Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
1.
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
2.
Environ Toxicol ; 36(5): 729-736, 2021 May.
Article in English | MEDLINE | ID: mdl-33336893

ABSTRACT

Oxidative stress is a major cause of aging related skin injuries. Hydrogen peroxide related ROS accumulation triggers increase in matrix metalloproteinases and elevated collagen degradation, which is a characteristic of skin aging. In this study, we investigated the protective effect of Poria cocos, used widely in the treatment of inflammatory diseases, against H2 O2 induced oxidative stress. The aqueous extract of dried P. cocos was obtained by heating 10 g in 500 ml of distilled water. The mixture was evaporated up to 400 ml and the remaining 100 ml was filtered through muslin cloth repeatedly to obtain a clear aqueous extract of the P. cocos. Hs68 human dermal fibroblast cells were challenged with 100 µM of H2 O2 for 24 h. Following H2 O2 challenge, the cells were treated with increasing concentration of P. cocos extract (100-400 µg/ml) for 24 h. P. cocos extract hindered the H2 O2 induced cell death significantly that was correlated with reduction in ROS accumulation. Western blot analysis show that P. cocos extract suppressed the expression of metallomatrix proteinases, inflammatory markers and skin aging markers, but increased TGF-ß1 levels and antioxidant related proteins. These data suggest that P. cocos is effective in attenuating oxidative stress associated skin aging effects and may be a potential agent in cosmetics products.


Subject(s)
Skin Aging , Wolfiporia , Antioxidants/pharmacology , Cell Line , Collagen , Fibroblasts , Humans , NF-E2-Related Factor 2 , Smad7 Protein , Transforming Growth Factor beta
3.
Environ Toxicol ; 35(4): 468-477, 2020 Apr.
Article in English | MEDLINE | ID: mdl-31794124

ABSTRACT

It has been well-documented that the consumption of deep sea water (DSW) has beneficial effects on myocardial hypertrophy and cardiac apoptosis induced by hypercholesterolemia. However, the molecular mechanisms for the anti-inflammatory effects of DSW on diabetic cardiomyopathy are still largely unclear. The main purpose of this present study was to test the hypothesis that DSW exerts anti-inflammatory effects through the suppression of the TNF-α-mediated signaling pathways. IP injection of streptozotocin (STZ) at the dose of 65 mg/kg was used to establish a diabetes rat model. DSW mineral extracts that diluted in desalinated water were prepared in three different dosages and administered to the rats through gavages for 4 weeks. These dosages are DSW-1X (equivalent to 37 mg Mg2+ /kg/day), 2X (equivalent to 74 mg Mg2+ /kg/day) and 3X (equivalent to 111 mg Mg2+ mg/kg/day). Immunofluorescence staining and Western blot showed that the protein expression level of TNF-α was markedly higher in the STZ-induced diabetic rat hearts than in the control group. Consequently, the phosphorylation levels of the TNF-α-modulated downstream signaling molecules and P38 mitogen-activated protein kinases (MAPKs) were notably elevated in heart tissues of STZ-induced diabetes. These higher phosphorylation levels subsequently upregulated NF-κB-modulated inflammatory mediators, such as cyclooxygenase (COX)-II and inducible nitric oxide synthase (iNOS). However, treatment with DSW as well as MgSO4 , the main mineral in DSW, significantly reversed all the alterations. These findings suggest that DSW has potential as a therapeutic agent for preventing diabetes-related cardiovascular diseases.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Diabetes Mellitus, Experimental/drug therapy , Diabetic Cardiomyopathies/prevention & control , Minerals/therapeutic use , Seawater/chemistry , Tumor Necrosis Factor-alpha/metabolism , Animals , Anti-Inflammatory Agents/administration & dosage , Diabetes Mellitus, Experimental/immunology , Diabetic Cardiomyopathies/immunology , Inflammation , Male , Minerals/administration & dosage , Myocardium/immunology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/immunology , Myocytes, Cardiac/pathology , Rats , Rats, Sprague-Dawley , Signal Transduction , Streptozocin
4.
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
5.
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
6.
Environ Toxicol ; 34(2): 210-218, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30450786

