Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 67
Filter
Add more filters

Country/Region as subject
Affiliation country
Publication year range
1.
J Cell Biochem ; 124(4): 619-632, 2023 04.
Article in English | MEDLINE | ID: mdl-36976911

ABSTRACT

Resistance to chemotherapy is the deadlock in cancer treatment. In this study, we used wild-type LOVO (LOVOWT ), a human colon cancer cell line, and the oxaliplatin-resistant sub-clone LOVOOR cells to investigate the molecular mechanisms of the development of drug resistance in colon cancer. Compared with LOVOWT cells, LOVOOR cells had a high proliferation capacity and a high percentage on the G2/M phase. The expression and activation of Aurora-A, a critical kinase in G2/M phase, were higher in LOVOOR cells than in LOVOWT cells. The results from immunofluorescence indicated an irregular distribution of Aurora-A in LOVOOR cells. To evaluate the importance of Aurora-A in oxaliplatin-resistant property of LOVOOR cells, overexpression of Aurora-A in LOVOWT cells and otherwise knockdown of Aurora-A in LOVOOR cells were performed and followed by administration of oxaliplatin. The results indicated that Aurora-A might contribute to the resistance of LOVOOR cells to oxaliplatin treatment by depressing p53 signaling. The specific findings in this study provide a possibility that targeting Aurora-A might be a solution for patients who have failed oxaliplatin treatment.


Subject(s)
Antineoplastic Agents , Colonic Neoplasms , Humans , Oxaliplatin/pharmacology , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Cell Line, Tumor , Colonic Neoplasms/drug therapy , Colonic Neoplasms/genetics , Colonic Neoplasms/metabolism , Drug Resistance, Neoplasm
2.
Environ Toxicol ; 38(1): 39-48, 2023 Jan.
Article in English | MEDLINE | ID: mdl-36124540

ABSTRACT

Parkinson's disease (PD) is a common neurodegenerative disorder associated with striatal dopaminergic neuronal loss in the Substantia nigra. Oxidative stress plays a significant role in several neurodegenerative diseases. Paraquat (PQ) is considered a potential neurotoxin that affects the brain leading to the death of dopaminergic neurons mimicking the PD phenotype. Various scientific reports have proven that cryptotanshinone possesses antioxidant and anti-inflammatory properties. We hypothesized that cryptotanshinone could extend its neuroprotective activity by exerting antioxidant effects. This study was designed to evaluate the effects of cryptotanshinone in both cellular and animal models of PQ-induced PD. Annexin V-PI double staining and immunoblotting were used to detect apoptosis and oxidative stress proteins, respectively. Reactive oxygen species kits were used to evaluate oxidative stress in cells. For in vivo studies, 18 B6 mice were divided into three groups. The rotarod data revealed the motor function and immunostaining showed the survival of TH+ neurons in SNpc region. Our study showed that cryptotanshinone attenuated paraquat-induced oxidative stress by upregulating anti-oxidant markers in vitro, and restored behavioral deficits and survival of dopaminergic neurons in vivo, demonstrating its therapeutic potential.


Subject(s)
Neuroprotective Agents , Parkinson Disease , Animals , Mice , Paraquat/toxicity , Parkinson Disease/drug therapy , Parkinson Disease/metabolism , Dopaminergic Neurons/metabolism , Oxidative Stress , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Disease Models, Animal
3.
Mol Cell Biochem ; 477(1): 143-152, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34586566

