Citrus/chemistry , Galectin 3/metabolism , Mucoproteins/pharmacology , Pancreatic Neoplasms/metabolism , Pectins/pharmacology , Blood Proteins , Carcinoma, Pancreatic Ductal/drug therapy , Carcinoma, Pancreatic Ductal/genetics , Cell Adhesion/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Galectin 3/genetics , Galectins , Gene Expression Regulation, Neoplastic/drug effects , Gene Knockdown Techniques , Humans , Neoplasm Metastasis , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/genetics , Plant Proteins/pharmacology
7.
Int J Biol Macromol ; 139: 486-499, 2019 Oct 15.
Article in English | MEDLINE | ID: mdl-31356934

ABSTRACT

Galectin-3 and galectin-3 binding proteins (G3BP) are implicated as key players in metastasis. In the current study, we evaluated the effect of pectic polysaccharides on galectin-3 and G3BP mediated metastasis in vitro (cells) and in vivo (tissues). In vitro study (double immunostaining) confirms the presence of galectin-3 on the cell surface and G3BP in the interlinking region of the cells confirming the role of G3BP in bridging galectin-3 molecules. Dietary carrot (Daucus carota) pectic polysaccharide (CRPP) blocked the expression of galectin-3 and G3BP more effectively (80%), whereas the expressions were reduced to 60% upon treatment with swallow root (Decalepis hamiltonii) pectic polysaccharide (SRPP), ß­carotene and deferoxamine (antiproliferative drug). Ginger (Gingiber officinale) pectic polysaccharide (GRPP) showed only 20% reduction. CRPP reduced 80% of tumor incidence followed by cyclophosphamide - a chemotherapeutic drug (77%), SRPP (67%) and GRPP (45%). Further 3-5 folds reduction in galectin-3/G3BP expression followed by infiltration of macrophages into the deeper layer of the skin by CRPP and SRPP suggested the anticancer property via immunomodulation. Surface Plasmon Resonance (SPR) studies confirm galectin-3 and G3BP interaction, which are disrupted during the treatment with dietary pectic polysaccharides (DPP) (Supplementary Scheme-1). Overall data demonstrate the role of DPPs as potential anticancer alternatives.


Subject(s)
Apoptosis , DNA Helicases/metabolism , Dietary Carbohydrates/metabolism , Galectin 3/genetics , Galectin 3/metabolism , Poly-ADP-Ribose Binding Proteins/metabolism , Polysaccharides/metabolism , RNA Helicases/metabolism , RNA Recognition Motif Proteins/metabolism , Animals , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Blood Proteins , Cell Line, Tumor , Cell Proliferation/drug effects , Dietary Carbohydrates/pharmacology , Fluorescent Antibody Technique , Galectins , Gene Expression , Immunohistochemistry , Melanoma, Experimental , Mice , Polysaccharides/pharmacology , Skin Neoplasms/etiology , Skin Neoplasms/metabolism
8.
Sci Rep ; 9(1): 9607, 2019 07 03.
Article in English | MEDLINE | ID: mdl-31270370

ABSTRACT

Although optimal therapy for myocardial infarction includes reperfusion to restore blood flow to the ischemic region, ischemia/reperfusion (IR) also initiates an inflammatory response likely contributing to adverse left ventricular (LV) extracellular matrix (ECM) remodeling. Galectin-3 (Gal-3), a ß-galactoside-binding-lectin, promotes cardiac remodeling and dysfunction. Our aim is to investigate whether Gal-3 pharmacological inhibition using modified citrus pectin (MCP) improves cardiac remodeling and functional changes associated with IR. Wistar rats were treated with MCP from 1 day before until 8 days after IR (coronary artery ligation) injury. Invasive hemodynamics revealed that both LV contractility and LV compliance were impaired in IR rats. LV compliance was improved by MCP treatment 8 days after IR. Cardiac magnetic resonance imaging showed decreased LV perfusion in IR rats, which was improved with MCP. There was no difference in LV hypertrophy in MCP-treated compared to untreated IR rats. However, MCP treatment decreased the ischemic area as well as Gal-3 expression. Gal-3 blockade paralleled lower myocardial inflammation and reduced fibrosis. These novel data showing the benefits of MCP in compliance and ECM remodeling in IR reinforces previously published data showing the therapeutic potential of Gal-3 inhibition.