ABSTRACT

IGF-IIR activation regulates cardiac remodeling leading to apoptosis. Here, we identified the novel IGF-IIRα (150 KDa), a truncated IGF-IIR transcript enhances cardiac apoptosis under high-salt uptake in transgenic rat model. Echocardiographic analysis revealed decline in ejection fraction and fractional shortening percentage in IGF-IIRα (TG) rats. We found that IGF-IIRα TG rats developed severe apoptosis and fibrosis as identified through TUNEL assay and Masson's trichrome staining. Importantly, the heart functioning, apoptosis, and fibrosis were significantly affected under high-salt conditions in IGF-IIRα (TG) rats. Significant upregulation of apoptosis was evident from decreased Bcl-2, p-AKT, and p-PI3K expressions with concomitant increase in Bad, cytochrome C, cleaved caspase 3 levels. We found that, IGF-IIRα highly induced tissue fibrosis through collagen accumulation (col I, col III) and up regulated various fibrotic markers such as tPA, uPA, TGF-ß, and vimentin expressions. The observed upregulation of fibrosis were significantly regulated under high-salt conditions and their over regulation under IGF-IIRα over expressions shows the key role of IGF-IIRα in promoting high-salt induced fibrosis. During IGF-IIRα over expression induced cardiotoxicity, under high salt condition, and it destroys the interaction between CHIP and HSF1, which promotes the degradation of HSF1 and results in upregulation of IGF-IIR/IGF-IIRα expressions. Altogether, the study unveils novel IGF-IIRα in the regulation of cardiac apoptosis and fibrosis under high-salt diet.


Subject(s)
Apoptosis/genetics , Gene Expression Regulation , Myocardium/pathology , Receptor, IGF Type 2/genetics , Sodium Chloride, Dietary/adverse effects , Ventricular Remodeling/genetics , Animals , Apoptosis/drug effects , Fibrosis , Gene Expression Regulation/drug effects , Myocardium/metabolism , Rats , Rats, Transgenic , Ventricular Remodeling/drug effects
7.
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
8.
Int J Mol Sci ; 20(12)2019 Jun 23.
Article in English | MEDLINE | ID: mdl-31234585

ABSTRACT

Hypertension is one of the growing risk factors for the progression of long-term memory loss. Hypertension-mediated memory loss and treatment remain not thoroughly elucidated to date. Plant-based natural compounds are an alternative solution to treating human diseases without side effects associated with commercial drugs. This study reveals that bioactive peptides extracted from soy hydrolysates mimic hypertension-mediated memory loss and neuronal degeneration and alters the memory molecular pathway in spontaneously hypertensive rats (SHR). The SHR animal model was treated with bioactive peptide VHVV (10 mg/kg/oral administration) and angiotensin-converting-enzyme (ACE) inhibitors (5 mg/kg/oral administration) for 24 weeks. We evaluated molecular level expression of brain-derived neurotrophic factor (BDNF), cAMP response element binding protein (CREB), and survival markers phospho-protein kinase B (P-AKT) and phosphoinositide 3-kinase (PI3K) after 24 weeks of treatment for SHR in this study. Western blotting, hematoxylin and eosin (H&E) staining, and immunohistochemistry showed long-term memory loss and neuronal degeneration in SHR animals. Bioactive peptide VHVV-treated animals upregulated the expression of long-term memory-relate proteins and neuronal survival. Spontaneously hypertensive rats treated with oral administration of bioactive peptide VHVV had activated CREB-mediated downstream proteins which may reduce hypertension-mediated long-term memory loss and maintain neuronal survival.


