Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 215
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cell Mol Life Sci ; 77(22): 4663-4673, 2020 Nov.
Article in English | MEDLINE | ID: mdl-31894360

ABSTRACT

The androgen receptor (AR) plays an important role in the pathogenesis and development of prostate cancer (PCa). Mostly, PCa progresses to androgen-independent PCa, which has activated AR signaling from androgen-dependent PCa. Thus, inhibition of AR signaling may be an important therapeutic target in androgen-dependent and castration-resistant PCa. In this study, we determined the anticancer effect of a newly found natural compound, sakurasosaponin (S-saponin), using androgen-dependent and castration-resistant PCa cell lines. S-saponin induces mitochondrial-mediated cell death in both androgen-dependent (LNCaP) and castration-resistant (22Rv1 and C4-2) PCa cells, via AR expression. S-saponin treatment induces a decrease in AR expression in a time- and dose-dependent manner and a potent decrease in the expression of its target genes, including prostate-specific antigen (PSA), transmembrane protease, serin 2 (TMPRSS2), and NK3 homeobox 1 (NKX3.1). Furthermore, S-saponin treatment decreases B-cell lymphoma-extra large (Bcl-xL) and mitochondrial membrane potential, thereby increasing the release of cytochrome c into the cytosol. Moreover, Bcl-xL inhibition and subsequent mitochondria-mediated cell death caused by S-saponin were reversed by Bcl-xL or AR overexpression. Interestingly, S-saponin-mediated cell death was significantly reduced by a reactive oxygen species (ROS) scavenger, N-acetylcystein. Animal xenograft experiments showed that S-saponin treatment significantly reduced tumor growth of AR-positive 22Rv1 xenografts but not AR-negative PC-3 xenografts. Taken together, for the first time, our results revealed that S-saponin induces mitochondrial-mediated cell death in androgen-dependent and castration-resistant cells through regulation of AR mechanisms, including downregulation of Bcl-xL expression and induction of ROS stress by decreasing mitochondrial membrane potential.


Subject(s)
Antineoplastic Agents/poisoning , Cell Death/drug effects , Prostatic Neoplasms/drug therapy , Receptors, Androgen/metabolism , Saponins/pharmacology , Androgens/metabolism , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Disease Models, Animal , Down-Regulation/drug effects , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Male , Membrane Potential, Mitochondrial , Mice , Mice, Nude , PC-3 Cells , Prostate/drug effects , Prostate/metabolism , Prostate-Specific Antigen/metabolism , Prostatic Neoplasms/metabolism , Reactive Oxygen Species/metabolism , bcl-X Protein/metabolism
2.
Cell Physiol Biochem ; 52(3): 468-485, 2019.
Article in English | MEDLINE | ID: mdl-30873822

ABSTRACT

BACKGROUND/AIMS: Breast cancer is a clinically and molecularly heterogeneous disease. Patients with triple-negative breast cancer (TNBC) have poorer outcomes than those with other breast cancer subtypes due to lack of effective molecular targets for therapy. The present study aimed to the identification of estrogen receptor (ER)ß as a novel mitochondrial target in TNBC cells, together with underlying mechanisms. METHODS: Expression of ERß in clinical breast samples were examined by qRT-PCR, immunohistochemistry and immunoblotting. Subcellular distribution and binding of ERß-Grp75 was determined by confocal microscopic analysis, co-immunoprecipitation experiments, and limited-detergent extraction of subcellular organelles. The effect of mitocondrial ERß(mitoERß) overexpression on cell proliferation and cell cycle distribution were assessed CCK-8 assays and FACS. Mitochondrial ROS, membrane potential, and Ca²âº level were measured using the specific fluorescent probes Mito-Sox, TMRE, and Rhod-2AM. The tumorigenic effect of mitoERß overexpression was assessed using an anchorage-independent growth assay, sphere formation and a mouse orthotopic xenograft model. RESULTS: ERß expression was lower in tumor tissue than in adjacent normal tissue of patients with breast cancer, and low levels of mitochondrial ERß (mitoERß) also were associated with increased tumor recurrence after surgery. Overexpression of mitoERß inhibited the proliferation of TNBC cells and tumor masses in an animal model. Moreover, overexpression of mitoERß increased ATP production in TNBC cells and normal breast MCF10A cells, with the latter completely reversed by mitoERß knockdown in MCF10A cells. Grp75 was found to positively regulate ERß translocation into mitochondria via a direct interaction. Coimmunoprecipitation and subcellular fractionation experiments revealed that ERß-Grp75 complex is stable in mitochondria. CONCLUSION: These results suggest that the up-regulation of mitoERß in TNBC cells ensures proper mitochondrial transcription, activating the OXPHOS system to produce ATP. Studying the effects of mitoERß on mitochondrial activity and specific mitochondrial gene expression in breast cancer might help predict tumor recurrence, inform clinical decision-making, and identify novel drug targets in the treatment of TNBC.