Subject(s)
Galectin 3/antagonists & inhibitors , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/pathology , Myocardium/metabolism , Myocardium/pathology , Pectins/pharmacology , Animals , Biomarkers , Blood Proteins , Disease Models, Animal , Galectin 3/genetics , Galectins , Gene Expression , Heart Failure/diagnosis , Heart Failure/drug therapy , Heart Failure/etiology , Heart Failure/metabolism , Heart Function Tests , Immunohistochemistry , Magnetic Resonance Imaging , Myocardial Infarction/diagnosis , Myocardial Infarction/drug therapy , Myocardial Infarction/etiology , Myocardial Infarction/metabolism , Myocardial Reperfusion Injury/drug therapy , Myocardial Reperfusion Injury/etiology , Rats
9.
Sci Rep ; 9(1): 2186, 2019 02 18.
Article in English | MEDLINE | ID: mdl-30778105

ABSTRACT

Galectin-3 is a carbohydrate binding protein which has important roles in cancer and immunity. Potent galectin-3 inhibitors have been synthesized, for experimental purposes and potential clinical use. As galectin-3 is implicated in both intra- and extracellular activities, permeability of galectin-3 inhibitors is an important parameter determining biological effects. We compared the cellular uptake of galectin-3 inhibitors and their potency in the intracellular or extracellular space. The inhibitors differed in their polar surface area (PSA), but had similar affinities for galectin-3. Using a well-established permeability assay, we confirmed that the uptake was significantly higher for the inhibitor with the lowest PSA, as expected. To analyze intracellular activity of the inhibitors, we developed a novel assay based on galectin-3 accumulation around damaged intracellular vesicles. The results show striking differences between the inhibitors intracellular potency, correlating with their PSAs. To test extracellular activity of the inhibitors, we analyzed their potency to block binding of galectin-3 to cell surfaces. All inhibitors were equally able to block galectin-3 binding to cells and this was proportional to their affinity for galectin-3. These inhibitors may serve as useful tools in exploring biological roles of galectin-3 and may further our understanding of intracellular versus extracellular roles of galectin-3.


Subject(s)
Galectin 3/antagonists & inhibitors , Animals , Binding Sites , Blood Proteins , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , CHO Cells , Caco-2 Cells , Cell Line, Tumor , Cell Membrane/drug effects , Cell Membrane/metabolism , Cell Membrane Permeability , Cell Proliferation/drug effects , Cricetulus , Drug Evaluation, Preclinical , Female , Galactosides/chemistry , Galactosides/pharmacokinetics , Galactosides/pharmacology , Galectin 3/chemistry , Galectin 3/genetics , Galectins , Humans , MCF-7 Cells , Molecular Structure , Thiogalactosides/chemistry , Thiogalactosides/pharmacokinetics , Thiogalactosides/pharmacology
10.
Phytother Res ; 33(2): 294-308, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30402951

ABSTRACT

Macrophage activation plays a central role in neoatherosclerosis and in-stent restenosis after percutaneous coronary intervention (PCI). Galectin-3, mainly expressed on macrophages, is an important regulator of inflammation. This study aimed to investigate the effects of berberine (BBR) on oxidized low-density lipoprotein (ox-LDL)-induced macrophage activation and galectin-3 expression and their underlying mechanisms. THP-1-derived macrophages were pretreated with BBR prior to stimulation with ox-LDL. Galectin-3 expression was measured by real-time PCR, Western blotting, and confocal microscopy. Macrophage activation was assessed by lipid accumulation, expression of inflammatory cytokines, and CD11b and CD86. Plasma galectin-3 levels were measured in patients undergoing PCI at baseline and after BBR treatment for 3 months. BBR suppressed ox-LDL-induced upregulation of galectin-3 and macrophage activation. Overexpression of galectin-3 intervened the inhibitory effect of BBR on macrophage activation. BBR activated phospho-AMPK and inhibited phospho-NF-κB p65 nuclear translocation. AMPK inhibition and NF-κB activation abolished the inhibitory effects of BBR on galectin-3 expression and macrophage activation. Combination of BBR and rosuvastatin exerted greater effects than BBR or rosuvastatin alone. However, BBR treatment did not further reduce plasma galectin-3 after PCI in patients receiving standard therapy. In conclusion, BBR alleviates ox-LDL-induced macrophage activation by downregulating galectin-3 via the NF-κB and AMPK signaling pathways.


Subject(s)
Berberine/pharmacology , Lipoproteins, LDL , Macrophage Activation/drug effects , Macrophages/drug effects , AMP-Activated Protein Kinases/metabolism , Aged , Aged, 80 and over , Cells, Cultured , Down-Regulation/drug effects , Down-Regulation/genetics , Female , Galectin 3/genetics , Galectin 3/metabolism , Gene Expression Regulation/drug effects , Humans , Inflammation/drug therapy , Inflammation/genetics , Inflammation/metabolism , Lipoproteins, LDL/metabolism , Macrophages/physiology , Male , Middle Aged , NF-kappa B/metabolism , Percutaneous Coronary Intervention , Signal Transduction/drug effects , Transcription Factor RelA/metabolism
11.
Biosci Rep ; 38(6)2018 12 21.
Article in English | MEDLINE | ID: mdl-30455396

ABSTRACT

Nephrotoxicity is a major toxic effect in chemotherapy, which constitutes up to 60% of hospitalized acute kidney injury (AKI). Very few treatment options exist to slow the transition from AKI to subsequent chronic kidney diseases (CKD). Here, we demonstrate that galectin-3 (Gal-3), a ß-galactoside binding lectin that plays an important role in kidney fibrosis and renal failure, is one of the key factors for renal injury progression. Ectopic overexpression of Gal-3 significantly decreased the viability of HEK293, simultaneously inducing of cell cycle arrest and apoptosis. However, inhibition of Gal-3, mediated by modified citrus pectin (MCP), predominantly antagonized the pro-apoptotic effects. Mice were pre-treated with normal or 1% MCP-supplemented drinking water 1 week before cisplatin injection. Analyses of serum creatinine and renal tissue damage indicated that MCP-treated mice demonstrated increased renal function and attenuated renal fibrosis after cisplatin-induced injury. MCP-treated mice also demonstrated decreased renal fibrosis and apoptosis, as revealed by masson trichrome staining and Western blot analysis of cleaved caspase-3. Additionally, the protective role of Gal-3 inhibition in the kidney injury was shown to be mediated by protein kinase C α (PKC-α), which promoted cell apoptosis and collagen I synthesis in HEK293 cells. These results demonstrated the potential Gal-3 and PKC-α as therapeutic targets for the treatment of AKI and CKD.


Subject(s)
Acute Kidney Injury/genetics , Cisplatin/adverse effects , Fibrosis/genetics , Galectin 3/genetics , Protein Kinase C-alpha/genetics , Acute Kidney Injury/blood , Acute Kidney Injury/chemically induced , Acute Kidney Injury/pathology , Animals , Apoptosis/genetics , Blood Proteins , Caspase 3/genetics , Cisplatin/administration & dosage , Creatinine/blood , Disease Models, Animal , Fibrosis/blood , Fibrosis/chemically induced , Fibrosis/pathology , Galectin 3/antagonists & inhibitors , Galectins , Gene Expression Regulation , Humans , Kidney/drug effects , Kidney/metabolism , Kidney/pathology , Mice , Neoplasms/complications , Neoplasms/drug therapy , Pectins/genetics , Renal Insufficiency, Chronic/blood , Renal Insufficiency, Chronic/genetics , Renal Insufficiency, Chronic/pathology
12.
J Agric Food Chem ; 66(35): 9248-9258, 2018 Sep 05.
Article in English | MEDLINE | ID: mdl-30113849

ABSTRACT

A library consisting of 429 food-source compounds was used to screen the natural products with anticancer properties in esophageal squamous cell carcinoma (ESCC). We demonstrated for the first time that synephrine, an active compound isolated from leaves of citrus trees, markedly suppressed cell proliferation (inhibition rate with 20 µM synephrine at day 5:71.1 ± 5.8% and 75.7 ± 6.2% for KYSE30 and KYSE270, respectively) and colony formation (inhibition rate with 10 µM synephrine: 86.5 ± 5.9% and 82.3 ± 4.5% for KYSE30 and KYSE270, respectively), as well as migration (inhibition rate with 10 µM synephrine: 76.9 ± 4.4% and 62.2 ± 5.8% for KYSE30 and KYSE270, respectively) and invasion abilities (inhibition rate with 10 µM synephrine: 73.3 ± 7.5% and 75.3 ± 3.4% for KYSE30 and KYSE270, respectively) of ESCC cells in a dose-dependent manner, without significant toxic effect on normal esophageal epithelial cells. Mechanistically, quantitative proteomics and bioinformatics analyses were performed to explore the synephrine-regulated proteins. Western blot and qRT-PCR data indicated that synephrine may downregulate Galectin-3 to inactivate AKT and ERK pathways. In addition, we found that the sensitivity of ESCC to fluorouracil (5-FU) could be enhanced by synephrine. Furthermore, in vivo experiments showed that synephrine had significant antitumor effect on ESCC tumor xenografts in nude mice (inhibition rate with 20 mg/kg synephrine is 61.3 ± 20.5%) without observed side effects on the animals. Taken together, synephrine, a food-source natural product, may be a potential therapeutic strategy in ESCC.


Subject(s)
Citrus/chemistry , Esophageal Neoplasms/drug therapy , Esophageal Neoplasms/physiopathology , MAP Kinase Signaling System , Plant Extracts/administration & dosage , Synephrine/administration & dosage , Animals , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Esophageal Neoplasms/genetics , Esophageal Neoplasms/metabolism , Extracellular Signal-Regulated MAP Kinases/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Galectin 3/genetics , Galectin 3/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Humans , Mice, Inbred BALB C , Mice, Nude , Neoplasm Metastasis , Plant Extracts/chemistry , Plant Leaves/chemistry , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , Synephrine/chemistry
13.
Acta Pharmacol Sin ; 39(12): 1885-1893, 2018 Dec.
Article in English | MEDLINE | ID: mdl-29769742

ABSTRACT

Modified citrus pectin (MCP) is a carbohydrate enriched complex, which has been implicated in cancer treatment and prevention. However, the effects of MCP on urinary bladder cancer (UBC) are unknown. In this study, MCP was first tested in T24 and J82 human UBC cells and showed the inhibition of cell viability by the sulforhodamine B (SRB) assay. The MCP-treated UBC cells exhibited G2/M phase arrest with the decrease of Cyclin B1 and phosphorylated Cdc2. Caspase-3 was also activated, leading to the cleavage of Caspase-3 and PARP. We further explored the possible molecular mechanisms upon MCP treatment in UBC cells. Reduction of galectin-3 was observed and followed with the inactivation of Akt signaling pathway. Of note, galectin-3 knockdown by RNA interference recapitulated the MCP-mediated anti-proliferation, cell cycle arrest and apoptosis. Moreover, oral administration of MCP to the T24 xenograft-bearing nude mice inhibited the tumor growth significantly (P < 0.05). Quantification analysis of immunohistochemistry staining for Ki67 and cleaved Caspase-3 confirmed the decrease of proliferation index (P < 0.05) and the increase of apoptosis index (P < 0.01) in 700 mg/kg MCP-fed UBC xenografts. Using the information from TCGA database, we revealed that the overexpression of galectin-3 was associated with high tumor grade with lymph node metastasis, poor overall survival in UBC patients. Considering the remarkable inhibitory effects of MCP on UBC cell proliferation and survival in vitro and in vivo mainly through galectin-3, which is upregulated in UBCs, MCP may become an attractive agent, as a natural dietary fiber, for prevention and therapy of UBCs.


Subject(s)
Antineoplastic Agents/therapeutic use , Down-Regulation , Galectin 3/genetics , Pectins/therapeutic use , Urinary Bladder Neoplasms/drug therapy , Animals , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Blood Proteins , Caspase 3/metabolism , Cell Line, Tumor , G2 Phase Cell Cycle Checkpoints/drug effects , Galectins , Humans , Male , Mice, Nude , Pectins/pharmacology , Urinary Bladder Neoplasms/genetics
14.
Am J Physiol Heart Circ Physiol ; 314(6): H1169-H1178, 2018 06 01.
Article in English | MEDLINE | ID: mdl-29424570

ABSTRACT

Preclinical studies have demonstrated that anti-galectin-3 (Gal-3) interventions are effective in attenuating cardiac remodeling, fibrosis, and dysfunction. We determined, in a transgenic (TG) mouse model of fibrotic cardiomyopathy, whether Gal-3 expression was elevated and whether Gal-3 played a critical role in disease development. We studied mice with fibrotic cardiomyopathy attributable to cardiac overexpression of human ß2-adrenoceptors (ß2-TG). Cardiac expression levels of Gal-3 and fibrotic or inflammatory genes were determined. The effect of Gal-3 inhibition in ß2-TG mice was studied by treatment with Gal-3 inhibitors ( N-acetyllactosamine and modified citrus pectin) or by deletion of Gal-3 through crossing ß2-TG and Gal-3 knockout mice. Changes in cardiomyopathy phenotypes were assessed by echocardiography and biochemical assays. In ß2-TG mice at 3, 6, and 9 mo of age, upregulation of Gal-3 expression was observed at mRNA (~6- to 15-fold) and protein (~4- to 8-fold) levels. Treatment of ß2-TG mice with N-acetyllactosamine (3 wk) or modified citrus pectin (3 mo) did not reverse cardiac fibrosis, inflammation, and cardiomyopathy. Similarly, Gal-3 gene deletion in ß2-TG mice aged 3 and 9 mo did not rescue the cardiomyopathy phenotype. In conclusion, the ß2-TG model of cardiomyopathy showed a robust upregulation of Gal-3 that correlated with disease severity, but Gal-3 inhibitors or Gal-3 gene deletion had no effect in halting myocardial fibrosis, remodeling, and dysfunction. Gal-3 may not be critical for cardiac fibrogenesis and remodeling in this cardiomyopathy model. NEW & NOTEWORTHY We showed a robust upregulation of cardiac galectin-3 (Gal-3) expression in a mouse model of cardiomyopathy attributable to cardiomyocyte-restricted transgenic activation of ß2-adrenoceptors. However, pharmacological and genetic inhibition of Gal-3 did not confer benefit in this model, implying that Gal-3 may not be a critical disease mediator of cardiac remodeling in this model.


Subject(s)
Cardiomyopathies/metabolism , Galectin 3/metabolism , Myocytes, Cardiac/metabolism , Receptors, Adrenergic, beta-2/metabolism , Ventricular Remodeling , Amino Sugars/pharmacology , Animals , Cardiomyopathies/etiology , Cardiomyopathies/genetics , Cardiomyopathies/physiopathology , Disease Models, Animal , Fibrosis , Galectin 3/antagonists & inhibitors , Galectin 3/deficiency , Galectin 3/genetics , Genetic Predisposition to Disease , Humans , Male , Mice, Inbred C57BL , Mice, Knockout , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , Pectins/pharmacology , Phenotype , Receptors, Adrenergic, beta-2/genetics , Severity of Illness Index , Up-Regulation , Ventricular Remodeling/drug effects
15.
Int J Biol Macromol ; 106: 207-217, 2018 Jan.
Article in English | MEDLINE | ID: mdl-28797814

ABSTRACT

Our previous paper reported the structure of ginseng pectic polysaccharides related to the cell migration inhibitory effects, but the underlying mechanisms are poorly understood. In this manuscript, rhamnogalacturonan I (RGI)-rich pectins prepared from ginseng pectin were investigated for their effect on cell migration. The results indicated that the combination of homogalacturonan (HG) and RGI-rich pectins exerted stronger effects than either HG- or RGI-rich pectin alone. Further studies revealed that the effects of HG- and RGI-rich pectins were dependent on pretreatment, which caused alterations in cell morphologies such as cell size and shape, focal adhesion, and the organization of actin filaments, suggesting that HG and RGI pectins exert synergistic effects on cell migration, likely through different ways. Morphological data and quantitative cell adhesion and spreading assays showed that HG- and RGI-rich pectin treatment decreased cell adhesion and cell spreading on the substratum, suggesting that HG- and RGI-rich pectins may exert their effects on cell migration via decreasing cell adhesion and cell spreading. Additionally, we showed that L-929 cells expressed little galectin-3 (Gal-3) and that lactose, an inhibitor of Gal-3 did not block the activities of HG- and RGI-rich pectins, implicating that cell migration inhibited by pectin did not correlate to Gal-3.


Subject(s)
Cell Movement/drug effects , Fibroblasts/drug effects , Pectins/pharmacology , Actin Cytoskeleton/metabolism , Actin Cytoskeleton/ultrastructure , Animals , Cell Adhesion/drug effects , Cell Line , Cell Shape/drug effects , Cell Size/drug effects , Drug Synergism , Fibroblasts/cytology , Fibroblasts/metabolism , Focal Adhesions/drug effects , Focal Adhesions/metabolism , Focal Adhesions/ultrastructure , Galectin 3/genetics , Galectin 3/metabolism , Gene Expression , Lactose/pharmacology , Mice , Panax/chemistry , Pectins/isolation & purification
16.
Biochem J ; 474(22): 3849-3868, 2017 11 09.
Article in English | MEDLINE | ID: mdl-28986508

ABSTRACT

Although pectin-derived polysaccharides can antagonize galectin function in various pathological disorders, the nature of their binding interactions needs to be better defined for developing them as drugs. Moreover, given their relatively large size and complexity, pectin-derived polysaccharides are also useful as model systems to assess inter-polysaccharide and protein-polysaccharide interactions. Here, we investigated interactions between galectin-3 (Gal-3) and pectin-derived polysaccharides: a rhamnogalacturonan (RG) and two homogalacturonans (HGs). BioLayer Interferometry and fluorescence-linked immunosorbent assays indicate that these polysaccharides bind Gal-3 with macroscopic or apparent KD values of 49 nM, 46 µM, and 138 µM, respectively. 15N-1H heteronuclear single quantum coherence (HSQC) NMR studies reveal that these polysaccharides interact primarily with the F-face of the Gal-3 carbohydrate recognition domain. Even though their binding to Gal-3 does not inhibit Gal-3-mediated T-cell apoptosis and only weakly attenuates hemagglutination, their combination in specific proportions increases activity synergistically along with avidity for Gal-3. This suggests that RG and HG polysaccharides act in concert, a proposal supported by polysaccharide particle size measurements and 13C-1H HSQC data. Our model has HG interacting with RG to promote increased avidity of RG for Gal-3, likely by exposing additional lectin-binding sites on the RG. Overall, the present study contributes to our understanding of how complex HG and RG polysaccharides interact with Gal-3.


Subject(s)
Galectin 3/metabolism , Pectins/pharmacology , Blood Proteins , Galectin 3/chemistry , Galectin 3/genetics , Galectins , Humans , Jurkat Cells , Pectins/chemistry , Pectins/genetics , Protein Binding
17.
Clin Sci (Lond) ; 131(22): 2707-2719, 2017 Nov 15.
Article in English | MEDLINE | ID: mdl-28982723

ABSTRACT

Abdominal aortic aneurysm (AAA) evolution is unpredictable and no specific treatment exists for AAA, except surgery to prevent aortic rupture. Galectin-3 has been previously associated with CVD, but its potential role in AAA has not been addressed. Galectin-3 levels were increased in the plasma of AAA patients (n=225) compared with the control group (n=100). In addition, galectin-3 concentrations were associated with the need for surgical repair, independently of potential confounding factors. Galectin-3 mRNA and protein expression were increased in human AAA samples compared with healthy aortas. Experimental AAA in mice was induced via aortic elastase perfusion. Mice were treated intravenously with the galectin-3 inhibitor modified citrus pectin (MCP, 10 mg/kg, every other day) or saline. Similar to humans, galectin-3 serum and aortic mRNA levels were also increased in elastase-induced AAA mice compared with control mice. Mice treated with MCP showed decreased aortic dilation, as well as elastin degradation, vascular smooth muscle cell (VSMC) loss, and macrophage content at day 14 postelastase perfusion compared with control mice. The underlying mechanism(s) of the protective effect of MCP was associated with a decrease in galectin-3 and cytokine (mainly CCL5) mRNA and protein expression. Interestingly, galectin-3 induced CCL5 expression by a mechanism involving STAT3 activation in VSMC. Accordingly, MCP treatment decreased STAT3 phosphorylation in elastase-induced AAA. In conclusion, increased galectin-3 levels are associated with AAA progression, while galectin-3 inhibition decreased experimental AAA development. Our data suggest the potential role of galectin-3 as a therapeutic target in AAA.


Subject(s)
Aorta, Abdominal/drug effects , Aortic Aneurysm, Abdominal/prevention & control , Galectin 3/antagonists & inhibitors , Galectin 3/blood , Pancreatic Elastase , Pectins/pharmacology , Animals , Aorta, Abdominal/enzymology , Aorta, Abdominal/pathology , Aortic Aneurysm, Abdominal/blood , Aortic Aneurysm, Abdominal/enzymology , Aortic Aneurysm, Abdominal/pathology , Blood Proteins , Case-Control Studies , Cells, Cultured , Chemokine CCL5/genetics , Chemokine CCL5/metabolism , Dilatation, Pathologic , Disease Models, Animal , Disease Progression , Galectin 3/genetics , Galectin 3/metabolism , Galectins , Humans , Mice, Inbred C57BL , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Phosphorylation , RNA, Messenger/blood , RNA, Messenger/genetics , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects , Time Factors , Up-Regulation
18.
J Am Heart Assoc ; 5(11)2016 11 04.
Article in English | MEDLINE | ID: mdl-27815266

ABSTRACT

BACKGROUND: Aortic stenosis (AS) is a chronic inflammatory disease, and calcification plays an important role in the progression of the disease. Galectin-3 (Gal-3) is a proinflammatory molecule involved in vascular osteogenesis in atherosclerosis. Therefore, we hypothesized that Gal-3 could mediate valve calcification in AS. METHODS AND RESULTS: Blood samples and aortic valves (AVs) from 77 patients undergoing AV replacement were analyzed. As controls, noncalcified human AVs were obtained at autopsy (n=11). Gal-3 was spontaneously expressed in valvular interstitial cells (VICs) from AVs and increased in AS as compared to control AVs. Positive correlations were found between circulating and valvular Gal-3 levels. Valvular Gal-3 colocalized with the VICs markers, alpha-smooth muscle actin and vimentin, and with the osteogenic markers, osteopontin, bone morphogenetic protein 2, runt-related transcription factor 2, and SRY (sex-determining region Y)-box 9. Gal-3 also colocalized with the inflammatory markers cd68, cd80 and tumor necrosis factor alpha. In vitro, in VICs isolated from AVs, Gal-3 induced expression of inflammatory, fibrotic, and osteogenic markers through the extracellular signal-regulated kinase 1 and 2 pathway. Gal-3 expression was blocked in VICs undergoing osteoblastic differentiation using its pharmacological inhibitor, modified citrus pectin, or the clustered regularly interspaced short palindromic repeats/Cas9 knockout system. Gal-3 blockade and knockdown decreased the expression of inflammatory, fibrotic, and osteogenic markers in differentiated VICs. CONCLUSIONS: Gal-3, which is overexpressed in AVs from AS patients, appears to play a central role in calcification in AS. Gal-3 could be a new therapeutic approach to delay the progression of AV calcification in AS.


Subject(s)
Aortic Valve Stenosis/metabolism , Aortic Valve/metabolism , Aortic Valve/pathology , Calcinosis/metabolism , Galectin 3/metabolism , Aged , Aged, 80 and over , Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , Aortic Valve/surgery , Aortic Valve Stenosis/surgery , B7-1 Antigen/metabolism , Blood Proteins , Blotting, Western , Bone Morphogenetic Protein 2/metabolism , CRISPR-Cas Systems , Calcinosis/surgery , Case-Control Studies , Cell Differentiation , Core Binding Factor Alpha 1 Subunit/metabolism , Female , Galectin 3/genetics , Galectin 3/pharmacology , Galectins , Gene Knockdown Techniques , Heart Valve Prosthesis Implantation , Humans , In Vitro Techniques , Male , Osteoblasts , Osteopontin/metabolism , Pectins/pharmacology , Prospective Studies , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , SOX9 Transcription Factor/metabolism , Tumor Necrosis Factor-alpha/metabolism
19.
Cancer Biol Ther ; 14(10): 897-906, 2013 Oct 01.
Article in English | MEDLINE | ID: mdl-23917726

ABSTRACT

The anti-tumor effects of arsenic trioxide (ATO) were well established in acute promyelocytic leukemia, but not in renal cell carcinoma (RCC). Recent evidences indicate that galectin-3 (Gal-3) plays an anti-apoptotic role in chemotherapy induced tumor cell death. This study was intended to clarify the exact roles of Gal-3 performed in ATO-induced apoptosis in RCC cells. Weak apoptosis was observed in Gal-3-positive RCC cells (Caki-1, Caki-2, 786-0, and ACHN) following ATO treatment. However, ATO treatment upregulated Gal-3 expression concurrently caused a Synexin-cooperated translocation of Gal-3 from the nucleus to the cytoplasm. Gal-3-knockdown cells were more sensitive to ATO treatment as indicated by a strong mitochondria-dependent apoptosis following ATO treatment. Meanwhile, Gal-3 was found to inhibit ATO-induced apoptosis through enhancing Bcl-2 expression and stabilizing mitochondria. To confirm the results obtained from genetic method, we employed a Gal-3 inhibitor, modified citrus prectin (MCP), and co-treated the RCC cells with ATO. The cells showed an increased apoptosis in the syngeneic application of Gal-3 inhibition and ATO compared with ATO application alone. Based on these results, we conclude that Gal-3 inhibition sensitizes human renal cell carcinoma cells to ATO treatment through increasing mitochondria-dependent apoptosis. Our studies implicate synergetic application of ATO and Gal-3 inhibition as a potential strategy for RCC treatment.


Subject(s)
Antineoplastic Agents/pharmacology , Arsenicals/pharmacology , Galectin 3/genetics , Oxides/pharmacology , Pectins/pharmacology , Annexin A7/metabolism , Apoptosis/drug effects , Arsenic Trioxide , Carcinoma, Renal Cell , Cell Line, Tumor , Drug Resistance, Neoplasm , Drug Synergism , Galectin 3/antagonists & inhibitors , Galectin 3/metabolism , Gene Knockdown Techniques , Humans , Kidney Neoplasms , Membrane Potential, Mitochondrial/drug effects , Protein Transport , RNA, Small Interfering/genetics
20.
Glycobiology ; 23(6): 654-63, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23426722

ABSTRACT

Atherosclerosis is a major risk factor for cardiovascular disease (CVD) and stroke. Galectin-3 is a carbohydrate-binding lectin implicated in the pathophysiology of CVD and is highly expressed within atherosclerotic lesions in mice and humans. The object of this present study was to use genetic deletion and pharmacological inhibition in a well-characterized mouse model of atherosclerosis to determine the role of galectin-3 in plaque development. Apolipoprotein-E/galectin-3 knockout mice were generated and fed a high-cholesterol "western" diet. Galectin-3 deletion had no consistent effect on the serum lipid profile but halved atherosclerotic lesion formation in the thoracic aorta (57% reduction), the aortic arch (50% reduction) and the brachiocephalic arteries. The aortic plaques were smaller, with reduced lipid core and less collagen. In apolipoprotein E-deficient (ApoE(-/-)) mice, there was a switch from high inducible nitric oxide expression in early lesions (6 weeks) to arginase-1 expression in later lesions (20 weeks), which was reversed in ApoE(-/-)/gal-3(-/-) mice. Administration of modified citrus pectin, an inhibitor of galectin-3, during the latter stage of the disease reduced plaque volume. We conclude that inhibiting galectin-3 causes decreased atherosclerosis. Strategies to inhibit galectin-3 function may reduce plaque progression and potentially represent a novel therapeutic strategy in the treatment of atherosclerotic disease.


Subject(s)
Apolipoproteins E/deficiency , Atherosclerosis/drug therapy , Galectin 3/antagonists & inhibitors , Pectins/pharmacology , Plaque, Atherosclerotic/prevention & control , Animals , Aorta, Thoracic/pathology , Apolipoproteins E/genetics , Arginase/metabolism , Arginine/metabolism , Atherosclerosis/blood , Cell Line , Cell Movement , Fatty Acids, Nonesterified/blood , Galectin 3/genetics , Galectin 3/metabolism , Humans , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Plaque, Atherosclerotic/blood , Plaque, Atherosclerotic/pathology , Triglycerides/blood , Weight Gain
SELECTION OF CITATIONS
SEARCH DETAIL