Subject(s)
Biomarkers , Memory, Long-Term/drug effects , Peptides/pharmacology , Angiotensin-Converting Enzyme Inhibitors/pharmacology , Animals , Blood-Brain Barrier/metabolism , Brain-Derived Neurotrophic Factor/metabolism , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , Hypertension/drug therapy , Hypertension/etiology , Hypertension/metabolism , Immunohistochemistry , Neurons/drug effects , Neurons/metabolism , Peptides/chemistry , Rats , Rats, Inbred SHR , Signal Transduction
9.
J Cell Physiol ; 233(11): 8458-8466, 2018 11.
Article in English | MEDLINE | ID: mdl-29856483

ABSTRACT

Heart failure (HF) remains a major cause of morbidity and mortality worldwide. The primary cause identified for HF is impaired left ventricular myocardial function, and clinical manifestations may lead to severe conditions like pulmonary congestion, splanchnic congestion, and peripheral edema. Development of new therapeutic strategies remains the need of the hour for controlling the problem of HF worldwide. Deeper insights into the molecular mechanisms involved in etiopathology of HF indicate the significant role of calcium signaling, autocrine signaling pathways, and insulin-like growth factor-1 signaling that regulates the physiologic functions of heart growth and development such as contraction, metabolism, hypertrophy, cytokine signaling, and apoptosis. In view of these facts, a transcription factor (TF) regulating the myriad of these signaling pathways may prove as a lead candidate for development of therapeutics. Adenovirus E4 promoter-binding protein (E4BP4), also known as nuclear-factor, interleukin 3 regulated (NFIL3), a type of basic leucine zipper TF, is known to regulate the signaling processes involved in the functioning of heart. The current review discusses about the expression, structure, and functional role of E4BP4 in signaling processes with emphasis on calcium signaling mechanisms, autocrine signaling, and insulin-like growth factor II receptor-mediated processes regulated by E4BP4 that may regulate the pathogenesis of HF. We propose that E4BP4, being the critical component for the regulation of the above signaling processes, may serve as a novel therapeutic target for HF, and scientific investigations are merited in this direction.


Subject(s)
Basic-Leucine Zipper Transcription Factors/genetics , DNA-Binding Proteins/genetics , Heart Failure/genetics , Apoptosis/genetics , Autocrine Communication/genetics , Calcium Signaling/genetics , Gene Expression Regulation/genetics , Heart Failure/pathology , Humans , Insulin-Like Growth Factor I/genetics , Promoter Regions, Genetic , Transcription Factors/genetics
10.
J Cell Biochem ; 119(9): 7855-7864, 2018 09.
Article in English | MEDLINE | ID: mdl-29932238

ABSTRACT

Aberrant expression of leucine zipper- and sterile ɑ motif-containing kinase (ZAK) observed in pathological human myocardial tissue is associated with the progression and elevation of hypertrophy. Our previous reports have correlated high levels of estrogen (E2) and abundant estrogen receptor (ER) α with a low incidence of pathological cardiac-hypertrophy and heart failure in the premenopause female population. However, the effect of elevated ERß expression is not well known yet. Therefore, in this study, we have analyzed the cardioprotective effects and mechanisms of E2 and/or ERß against ZAK overexpression-induced cellular hypertrophy. We have used transient transfection to overexpress ERß into the ZAK tet-on H9c2 cells that harbor the doxycycline-inducible ZAK plasmid. The results show that ZAK overexpression in H9c2 cells resulted in hypertrophic effects, which was correlated with the upregulation of p-JNK and p-p38 MAPKs and their downstream transcription factors c-Jun and GATA-4. However, ERß and E2 with ERß overexpressions totally suppressed the effects of ZAK overexpression and inhibited the levels of p-JNK, p-p38, c-Jun, and GATA-4 effectively. Our results further reveal that ERß directly binds with ZAK under normal conditions; however, ZAK overexpression reduced the association of ZAK-ERß. Interestingly, increase in ERß and E2 along with ERß overexpression both enhanced the binding strengths of ERß and ZAK and reduced the ZAK protein level. ERß overexpression also suppressed the E3 ligase-casitas B-lineage lymphoma (CBL) and attenuated CBL-phosphoinositide 3-kinase (PI3K) protein association to prevent PI3K protein degradation. Moreover, ERß and/or E2 blocked ZAK nuclear translocation via the inhibition of small ubiquitin-like modifier (SUMO)-1 modification. Taken together, our results further suggest that ERß overexpression strongly suppresses ZAK-induced cellular hypertrophy and myocardial damage.


Subject(s)
Estrogen Receptor beta/genetics , Myoblasts, Cardiac/cytology , Protein Kinases/metabolism , SUMO-1 Protein/metabolism , Animals , Cell Enlargement , Cell Line , Estrogen Receptor beta/metabolism , Estrogens/pharmacology , Gene Expression Regulation , Myoblasts, Cardiac/drug effects , Myoblasts, Cardiac/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Protein Kinases/genetics , Proto-Oncogene Proteins c-cbl , Rats
11.
J Cell Biochem ; 119(3): 2742-2749, 2018 03.
Article in English | MEDLINE | ID: mdl-29052864

ABSTRACT

Hypertension-stimulated cardiac hypertrophy and apoptosis play critical roles in the progression of heart failure. Our previous study suggested that hypertensive angiotensin II (Ang II) enhanced insulin-like growth factor receptor II (IGF-IIR) expression and cardiomyocyte apoptosis, which are involved JNK activation, sirtuin1 (SIRT1) degradation, and heat-shock transcription factor 1 (HSF1) acetylation. Moreover, previous studies have implied that short-term hypoxia (STH) might exert cardioprotective effects. However, the effects of STH on Ang II-induced cardiomyocyte apoptosis remain unknown. In this study, we found that STH reduced myocardial apoptosis caused by Ang II via upregulation of the Mas receptor (MasR) to inhibit the AT1 R signaling pathway. STH activates MasR to counteract the Ang II pro-apoptotic signaling cascade by inhibiting IGF-IIR expression via downregulation of JNK activation and reduction of SIRT1 degradation. Hence, HSF could remain deacetylated, and repress IGF-IIR expression. These effects decrease the activation of downstream pro-apoptotic and hypertrophic cascades and protect cardiomyocytes from Ang II-induced injury. In addition, we also found that silencing MasR expression enhanced Ang II-induced cardiac hypertrophy and the apoptosis signaling pathway. These findings suggest a critical role for MasR in cardiomyocyte survival. Altogether, our findings indicate that STH protects cardiomyocytes from Ang II-stimulated apoptosis. The protective effects of STH are associated with the upregulation of MasR to inhibit AT1 R signaling. STH could be a potential therapeutic strategy for cardiac diseases in hypertensive patients.


Subject(s)
Angiotensin II/pharmacology , Apoptosis/drug effects , Myocytes, Cardiac/metabolism , Proto-Oncogene Proteins/biosynthesis , Receptor, Angiotensin, Type 1/metabolism , Receptors, G-Protein-Coupled/biosynthesis , Signal Transduction/drug effects , Up-Regulation/drug effects , Animals , Cell Hypoxia/drug effects , Cell Line , Myocytes, Cardiac/pathology , Proto-Oncogene Mas , Rats
12.
Environ Toxicol ; 33(7): 789-797, 2018 Jul.
Article in English | MEDLINE | ID: mdl-29708300

ABSTRACT

Cardiomyocyte apoptosis is the major risk factor for the development of heart failure (HF). The purpose of this study was to evaluate the effects of Gamma-aminobutyric acid (GABA) tea on hypertension-induced cardiac apoptotic pathways in spontaneously hypertensive rats (SHR). In order to reveal the mechanisms, 36 male SHR at eight weeks of age, 200 g were divided into six groups. One group was fed water as a control group. Other rats were administered one of the following treatments: GABA tea at dose 150 and 300 mg/kg/day as low GABA tea (LGT) and high GABA tea (HGT) groups, respectively, pure GABA at dose 150 and 300 mg/kg/day as LG and HG groups, respectively, green tea (GT) as control of LGT and HGT groups. After 12 weeks, cardiac tissues were analyzed by histological analysis, western blotting, and TUNEL assays. GABA tea, GT, and pure GABA decreased hypertension-induced cardiac abnormalities, including abnormal myocardial architecture. In addition, GABA tea, GT, and pure GABA dramatically increased anti-apoptotic protein, Bcl2. Furthermore, GABA tea, GT, and pure GABA also decreased activated-caspase 9 and activated-caspase 3. Additionally, the survival associated protein IGF-I and PI3K/Akt were enhanced in cardiac tissues upon treatment. Our results showed an optimistic anti-apoptotic and pro-survival effects of GABA tea treatment against hypertensive rat hearts.


Subject(s)
Apoptosis/drug effects , Signal Transduction/drug effects , Tea/chemistry , gamma-Aminobutyric Acid/pharmacology , Animals , Caspase 3/metabolism , Heart Ventricles/drug effects , Heart Ventricles/metabolism , Hypertension/drug therapy , Hypertension/metabolism , Hypertension/pathology , Insulin-Like Growth Factor I/metabolism , Male , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Inbred SHR , Receptors, Somatomedin/metabolism , Tea/metabolism , bcl-2 Homologous Antagonist-Killer Protein/metabolism , bcl-2-Associated X Protein/metabolism , gamma-Aminobutyric Acid/therapeutic use
13.
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
14.
Growth Factors ; 33(4): 275-81, 2015.
Article in English | MEDLINE | ID: mdl-26340107

ABSTRACT

Myocardial infarction (MI) usually results in myocardial ischemia, remodeling and hypoxia that lead to cell death. To date, the insulin-like growth factor binding protein-3 (IGFBP3) is known to play an important role in insulin growth factor (IGF) bioavailability. Previous studies have found that hypoxia results in cell apoptosis. However, the detailed mechanism and roles of IGFBP3 in long-term hypoxia (LTH) regulated heart cell apoptosis remains unknown. In this study H9c2 cardiomyoblast cells were treated with investigated long-term hypoxic exposure with the possible mechanisms involved. The results showed that LTH enhanced IGFBP3 protein synthesis and induced its secretion. The accumulated IGFBP3 sequestered Insulin growth factor 1 (IGF-1) away from the type I IGF receptor (IGF-1 R), which blocked the IGF1R/PI3K/Akt survival signaling pathway, resulting in cell apoptosis. According to our findings, IGFBP3 could be a valuable target for developing treatments for cardiac diseases in long-term hypoxia exposure patients.


Subject(s)
Apoptosis , Insulin-Like Growth Factor Binding Protein 3/metabolism , Myocytes, Cardiac/metabolism , Oxygen/metabolism , Animals , Cell Hypoxia , Cell Line , Exocytosis , Insulin-Like Growth Factor Binding Protein 3/genetics , Insulin-Like Growth Factor I/metabolism , Rats
15.
Food Res Int ; 129: 108862, 2020 03.
Article in English | MEDLINE | ID: mdl-32036911

ABSTRACT

Hypertension, which is known as a silent killer, is the second leading cause of kidney failure worldwide. Elevated blood pressure causes approximately 7.6 million deaths, which account for ~13.5% of the total deaths and will continue to rise. High blood pressure is the prime risk factor associated with complications in major organs, including the heart, brain and kidney. High blood pressure accelerates oxidative stress and thereby causes organ dysfunction through the production of reactive oxygen species. In this study, we investigated the renal-protective effects of the bioactive peptide IF from alcalase potato protein hydrolysate in spontaneously hypertensive rat kidney. Sixteen-week-old spontaneously hypertensive rats were divided into three groups (n = 6), and Sixteen-week-old Wistar Kyoto rats (n = 6) served as the control group. The rats were administered IF and captopril via oral gavage for 8 weeks and then sacrificed, and their kidneys were harvested. The kidney sections from the rats treated with IF showed restoration of the structure of the glomerulus and Bowman's capsule. The expression levels of Nrf2-mediated antioxidants were also increased, as confirmed by 4-hydroxynonenal immunohistochemical staining. The TUNEL assay revealed a significant reduction in the number of apoptotic cells in the IF-treated groups, which was consistent with the western blot results. Thus, the bioactive peptide IF exerts potential protective effects against hypertension-associated ROS-mediated renal damage via the Nrf2-dependent antioxidant pathway along the DJ-1 and AKT axes. Hence, we speculate that IF might have promising therapeutic effects on renal damage associated with hypertension.