ABSTRACT

The heart is a very dynamic pumping organ working perpetually to maintain a constant blood supply to the whole body to transport oxygen and nutrients. Unfortunately, it is also subjected to various stresses based on physiological or pathological conditions, particularly more vulnerable to damages caused by oxidative stress. In this study, we investigate the molecular mechanism and contribution of IGF-IIRα in endoplasmic reticulum stress induction in the heart under doxorubicin-induced cardiotoxicity. Using in vitro H9c2 cells, in vivo transgenic rat cardiac tissues, siRNAs against CHOP, chemical ER chaperone PBA, and western blot experiments, we found that IGF-IIRα overexpression enhanced ER stress markers ATF4, ATF6, IRE1α, and PERK which were further aggravated by DOX treatment. This was accompanied by a significant perturbation in stress-associated MAPKs such as p38 and JNK. Interestingly, PARKIN, a stress responsive cellular protective mediator was significantly downregulated by IGF-IIRα concomitant with decreased expression of ER chaperone GRP78. Furthermore, ER stress-associated pro-apoptotic factor CHOP was increased considerably in a dose-dependent manner followed by elevated c-caspase-12 and c-caspase-3 activities. Conversely, treatment of H9c2 cells with chemical ER chaperone PBA or siRNA against CHOP abolished the IGF-IIRα-induced ER stress responses. Altogether, these findings suggested that IGF-IIRα contributes to ER stress induction and inhibits cellular stress coping proteins while increasing pro-apoptotic factors feeding into a cardio myocyte damage program that eventually paves the way to heart failure.


Subject(s)
Endoplasmic Reticulum Stress , Endoplasmic Reticulum/metabolism , Myocardium/metabolism , Receptor, IGF Type 2/metabolism , Animals , Cell Line , Cytotoxins/adverse effects , Cytotoxins/pharmacology , Doxorubicin/adverse effects , Doxorubicin/pharmacology , Endoplasmic Reticulum/genetics , Rats , Rats, Transgenic , Receptor, IGF Type 2/genetics
4.
J Cell Physiol ; 236(6): 4290-4302, 2021 06.
Article in English | MEDLINE | ID: mdl-33421145

ABSTRACT

High-glucose (HG) suppresses mesenchymal stem cell (MSC) functions, resulting in a decrease in cardiac regenerative capability for MSC in diabetes mellitus (DM). Resveratrol enhances MSC functions under stress. This study explores if cardiac regenerative capability can be enhanced in MSCs pretreated with resveratrol in DM rats receiving MSCs. In vitro evidence confirms that HG decreases MSCs capability through suppression of survival markers, AMP-activated protein kinase (AMPK)/Sirtuin 1 (Sirt1) axis, and expression of apoptotic markers. All of these markers are improved when MSCs are cocultured with resveratrol. Wistar male rats were randomly divided into Sham, DM (DM rats), DM rats with autologous transplantation of adipose-derived stem cells (DM + ADSC), and DM rats with resveratrol pretreated ADSC (DM + RSVL-ADSC). Compared to the Sham, DM induces pathological pathways (including fibrosis, hypertrophy, and apoptosis) and suppresses survival as well as the AMPK/Sirt1 axis in the DM group. DM + ADSC slightly improves the above pathways whereas DM + RSVL-ADSC significantly improves the above pathways when compared to the DM group. These results illustrate that resveratrol pretreated with MSCs may show clinical potential in the treatment of heart failure in patients with DM.


Subject(s)
Antioxidants/pharmacology , Diabetic Cardiomyopathies/therapy , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/drug effects , Myocytes, Cardiac/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Regeneration , Resveratrol/pharmacology , Sirtuin 1/metabolism , AMP-Activated Protein Kinases/metabolism , Adipose Tissue/cytology , Animals , Cell Communication , Cell Line , Cell Proliferation , Coculture Techniques , Diabetic Cardiomyopathies/enzymology , Diabetic Cardiomyopathies/pathology , Diabetic Cardiomyopathies/physiopathology , Disease Models, Animal , Male , Mesenchymal Stem Cells/metabolism , Myocytes, Cardiac/pathology , Rats, Wistar , Signal Transduction , Ventricular Function, Left
5.
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
6.
Mol Biol Rep ; 48(3): 2629-2637, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33791907

ABSTRACT

Obesity in aged population have surges the occurrence of various metabolic disorders including Nonalcoholic fatty liver disease (NAFLD). Apoptosis in the liver is one of the causative factors for NAFLD-induced liver damage. Plants derived bioactive peptides have been shown as an alternative treatment approach for the treating NAFLD due to its less toxicity. Moderate exercise has been reported to improve cellular physiological function prevent age associated metabolic disorders. In the present study, we evaluate the effects of bioactive dipeptide (IF) derived from alcalase potato-protein hydrolysates and swimming exercise in preventing High Fat Diet (HFD)-induced liver damage in senescence accelerated mouse-prone 8 (SAMP8) mice model. Mouse were fed with HFD for 6 weeks followed by oral IF administration or swimming exercise and both for 8 weeks. HFD induces significant structural changes in liver of HFD fed SAMP8 mouse. Both IF administration and exercise prevent the structural abnormalities induced by HFD, however, combined IF treatment and exercise offer better protection. Combined IF treatment and exercise activate PI3K/Akt cell survival protein and effectively inhibit Fas-FADD-induced apoptosis in HFD fed aged mouse. Oral supplementation of bioactive peptide IF combined with moderate swimming exercise effectively alleviate HFD-induced hepatic injury in aged mice.


Subject(s)
Apoptosis , Dipeptides/pharmacology , Hepatocytes/pathology , Phosphatidylinositol 3-Kinases/metabolism , Physical Conditioning, Animal , Protein Hydrolysates/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Swimming , Animals , Apoptosis/drug effects , Biomarkers/metabolism , Cell Survival/drug effects , Diet, High-Fat , Hepatocytes/drug effects , Mice , Solanum tuberosum/chemistry
7.
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
8.
Environ Toxicol ; 35(10): 1043-1049, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32415908

ABSTRACT

In this study, we used ICI 182 780 (ICI), an estrogen receptor (ER) antagonist, to investigate the estrogenic activity of Danshen, and to further explored whether Danshen extract can block Leu27IGF-II-induced hypertrophy in H9c2 cardiomyoblast cells. We first used an IGF-II analog Leu27IGF-II, which specifically activates IGF2R signaling cascades and induces H9c2 cardiomyoblast cell hypertrophy. However, Danshen extract completely inhibited Leu27IGF-II-induced cell size increase, ANP and BNP hypertrophic marker expression, and IGF2R induction. We also observed that Danshen extract inhibited calcineurin protein expression and NFAT3 nuclear translocation, leading to suppression of Leu27IGF-II-induced cardiac hypertrophy. Moreover, the anti-Leu27IGF-II-IGF2R signaling effect of Danshen was totally reversed by ICI, which suggest the cardio protective effect of Danshen is mediated through estrogen receptors. Our study suggests that, Danshen exerts estrogenic activity, and thus, it could be used as a selective ER modulator in IGFIIR induced hypertrophy model.


Subject(s)
Cell Enlargement/drug effects , Drugs, Chinese Herbal/pharmacology , Insulin-Like Growth Factor II/analogs & derivatives , Myoblasts, Cardiac/drug effects , Receptor, IGF Type 2/metabolism , Salvia miltiorrhiza/chemistry , Animals , Calcineurin/metabolism , Cardiomegaly/prevention & control , Cell Line , Cell Survival/drug effects , Drugs, Chinese Herbal/isolation & purification , Estrogen Receptor Antagonists/pharmacology , Fulvestrant/pharmacology , Insulin-Like Growth Factor II/pharmacology , Myoblasts, Cardiac/metabolism , Myoblasts, Cardiac/pathology , Protein Transport , Rats , Receptors, Estrogen/metabolism , Signal Transduction
9.
Environ Toxicol ; 35(2): 145-151, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31714667

ABSTRACT

The use of herbs as alternative cardiovascular disease treatment has attracted a great deal of attention owing to their lower toxicity. Whether Carthamus tinctorius extract prevent cardiomyoblast cell hypertrophy remains unclear. The present study was performed to investigate the effect of C tinctorius extract (CTF) on rat cardiomyoblast cell H9c2 and the possible molecular mechanisms. H9c2 cells were treated with lipopolysaccharide (LPS; 2 µg/mL) for 12 hours, subsequently treated with CTF (1-25 µg/mL) The incubation continued for another 24 hours, and the cells were analyzed with actin staining assay, western blot analysis, and siRNA transfection assays. In the present study, the increased cell size induced by LPS was significantly decreased by pretreating at a concentration of 1-25 µg/mL CTF. It was found that CTF could inhibit cardiac hypertrophy induced by LPS and decrease hypertrophic proteins calcineurin, p-GATA-4, GATA-4, atrial natriuretic peptide, and B-type natriuretic peptide levels in H9c2 cells. Additionally, LPS-induced insulin-like growth factor-II receptor (IGF-IIR) hypertrophy pathway was downregulated by CTF. Moreover, IGF-IR siRNA or inhibitors both reversed the CTF effects, confirming that CTF activates IGF-1R to prevent LPS-induced H9c2 cardiomyoblast cell hypertrophy. The current findings indicate that CTF activates IGF-IR to inhibit IGF-IIR signaling pathway which resulted in reducing H9c2 cardiomyoblast cell hypertrophy induced by LPS.


Subject(s)
Carthamus tinctorius/chemistry , Lipopolysaccharides/pharmacology , Myocytes, Cardiac/drug effects , Plant Extracts/pharmacology , Receptor, IGF Type 1/metabolism , Receptor, IGF Type 2/metabolism , Animals , Cardiomegaly/prevention & control , Cell Size , Dose-Response Relationship, Drug , Gene Knockdown Techniques , Myocytes, Cardiac/metabolism , Plant Extracts/isolation & purification , Rats , Receptor, IGF Type 1/genetics , Receptor, IGF Type 2/genetics , Signal Transduction
10.
Chin J Physiol ; 63(3): 122-127, 2020.
Article in English | MEDLINE | ID: mdl-32594065

ABSTRACT

Pancreatic damage is the major causative agent in type 1 diabetes mellitus (DM). Several strategies have been suggested to regenerate pancreatic functions, such as stem cell transplantation and administration of active components isolating from natural herbals. This study aims to investigate if the synergistically protective effect on damaged pancreatic tissues can be observed in STZ-induced DM rats with autologous transplantation of adipose-derived stem cells (ADSC) coupling with oral administration of resveratrol. Pathological conditions can be recognized in DM rats with pancreatic damage, including reduction of islet size, suppression of survival markers, downregulation of AMPK/Sirt1 axis, and activation of apoptotic signaling. Autologous transplantation of ADSC slightly improves pancreatic functions, whereas autologous transplantation of ADSC coupling with oral administration of resveratrol significantly improves pancreatic functions in DM rats. We suggest that oral administration of resveratrol may enhance the therapeutic effect on DM patients receiving autologous transplantation of ADSC.


Subject(s)
Diabetes Mellitus , Adipose Tissue , Animals , Pancreas , Rats , Regeneration , Resveratrol , Transplantation, Autologous
11.
Int J Mol Sci ; 21(9)2020 May 09.
Article in English | MEDLINE | ID: mdl-32397561

ABSTRACT

Although various advancements in radical surgery and neoadjuvant chemotherapy have been developed in treating osteosarcoma (OS), their clinical prognosis remains poor. A synthetic chemical compound, 3-hydroxylflavone, that is reported to regulate ROS production is known to inhibit human bone osteosarcoma cells. However, its role and mechanism in human OS cells remains unclear. In this study, we have determined the potential of 3-Hydroxy-2-phenylchromone (3-HF) against OS using human osteosarcoma (HOS) cells. Our previous studies showed that Zipper sterile-alpha-motif kinase (ZAK), a kinase member of the MAP3K family, was involved in various cellular events such as cell proliferation and cell apoptosis, and encoded two transcriptional variants, ZAKα and ß. In this study, we show that 3-HF induces the expression of ZAK and thereby enhances cellular apoptosis. Using gain of function and loss of function studies, we have demonstrated that ZAK activation by 3-HF in OS cells is confined to a ZAKß form that presumably plays a leading role in triggering ZAKα expression, resulting in an aggravated cancer apoptosis. Our results also validate ZAKß as the predominant form of ZAK to drive the anticancer mechanism in HOS cells.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Bone Neoplasms/pathology , Flavonoids/pharmacology , MAP Kinase Kinase 4/drug effects , MAP Kinase Kinase Kinases/drug effects , Osteosarcoma/pathology , Caspase 3/biosynthesis , Caspase 3/genetics , Cell Line, Tumor , Enzyme Activation/drug effects , Gain of Function Mutation , Humans , Loss of Function Mutation , MAP Kinase Kinase Kinases/genetics , Membrane Potential, Mitochondrial/drug effects , Protein Isoforms/drug effects , Protein Isoforms/genetics , Signal Transduction/drug effects , Up-Regulation/drug effects
12.
Am J Physiol Cell Physiol ; 317(2): C235-C243, 2019 08 01.
Article in English | MEDLINE | ID: mdl-31116582

ABSTRACT

Doxorubicin (DOX) is an anthracycline antibiotic commonly employed for the treatment of various cancers. However, its therapeutic uses are hampered by side effects associated with cumulative doses during the course of treatment. Whereas deregulation of autophagy in the myocardium has been involved in a variety of cardiovascular diseases, the role of autophagy in DOX-induced cardiomyopathy remains debated. Our earlier studies have shown that DOX treatment in a rat animal model leads to increased expression of the novel stress-inducible protein insulin-like growth factor II receptor-α (IGF-IIRα) in cardiac tissues, which exacerbated the cardiac injury by enhancing oxidative stress and p53-mediated mitochondria-dependent cardiac apoptosis. Through this study, we investigated the contribution of IGF-IIRα to dysregulation of autophagy in heart using both in vitro H9c2 cells (DOX treated, 1 µM) and in vivo transgenic rat models (DOX treated, 5 mg/kg ip for 6 wk) overexpressing IGF-IIRα specifically in the heart. We found that IGF-IIRα primarily localized to mitochondria, causing increased mitochondrial oxidative stress that was severely aggravated by DOX treatment. This was accompanied by a significant perturbation in mitochondrial membrane potential and increased leakage of cytochrome c, causing increased cleaved caspase-3 activity. There were significant alterations in phosphorylated AMP-activated protein kinase (p-AMPK), phosphorylated Unc-51 like kinase-1 (p-ULK1), PARKIN, PTEN-induced kinase 1 (PINK1), microtubule-associated protein 1 light chain 3 (LC3), and p62 proteins, which were more severely disrupted under the combined effect of IGF-IIRα overexpression plus DOX. Finally, LysoTracker Red staining showed that IGF-IIRα overexpression causes lysosomal impairment, which was rescued by rapamycin treatment. Taken together, we found that IGF-IIRα leads to mitochondrial oxidative stress, decreased antioxidant levels, disrupted mitochondrial membrane potential, and perturbed mitochondrial autophagy contributing to DOX-induced cardiomyopathy.


Subject(s)
Antibiotics, Antineoplastic/toxicity , Doxorubicin/toxicity , Heart Diseases/chemically induced , Mitochondria, Heart/drug effects , Mitophagy/drug effects , Myocytes, Cardiac/drug effects , Oxidative Stress/drug effects , Receptor, IGF Type 2/metabolism , Animals , Apoptosis Regulatory Proteins/metabolism , Autophagy-Related Proteins/metabolism , Cardiotoxicity , Cell Line , Heart Diseases/genetics , Heart Diseases/metabolism , Heart Diseases/pathology , Lysosomes/drug effects , Lysosomes/metabolism , Lysosomes/pathology , Membrane Potential, Mitochondrial/drug effects , Mitochondria, Heart/metabolism , Mitochondria, Heart/pathology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Rats, Sprague-Dawley , Rats, Transgenic , Receptor, IGF Type 2/genetics , Signal Transduction/drug effects
13.
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
14.
Environ Toxicol ; 34(1): 5-12, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30240538

ABSTRACT

Consumption of high fat diet (HFD) is associated with increased cardiovascular risk factors among elderly people. Aging and obesity induced-cardiac remodeling includes hypertrophy and fibrosis. Gelsolin (GSN) induces cardiac hypertrophy and TGF-ß, a key cytokine, which induces fibrosis. The relationship between TGF-ß and GSN in aging induced cardiac remodeling is still unknown. We evaluated the expressions of TGF-ß and GSN in HFD fed 22 months old aging SD rats, followed by the administration of either probucol or alcalase potato protein hydrolysate (APPH). Western blotting and Masson trichrome staining showed that APPH (45 and 75 mg/kg/day) and probucol (500 mg/kg/day) treatments significantly reduced the aging and HFD-induced hypertrophy and fibrosis. Echocardiograph showed that the performance of the hearts was improved in APPH, and probucol treated HFD aging rats. Serum from all rats was collected and H9c2 cells were cultured with collected serums separately. The GSN dependent hypertrophy was inhibited with an exogenous TGF-ß in H9c2 cells cultured in HFD+ APPH treated serum. Thus, we propose that along with its role in cardiac fibrosis, TGF-ß also acts as an upstream activator of GSN dependent hypertrophy. Hence, TGF-ß in serum could be a promising therapeutic target for cardiac remodeling in aging and/or obese subjects.


Subject(s)
Diet, High-Fat/adverse effects , Heart Diseases/prevention & control , Obesity/diet therapy , Protein Hydrolysates/administration & dosage , Solanum tuberosum/anatomy & histology , Subtilisins/administration & dosage , Administration, Oral , Aging/metabolism , Aging/pathology , Animals , Cells, Cultured , Gelsolin/metabolism , Heart Diseases/etiology , Myocardium/metabolism , Myocardium/pathology , Obesity/complications , Obesity/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction , Subtilisins/chemistry , Transforming Growth Factor beta/metabolism
15.
Environ Toxicol ; 33(12): 1254-1260, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30208247

ABSTRACT

Human hepatocellular carcinoma (HCC) is currently the second most common cancer and one of the leading causes of cancer-related mortality in Taiwan. Previous reports show that the expression of (E-type prostaglandin 2) EP2 and (E-type prostaglandin 4) EP4 are elevated in HCC and further demonstrate that Prostaglandin E2 (PGE2) induces HA22T cell proliferation and metastasis through EP2 and EP4 receptor. Danshen (root of Salvia miltiorrhiza Bunge) is a very important and popular traditional Chinese herbal medicine which is widely and successfully used against breast cancer, leukemia, pancreatic cancer, and head and neck squamous carcinoma cells. In this study, we used Cryptotansinone (Dsh-003) (MW 269.14) from Danshen to investigate their effect and corresponding mechanism of action in PGE2-treated HA22T cells. Dsh-003 inhibited HA22T cell viability and further induced cell apoptosis in PGE2-treated HA22T cells. Furthermore, Dsh-003 inhibited EP2, EP4, and their downstream effector such as p-PI3K and p-Akt expression in HA22T hepatocellular carcinoma cells. We also observed that Dsh-003 blocked PGE2-induced cell migration by down-regulating PGE2-induced ß-catenin expression and by up-regulating E-cadherin and GSK3-ß expression. All these findings suggest that Dsh-003 inhibit human HCC cell lines and could potentially be used as a novel drug for HCC treatment.


Subject(s)
Carcinoma, Hepatocellular/pathology , Cell Movement/drug effects , Dinoprostone/pharmacology , Liver Neoplasms/pathology , Phenanthrenes/isolation & purification , Phenanthrenes/pharmacology , Salvia miltiorrhiza/chemistry , Cell Proliferation/drug effects , Cell Survival/drug effects , Humans , Signal Transduction/drug effects , Tumor Cells, Cultured
16.
Environ Toxicol ; 32(3): 754-763, 2017 Mar.
Article in English | MEDLINE | ID: mdl-27098997

ABSTRACT

Severe and potentially fatal hypotension and cardiac contractile dysfunction are common symptoms in patients with sepsis. LPS was previously found to dramatically upregulate expression of fibrosis-related factors FGF-2, uPA, MMP-2, and MMP-9 in primary cardiac fibroblasts. MMPs are capable of denaturing and degrading fibrillar collagens and other components of the extracellular matrix (ECM). Studies have shown that dysregulation of expression of MMPs is associated with development of myocardial extracellular matrix remodeling and cardiac fibrosis, which contribute to progression of heart failure. In this study, H9c2 cells and cardiac fibroblasts were divided into five treatment groups: control, LPS (1 µg/mL) and three concentrations of FCEtOH (Carthami Flos ethanolic extract) (31.25, 62.5, and 125 µg/mL). Phosphorylation of ERK-1/2 was observed to be rapidly induced upon treatment with LPS. In contrast, it was significantly suppressed by the administration of FCEtOH (125 µg/mL). Effects of FCEtOH on LPS-induced MMP-2 and MMP-9 expression in H9c2 cells occurred directly through ERK1/2 were determined. H9c2 cells were therefore pretreated with EGF-R to activate ERK pathway. Both protein levels of MMP-2 and MMP-9 and immunefluorescent signals of MMP-9 were significantly enhanced by EGFR. In contrast, MMP-2 and MMP-9 were significantly reduced after FCEtOH administration. Based on these findings, the authors concluded that FCEtOH elicits a protective effect against LPS-induced cardio-fibrosis through the ERK1/2 pathway. Carthamus tinctorius L may potentially serve as a cardio-protective agent against LPS- induced cardiac fibrosis. © 2016 Wiley Periodicals, Inc. Environ Toxicol 32: 754-763, 2017.


Subject(s)
Carthamus tinctorius/chemistry , Lipopolysaccharides/toxicity , MAP Kinase Signaling System/drug effects , Plant Extracts/pharmacology , Up-Regulation/drug effects , Animals , Carthamus tinctorius/metabolism , Cells, Cultured , Down-Regulation/drug effects , Fibroblast Growth Factor 2/metabolism , Humans , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Mice , Microscopy, Fluorescence , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Phosphorylation/drug effects , Plant Extracts/chemistry , Rats , Rats, Sprague-Dawley , Urokinase-Type Plasminogen Activator/metabolism
17.
Growth Factors ; 34(1-2): 11-8, 2016 02.
Article in English | MEDLINE | ID: mdl-27055565

ABSTRACT

Extreme hypoxia often leads to myocardial apoptosis and causes heart failure. Angiotensin-(1-7)Ang-(1-7) is well known for its cardio-protective effects. However, the effects of Ang-(1-7) on long-term hypoxia (LTH)-induced apoptosis remain unknown. In this study, we found that Ang-(1-7) reduced myocardial apoptosis caused by hypoxia through the Mas receptor. Activation of the Ang-(1-7)/Mas axis down-regulated the hypoxia pro-apoptotic signaling cascade by decreasing the protein levels of hypoxia-inducible factor 1α (HIF-1α) and insulin-like growth factor binding protein-3 (IGFBP3). Moreover, the Ang-(1-7)/Mas axis further inhibited HIF-1α nuclear translocation. On the other hand, Ang-(1-7) activated the IGF1R/PI3K/Akt signaling pathways, which mediate cell survival. However, the above effects were abolished by A779 treatment or silencing of Mas expression. Taken together, our findings indicate that the Ang-(1-7)/Mas axis protects cardiomyocytes from LTH-stimulated apoptosis. The protective effect of Ang-(1-7) is associated with the inhibition of HIF-1α nuclear translocation and the induction of IGF1R and Akt phosphorylation.


Subject(s)
Angiotensin I/pharmacology , Apoptosis/drug effects , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Myocytes, Cardiac/drug effects , Peptide Fragments/pharmacology , Receptor, Angiotensin, Type 1/metabolism , Active Transport, Cell Nucleus/drug effects , Animals , Cell Hypoxia , Cell Nucleus/metabolism , Cells, Cultured , Myocytes, Cardiac/metabolism , Rats , Rats, Sprague-Dawley
18.
Int J Med Sci ; 13(8): 569-77, 2016.
Article in English | MEDLINE | ID: mdl-27499689

ABSTRACT

High-calorie diet-induced obesity leads to cardiomyocyte dysfunction and apoptosis. Impaired regulation of epididymal fat content in obese patients has been known to increase the risk of cardiac injury. In our study, a lactic acid bacteria, Lactobacillus reuteri GMNL-263, was evaluated for its potential to reduce body weight and body fat ratio and to prevent heart injury in rats with high-fat diet-induced obesity. Lactic acid bacteria supplementation restored the cardiac function and decreased the physiological changes in the heart of the obese rats. In addition, the Fas/Fas-associated protein pathway-induced caspase 3/e Poly polymerase mediated apoptosis in the cardiomyocytes of the obese rats was reversed in the Lr263-treated rats. These results reveal that fed with Lr-263 reduces body fat ratio, inhibits caspase 3-mediated apoptosis and restores cardiac function in obese rats through recovery of ejection fraction and fractional shortening. Our results indicated that the administration of Lr263 lactic acid bacteria can significantly down-regulate body fat and prevent cardiomyocyte injury in obese rats.


Subject(s)
Epididymis/physiopathology , Limosilactobacillus reuteri/metabolism , Obesity/therapy , Probiotics/administration & dosage , Adipose Tissue/growth & development , Adipose Tissue/microbiology , Animals , Apoptosis/drug effects , Body Weight/drug effects , Diet, High-Fat , Dietary Supplements , Epididymis/drug effects , Epididymis/growth & development , Epididymis/microbiology , Hot Temperature , Male , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Obesity/metabolism , Obesity/microbiology , Rats
19.
BMC Complement Altern Med ; 16: 150, 2016 May 27.
Article in English | MEDLINE | ID: mdl-27234802

ABSTRACT

BACKGROUND: San Huang Shel Shin Tang (SHSST) is a traditional herbal decoction used as a hepato-protective agent and is composed of Rheum officinale Baill, Scutellaria baicalnsis Geprgi and Coptis chinensis Franch (2:1:1 w/w). Beta-cyclodextrin (ß-CD) modification may potentially increase the solubility and spectral properties of SHSST. METHODS: In this research, the hepato-protective effects of unmodified SHSST, ß-CD modified SHSST complex (SHSSTc) and silymarin were evaluated in carbon tetrachloride (CCl4) induced acute hepatotoxicity in rats. RESULTS: SHHSTc (40 mg/kg/day) and silymarin (100 mg/kg/day) both decreased the CCl4-induced cirrhosis pathway-related transforming growth factor beta (TGF-ß) and apoptosis pathway-related caspase-8 protein expressions, but SHSST (40 mg/kg/day) did not reduce TGF-ß and caspase-8 significantly . Moreover, SHHSTc (40 mg/kg/day) enhanced the activation of insulin-like growth factor 1 receptor (IGF1R) mediated survival pathway than the silymarin (100 mg/kg/day) to protect the liver from damage induced by CCl4. CONCLUSIONS: ß-CD modification promotes hepato-protective effects of SHSST and reduces the required-dosage of the SHSST.


Subject(s)
Chemical and Drug Induced Liver Injury/prevention & control , Drugs, Chinese Herbal/pharmacology , Plant Extracts/pharmacology , Protective Agents/pharmacology , beta-Cyclodextrins/pharmacology , Animals , Carbon Tetrachloride , Drug Synergism , Rats
20.
Chin J Physiol ; 59(6): 323-330, 2016 Dec 31.
Article in English | MEDLINE | ID: mdl-27817194

ABSTRACT

Increased serum norepinephrine level is one of pathological processes relating to heart disease (HD). Estrogens are considered as potential therapeutics for the treatment of HD; however, estrogen supplementation shows some side-effects, such as increasing the risk of developing breast, endometrial and ovarian cancers. This study investigated the cardio-protective effects of daidzein (Dai), a selective estrogen receptor modulator (SERM) from soy bean extract, in H9c2 cardiomyoblast cells treated with isoproterenol (ISO), a norepinephrine analog. In this in vitro model, H9c2 cells treated with Dai at different concentrations showed no statistical difference in cell viability. TdT-mediated digoxigenin-dUTP nick-end labeling (TUNEL) data and western blotting results indicated that Dai treated-H9c2 cells recovered from the damage induced by ISO. The recovery effects of Dai on ISO-induced damage were blocked by inhibition of Akt activation through adding Akt inhibitor. On the other hand, the fold changes of phosphorylated Akt (p-Akt)/Akt normalized with the control for con, 0.25, 0.5, 1, 3 and 24 h of treatment were 1, 2, 5, 13, 11 and 10, respectively. In conclusion, Dai ameliorates apoptosis of cardiomyoblasts induced by ISO through Akt signaling pathway.


Subject(s)
Apoptosis/drug effects , Isoflavones/pharmacology , Myoblasts, Cardiac/drug effects , Phytoestrogens/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Animals , Cell Line , In Situ Nick-End Labeling , Isoproterenol , Myoblasts, Cardiac/metabolism , Rats
SELECTION OF CITATIONS
SEARCH DETAIL