Subject(s)
Adenosine Triphosphate/biosynthesis , Estrogen Receptor beta/genetics , Gene Expression Regulation, Neoplastic , HSP70 Heat-Shock Proteins/genetics , Mitochondria/metabolism , Mitochondrial Proteins/genetics , Triple Negative Breast Neoplasms/genetics , Animals , Calcium/metabolism , Cell Cycle , Cell Line, Tumor , Cell Proliferation , Estrogen Receptor beta/antagonists & inhibitors , Estrogen Receptor beta/metabolism , Female , Fluorescent Dyes/chemistry , HSP70 Heat-Shock Proteins/antagonists & inhibitors , HSP70 Heat-Shock Proteins/metabolism , Humans , Mice , Mitochondria/genetics , Mitochondria/pathology , Mitochondrial Proteins/antagonists & inhibitors , Mitochondrial Proteins/metabolism , Neoplasm Staging , Oxidative Phosphorylation , Protein Binding , Protein Transport , RNA, Small Interfering/biosynthesis , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction , Survival Analysis , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/mortality , Triple Negative Breast Neoplasms/pathology , Xenograft Model Antitumor Assays
3.
Pflugers Arch ; 470(2): 263-275, 2018 02.
Article in English | MEDLINE | ID: mdl-29032504

ABSTRACT

Metabolic disturbance and mitochondrial dysfunction are a hallmark of diabetic cardiomyopathy (DC). Resistance exercise (RE) not only enhances the condition of healthy individuals but could also improve the status of those with disease. However, the beneficial effects of RE in the prevention of DC and mitochondrial dysfunction are uncertain. Therefore, this study investigated whether RE attenuates DC by improving mitochondrial function using an in vivo rat model of diabetes. Fourteen Otsuka Long-Evans Tokushima Fatty rats were assigned to sedentary control (SC, n = 7) and RE (n = 7) groups at 28 weeks of age. Long-Evans Tokushima Otsuka rats were used as the non-diabetic control. The RE rats were trained by 20 repetitions of climbing a ladder 5 days per week. RE rats exhibited higher glucose uptake and lower lipid profiles, indicating changes in energy metabolism. RE rats significantly increased the ejection fraction and fractional shortening compared with the SC rats. Isolated mitochondria in RE rats showed increase in mitochondrial numbers, which were accompanied by higher expression of mitochondrial biogenesis proteins such as proliferator-activated receptor-γ coactivator-1α and TFAM. Moreover, RE rats reduced proton leakage and reactive oxygen species production, with higher membrane potential. These results were accompanied by higher superoxide dismutase 2 and lower uncoupling protein 2 (UCP2) and UCP3 levels in RE rats. These data suggest that RE is effective at ameliorating DC by improving mitochondrial function, which may contribute to the maintenance of diabetic cardiac contractility.


Subject(s)
Diabetic Cardiomyopathies/prevention & control , Energy Metabolism , Mitochondria, Muscle/metabolism , Myocardial Contraction , Physical Conditioning, Animal/methods , Animals , Diabetic Cardiomyopathies/physiopathology , Lipid Metabolism , Male , Rats , Rats, Long-Evans
4.
Gastroenterology ; 149(4): 1006-16.e9, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26091938

ABSTRACT

BACKGROUND & AIMS: Reagents designed to target cancer stem cells (CSCs) could reduce tumor growth, recurrence, and metastasis. We investigated the mitochondrial features of CSCs. METHODS: Colon adenocarcinoma fragments were obtained from 8 patients during surgery at Busan Paik Hospital in Korea. We used immunohistochemistry and quantitative polymerase chain reaction to compare expression of mitochondrial peroxiredoxin 3 (PRX3) in CD133(+)CD44(+) Lgr5(+)cells (CSCs) vs CD133(-)CD44(-)Lgr5(-) colon tumor cells (non-CSCs). Cell survival and expression of mitochondrial-related genes were analyzed in the presence of 5-fluorouracil and/or antimycin A. We used small-interfering and short-hairpin RNAs and an overexpression vector to study PRX3, which functions in the mitochondria. CD133(+) cells with PRX3 knockdown or overexpressing PRX3 were grown as xenograft tumors in immunocompromised mice. Metastasis was studied after injection of tumor cells in spleens of mice. We used chromatin immunoprecipitation and reporter assays to characterize transcriptional regulation of PRX3 by forkhead box protein 1. RESULTS: CSCs had a higher mitochondrial membrane potential and increased levels of adenosine triphosphate, Ca(2+), reactive oxygen species, and oxygen consumption than non-CSCs. Levels of PRX3 were increased in colon CSCs compared with non-CSCs. PRX3 knockdown reduced the viability of CSCs, but non non-CSCs, by inducing mitochondrial dysfunction. PRX3 knockdown reduced growth of CSCs as xenograft tumors or metastases in mice. The expression of FOXM1 activated transcription of PRX3 and expression of CD133 in colon CSCs. CONCLUSIONS: Human colon CSCs have increased mitochondrial function compared with colon tumor cells without stem cell properties. Colon CSCs overexpress the mitochondrial gene PRX3, which is required for maintenance of mitochondrial function and tumorigenesis, and is regulated by forkhead box protein 1, which also regulates expression of CD133 in these cells. These proteins might be therapeutic targets for colorectal cancer.