Subject(s)
Antioxidants/pharmacology , Kidney Diseases/prevention & control , NF-E2-Related Factor 2/metabolism , Plant Proteins/pharmacology , Solanum tuberosum/chemistry , Animals , Antioxidants/chemistry , Gene Expression Regulation/drug effects , Male , NF-E2-Related Factor 2/genetics , Plant Proteins/chemistry , Rats , Rats, Inbred SHR , Rats, Inbred WKY
16.
Nutrients ; 11(7)2019 Jul 19.
Article in English | MEDLINE | ID: mdl-31331066

ABSTRACT

Vasicinone is a quinazoline alkaloid isolated from the Adhatoda vasica plant. In this study, we explored the neuroprotective effect and underlying molecular mechanism of vasicinone against paraquat-induced cellular apoptosis in SH-SY5Y cells. Vasicinone reduced the paraquat-induced loss of cell viability, rescued terminal deoxynucleotide transferase-mediated dUTP nick end-labeling (TUNEL)-positive apoptotic nuclei, and suppressed generation of reactive oxygen species (ROS) in a dose-dependent manner. Western blotting analysis revealed that vasicinone increased the phosphorylation of IGF1R/PI3K/AKT cell survival signaling molecules and downregulated the paraquat-induced, mitogen-activated protein kinase (MAPK)/c-Jun N-terminal kinase (JNK)-mediated apoptotic pathways compared to that observed in cells not treated with vasicinone. This protection depended critically on the activation of IGF1R, and the silencing of IGF1R by siRNA completely abrogated the protective effect of vasicinone in SH-SY5Y cells. Our findings indicated that vasicinone is a potential candidate for the treatment of Parkinson's disease and possibly other oxidative stress-related neurodegenerative disorders.


Subject(s)
Alkaloids/pharmacology , Apoptosis/drug effects , Paraquat/pharmacology , Parkinson Disease , Phosphatidylinositol 3-Kinases/metabolism , Receptor, IGF Type 1/metabolism , Cell Line , Cell Survival/drug effects , Humans , In Situ Nick-End Labeling , MAP Kinase Signaling System/drug effects , Neuroprotective Agents/pharmacology , Parkinson Disease/drug therapy , Parkinson Disease/metabolism , Parkinson Disease/pathology , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Reactive Oxygen Species/antagonists & inhibitors , Receptor, IGF Type 1/genetics , Signal Transduction/drug effects , Tumor Suppressor Protein p53/drug effects , Tumor Suppressor Protein p53/metabolism
17.
J Tissue Eng Regen Med ; 13(9): 1629-1640, 2019 09.
Article in English | MEDLINE | ID: mdl-31216096

ABSTRACT

Previous studies stated that stem cell functions are reduced under high glucose environment, leading to reduce stem cell capability of tissue regeneration. This study aimed to investigate if stem cells preconditioned with resveratrol show better therapeutic effect on the treatment of liver dysfunction in diabetic rats than stem cells without resveratrol precondition. Male Wistar rats were divided into four groups including sham, DM (diabetic rats), DM + ADSC (DM rats receiving autologous transplantation of adipose-derived stem cells), and DM + pre-R-ADSC (DM rats receiving ADSC preconditioned with resveratrol). Compared with sham group, experimental results showed that DM group induced suppression of survival, suppression of Sirt1, activation of apoptotic, and activation of fibrotic pathways, leading to liver dysfunction. Autologous transplantation of ADSC (DM + ADSC) improved above pathways except for fibrotic signaling. By contrast, transplantation of resveratrol preconditioned ADSC (DM + pre-R-ADSC) significantly improved above pathways including fibrosis. Supplemental evidences suggest that resveratrol precondition increases ADSC viability under high glucose stress via Sirt1 and IGF1R expressions. Furthermore, increased secretion of IGF1 via paracrine route also confirmed in ADSC preconditioned with resveratrol. The experimental results imply that ADSC preconditioned with resveratrol shows potential in the treatment of liver dysfunction in DM patients with liver dysfunction.


Subject(s)
Adipose Tissue/cytology , Diabetes Mellitus, Experimental/physiopathology , Diabetes Mellitus, Experimental/therapy , Liver Diseases/therapy , Protective Agents/therapeutic use , Resveratrol/therapeutic use , Stem Cell Transplantation , Stem Cells/cytology , Animals , Apoptosis , Cell Survival/drug effects , Clone Cells , Culture Media, Conditioned/pharmacology , Disease Models, Animal , Glucose/toxicity , Liver/drug effects , Liver/pathology , Liver/physiopathology , Liver Cirrhosis/pathology , Liver Cirrhosis/physiopathology , Male , Rats, Wistar , Resveratrol/pharmacology , Signal Transduction , Sirtuin 1/metabolism , Tissue Survival/drug effects , Transplantation, Autologous
18.
J Appl Physiol (1985) ; 127(2): 356-364, 2019 08 01.
Article in English | MEDLINE | ID: mdl-31095463

ABSTRACT

We previously reported that deep sea water (DSW) prolongs the life span of streptozotocin (STZ)-induced diabetic rats by the compensatory augmentation of the insulin like growth factor (IGF)-I survival signaling and inhibition of apoptosis. Here, we investigated the effects of DSW on cardiac hypertrophy in diabetic rats. Cardiac hypertrophy was induced in rats by using STZ (65 mg/kg) administered via IP injection. DSW was prepared by mixing DSW mineral extracts and desalinated water. Different dosages of DSW-1X (equivalent to 37 mg Mg2+·kg-1·day-1), 2X (equivalent to 74 mg Mg2+·kg-1·day-1) and 3X (equivalent to 111 mg Mg2+·kg-1·day-1) were administered to the rats through gavage for 4 wk. Cardiac hypertrophy was evaluated by the heart weight-to-body weight ratio and the cardiac tissue cross-sectional area after hematoxylin and eosin staining. The protein levels of the cardiac hypertrophy signaling molecules were determined by Western blot. Our results showed that the suppressive effects of the DSW treatment on STZ-induced cardiac hypertrophy were comparable to those of MgSO4 administration and that the hypertrophic marker brain natriuretic peptide (BNP) was decreased by DSW. In addition, DSW attenuated both the eccentric hypertrophy signaling pathway, IL-6-MEK-STAT3, and the concentric signaling pathway, IGF-II-PKCα-CaMKII, in DM rat hearts. The cardiac hypertrophy-associated activation of extracellular signal-regulated kinase (ERK) and the upregulation of the transcription factor GATA binding protein 4 (GATA4) were also negated by treatment with DSW. The results from this study suggest that DSW could be a potential therapeutic agent for the prevention and treatment of diabetic cardiac hypertrophy.NEW & NOTEWORTHY Deep sea water, containing high levels of minerals, improve cardiac hypertrophy in diabetic rats through attenuating the eccentric signaling pathway, IL-6-MEK5-STAT3, and concentric signaling pathway, IGF2-PKCα-CaMKII. The results from this study suggest that deep sea water could be a potential therapeutic agent for the prevention and treatment of diabetic cardiac hypertrophy.


Subject(s)
Cardiomegaly/metabolism , Diabetes Mellitus, Experimental/metabolism , Heart/physiopathology , Interleukin-6/metabolism , Receptor, IGF Type 2/metabolism , Signal Transduction/physiology , Animals , Apoptosis/physiology , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , MAP Kinase Signaling System/physiology , Male , Protein Kinase C-alpha/metabolism , Rats , Rats, Sprague-Dawley , STAT3 Transcription Factor/metabolism
19.
PLoS One ; 14(6): e0216285, 2019.
Article in English | MEDLINE | ID: mdl-31211784

ABSTRACT

Stress-induced cardiac hypertrophy leads to heart failure. Our previous studies demonstrate that insulin-like growth factor-II receptor (IGF-IIR) signaling is pivotal to hypertrophy regulation. In this study, we show a novel IGF-IIR alternative spliced transcript, IGF-IIRα (150 kDa) play a key role in high-salt induced hypertrophy mechanisms. Cardiac overexpression of IGF-IIRα and high-salt diet influenced cardiac dysfunction by increasing pathophysiological changes with up-regulation of hypertrophy markers, atrial natriuretic peptide (ANP) and brain natriuretic peptide (BNP). We found that, cardiac hypertrophy under high-salt conditions were amplified in the presence of IGF-IIRα overexpression. Importantly, high-salt induced angiotensin II type I receptor (AT1R) up regulation mediated IGF-IIR expressions via upstream mitogen activated protein kinase (MAPK)/silent mating type information regulation 2 homolog 1 (SIRT1)/heat shock factor 1 (HSF1) pathway. Further, G-coupled receptors (Gαq) activated calcineurin/nuclear factor of activated T-cells, cytoplasmic 3 (NFATc3)/protein kinase C (PKC) signaling was significantly up regulated under high-salt conditions. All these effects were observed to be dramatically over-regulated in IGF-IIRα transgenic rats fed with a high-salt diet. Altogether, from the findings, we demonstrate that IGF-IIRα plays a crucial role during high-salt conditions leading to synergistic cardiac hypertrophy.


Subject(s)
Cardiomegaly/pathology , Receptor, IGF Type 2/genetics , Sodium Chloride, Dietary/adverse effects , Alternative Splicing , Animals , Atrial Natriuretic Factor/metabolism , Cardiomegaly/chemically induced , Cardiomegaly/genetics , Cardiomegaly/veterinary , Female , MAP Kinase Signaling System , Male , Natriuretic Peptide, Brain/metabolism , Organ Specificity , Rats , Rats, Sprague-Dawley , Rats, Transgenic
20.
Biomed Pharmacother ; 97: 880-885, 2018 Jan.
Article in English | MEDLINE | ID: mdl-29136764

ABSTRACT

The insulin-like growth factor type 2 receptor (IGF2R) overexpression has been implicated in heart disease progression. Unregulated IGF2R signaling triggers cardiac hypertrophy, apoptosis, and cardiomyopathies. The present study investigated the role of IGF2R in cardiomyocyte apoptosis under high glucose (HG) levels and in streptozotocin (STZ) induced diabetic rat hearts. We found that IGF2 and IGF2R protein expression were highly upregulated under high glucose condition in H9c2 cells as well as in STZ induced diabetic rat hearts. Using immunoblotting and TUNEL assay, we found that elevated glucose condition induced IGF2R expression leads to activation of Gαq mediated calcineurin-dependent signaling pathway, which further leads to downstream activation and expression of cardiac hypertrophy related proteins, ANP and BNP. Further, we found that glucose-induced IGF2R expression downregulated survival protein p-Akt, p-Bad (Ser 155) and enhanced the expression of apoptosis-inducing proteins cytochrome c and cleaved Caspase-3. Our results suggested that hyperglycemic condition leads to cellular cardiomyocyte apoptosis both in vitro and in vivo models, via abnormally increased activation of the IGF2R signaling pathway.


Subject(s)
Apoptosis/physiology , Diabetes Mellitus, Experimental/physiopathology , Myocytes, Cardiac/pathology , Receptor, IGF Type 2/genetics , Animals , Cardiomegaly/physiopathology , Caspase 3/metabolism , Cell Line , Down-Regulation , Glucose/metabolism , Hyperglycemia/complications , In Situ Nick-End Labeling , Insulin-Like Growth Factor II/genetics , Insulin-Like Growth Factor II/metabolism , Male , Rats , Rats, Wistar , Receptor, IGF Type 2/metabolism , Signal Transduction , Streptozocin , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL