Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
Add more filters










Publication year range
1.
Article in English | MEDLINE | ID: mdl-38310451

ABSTRACT

Esophageal cancer is a complex disease influenced by genetic and environmental factors. Single nucleotide polymorphisms [SNPs] in non-coding regions of the genome have emerged as crucial contributors to esophageal cancer susceptibility. This review provides a comprehensive overview of the role of SNPs in non-coding regions and their association with esophageal cancer. The accumulation of SNPs in the genome has been implicated in esophageal cancer risk. Various studies have identified specific locations in the genome where SNPs are more likely to occur, suggesting a location-specific response. Chromatin conformational studies have shed light on the localization of SNPs and their impact on gene transcription, posttranscriptional modifications, gene expression regulation, and histone modification. Furthermore, miRNA-related SNPs have been found to play a significant role in esophageal squamous cell carcinoma [ESCC]. These SNPs can affect miRNA binding sites, thereby altering target gene regulation and contributing to ESCC development. Additionally, the risk of ESCC has been linked to base excision repair, suggesting that SNPs in this pathway may influence disease susceptibility. Somatic DNA segment alterations and modified expression quantitative trait loci [eQTL] have also been associated with ESCC. These alterations can lead to disrupted gene expression and cellular processes, ultimately contributing to cancer development and progression. Moreover, SNPs have been found to be associated with the long non-coding RNA HOTAIR, which plays a crucial role in ESCC pathogenesis. This review concludes with a discussion of the current and future perspectives in the field of SNPs in non-coding regions and their relevance to esophageal cancer. Understanding the functional implications of these SNPs may lead to the identification of novel therapeutic targets and the development of personalized approaches for esophageal cancer prevention and treatment.

2.
Aging (Albany NY) ; 15(23): 14019-14038, 2023 Dec 11.
Article in English | MEDLINE | ID: mdl-38085649

ABSTRACT

Aging-associated cardiovascular diseases depend on the longitudinal deterioration of stem cell dynamics. The entire mechanism behind it is not completely understood. However, many studies suggest that endocrine pathways, particularly the insulin-like growth factor-1(IGF1) signaling pathway are involved in cardioprotection, especially in stem-cell treatments. Here, we investigated the role of a co-chaperone, carboxyl-terminus of Hsp70 interacting protein (CHIP) in the aspects of growth factor secretion and receptor stabilization in mesenchymal stem cells (MSCs). Briefly, we overexpressed CHIP in rat adipose-derived stem cells (rADSCs) and explored the consequences in vitro, and in vivo, in spontaneously hypertensive rats (SHR). Our data revealed that CHIP overexpression in rADSCs promoted the secretion of insulin-like growth factor-1 (IGF1) and IGF binding protein-3 (IGFBP3) as per immunoblot/cytokine array analysis. We also found that these results were dependent on the nuclear translocation of signal transducer and activator of transcription 3 (STAT3) in rADSCs. Further, the CHIP co-chaperone was also involved in the stabilization of the receptor of IGF1 (IGF1R); interactions between the beta transmembrane region of IGF1R, and the tetracopeptide repeat (TPR) domain of CHIP were evident. Importantly, after the transplantation of lentiviral CHIP overexpression of rADSCs (rADSCsCHIP-WT) into nine months aging-SHR led to an increase in their cardiac function - increased ejection fraction and fractional shortening (≈15% vs. control SHR) - as well as a decrease in their heart size and heart rate, respectively. Altogether, our results support the use of CHIP overexpressing stem cells for the mitigation of cardiac hypertrophy and remodeling associated with late-stage hypertension.


Subject(s)
Hypertension , Receptor, IGF Type 1 , Animals , Rats , Adipocytes/metabolism , Aging , Insulin-Like Growth Factor I/metabolism , Receptor, IGF Type 1/metabolism , Signal Transduction , Stem Cells/metabolism
3.
Neurotox Res ; 41(6): 648-659, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37707697

ABSTRACT

Patients with Alzheimer's disease have increased risk of developing heart disease, which therefore highlights the need for strategies aiming at reducing Alzheimer's disease-related cardiovascular disease. Folic acid and folinic acid are beneficial to the heart. We aimed to investigate the benefits of folic acid and folinic acid in heart of patients with late-stage Alzheimer's disease. Twelve 16-month-old mice of triple-transgenic late-stage Alzheimer's disease were divided into three groups: Alzheimer's disease group, Alzheimer's disease + folic acid group, and Alzheimer's disease + folinic acid group. The mice were administered 12 mg/kg folic acid or folinic acid once daily via oral gavage for 3 months. In the folic acid and folinic acid treatment groups, the intercellular space was reduced, compared with the Alzheimer's disease group. TUNEL assay and western blot images showed that the number of apoptotic cells and the apoptosis-related protein expression were higher in the Alzheimer's disease group than in other two treated groups. Folic acid and folinic acid induced the IGF1R/PI3K/AKT and SIRT1/ AMPK pathways in the hearts of mice with Alzheimer's disease. Our results showed that folic acid and folinic acid treatment increased survival and SIRT1 expression to reduce apoptotic proteins in the heart. The aging mice treated with folinic acid had more IGF1R and SIRT1/AMPK axes to limit myocardial cell apoptosis. In conclusion, folic acid and folinic acid promote cardiac cell survival and prevent apoptosis to inhibit heart damage in aging mice with triple-transgenic late-stage Alzheimer's disease. In particular, folinic acid provides a better curative effect than folic acid.


Subject(s)
Alzheimer Disease , Folic Acid , Humans , Mice , Animals , Folic Acid/pharmacology , Folic Acid/therapeutic use , Leucovorin/pharmacology , Leucovorin/therapeutic use , AMP-Activated Protein Kinases , Phosphatidylinositol 3-Kinases , Proto-Oncogene Proteins c-akt , Mice, Transgenic , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Sirtuin 1 , Aging , Receptor, IGF Type 1
4.
Curr Pharm Biotechnol ; 24(15): 1847-1858, 2023.
Article in English | MEDLINE | ID: mdl-37069718

ABSTRACT

Lipids have received less attention than nucleic acids and proteins, which play a major role in building up the cell. They are a complex group of biomolecules varying in structure and function whose complexity can only be revealed by refining the present analytical tools. Lipogenesis is critical for tumor growth as it has been observed that FA (Fatty Acid) synthesis increases in many cancers. In this review, we have detailed the causes and concerns for considering lipids as a trademark for cancer, including other events such as mutations, epigenetic changes, chromosomal rearrangements, and hormonal stimulations. The process of biomarker development can be heightened from the critical changes observed in lipid profiling that occur in the reprogramming of lipid metabolism. The cancer alterations that occur during lipid metabolism and the expression of various genes during this process have been discussed in detail. The routes through which cancer cells source lipids for their nourishment and energy need and how FA synthesis contributes to this are discussed. The various pathways involved in the metabolism of lipid, which has the potential to be therapeutic targets, are highlighted. Also, the various driving factors critical for lipid metabolism alterations and the major role played by lipids in cancer and ways of targeting it are critically analyzed.


Subject(s)
Lipid Metabolism , Neoplasms , Humans , Fatty Acids/metabolism , Neoplasms/therapy , Neoplasms/drug therapy , Lipogenesis , Biomarkers/metabolism
5.
J Ethnopharmacol ; 284: 114728, 2022 Feb 10.
Article in English | MEDLINE | ID: mdl-34634367

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Solanum nigrum, commonly known as Makoi or black shade has been traditionally used in Asian countries and other regions of world to treat liver disorders, diarrhoea, inflammatory conditions, chronic skin ailments (psoriasis and ringworm), fever, hydrophobia, painful periods, eye diseases, etc. It has been observed that S. nigrum contains substances, like steroidal saponins, total alkaloid, steroid alkaloid, and glycoprotein, which show anti-tumor activity. However; there is no scientific evidence of the efficacy of S. nigrum in the treatment of cardiac hypertrophy. AIM: To investigate the ability of S. nigrum to attenuate Angiotensin II - induced cardiac hypertrophy and improve cardiac function through the suppression of protein kinase PKC-ζ and Mel-18-IGF-IIR signaling leading to the restoration of HSF2 desumolyation. MATERIALS AND METHODS: Cardiomyoblast cells (H9c2) were challenged with 100 nM Angiotensin-II (AngII) for 24 h and were then treated with different concentration of S.nigrum or Calphostin C for 24 h. The hypertrophic effect in cardiomyoblast cells were determined by immunofluorescence staining and the modulations in hypertrophic protein marker along with Protein Kinase C-ζ, MEL18, HSF2, and Insulin like growth factor II (IGFIIR), markers were analyzed by western blotting. In vivo experiments were performed using 12 week old male Wistar Kyoto rats (WKY) and Spontaneously hypertensive rats (SHR) separated into five groups. [1]Control WKY, [2] WKY -100 mg/kg of S.nigrum treatment, [3] SHR, [4] SHR-100 mg/kg of S.nigrum treatment, [5] SHR-300 mg/kg of S.nigrum treatment. S. nigrum was administered intraperitoneally for 8 week time interval. RESULTS: Western blotting results indicate that S. nigrum significantly attenuates AngII induced cardiac hypertrophy. Furthermore, actin staining confirmed the ability of S. nigrum to ameliorate AngII induced cardiac hypertrophy. Moreover, S. nigrum administration suppressed the hypertrophic signaling mediators like Protein Kinase C-ζ, Mel-18, and IGFIIR in a dose-dependent manner and HSF2 activation (restore deSUMOlyation) that leads to downregulation of IGF-IIR expression. Additionally in vivo experiments demonstrate the reduced heart sizes of S. nigrum treated SHRs rats when compared to control WKY rats. CONCLUSION: Collectively, the data reveals the cardioprotective effect of S. nigrum inhibiting PKC-ζ with alleviated IGF IIR level in the heart that profoundly remits cardiac hypertrophy for hypertension-induced heart failure.


Subject(s)
Cardiomegaly/drug therapy , Cardiotonic Agents/pharmacology , Plant Extracts/pharmacology , Solanum nigrum/chemistry , Angiotensin II , Animals , Cardiotonic Agents/administration & dosage , Cardiotonic Agents/isolation & purification , Cell Line , Disease Models, Animal , Dose-Response Relationship, Drug , Heat-Shock Proteins/metabolism , Hypertension/drug therapy , Male , Myoblasts, Cardiac/drug effects , Myoblasts, Cardiac/pathology , Plant Extracts/administration & dosage , Protein Kinase C/metabolism , Rats , Rats, Inbred SHR , Rats, Inbred WKY , Receptor, IGF Type 2/metabolism , Transcription Factors/metabolism
6.
Int J Biol Macromol ; 197: 77-87, 2022 Feb 01.
Article in English | MEDLINE | ID: mdl-34920072

ABSTRACT

Self assembling peptidebased hydrogel has been explored for delivering growth factors, anticancer drugs, antibiotics etc. Here, RADA 16-I (RADARADARADARADA), an ionic self complementary peptide that forms a well defined nanohydrogel has been studied for its ability to deliver PDGF-BB in a sustained manner and to destruct biofilm formed by wound specific pathogens. Results of the structural analysis of the nanohydrogel studied through AFM, FeSEM, CD, FT-IR and Rheometry, revealed the hydrogel forming ability of RADA 16-I with stable ß-sheet structure at room temperature. The nanohydrogel was also found to destruct the biofilm formed under in vitro condition using S. aureus, E. coli and P. aeruginosa. The growth factor incorporated in the nanohydrogel followed first order release kinetics and showed sustained release up to 48 h. Angiogenic potential and wound healing ability of PDGF-BB incorporated nanohydrogel was confirmed in both in vitro and in vivo conditions. The animals treated with PDGF-BB incorporated nanohydrogel exhibited 99.5% wound closure at day 21. The content of hydroxyproline and ascorbic acid was significantly high in the treated animals when compared to control and untreated animals. Overall, the study provides the essential information and data for using RADA 16-I for treating chronic wounds.


Subject(s)
Hydrogels
7.
Environ Toxicol ; 36(8): 1567-1575, 2021 Aug.
Article in English | MEDLINE | ID: mdl-33929070

ABSTRACT

Habitual chewing of areca nut increases the risk of cardiovascular disease mortality, but less report demonstrate the toxic mechanism of areca nut on heart. To investigate toxicity of areca nut on cardiomyocytes, we induced the heart injury with arecoline to evaluate the acute damage of areca nut on heart. Different concentrations of are coline (lowdosage: 5 mg/kg/day and high dosage 50 mg/kg/day) were injected into Sprague-Dawley rat via intra-peritoneal method for 21 days to create negative effects of arecoline on cardiomyocyte. Themyocardial architecture of the rat heart was observed. The arecoline-induced apoptotic proteins were analysed via western blotting. The myocardialarchitecture of heart was injured with arecoline and TUNEL stain was also shown are coline-induced cardiac apoptosis. Arecoline promoted the protein expression of both Fas dependent snd mitochondrial dependent apoptosis. In summary, arecoline induces cardiac toxicity and apoptosis by inducing both death receptor and mitochondria-dependent apoptotic pathways on heart.


Subject(s)
Areca , Arecoline , Animals , Fas Ligand Protein , Plant Extracts , Rats , Rats, Sprague-Dawley
8.
Environ Toxicol ; 36(7): 1466-1475, 2021 Jul.
Article in English | MEDLINE | ID: mdl-33881220

ABSTRACT

In aging hypertensive conditions, deterioration of insulin-like growth factor 1 receptor (IGF1R) cause a pathological impact on hypertensive hearts with an increased Ang II level. Recovering these adverse conditions through transplanted adipose-derived stem cells is a challenging approach. Moreover, Danggui, a Traditional Chinese medicine (TCM), is used for the treatment of cardioprotective effects. In this study, to evaluate whether the combined effect of MSCs and TCM can recover the cardiac function in late-stage hypertension rats. We observed that lower dose of Danggui crude extract treatment showed an increased level of cell viability with maintained stemness properties and growth rate in rat adipose-derived stem cells (rADSCs). Further, we cocultured the H9c2 cells with rADSCs and the results revealed that Danggui-treated MSCs enhanced the IGF1R expression and attenuated the hypertrophy in H9c2 cells against Ang II challenge by immunoblot and rhodamine-phalloidin staining. In addition, Danggui crude extract was also quantified and characterized by HPLC and LC-MS analysis. Furthermore, the in vivo study was performed by considering 11 months old rats (n = 7). Importantly, the oral administration of Danggui crude extract with stem cells intravenous injection in SHR-D-ADSCs group showed a combination effect to augment the cardiac function through enhancement of ejection fraction, fractional shortening, contractility function in the late-stage hypertension conditions. We have also observed a decreased apoptosis rate in the heart tissue of SHR-D-ADSCs group. Taken together, these results indicate that the combinatorial effects of Danggui crude extract and stem cell therapy enhanced cardiac function in late-stage SHR rats.


Subject(s)
Hypertension , Insulin-Like Growth Factor I , Animals , Rats , Rats, Inbred SHR , Stem Cells , Up-Regulation
9.
Mol Cell Biochem ; 476(9): 3253-3260, 2021 Sep.
Article in English | MEDLINE | ID: mdl-33886061

ABSTRACT

Pathological cardiac hypertrophy is associated with many diseases including hypertension. Recent studies have identified important roles for microRNAs (miRNAs) in many cardiac pathophysiological processes, including the regulation of cardiomyocyte hypertrophy. However, the role of miR-145-5p in the cardiac setting is still unclear. In this study, H9C2 cells were overexpressed with microRNA-145-5p, and then treated with Ang-II for 24 h, to study the effect of miR-145-5p on Ang-II-induced myocardial hypertrophy in vitro. Results showed that Ang-II treatment down-regulated miR-145-5p expression were revered after miR-145-5p overexpression. Based on results of bioinformatics algorithms, paxillin was predicted as a candidate target gene of miR-145-5p, luciferase activity assay revealed that the luciferase activity of cells was substantial downregulated the following co-transfection with wild paxillin 3'UTR and miR-145-5p compared to that in scramble control, while the inhibitory effect of miR-145-5p was abolished after transfection of mutant paxillin 3'UTR. Additionally, overexpression of miR-145-5p markedly inhibited activation of Rac-1/ JNK /c-jun/ NFATc3 and ANP expression and induced SIRT1 expression in Ang-II treated H9c2 cells. Jointly, our study suggested that miR-145-5p inhibited cardiac hypertrophy by targeting paxillin and through modulating Rac-1/ JNK /c-jun/ NFATc3/ ANP / Sirt1 signaling, therefore proving novel downstream molecular pathway of miR-145-5p in cardiac hypertrophy.


Subject(s)
Angiotensin II/toxicity , Cardiomegaly/drug therapy , Gene Expression Regulation/drug effects , MicroRNAs/genetics , Myoblasts, Cardiac/drug effects , Paxillin/antagonists & inhibitors , Animals , Atrial Natriuretic Factor/genetics , Atrial Natriuretic Factor/metabolism , Cardiomegaly/chemically induced , Cardiomegaly/metabolism , Cardiomegaly/pathology , Cells, Cultured , MAP Kinase Kinase 4/genetics , MAP Kinase Kinase 4/metabolism , Myoblasts, Cardiac/metabolism , Myoblasts, Cardiac/pathology , NFATC Transcription Factors/genetics , NFATC Transcription Factors/metabolism , Proto-Oncogene Proteins c-jun/genetics , Proto-Oncogene Proteins c-jun/metabolism , Rats , Sirtuin 1/genetics , Sirtuin 1/metabolism , Vasoconstrictor Agents/toxicity , rac1 GTP-Binding Protein/genetics , rac1 GTP-Binding Protein/metabolism
10.
Aging (Albany NY) ; 13(8): 12046-12057, 2021 04 26.
Article in English | MEDLINE | ID: mdl-33901009

ABSTRACT

Hepatocellular carcinoma is a common type of liver cancer. Resistance to chemotherapeutic agents is a major problem in cancer therapy. MicroRNAs have been reported in cancer development and tumor growth; however, the relationship between chemoresistance and hepatocellular carcinoma needs to be fully investigated. Here, we treated hepatocellular carcinoma cell line (HA22T) with a histone deacetylase inhibitor to establish hepatocellular carcinoma-resistant cells (HDACi-R) and investigated the molecular mechanisms of chemoresistance in HCC cells. Although histone deacetylase inhibitor could not enhance cell death in HDACi-R but upregulation of miR-107 decreased cell viability both in parental cells and resistance cells, decreased the expression of cofilin-1, enhanced ROS-induced cell apoptosis, and dose-dependently sensitized HDACi-R to HDACi. Further, miR-107 upregulation resulted in tumor cell disorganization in both HA22T and HDACi-R in a mice xenograft model. Our findings demonstrated that miR-107 downregulation leads to hepatocellular carcinoma cell resistance in HDACi via a cofilin-1-dependent molecular mechanism and ROS accumulation.


Subject(s)
Carcinoma, Hepatocellular/drug therapy , Drug Resistance, Neoplasm/genetics , Histone Deacetylase Inhibitors/pharmacology , Liver Neoplasms/drug therapy , MicroRNAs/metabolism , Animals , Apoptosis/drug effects , Apoptosis/genetics , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/genetics , Cofilin 1/genetics , Drug Resistance, Neoplasm/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Gene Knockdown Techniques , Histone Deacetylase Inhibitors/therapeutic use , Humans , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Male , Mice , MicroRNAs/agonists , MicroRNAs/genetics , Reactive Oxygen Species/metabolism , Xenograft Model Antitumor Assays
11.
Probiotics Antimicrob Proteins ; 13(4): 1044-1053, 2021 08.
Article in English | MEDLINE | ID: mdl-33527184

ABSTRACT

Diabetes-induced cardiomyocyte apoptosis is one of the major causes of mortality in patients with diabetes. Numerous studies have indicated the beneficial effects of Lactobacillus reuteri GMNL-263. However, the protective effect of Lactobacillus reuteri GMNL-263 in cardiac damage associated with diabetes remains poorly understood. In this study, we aimed to investigate the protective effect of Lactobacillus reuteri GMNL-263 on cardiomyocytes in diabetic rats. Five-week-old male Wistar rats were categorized into normal control group, diabetes group (55 mg/kgw STZ-induced diabetes via intraperitoneal injection), and diabetic animals treated with Lactobacillus reuteri GMNL-263 (109 CFU/rat/day, oral administration for 4 weeks). The results were presented that oral administration of a high dose of Lactobacillus reuteri GMNL-263 in diabetic rats activated IGF1R cell survival pathways to decrease the Fas-dependent and mitochondrial-dependent apoptotic pathways induced by hyperglycemia. We found that GMNL-263 significantly attenuated cell apoptosis via the IGF1R survival pathway in diabetic rats. The findings of this study suggest that GMNL-263 treatment maybe an effective therapeutic approach for the prevention of cardiac apoptosis in patients with diabetes.


Subject(s)
Cardiotonic Agents , Diabetes Mellitus, Experimental , Hyperglycemia , Limosilactobacillus reuteri , Myocytes, Cardiac , Animals , Hot Temperature , Hyperglycemia/complications , Male , Rats , Rats, Wistar , Receptor, IGF Type 1
12.
Eur J Nutr ; 60(6): 3211-3223, 2021 Sep.
Article in English | MEDLINE | ID: mdl-33555373

ABSTRACT

PURPOSE: Diabetes mellitus (DM) leads to disorders such as cardiac hypertrophy, cardiac myocyte apoptosis, and cardiac fibrosis. Previous studies have shown that Lactobacillus reuteri GMNL-263 decreases cardiomyopathy by reducing inflammation. In this study, we investigated the potential benefit of GMNL-263 supplementation in treating diabetes-induced cardiomyocytes in rats with DM. METHODS: Five-week-old male Wistar rats were randomly divided into three groups, control, DM, and rats with DM treated with different dosages of L. reuteri GMNL-263. After undergoing treatment for 4 weeks, all rats were euthanized for further analysis. RESULTS: We observed that cardiac function and structure of rats with DM was rescued by GMNL-263. Activation of toll-like receptor 4 (TLR4)-related inflammatory, hypertrophic, and fibrotic signaling pathways in the hearts of rats with DM was reduced by treatment with GMNL-263. CONCLUSION: Our findings demonstrate that GMNL-263 inhibited diabetes-induced cardiomyocytes via the repression of the TLR4 pathway. Moreover, these findings suggest that treatment with high-dose GMNL-263 could be a precautionary therapy for reducing the diabetes-induced cardiomyopathy.


Subject(s)
Cardiomyopathies , Diabetes Mellitus, Experimental , Limosilactobacillus reuteri , Probiotics , Animals , Cardiomyopathies/therapy , Diabetes Mellitus, Experimental/therapy , Hot Temperature , Male , Myocytes, Cardiac , Rats , Rats, Wistar , Toll-Like Receptor 4/genetics
13.
Environ Toxicol ; 36(6): 1021-1030, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33475235

ABSTRACT

The aim of this study was to investigate the effects of longan flower (LF) water extract on cardiac apoptotic and survival pathways in rat models of fructose-induced metabolic syndrome. The study findings revealed that the levels of glucose, insulin, triglyceride, and cholesterol and TUNEL-positive apoptotic cells were significantly increased in the HF group compared with the control group; whereas, the levels were decreased in the HFLF group. The expressions of Fas, FADD, and activated caspases 8 and 3, as well as the expressions of Bax, Bak, Bax/Bcl-2, Bak/Bcl-xL, cytosolic cytochrome c, and activated caspases 9 and 3 were increased in the HF group were significantly reversed in HFLF administrated group. Furthermore, LF extract increased IGF-1R, p-PI3K, p-Akt, Bcl-2, and Bcl-xL expression compared to HF group. Taken together, the present findings help identify LF as a potential cardioprotective agent that can be effectively used in treating fructose-induced metabolic syndrome.


Subject(s)
Metabolic Syndrome , Animals , Apoptosis , Flowers , Fructose/toxicity , Metabolic Syndrome/chemically induced , Myocardium , Proto-Oncogene Proteins c-bcl-2 , Rats , Sapindaceae , bcl-2-Associated X Protein , fas Receptor
14.
Phytomedicine ; 82: 153442, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33412494

ABSTRACT

BACKGROUND: Chemoresistance remains the main obstacle in hepatocellular carcinoma (HCC) therapy. Despite significant advances in HCC therapy, HCC still has a poor prognosis. Thus, there is an urgent need to identify a treatment target to reverse HCC chemotherapy resistance. Platycodon grandiflorus (PG) is a perennial herb that has been used as food and traditional Chinese medicine for thousands of years in Northeast Asia. Platycodin D (PD), a main active triterpenoid saponin found in the root of PG, has been reported to possess anticancer properties in several cancer cell lines, including HCC; however, the reversal effect of this molecule on HCC chemoresistance remains largely unknown. PURPOSE: This study aimed to investigate the role and the mechanism of PD-mediated reversal of the histone deacetylase inhibitor (HDACi) resistance in HCC cells. METHODS: Human HCC cells (HA22T) and HDACi-resistant (HDACi-R) cells were used. Cell viability was measured using the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay. Combination index was used to calculate the synergism potential. Expression of ERK1/2 (total/phospho), cofilin-1 (total/phospho) and apoptosis-related protein was determined using western blotting. Mitochondrial membrane potential was assessed using the JC-1 (5,5',6,6'-tetrachloro-1,1',3,3'-tetraethylbenzimidazolocarbocyanine iodide) probe. Apoptosis was detected using the terminal deoxynucleotidyl transferase dUTP nick end labeling assay. Mitochondrial reactive oxygen species generation was measured using the MitoSOX Red fluorescent probe. RESULTS: We found that PD treatment inhibited cell viability both in HA22T HCC and HDACi-R cells. Inhibition of ERK1/2 by PD98059 could reverse drug resistance in HDACi-R cells treated with PD98059 and PD. Nevertheless, pre-treatment with U46619, an ERK1/2 activator, rescued PD-induced apoptosis by decreasing levels of apoptosis-related proteins in HCC cells. The combined treatment of PD with apicidin a powerful HDACi, dramatically enhanced the apoptotic effect in HDACi-R cells. CONCLUSION: For the first time, we showed that PD reversed HDACi resistance in HCC by repressing ERK1/2-mediated cofilin-1 phosphorylation. Thus, PD can potentially be a treatment target to reverse HCC chemotherapy resistance in future therapeutic trials.


Subject(s)
Carcinoma, Hepatocellular/drug therapy , Cofilin 1/metabolism , Drug Resistance, Neoplasm/drug effects , Histone Deacetylase Inhibitors/therapeutic use , Liver Neoplasms/drug therapy , MAP Kinase Signaling System/drug effects , Saponins/pharmacology , Triterpenes/pharmacology , Apoptosis/drug effects , Cell Line, Tumor , Cofilin 2/metabolism , Humans , Membrane Potential, Mitochondrial/drug effects , Mitochondria/drug effects , Phosphorylation
15.
Arch Physiol Biochem ; 127(1): 82-89, 2021 Feb.
Article in English | MEDLINE | ID: mdl-31112046

ABSTRACT

BACKGROUND: Cardio-dysfunction is one of the complications in patients with diabetes mellitus (DM). This paper aimed to investigate if oral administration of green tea Epigallocatechin-3-gallate (EGCG, E) and transplantation of adipose-derived stem cells (ADSC) show cross effects on the treatment of cardiomyopathy in rats with type 1 DM. MATERIALS AND METHODS: Wistar male rats were divided into four groups (each group contained 8 animals) including sham, DM (diabetic group), DM + ADSC (DM group with ADSC treatment) and DM + ADSC + E (DM + ADSC group with oral administration of EGCG). RESULTS: Pathological parameters including hypertrophy, inflammation, and fibrosis were activated in DM group. By contrast, all parameters were significantly improved in treatment group (DM + ADSC group). In addition, improvement of pathological parameters in DM + ADSC + E was significantly better than DM + ADSC. CONCLUSION: We found that EGCG can increase expression of survival marker in ADSC under high glucose environment and reduce serum oxidative stress in DM rats.


Subject(s)
Adipocytes/drug effects , Cardiomyopathies/drug therapy , Catechin/analogs & derivatives , Oxidative Stress , Stem Cells/drug effects , Tea , Adipocytes/cytology , Adipose Tissue/drug effects , Administration, Oral , Animals , Blood Glucose/metabolism , Catechin/pharmacology , Diabetes Mellitus, Experimental , Echocardiography , Inflammation , Male , Rats , Rats, Wistar , Stem Cell Transplantation , Stem Cells/cytology , Transplantation, Autologous
16.
Biochem Biophys Res Commun ; 532(3): 347-354, 2020 11 12.
Article in English | MEDLINE | ID: mdl-32888650

ABSTRACT

Hypoxic preconditioning is a well-known strategy to improve the survival and therapeutic potential of stem cells against various challenges including hemodynamic and neurohormonal modulations. However, the mechanism involved in hypoxia-induced benefits on stem cells is still ambiguous. In pathological hypertension, the elevation of the neurohormonal mediator Angiotensin II (Ang II) causes the adverse effects to stem cells. In this study, we investigate the effect and mechanism of action of short term hypoxia-inducible miRNA in suppressing the effects of AngII on stem cells. According to the results obtained, Ang II affects the normal cell cycle and triggers apoptosis in rADSCs with a corresponding increase in the expression of cell death-inducing p53 target 1 (CDIP1) protein. However, the short term hypoxia-inducible miRNA-miR-210-3p was found to target CDIP1 and reduce their levels upon the Ang II challenge. CDIP1 induces stress-mediated apoptosis involving the extrinsic apoptosis pathway via Bid/Bax/cleaved caspase3 activation. Administration of mimic miR-210-3p targets CDIP1 mRNA by binding to the 3' UTR region as confirmed by dual luciferase assay and also reduced Ang II-induced mitochondrial ROS accumulation as analyzed by MitoSOX staining. Moreover, the present study demonstrates the mechanism of miR-210-3p in the regulation of Ang II-induced CDIP1-associated apoptotic pathway in rADSCs.


Subject(s)
Angiotensin II/pharmacology , Apoptosis Regulatory Proteins/antagonists & inhibitors , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , MicroRNAs/genetics , Adipose Tissue/cytology , Animals , Apoptosis/drug effects , Apoptosis/genetics , Apoptosis/physiology , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Cell Cycle Checkpoints , Cell Hypoxia , Cell Proliferation , Mesenchymal Stem Cells/cytology , MicroRNAs/metabolism , Rats , Reactive Oxygen Species/metabolism , Up-Regulation
17.
J Cell Physiol ; 235(4): 3539-3547, 2020 04.
Article in English | MEDLINE | ID: mdl-31584202

ABSTRACT

Cardiac hypertrophy is a common phenomenon observed in progressive heart disease associated with heart failure. Insulin-like growth factor receptor II (IGF-IIR) has been much implicated in myocardial hypertrophy. Our previous studies have found that increased activities of signaling mediators, such as calcium/calmodulin-dependent protein kinase II (CaMKII) and calcineurin induces pathological hypertrophy. Given the critical roles played by CaMKII and calcineurin signaling in the progression of maladaptive hypertrophy, we anticipated that inhibition of CaMKII and calcineurin signaling may attenuate IGF-IIR-induced cardiac hypertrophy. The current study, therefore, investigated the effects of IGF-IIR activation on the CaMKII and calcineurin signaling and whether the combinatorial inhibition of the CaMKIIδ and calcineurin signaling could ameliorate IGF-IIR-induced pathological hypertrophy. In the present study, we induced IGF-IIR through the cardiomyocyte-specific transduction of IGFIIY27L via adeno-associated virus 2 (AAV2) to evaluate its effects on cardiac hypertrophy. Interestingly, it was observed that the activation of IGF-IIR signaling through IGFIIY27L induces significant hypertrophy of the myocardium and increased cardiac apoptosis and fibrosis. Moreover, we found that Leu27 IGF-II significantly induced calcineurin and CaMKII expression. Furthermore and importantly, the combinatorial treatment with CaMKII and calcineurin inhibitors significantly alleviates IGF-IIR-induced hypertrophic responses. Thus, it could be envisaged that the inhibition of IGF-IIR may serve as a promising candidate for attenuating maladaptive hypertrophy. Both calcineurin and CaMKII could be valuable targets for developing treatment strategies against hypertension-induced cardiomyopathies.


Subject(s)
Calcineurin/genetics , Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Cardiomegaly/drug therapy , Heart Failure/drug therapy , Receptor, IGF Type 2/genetics , Animals , Apoptosis/genetics , Calcineurin/drug effects , Calcineurin Inhibitors/pharmacology , Calcium-Calmodulin-Dependent Protein Kinase Type 2/antagonists & inhibitors , Cardiomegaly/genetics , Cardiomegaly/pathology , Disease Models, Animal , Heart Failure/genetics , Heart Failure/pathology , Humans , Hypertension/drug therapy , Hypertension/genetics , Hypertension/pathology , Insulin-Like Growth Factor II/genetics , Myocardium/metabolism , Myocardium/pathology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , Rats , Signal Transduction/drug effects
18.
Int J Mol Sci ; 20(17)2019 Sep 02.
Article in English | MEDLINE | ID: mdl-31480672

ABSTRACT

Cardiovascular diseases have a high prevalence worldwide and constitute the leading causes of mortality. Recently, malfunctioning of ß-catenin signaling has been addressed in hypertensive heart condition. Ang-II is an important mediator of cardiovascular remodeling processes which not only regulates blood pressure but also leads to pathological cardiac changes. However, the contribution of Ang-II/ß-catenin axis in hypertrophied hearts is ill-defined. Employing in vitro H9c2 cells and in vivo spontaneously hypertensive rats (SHR) cardiac tissue samples, western blot analysis, luciferase assays, nuclear-cytosolic protein extracts, and immunoprecipitation assays, we found that under hypertensive condition ß-catenin gets abnormally induced that co-activated LEF1 and lead to cardiac hypertrophy changes by up-regulating the IGF-IIR signaling pathway. We identified putative LEF1 consensus binding site on IGF-IIR promoter that could be regulated by ß-catenin/LEF1 which in turn modulate the expression of cardiac hypertrophy agents. This study suggested that suppression of ß-catenin expression under hypertensive condition could be exploited as a clinical strategy for cardiac pathological remodeling processes.


Subject(s)
Cardiomegaly/chemically induced , Cardiomegaly/metabolism , Receptor, IGF Type 2/metabolism , Signal Transduction , beta Catenin/metabolism , Angiotensin II , Animals , Biomarkers/metabolism , Cardiomegaly/pathology , Cell Nucleus/metabolism , GATA4 Transcription Factor/metabolism , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , Lymphoid Enhancer-Binding Factor 1/metabolism , Myocytes, Cardiac/metabolism , NFATC Transcription Factors/metabolism , Promoter Regions, Genetic/genetics , Protein Kinase C-alpha/metabolism , Rats, Inbred SHR
19.
J Cell Biochem ; 120(10): 16956-16966, 2019 10.
Article in English | MEDLINE | ID: mdl-31104312

ABSTRACT

Cardiotoxicity by doxorubicin hampers its therapeutic potential as an anticancer drug, but mechanisms leading to cardiotoxicity remain contentious. Through this study, the functional contribution of insulin-like growth factor receptor type II α (IGF-IIRα) which is a novel stress-inducible protein was explored in doxorubicin-induced cardiac stress. Employing both in vitro H9c2 cells and in vivo transgenic rat models (SD-TG [IGF-IIRα]) overexpressing IGF-IIRα specifically in heart, we found that IGF-IIRα leads to cardiac structural abnormalities and functional perturbations that were severely aggravated by doxorubicin-induced cardiac stress. Overexpression of IGF-IIRα leads to cumulative elevation of stress associated cardiac hypertrophy and apoptosis factors. There was a significant reduction of survival associated proteins p-Akt and estrogen receptor ß/α, and abnormal elevation of cardiac hypertrophy markers such as atrial natriuretic peptide, cardiac troponin-I, and apoptosis-inducing agents such as p53, Bax, and cytochrome C, respectively. IGF-IIRα also altered the expressions of AT1R, ERK1/2, and p38 proteins. Besides, IGF-IIRα also increased the reactive oxygen species production in H9c2 cells which were markedly aggravated by doxorubicin treatment. Together, we showed that IGF-IIRα is a novel stress-induced protein that perturbed cardiac homeostasis and cumulatively exacerbated the doxorubicin-induced cardiac injury that perturbed heart functions and ensuing cardiomyopathy.


Subject(s)
Antibiotics, Antineoplastic/toxicity , Cardiomegaly/chemically induced , Cardiomyopathies/chemically induced , Doxorubicin/toxicity , Heart Defects, Congenital/chemically induced , Receptor, IGF Type 2/biosynthesis , Animals , Apoptosis/drug effects , Cardiotoxicity/pathology , Cell Line , Estrogen Receptor alpha/metabolism , Estrogen Receptor beta/metabolism , Heart/anatomy & histology , Myocytes, Cardiac/metabolism , Oxidative Stress/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Transgenic , Reactive Oxygen Species/metabolism , Receptor, IGF Type 2/genetics , Signal Transduction/drug effects
20.
J Cell Physiol ; 233(9): 7134-7142, 2018 09.
Article in English | MEDLINE | ID: mdl-29574877

ABSTRACT

Irinotecan (CPT11) and Oxaliplatin have been used in combination with fluorouracil and leucovorin for treating colorectal cancer. However, the efficacy of these drugs is reduced due to various side effects and drug resistance. Fisetin, a hydroxyflavone possess anti-proliferative, anti-cancer, anti-inflammatory, and antioxidant activity against various types of cancers. Apart from that, fisetin has been shown to induce cytotoxic effects when combined with other known chemotherapeutic drugs. In this study, we aimed to investigate whether Fisetin was capable of sensitizing both Irinotecan and Oxaliplatin resistance colon cancer cells and explored the possible signaling pathways involved using In vitro and In vivo models. The results showed that Fisetin treatment effectively inhibited cell viability and apoptosis of CPT11-LoVo cells than Oxaliplatin (OR) and parental LoVo cancer cells. Western blot assays suggested that apoptosis was induced by fisetin administration, promoting Caspase-8, and Cytochrome-C expressions possibly by inhibiting aberrant activation of IGF1R and AKT proteins. Furthermore, fisetin inhibited tumor growth in athymic nude mouse xenograft model. Overall, our results provided a basis for Fisetin as a promising agent to treat parental as well as chemoresistance colon cancer.


Subject(s)
Apoptosis/drug effects , Colorectal Neoplasms/pathology , Drug Resistance, Neoplasm/drug effects , Flavonoids/pharmacology , Irinotecan/pharmacology , Oxaliplatin/pharmacology , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Colonic Neoplasms , Flavonols , Male , Mice, Nude , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...