Subject(s)
Adenocarcinoma/metabolism , Antineoplastic Agents/pharmacology , Colonic Neoplasms/metabolism , Forkhead Transcription Factors/metabolism , Mitochondria/metabolism , Neoplastic Stem Cells/metabolism , Peroxiredoxin III/metabolism , AC133 Antigen , Adenocarcinoma/genetics , Adenocarcinoma/secondary , Adenocarcinoma/therapy , Adenosine Triphosphate/metabolism , Adult , Aged , Animals , Antigens, CD/genetics , Antigens, CD/metabolism , Calcium/metabolism , Cell Survival , Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , Colonic Neoplasms/therapy , Dose-Response Relationship, Drug , Drug Resistance, Neoplasm , Energy Metabolism , Female , Forkhead Box Protein M1 , Forkhead Transcription Factors/genetics , Gene Expression Regulation, Neoplastic , Glycoproteins/genetics , Glycoproteins/metabolism , HCT116 Cells , HT29 Cells , Humans , Membrane Potential, Mitochondrial , Mice, Inbred NOD , Mice, SCID , Middle Aged , Mitochondria/drug effects , Mitochondria/pathology , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/pathology , Oxygen Consumption , Peptides/genetics , Peptides/metabolism , Peroxiredoxin III/genetics , RNA Interference , RNAi Therapeutics , Reactive Oxygen Species/metabolism , Signal Transduction , Time Factors , Transcription, Genetic , Transfection , Xenograft Model Antitumor Assays
5.
Tumour Biol ; 37(11): 14577-14584, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27612479

ABSTRACT

Epithelial to mesenchymal transition (EMT) is a critical step in the metastasis of epithelial cancer cells. Butyrate, which is produced from dietary fiber by colonic bacterial fermentation, has been reported to influence EMT. However, some studies have reported that butyrate promotes EMT, while others have reported an inhibitory effect. To clarify these controversial results, it is necessary to elucidate the mechanism by which butyrate can influence EMT. In this study, we examined the potential role of annexin A1 (ANXA1), which was previously reported to promote EMT in breast cancer cells, as a mediator of EMT regulation by butyrate. We found that ANXA1 mRNA and protein were expressed in highly invasive melanoma cell lines (A2058 and A375), but not in SK-MEL-5 cells, which are less invasive. We also showed that butyrate induced ANXA1 mRNA and protein expression and promoted EMT-related cell invasion in SK-MEL-5 cells. Downregulation of ANXA1 expression using specific small interfering RNAs in butyrate-treated SK-MEL-5 cells resulted in increased expression of the epithelial marker E-cadherin and decreased cell invasion. Moreover, overexpressing ANXA1 decreased the expression of the E-cadherin. Collectively, these results indicate that butyrate induces the expression of ANXA1 in human melanoma cells, which then promotes invasion through activating the EMT signaling pathway.


Subject(s)
Annexin A1/biosynthesis , Butyrates/pharmacology , Cadherins/biosynthesis , Epithelial-Mesenchymal Transition/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Melanoma/pathology , Annexin A1/genetics , Cell Line, Tumor , Cell Movement/drug effects , Humans , Melanoma/genetics , Neoplasm Invasiveness/pathology , RNA Interference , RNA, Messenger/biosynthesis , RNA, Small Interfering/genetics , Skin Neoplasms , Up-Regulation/drug effects , Melanoma, Cutaneous Malignant
6.
Korean J Physiol Pharmacol ; 20(2): 201-11, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26937217

ABSTRACT

Although the antioxidant and cardioprotective effects of NecroX-5 on various in vitro and in vivo models have been demonstrated, the action of this compound on the mitochondrial oxidative phosphorylation system remains unclear. Here we verify the role of NecroX-5 in protecting mitochondrial oxidative phosphorylation capacity during hypoxia-reoxygenation (HR). Necrox-5 treatment (10 µM) and non-treatment were employed on isolated rat hearts during hypoxia/reoxygenation treatment using an ex vivo Langendorff system. Proteomic analysis was performed using liquid chromatography-mass spectrometry (LC-MS) and non-labeling peptide count protein quantification. Real-time PCR, western blot, citrate synthases and mitochondrial complex activity assays were then performed to assess heart function. Treatment with NecroX-5 during hypoxia significantly preserved electron transport chain proteins involved in oxidative phosphorylation and metabolic functions. NecroX-5 also improved mitochondrial complex I, II, and V function. Additionally, markedly higher peroxisome proliferator-activated receptor-gamma coactivator-1α (PGC1α) expression levels were observed in NecroX-5-treated rat hearts. These novel results provide convincing evidence for the role of NecroX-5 in protecting mitochondrial oxidative phosphorylation capacity and in preserving PGC1α during cardiac HR injuries.

7.
Pflugers Arch ; 467(10): 2151-63, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25410495

ABSTRACT

Echinochrome A (Ech A), a marine bio-product isolated from sea urchin eggs, is known to have cardioprotective effects through its strong antioxidant and ATP-sparing capabilities. However, the effects of Ech A on cardiac excitation-contraction (E-C) are not known. In this study, we investigated the effects of Ech A on cardiac contractility and Ca(2+) handling in the rat heart. In ex vivo Langendorff hearts, Ech A (3 µM) decreased left ventricular developing pressure to 77.7 ± 6.5 % of basal level. In isolated ventricular myocytes, Ech A reduced the fractional cell shortening from 3.4 % at baseline to 2.1 %. Ech A increased both diastolic and peak systolic intracellular Ca(2+) ([Ca(2+)]i). However, the ratio of peak [Ca]i to resting [Ca]i was significantly decreased. Ech A did not affect the L-type Ca(2+) current. Inhibiting the Na(+)/Ca(2+) exchanger with either NiCl2 or SEA400 did not affect the Ech A-dependent changes in Ca(2+) handling. Our data demonstrate that treatment with Ech A results in a significant reduction in the phosphorylation of phospholamban at both serine 16 and threonine 17 leading to a significant inhibition of SR Ca(2+)-ATPase 2A (SERCA2A) and subsequent reduced Ca(2+) uptake into the intracellular Ca(2+) store. Taken together, our data show that Ech A negatively regulates cardiac contractility by inhibiting SERCA2A activity, which leads to a reduction in internal Ca(2+) stores.


Subject(s)
Calcium Signaling , Calcium-Binding Proteins/metabolism , Cardiotonic Agents/pharmacology , Myocytes, Cardiac/metabolism , Naphthoquinones/pharmacology , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Animals , Cells, Cultured , Heart Ventricles/cytology , Heart Ventricles/drug effects , Heart Ventricles/metabolism , Male , Myocardial Contraction , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/physiology , Phosphorylation , Rats , Rats, Wistar , Serine/metabolism , Threonine/metabolism , Ventricular Function
8.
Stem Cells ; 32(4): 998-1011, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24715692

ABSTRACT

Redox balance has been suggested as an important determinant of "stemness" in embryonic stem cells (ESCs). In this study, we demonstrate that peroxiredoxin (Prx) plays a pivotal role in maintenance of ESC stemness during neurogenesis through suppression of reactive oxygen species (ROS)-sensitive signaling. During neurogenesis, Prx I and Oct4 are expressed in a mutually dependent manner and their expression is abruptly downregulated by an excess of ROS. Thus, in Prx I(-/-) or Prx II(-/-) ESCs, rapid loss of stemness can occur due to spontaneous ROS overload, leading to their active commitment into neurons; however, stemness is restored by the addition of an antioxidant or an inhibitor of c-Jun N-terminal kinase (JNK). In addition, Prx I and Prx II appear to have a tight association with the mechanism underlying the protection of ESC stemness in developing teratomas. These results suggest that Prx functions as a protector of ESC stemness by opposing ROS/JNK cascades during neurogenesis. Therefore, our findings have important implications for understanding of maintenance of ESC stemness through involvement of antioxidant enzymes and may lead to development of an alternative stem cell-based therapeutic strategy for production of high-quality neurons in large quantity.


Subject(s)
Embryonic Stem Cells/enzymology , MAP Kinase Kinase 4/metabolism , Neurogenesis/physiology , Peroxiredoxins/metabolism , Reactive Oxygen Species/metabolism , Animals , Embryonic Stem Cells/cytology , MAP Kinase Kinase 4/genetics , Mice , Mice, Knockout , Peroxiredoxins/genetics
9.
Cell Biol Int ; 39(7): 865-72, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25825022

ABSTRACT

Mutation or depletion of mitochondrial DNA (mtDNA) can cause severe mitochondrial malfunction, originating from the mitochondrion itself, or from the crosstalk between nuclei and mitochondria. However, the changes that would occur if the amount of mtDNA is diminished are less known. Thus, we generated rat myoblast H9c2 cells containing lower amounts of mtDNA via ethidium bromide and uridine supplementation. After confirming the depletion of mtDNA by quantitative PCR and gel electrophoresis analysis, we investigated the changes in mitochondrial physical parameters by using flow cytometry. We also evaluated the resistance of these cells to serum starvation and sodium nitroprusside. H9c2 cells with diminished mtDNA contents showed decreased mitochondrial membrane potential, mass, free calcium, and zinc ion contents as compared to naïve H9c2 cells. Furthermore, cytosolic and mitochondrial reactive oxygen species levels were significantly higher in mtDNA-lowered H9c2 cells than in the naïve cells. Although the oxygen consumption rate and cell proliferation were decreased, mtDNA-lowered H9c2 cells were more resistant to serum deprivation and nitroprusside insults than the naïve H9c2 cells. Taken together, we conclude that the low abundance of mtDNA cause changes in cellular status, such as changes in reactive oxygen species, calcium, and zinc ion levels inducing resistance to stress.


Subject(s)
DNA, Mitochondrial/genetics , Gene Dosage , Myocytes, Cardiac/metabolism , Nitroprusside/metabolism , Serum/metabolism , Animals , Cell Line , Cell Proliferation , Membrane Potential, Mitochondrial , Mitochondria/genetics , Mitochondria/metabolism , Myocytes, Cardiac/cytology , Oxidative Stress , Oxygen Consumption , Rats , Reactive Oxygen Species
10.
Mar Drugs ; 13(2): 1051-67, 2015 Feb 16.
Article in English | MEDLINE | ID: mdl-25690093

ABSTRACT

Fucoidan is an l-fucose-enriched sulfated polysaccharide isolated from brown algae and marine invertebrates. In this study, we investigated the protective effect of fucoidan from Fucus vesiculosus on alcohol-induced murine liver damage. Liver injury was induced by oral administration of 25% alcohol with or without fucoidan (30 mg/kg or 60 mg/kg) for seven days. Alcohol administration increased serum aspartate aminotransferase and alanine aminotransferase levels, but these increases were suppressed by the treatment of fucoidan. Transforming growth factor beta 1 (TGF-ß1), a liver fibrosis-inducing factor, was highly expressed in the alcohol-fed group and human hepatoma HepG2 cell; however, the increase in TGF-ß1 expression was reduced following fucoidan administration. Treatment with fucoidan was also found to significantly reduce the production of inflammation-promoting cyclooygenase-2 and nitric oxide, while markedly increasing the expression of the hepatoprotective enzyme, hemeoxygenase-1, on murine liver and HepG2 cells. Taken together, the antifibrotic and anti-inflammatory effects of fucoidan on alcohol-induced liver damage may provide valuable insights into developing new therapeutics or interventions.


Subject(s)
Fucus/chemistry , Hepatitis, Alcoholic/prevention & control , Inflammation Mediators/metabolism , Polysaccharides/pharmacology , Alanine Transaminase/blood , Animals , Aspartate Aminotransferases/blood , Cell Line , Cyclooxygenase 2/metabolism , Cyclooxygenase 2 Inhibitors/pharmacology , Heme Oxygenase-1/biosynthesis , Hep G2 Cells , Hepatitis, Alcoholic/metabolism , Humans , Male , Mice , Mice, Inbred BALB C , Nitric Oxide/antagonists & inhibitors , Nitric Oxide/biosynthesis , Organ Size/drug effects , Polysaccharides/isolation & purification , Transforming Growth Factor beta1/metabolism
11.
Korean J Physiol Pharmacol ; 19(5): 389-99, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26330751

ABSTRACT

Zinc has been considered as a vital constituent of proteins, including enzymes. Mobile reactive zinc (Zn(2+)) is the key form of zinc involved in signal transductions, which are mainly driven by its binding to proteins or the release of zinc from proteins, possibly via a redox switch. There has been growing evidence of zinc's critical role in cell signaling, due to its flexible coordination geometry and rapid shifts in protein conformation to perform biological reactions. The importance and complexity of Zn(2+) activity has been presumed to parallel the degree of calcium's participation in cellular processes. Whole body and cellular Zn(2+) levels are largely regulated by metallothioneins (MTs), Zn(2+) importers (ZIPs), and Zn(2+) transporters (ZnTs). Numerous proteins involved in signaling pathways, mitochondrial metabolism, and ion channels that play a pivotal role in controlling cardiac contractility are common targets of Zn(2+). However, these regulatory actions of Zn(2+) are not limited to the function of the heart, but also extend to numerous other organ systems, such as the central nervous system, immune system, cardiovascular tissue, and secretory glands, such as the pancreas, prostate, and mammary glands. In this review, the regulation of cellular Zn(2+) levels, Zn(2+)-mediated signal transduction, impacts of Zn(2+) on ion channels and mitochondrial metabolism, and finally, the implications of Zn(2+) in health and disease development were outlined to help widen the current understanding of the versatile and complex roles of Zn(2+).

12.
Pflugers Arch ; 466(12): 2323-38, 2014 Dec.
Article in English | MEDLINE | ID: mdl-24658911

ABSTRACT

B7-H4 is a B7 family coregulatory protein that inhibits T cell-mediated immunity. B7-H4 is overexpressed in various cancers; however, the functional role of B7-H4 in cancer metabolism is poorly understood. Because mitochondria play pivotal roles in development, proliferation, and death of cancer cells, we investigated molecular and functional alterations of mitochondria in B7-H4-depleted HeLa cells. In a human study, overexpression of B7-H4 was confirmed in the cervices of adenocarcinoma patients (n = 3) compared to noncancer patients (n = 3). In the cell line model, B7-H4 depletion was performed by transfection with small interfering RNA (siRNA). B7-H4 depletion suppressed oxygen consumption rate, ATP production, and mitochondrial membrane potential and mass and increased reactive oxygen species production. In particular, electron transport complex III activity was significantly impaired in siB7-H4-treated cells. Coincidently, depletion of B7-H4 suppressed major mitochondrial regulators (peroxisome proliferator-activated receptor gamma coactivator 1-alpha [PGC1-α] and mitochondrial transcription factor A), a component of oxidative phosphorylation (ubiquinol-cytochrome c reductase core protein 1), and an antiapoptosis protein (Bcl-XL). Mitochondrial dysfunction in siRNA-treated cells significantly augmented oxidative stress, which strongly activated the JNK/P38/caspase axis in the presence of doxorubicin, resulting in increased apoptotic cell death. Investigating the mechanism of B7-H4-mediated mitochondrial modulation, we found that B7-H4 depletion significantly downregulated the cAMP/cAMP response element-binding protein/PGC1-α signaling pathway. Based on these findings, we conclude that B7-H4 has a role in the regulation of mitochondrial function, which is closely related to cancer cell physiology and drug sensitivity.


Subject(s)
Adenocarcinoma/metabolism , Down-Regulation , Mitochondria/metabolism , Signal Transduction , Uterine Cervical Neoplasms/metabolism , V-Set Domain-Containing T-Cell Activation Inhibitor 1/metabolism , Aged , Antibiotics, Antineoplastic/pharmacology , Apoptosis , Cyclic AMP/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , Doxorubicin/pharmacology , Female , HeLa Cells , Humans , Middle Aged , Mitochondria/drug effects , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Transcription Factors/metabolism , V-Set Domain-Containing T-Cell Activation Inhibitor 1/genetics
13.
Mar Drugs ; 12(8): 4602-15, 2014 Aug 21.
Article in English | MEDLINE | ID: mdl-25196935

ABSTRACT

Echinochrome A (Ech A) is a natural pigment from sea urchins that has been reported to have antioxidant properties and a cardio protective effect against ischemia reperfusion injury. In this study, we ascertained whether Ech A enhances the mitochondrial biogenesis and oxidative phosphorylation in rat cardio myoblast H9c2 cells. To study the effects of Ech A on mitochondrial biogenesis, we measured mitochondrial mass, level of oxidative phosphorylation, and mitochondrial biogenesis regulatory gene expression. Ech A treatment did not induce cytotoxicity. However, Ech A treatment enhanced oxygen consumption rate and mitochondrial ATP level. Likewise, Ech A treatment increased mitochondrial contents in H9c2 cells. Furthermore, Ech A treatment up-regulated biogenesis of regulatory transcription genes, including proliferator-activated receptor gamma co-activator (PGC)-1α, estrogen-related receptor (ERR)-α, peroxisome proliferator-activator receptor (PPAR)-γ, and nuclear respiratory factor (NRF)-1 and such mitochondrial transcription regulatory genes as mitochondrial transcriptional factor A (TFAM), mitochondrial transcription factor B2 (TFB2M), mitochondrial DNA direct polymerase (POLMRT), single strand binding protein (SSBP) and Tu translation elongation factor (TUFM). In conclusion, these data suggest that Ech A is a potentiated marine drug which enhances mitochondrial biogenesis.


Subject(s)
Mitochondria/drug effects , Mitochondria/genetics , Mitochondrial Turnover/drug effects , Mitochondrial Turnover/genetics , Myoblasts, Cardiac/drug effects , Naphthoquinones/pharmacology , Adenosine Triphosphate/metabolism , Animals , Cell Line , DNA, Mitochondrial/genetics , Mitochondria/metabolism , Myoblasts, Cardiac/metabolism , Oxidative Phosphorylation/drug effects , Oxygen Consumption/drug effects , Rats , Transcription, Genetic/drug effects , Transcription, Genetic/genetics , Up-Regulation/drug effects , Up-Regulation/genetics
14.
Mar Drugs ; 12(5): 2922-36, 2014 May 13.
Article in English | MEDLINE | ID: mdl-24828295

ABSTRACT

Echinochrome A (Ech A) is a naphthoquinoid pigment from sea urchins that possesses antioxidant, antimicrobial, anti-inflammatory and chelating abilities. Although Ech A is the active substance in the ophthalmic and cardiac drug Histochrome®, its underlying cardioprotective mechanisms are not well understood. In this study, we investigated the protective role of Ech A against toxic agents that induce death of rat cardiac myoblast H9c2 cells and isolated rat cardiomyocytes. We found that the cardiotoxic agents tert-Butyl hydroperoxide (tBHP, organic reactive oxygen species (ROS) inducer), sodium nitroprusside (SNP; anti-hypertension drug), and doxorubicin (anti-cancer drug) caused mitochondrial dysfunction such as increased ROS level and decreased mitochondrial membrane potential. Co-treatment with Ech A, however, prevented this decrease in membrane potential and increase in ROS level. Co-treatment of Ech A also reduced the effects of these cardiotoxic agents on mitochondrial oxidative phosphorylation and adenosine triphosphate level. These findings indicate the therapeutic potential of Ech A for reducing cardiotoxic agent-induced damage.


Subject(s)
Cardiotonic Agents/pharmacology , Cardiotoxins/antagonists & inhibitors , Mitochondria, Heart/drug effects , Myocytes, Cardiac/drug effects , Naphthoquinones/pharmacology , Adenosine Triphosphate/metabolism , Animals , Cardiotoxins/toxicity , Cell Death/drug effects , Membrane Potential, Mitochondrial/drug effects , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinases/metabolism , Rats , Reactive Oxygen Species/metabolism , Sea Urchins
15.
J Korean Med Sci ; 29(12): 1639-45, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25469063

ABSTRACT

Gastric cancer is one of the most common cancers, especially among the elderly. However little is known about gastric cancer in elderly patients. This study was designed to evaluate the specific features of gastric cancer in elderly patients. Medical records of 1,107 patients who had radical gastrectomy for gastric cancer between June 2005 and December 2009 were reviewed. They were divided into young (<65 yr, n=676), young-old (65-74 yr, n=332), and old-old age group (≥75 yr, n=99). Increased CA 19-9 (5.6%, 13.4%, 14.6%, P=0.001), advanced diseases (42.5%, 47.0%, and 57.6, P=0.014), and node metastasis (37.6%, 38.9%, 51.5%, P=0.029) were more common in the young-old and old-old age groups. There were no significant differences in Helicobacter pylori status (63.6%, 56.7%, 61.2%, P=0.324) between the three groups. Surgery-related complication rates were similar in the three groups (5.3%, 5.1%, 8.1%, P=0.497). Microsatellite instability (P<0.001) and p53 overexpression (P<0.001) were more common among the elderly. The elderly group had more synchronous tumors (7.5%, 10.2%, 17.2%; P=0.006). Surgery can be applied to elderly gastric cancer without significant risk of complications. However, considering the more advanced disease and synchronous tumors among the elderly, care should be taken while deciding the extent of surgery for elderly gastric cancer.


Subject(s)
Gastrectomy/statistics & numerical data , Postoperative Complications/epidemiology , Stomach Neoplasms/epidemiology , Stomach Neoplasms/surgery , Adult , Age Distribution , Aged , Aged, 80 and over , Causality , Comorbidity , Female , Humans , Male , Middle Aged , Patient Safety , Prevalence , Republic of Korea/epidemiology , Risk Factors , Sex Distribution , Treatment Outcome
16.
Gastroenterology ; 143(5): 1341-1351, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22841785

ABSTRACT

BACKGROUND & AIMS: The TOR signaling pathway regulator-like (TIPRL) protein, the mammalian ortholog of yeast TIP41, was identified in an expression profiling screen for factors that regulate human liver carcinogenesis. We investigated the role of human TIPRL protein in hepatocellular carcinoma (HCC). METHODS: We measured the level of TIPRL in HCC and adjacent nontumor tissues from patients. We used small interfering RNAs and zebrafish to study the function of TIPRL. We used annexin V propidium iodide staining and immunoblot analyses to measure apoptosis and activation of apoptotic signaling pathways. We used confocal microscopy, coimmunoprecipitation, and glutathione-S transferase pull-down analyses to determine interactions among mitogen-activated protein kinase kinase 7 (MKK7 or MAP2K7), TIPRL, and the protein phosphatase type 2A (PP2Ac). We studied the effects of TIPRL in tumor xenografts in mice. RESULTS: Levels of TIPRL were higher in HCC tissues and cell lines than nontumor tissues and primary hepatocytes. Knockdown of tiprl expression in zebrafish led to large amounts of apoptosis throughout the embryos. Incubation of HCC cells, but not primary human hepatocytes, with small interfering RNA against TIPRL (siTIPRL) and tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) caused prolonged activation (phosphorylation) of MKK7 and c-Jun N-terminal kinase (JNK) and led to apoptosis, indicated by cleavage of procaspase-8,-3 and of poly-(adenosine diphosphate-ribose) polymerase. TIPRL bound to MKK7 and PP2Ac and promoted the interaction between MKK7 and PP2Ac. In mice, injection of HCC xenograft tumors with siTIPRL and TRAIL led to tumor apoptosis and regression. CONCLUSIONS: TIPRL is highly up-regulated in human HCC samples and cell lines, compared with noncancerous liver tissues. TIPRL prevents prolonged activation of MKK7 and JNK and TRAIL-induced apoptosis by mediating the interaction between MKK7 and PP2Ac.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Liver Neoplasms/metabolism , MAP Kinase Kinase 7/metabolism , Animals , Apoptosis , Carcinoma, Hepatocellular/genetics , Female , Gene Knockdown Techniques , Hep G2 Cells , Hepatocytes/metabolism , Humans , Intracellular Signaling Peptides and Proteins/genetics , Liver/metabolism , Liver Neoplasms/genetics , Mice , Mice, Nude , Neoplasm Transplantation , Protein Phosphatase 2/metabolism , RNA, Messenger/metabolism , RNA, Small Interfering , Signal Transduction , TNF-Related Apoptosis-Inducing Ligand/metabolism , Up-Regulation , Zebrafish/embryology , Zebrafish/genetics , Zebrafish/metabolism
17.
Helicobacter ; 18(3): 180-6, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23305083

ABSTRACT

BACKGROUND: The eradication rate with PPI-based standard triple therapy for Helicobacter pylori infection has fallen considerably. One recent innovation is sequential therapy with PPI and three antibiotics, but the complexity of this regimen may reduce its usability. Concomitant administration of nonbismuth quadruple drugs (concomitant therapy) is also an effective treatment strategy. To investigate which regimen is a reasonable choice for Korean population, we performed two pilot studies with sequential and concomitant therapies. METHODS: A total of 164 patients with proven H. pylori infection randomly received 14 days of sequential (n = 86) or concomitant (n = 78) therapies. The sequential group received 20 mg rabeprazole and 1 g amoxicillin (first week), followed by 20 mg rabeprazole, 500 mg clarithromycin, and 500 mg metronidazole (second week). The concomitant group received 20 mg rabeprazole, 1 g amoxicillin, 500 mg clarithromycin, and 500 mg metronidazole for 2 weeks. All drugs were administered BID. Helicobacter pylori status was confirmed 4 weeks later, after completion of treatment by (13) C-urea breath test. RESULTS: The intention-to-treat and per-protocol eradication rates were 75.6% (95% CI, 66.3-84.9) and 76.8% (95% CI, 67.1-85.5) in the sequential group, and 80.8% (95% CI, 71.8-88.5) and 81.3% (95% CI, 71.6-90.7) in the concomitant group. There were no significant between-group differences, in regard to the eradication rates, compliance, or side effects. The most common side effects were bitter taste, epigastric soreness, and diarrhea. CONCLUSIONS: Two-week concomitant and sequential therapies showed suboptimal efficacies. However, considering high antibiotics resistance, either of these two regimens may be a reasonable choice for Korean population.


Subject(s)
2-Pyridinylmethylsulfinylbenzimidazoles/therapeutic use , Anti-Bacterial Agents/therapeutic use , Helicobacter Infections/drug therapy , Helicobacter pylori/drug effects , Proton Pump Inhibitors/therapeutic use , 2-Pyridinylmethylsulfinylbenzimidazoles/administration & dosage , 2-Pyridinylmethylsulfinylbenzimidazoles/adverse effects , Aged , Amoxicillin/administration & dosage , Amoxicillin/adverse effects , Amoxicillin/therapeutic use , Anti-Bacterial Agents/administration & dosage , Anti-Bacterial Agents/adverse effects , Anti-Bacterial Agents/pharmacology , Anti-Ulcer Agents/adverse effects , Anti-Ulcer Agents/pharmacology , Anti-Ulcer Agents/therapeutic use , Breath Tests , Clarithromycin/administration & dosage , Clarithromycin/adverse effects , Drug Administration Schedule , Drug Therapy, Combination , Female , Helicobacter Infections/microbiology , Humans , Male , Metronidazole/administration & dosage , Metronidazole/adverse effects , Metronidazole/therapeutic use , Middle Aged , Pilot Projects , Proton Pump Inhibitors/administration & dosage , Proton Pump Inhibitors/adverse effects , Proton Pump Inhibitors/pharmacology , Rabeprazole , Treatment Outcome , Urea/therapeutic use
18.
Bioorg Med Chem Lett ; 23(14): 4225-9, 2013 Jul 15.
Article in English | MEDLINE | ID: mdl-23735745

ABSTRACT

Resveratrol is known to exert a cardioprotective effect against hypoxia/reoxygenation (H/R) injury. HS-1793 is a novel, more stable resveratrol analog, but its cardioprotective effects were unknown. The present study aimed to test the cardioprotective effect of HS-1793 against H/R injury and investigate the role of mitochondria in Sprague Dawley rat heart damage using an ex vivo Langendorff system. HS-1793 ameliorated H/R-induced mitochondrial dysfunction by reducing mitochondrial reactive oxygen species production, improving mitochondrial oxygen consumption and suppressing mitochondrial calcium (Ca(2+)) overload during reperfusion. Moreover, HS-1793-treated rat heart showed reduced infarct size. Our data suggest that HS-1793 can protect cardiac against mitochondrial damage following H/R, thereby suppressing injury.


Subject(s)
Naphthols/chemistry , Resorcinols/chemistry , Stilbenes/chemistry , Animals , Calcium/metabolism , Heart/physiopathology , Hypoxia , Mitochondria/drug effects , Mitochondria/metabolism , Myocardial Reperfusion Injury/drug therapy , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/pathology , Naphthols/pharmacology , Naphthols/therapeutic use , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Resorcinols/pharmacology , Resorcinols/therapeutic use , Resveratrol
19.
J Pathol ; 226(4): 654-65, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22252584

ABSTRACT

Aberrant DNA methylation is frequently found during gastric carcinogenesis. Recently, we identified potential methylation markers important for Helicobacter pylori-induced gastric carcinogenesis using an Illumina methylation chip assay. In this study, we evaluated the candidate genes as markers for gastric cancer (GC) in a large Korean population. DNA methylation of PTPN6, MOS, DCC, CRK, and VAV1 was evaluated in non-neoplastic gastric specimens using quantitative methylation-specific PCR in patients with GC (n = 207) and their age- and gender-matched controls (n = 207). Methylation levels in 125 GC samples were also compared. H. pylori infection status was categorized as negative, active, or past infection according to the results of endoscopy-based tests (CLOtest, histology, and culture), H. pylori serology, and serum pepsinogen test. In the controls, active H. pylori infection increased methylation levels in DCC, CRK, MOS, and VAV1 but decreased methylation levels in PTPN6 (all p < 0.05); the methylation levels in MOS remained increased in patients with past H. pylori infection compared to H. pylori-negative subjects (p < 0.001). Methylation levels in MOS in non-neoplastic gastric mucosae increased in the presence of GC, regardless of H. pylori infection status (p < 0.01). Methylation levels in all genes but DCC decreased significantly in GC specimens compared to neoplastic gastric mucosae (p < 0.01); however, methylation levels in GC tissues were not correlated with those in their background gastric mucosae. Hypomethylation of MOS in GC tissues was associated with tumour invasion, nodal metastasis, and undifferentiated histology (p < 0.05). To summarize, among the candidate genes, DNA methylation of MOS may reflect the duration of H. pylori exposure and may be a marker for the development of GC.


Subject(s)
DNA Methylation , Gastric Mucosa/pathology , Gastritis/pathology , Genetic Predisposition to Disease , Precancerous Conditions/pathology , Stomach Neoplasms/pathology , Female , Gastric Mucosa/microbiology , Gastritis/genetics , Gastritis/microbiology , Genes, mos/genetics , Helicobacter Infections/genetics , Helicobacter Infections/microbiology , Helicobacter Infections/pathology , Helicobacter pylori , Humans , Male , Middle Aged , Precancerous Conditions/genetics , Precancerous Conditions/microbiology , Predictive Value of Tests , Risk , Stomach Neoplasms/genetics , Stomach Neoplasms/microbiology
20.
Hepatogastroenterology ; 60(124): 776-80, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23165228

ABSTRACT

BACKGROUND/AIMS: Although some studies have shown improvement of precancerous lesions and a decrease of metachronous gastric cancer after eradication of H. pylori, this is still controversial. METHODOLOGY: We identified 74 patients with early gastric cancer and who had their H. pylori eradicated after undergoing endoscopic resection between September, 2003 and September, 2010. The endoscopic biopsy specimens, campylobacter-like organism test and urea breath test were reviewed. Relapse of gastric cancer was assessed from medical records. RESULTS: Among the 74 patients, 61 (82.4%) were successfully eradicated. The mean duration of follow-up was 27.2±18.7 months. H. pylori colonization, neutrophil infiltration, mononuclear cell infiltration and intestinal metaplasia decreased after eradication (all p<0.05). For all the patients, metachronous gastric cancer showed a decrease in the eradicated group, but this did not reach statistical significance (odds ratio: 0.36, 95% CI: 0.08-1.70, p=0.189). However, when restricted to those who were followed-up for more than 18 months, metachronous gastric cancer was significantly decreased in the eradicated group (odds ratio: 0.108, 95% CI: 0.016-0.726, p=0.035). CONCLUSIONS: Eradication of H. pylori decreased precancerous lesions, and when following-up for more than 18 months, eradication also reduced metachronous gastric cancer.


Subject(s)
Gastroscopy , Helicobacter Infections/prevention & control , Helicobacter pylori , Neoplasms, Second Primary/prevention & control , Precancerous Conditions/prevention & control , Stomach Neoplasms/microbiology , Stomach Neoplasms/surgery , Biopsy , Breath Tests , Female , Humans , Male , Middle Aged , Neoplasm Recurrence, Local/pathology , Neoplasms, Second Primary/pathology , Precancerous Conditions/pathology , Retrospective Studies , Stomach Neoplasms/pathology , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL