Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
Cell Mol Life Sci ; 80(3): 69, 2023 Feb 23.
Article in English | MEDLINE | ID: mdl-36821008

ABSTRACT

Animal models have been utilized to understand the pathogenesis of Zellweger spectrum disorders (ZSDs); however, the link between clinical manifestations and molecular pathways has not yet been clearly established. We generated peroxin 5 homozygous mutant zebrafish (pex5-/-) to gain insight into the molecular pathogenesis of peroxisome dysfunction. pex5-/- display hallmarks of ZSD in humans and die within one month after birth. Fasting rapidly depletes lipids and glycogen in pex5-/- livers and expedites their mortality. Mechanistically, deregulated mitochondria and mechanistic target of rapamycin (mTOR) signaling act together to induce metabolic alterations that deplete hepatic nutrients and accumulate damaged mitochondria. Accordingly, chemical interventions blocking either the mitochondrial function or mTOR complex 1 (mTORC1) or a combination of both improve the metabolic imbalance shown in the fasted pex5-/- livers and extend the survival of animals. In addition, the suppression of oxidative stress by N-acetyl L-cysteine (NAC) treatment rescued the apoptotic cell death and early mortality observed in pex5-/-. Furthermore, an autophagy activator effectively ameliorated the early mortality of fasted pex5-/-. These results suggest that fasting may be detrimental to patients with peroxisome dysfunction, and that modulating the mitochondria, mTORC1, autophagy activities, or oxidative stress may provide a therapeutic option to alleviate the symptoms of peroxisomal diseases associated with metabolic dysfunction.


Subject(s)
Fasting , Mitochondria , Peroxisome-Targeting Signal 1 Receptor , Zebrafish , Animals , Humans , Autophagy/physiology , Mechanistic Target of Rapamycin Complex 1/metabolism , Mitochondria/metabolism , Peroxisomes/metabolism , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism , Zebrafish/genetics , Zebrafish/metabolism , Peroxisome-Targeting Signal 1 Receptor/genetics , Peroxisome-Targeting Signal 1 Receptor/metabolism
2.
J Biochem ; 173(1): 53-63, 2022 Dec 27.
Article in English | MEDLINE | ID: mdl-36270274

ABSTRACT

The Nudt family has been identified as enzymes performing Coenzyme A to 3'5'-ADP + 4'-phospho pantetheine catalysis. The members of this family have been shown to be particularly involved in lipid metabolism, while their involvement in gene regulation through regulating transcription or mRNA metabolism has also been suggested. Here, we focused on peroxisomal NUDT7, possessing enzymatic activity similar to that of its paralog, peroxisomal NUDT19, which is involved in mRNA degradation. No reports have been published about the Nudt family in zebrafish. Our transcriptomic data showed that the Nudt family members are highly expressed around zygotic gene activation (ZGA) in developing zebrafish embryos. Therefore, we confirmed the computational prediction that the products of the nudt7 gene in zebrafish were localized in the peroxisome and highly expressed in early embryogenesis. The depletion of nudt7 genes by the CRISPR/Cas9 system did not affect development; however, it decreased the rate of transcription in ZGA. In addition, H3K27ac ChIP-seq analysis demonstrated that this decrease in transcription was correlated with the genome-wide decrease of H3K27ac level. This study suggests that peroxisomal Nudt7 functions in regulating transcription in ZGA via formation of the H3K27ac domain in active chromatin.


Subject(s)
Transcriptome , Zebrafish , Animals , Zebrafish/genetics , Chromatin , Genome , Gene Expression Profiling
3.
Article in English | MEDLINE | ID: mdl-34517131

ABSTRACT

Non-shivering thermogenesis (NST) is a heat generating process controlled by the mitochondria of brown adipose tissue (BAT). In the recent decade, 'functionally' acting brown adipocytes in white adipose tissue (WAT) has been identified as well: the so-called process of the 'browning' of WAT. While the importance of uncoupling protein 1 (UCP1)-oriented mitochondrial activation has been intensely studied, the role of peroxisomes during the browning of white adipocytes is poorly understood. Here, we assess the change in peroxisomal membrane proteins, or peroxins (PEXs), during cold stimulation and importantly, the role of PEX13 in the cold-induced remodeling of white adipocytes. PEX13, a protein that originally functions as a docking factor and is involved in protein import into peroxisome matrix, was highly increased during cold-induced recruitment of beige adipocytes within the inguinal WAT of C57BL/6 mice. Moreover, beige-induced 3 T3-L1 adipocytes and stromal vascular fraction (SVF) cells by exposure to the peroxisome proliferator-activated receptor gamma (PPARγ) agonist rosiglitazone showed a significant increase in mitochondrial thermogenic factors along with peroxisomal proteins including PEX13, and these were confirmed in SVF cells with the beta 3 adrenergic receptor (ß3AR)-selective agonist CL316,243. To verify the relevance of PEX13, we used the RNA silencing method targeting the Pex13 gene and evaluated the subsequent beige development in SVF cells. Interestingly, siPex13 treatment suppressed expression of thermogenic proteins such as UCP1 and PPARγ coactivator 1 alpha (PGC1α). Overall, our data provide evidence supporting the role of peroxisomal proteins, in particular PEX13, during beige remodeling of white adipocytes.


Subject(s)
Adipose Tissue, White/metabolism , Membrane Proteins/genetics , PPAR gamma/genetics , Thermogenesis/genetics , Uncoupling Protein 1/genetics , 3T3-L1 Cells , Adipose Tissue, Brown/metabolism , Adrenergic beta-3 Receptor Agonists/pharmacology , Animals , Dioxoles/pharmacology , Mice , Mitochondria/genetics , Mitochondria/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , Peroxisomes/genetics , RNA Interference , Receptors, Adrenergic, beta-3/genetics , Stromal Vascular Fraction/genetics , Stromal Vascular Fraction/metabolism
4.
Int J Mol Sci ; 22(8)2021 Apr 13.
Article in English | MEDLINE | ID: mdl-33924375

ABSTRACT

Zebrafish have become a popular animal model for studying various biological processes and human diseases. The metabolic pathways and players conserved among zebrafish and mammals facilitate the use of zebrafish to understand the pathological mechanisms underlying various metabolic disorders in humans. Adipocytes play an important role in metabolic homeostasis, and zebrafish adipocytes have been characterized. However, a versatile and reliable zebrafish model for long-term monitoring of adipose tissues has not been reported. In this study, we generated stable transgenic zebrafish expressing enhanced green fluorescent protein (EGFP) in adipocytes. The transgenic zebrafish harbored adipose tissues that could be detected using GFP fluorescence and the morphology of single adipocyte could be investigated in vivo. In addition, we demonstrated the applicability of this model to the long-term in vivo imaging of adipose tissue development and regulation based on nutrition. The transgenic zebrafish established in this study may serve as an excellent tool to advance the characterization of white adipose tissue in zebrafish, thereby aiding the development of therapeutic interventions to treat metabolic diseases in humans.


Subject(s)
Adipocytes/cytology , Adipocytes/metabolism , Zebrafish/genetics , Zebrafish/metabolism , Adipose Tissue/metabolism , Animal Nutritional Physiological Phenomena , Animals , Animals, Genetically Modified , Cell Shape , Green Fluorescent Proteins/metabolism , Larva/genetics , Larva/metabolism , Promoter Regions, Genetic/genetics , Transgenes , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
5.
J Cell Physiol ; 235(1): 151-165, 2020 01.
Article in English | MEDLINE | ID: mdl-31187491

ABSTRACT

Slc25a17 is known as a peroxisomal solute carrier, but the in vivo role of the protein has not been demonstrated. We found that the zebrafish genome contains two slc25a17 genes that function redundantly, but additively. Notably, peroxisome function in slc25a17 knockdown embryos is severely compromised, resulting in an altered lipid composition. Along the defects found in peroxisome-associated phenotypic presentations, we highlighted that development of the swim bladder is also highly dependent on Slc25a17 function. As Slc25a17 showed substrate specificity towards coenzyme A (CoA), injecting CoA, but not NAD+ , rescued the defective swim bladder induced by slc25a17 knockdown. These results indicated that Slc25a17 acts as a CoA transporter, involved in the maintenance of functional peroxisomes that are essential for the development of multiple organs during zebrafish embryogenesis. Given high homology in protein sequences, the role of zebrafish Slc25a17 may also be applicable to the mammalian system.


Subject(s)
Coenzyme A/metabolism , Gene Expression Regulation, Developmental/physiology , Membrane Proteins/metabolism , Air Sacs/growth & development , Air Sacs/metabolism , Amino Acid Sequence , Animals , Coenzyme A/genetics , Conserved Sequence , Evolution, Molecular , Membrane Proteins/genetics , Zebrafish
6.
Biochem Biophys Res Commun ; 514(4): 1264-1269, 2019 07 05.
Article in English | MEDLINE | ID: mdl-31113616

ABSTRACT

ABCD4, a member of the ATP-binding cassette transporter superfamily, is associated with the transport of vitamin B12 which is crucial for the development of red blood cells (RBCs) and may also be involved in its metabolism. However, the molecular function of ABCD4 during RBC development in zebrafish is mostly unknown. Using a morpholino-based knockdown approach, we found that abcd4-knockdown resulted in abnormal RBCs of irregular shapes and various sizes. o-Dianisidine staining, as an indicator of hemoglobin in RBCs, further confirmed that abcd4 morphants possessed fewer hemoglobinized cells and impaired blood circulation. Multiple protein sequence alignment revealed that the amino acid sequence for residues 13-292, which is the domain of vitamin B12 transport, of the zebrafish Abcd4 was highly conserved compared to that of other species. Accordingly, the abcd4 morphants can be rescued with human ABCD4, demonstrating a conserved role of ABCD4 in vertebrates. Notably, the vitamin B12-deficient phenotype in abcd4 morphants, which causes anemia, was recapitulated in the newly-established abcd4 mutant, indicating the possibility that the abcd4 mutant could be used as a disease model of vitamin B12-deficiency anemia. Our study provides an insight that the analysis of the newly-established abcd4 mutant may contribute to understanding its roles in ABCD4-related vitamin B12-deficiency anemia and the associated pathogeneses in humans.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Anemia/metabolism , Vitamin B 12 Deficiency/metabolism , ATP-Binding Cassette Transporters/deficiency , ATP-Binding Cassette Transporters/genetics , Animals , Mutation , Zebrafish
7.
Exp Mol Med ; 50(4): 1-12, 2018 04 06.
Article in English | MEDLINE | ID: mdl-29622767

ABSTRACT

Defects in the PEX5 gene impair the import of peroxisomal matrix proteins, leading to nonfunctional peroxisomes and other associated pathological defects such as Zellweger syndrome. Although PEX5 regulates autophagy process in a stress condition, the mechanisms controlling autophagy by PEX5 under nutrient deprivation are largely unknown. Herein, we show a novel function of PEX5 in the regulation of autophagy via Transcription Factor EB (TFEB). Under serum-starved conditions, when PEX5 is depleted, the mammalian target of rapamycin (mTORC1) inhibitor TSC2 is downregulated, which results in increased phosphorylation of the mTORC1 substrates, including 70S6K, S6K, and 4E-BP-1. mTORC1 activation further suppresses the nuclear localization of TFEB, as indicated by decreased mRNA levels of TFEB, LIPA, and LAMP1. Interestingly, peroxisomal mRNA and protein levels are also reduced by TFEB inactivation, indicating that TFEB might control peroxisome biogenesis at a transcriptional level. Conversely, pharmacological inhibition of mTOR resulting from PEX5 depletion during nutrient starvation activates TFEB by promoting nuclear localization of the protein. In addition, mTORC1 inhibition recovers the damaged-peroxisome biogenesis. These data suggest that PEX5 may be a critical regulator of lysosomal gene expression and autophagy through the mTOR-TFEB-autophagy axis under nutrient deprivation.


Subject(s)
Autophagy , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , Peroxisome-Targeting Signal 1 Receptor/metabolism , Autophagy/genetics , Cell Line, Tumor , Energy Metabolism , Gene Expression Regulation , Humans , Lysosomes/metabolism , Peroxisome-Targeting Signal 1 Receptor/genetics , Peroxisomes/metabolism , Protein Transport
8.
Organogenesis ; 12(2): 78-93, 2016 04 02.
Article in English | MEDLINE | ID: mdl-27078170

ABSTRACT

Very long chain fatty acids are required for sphingolipid synthesis, lipid homeostasis, myelin formation, epidermal permeability, and retinal function. Seven different enzymes are known to be involved in the elongation cycle of fatty acids, with different chain-length specificities. Elovl1 is one of those enzymes whose function has been linked mainly to the synthesis of sphingolipids and the epidermal barrier. However, the role of Elovl1 in organogenesis is not clear. In zebrafish, 2 Elovl1 genes, elovl1a and elovl1b, are highly expressed in the swim bladder, and elovl1b is also expressed in the kidney. We found that both elovl1 knockdown embryos contain increased levels of long chain fatty acids from carbon number 14 to 20 as compared to control embryos. Oil-Red-O staining shows that yolk lipid consumption is greatly reduced, whereas lipid droplets accumulate within the swim bladder. Notably, knockdown of either elovl1a or elovl1b affects the expression of genes involved in swim bladder development and impairs inflation of the swim bladder. Consistent with its expression in the pronephros, knockdown of elovl1b alone affects the expression of genes required for kidney development and reduces renal clearance. Our findings strongly suggest that both elovl1 genes are a key determinant of swim bladder and kidney development in zebrafish, which may be comparatively applicable to lung and kidney development in humans.


Subject(s)
Acetyltransferases/metabolism , Air Sacs/embryology , Air Sacs/enzymology , Embryonic Development , Kidney/embryology , Kidney/enzymology , Zebrafish Proteins/metabolism , Zebrafish/embryology , Zebrafish/metabolism , Acetyltransferases/chemistry , Acetyltransferases/genetics , Amino Acid Sequence , Animals , Conserved Sequence , Egg Yolk/metabolism , Embryo, Nonmammalian/metabolism , Embryonic Development/genetics , Gene Duplication , Gene Expression Regulation, Developmental , Gene Knockdown Techniques , Genome , Kidney/physiology , Lipid Metabolism , Mammals , Myelin Sheath/metabolism , Neurons/metabolism , Zebrafish/genetics , Zebrafish Proteins/chemistry , Zebrafish Proteins/genetics
9.
Sci Rep ; 5: 18011, 2015 Dec 10.
Article in English | MEDLINE | ID: mdl-26657540

ABSTRACT

Estrogen-related receptor alpha (ESRRa) regulates a number of cellular processes including development of bone and muscles. However, direct evidence regarding its involvement in cartilage development remains elusive. In this report, we establish an in vivo role of Esrra in cartilage development during embryogenesis in zebrafish. Gene expression analysis indicates that esrra is expressed in developing pharyngeal arches where genes necessary for cartilage development are also expressed. Loss of function analysis shows that knockdown of esrra impairs expression of genes including sox9, col2a1, sox5, sox6, runx2 and col10a1 thus induces abnormally formed cartilage in pharyngeal arches. Importantly, we identify putative ESRRa binding elements in upstream regions of sox9 to which ESRRa can directly bind, indicating that Esrra may directly regulate sox9 expression. Accordingly, ectopic expression of sox9 rescues defective formation of cartilage induced by the knockdown of esrra. Taken together, our results indicate for the first time that ESRRa is essential for cartilage development by regulating sox9 expression during vertebrate development.


Subject(s)
Chondrogenesis , Gene Expression Regulation, Developmental , Receptors, Estrogen/metabolism , SOX9 Transcription Factor/genetics , Zebrafish/genetics , Zebrafish/metabolism , Animals , Branchial Region/embryology , Cartilage/embryology , Cartilage/metabolism , Cell Survival/genetics , Chondrocytes/metabolism , Chondrogenesis/genetics , Embryonic Development/genetics , Gene Knockdown Techniques , Neural Crest/embryology , Nucleotide Motifs , Protein Binding , Receptors, Estrogen/genetics , Response Elements , Zebrafish/embryology , ERRalpha Estrogen-Related Receptor
10.
Int J Biol Sci ; 10(10): 1108-15, 2014.
Article in English | MEDLINE | ID: mdl-25332685

ABSTRACT

Caldesmon, an inhibitory actin binding protein, binds to actin and inhibits actin-myosin interactions, whereas caldesmon phosphorylation reverses the inhibitory effect of caldesmon on actin-myosin interactions, potentially leading to enhanced contraction. The goal of this study was to investigate the cellular signaling pathway responsible for caldesmon phosphorylation, which is involved in the regulation of the contraction induced by dexmedetomidine (DMT), an alpha-2 adrenoceptor agonist, in endothelium-denuded rat aortas. SP600125 (a c-Jun NH2-terminal kinase [JNK] inhibitor) dose-response curves were generated in aortas that were pre-contracted with DMT or phorbol 12,13-dibutyrate (PDBu), a protein kinase C (PKC) activator. Dose-response curves to the PKC inhibitor chelerythrine were generated in rat aortas pre-contracted with DMT. The effects of SP600125 and rauwolscine (an alpha-2 adrenoceptor inhibitor) on DMT-induced caldesmon phosphorylation in rat aortic vascular smooth muscle cells (VSMCs) were investigated by western blot analysis. PDBu-induced caldesmon and DMT-induced PKC phosphorylation in rat aortic VSMCs was investigated by western blot analysis. The effects of GF109203X (a PKC inhibitor) on DMT- or PDBu-induced JNK phosphorylation in VSMCs were assessed. SP600125 resulted in the relaxation of aortas that were pre-contracted with DMT or PDBu, whereas rauwolscine attenuated DMT-induced contraction. Chelerythrine resulted in the vasodilation of aortas pre-contracted with DMT. SP600125 and rauwolscine inhibited DMT-induced caldesmon phosphorylation. Additionally, PDBu induced caldesmon phosphorylation, and GF109203X attenuated the JNK phosphorylation induced by DMT or PDBu. DMT induced PKC phosphorylation in rat aortic VSMCs. These results suggest that alpha-2 adrenoceptor-mediated, DMT-induced contraction involves caldesmon phosphorylation that is mediated by JNK phosphorylation by PKC.


Subject(s)
Aorta/physiology , Calmodulin-Binding Proteins/metabolism , Dexmedetomidine/pharmacology , Muscle Contraction/drug effects , Muscle, Smooth, Vascular/drug effects , Protein Kinase C/metabolism , Signal Transduction/physiology , Analysis of Variance , Animals , Anthracenes/pharmacology , Aorta/metabolism , Benzophenanthridines/pharmacology , Blotting, Western , Cells, Cultured , Dexmedetomidine/metabolism , Dose-Response Relationship, Drug , Muscle Contraction/physiology , Muscle, Smooth, Vascular/physiology , Phosphorylation , Rats , Receptors, Adrenergic, alpha-2/metabolism , Yohimbine/pharmacology
11.
Clin Exp Pharmacol Physiol ; 41(12): 1014-22, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25224579

ABSTRACT

Vasoconstriction induced by dexmedetomidine, a highly selective alpha-2 adrenoceptor agonist, mainly involves c-Jun NH2 -terminal kinase (JNK) phosphorylation in the isolated endothelium-denuded aorta. We carried out an in vitro study to determine the main arachidonic acid metabolic pathway that is involved in dexmedetomidine-induced JNK activation. Cumulative dexmedetomidine concentration-contractile response curves were generated in the endothelium-denuded rat aorta in the presence or absence of the following inhibitors: the JNK inhibitor SP600125, the phospholipase A2 inhibitor quinacrine dihydrochloride, the non-specific lipoxygenase (LOX) inhibitor nordihydroguaiaretic acid, the 5-LOX inhibitor AA-861, the dual 5-LOX and cyclooxygenase (COX) inhibitor phenidone, the non-specific COX inhibitor indomethacin, the cytochrome p450 epoxygenase inhibitor fluconazole, the COX-1 inhibitor SC-560, and the COX-2 inhibitor NS-398. The effect of the alpha-2 adrenoceptor inhibitor rauwolscine and other inhibitors, such as quinacrine dihydrochloride, nordihydroguaiaretic acid, AA-861, phenidone, indomethacin and the protein kinase C inhibitor GF 109203X, on dexmedetomidine-induced JNK phosphorylation was investigated in rat aortic vascular smooth muscle cells with western blotting. The effect of dexmedetomidine on 5-LOX and COX-2 expression was investigated in vascular smooth muscle cells. SP600125, quinacrine dihydrochloride, nordihydroguaiaretic acid, AA-861, phenidone, rauwolscine and chelerythrine attenuated dexmedetomidine-induced contraction. Indomethacin slightly attenuated dexmedetomidine-induced contraction. Fluconazole and SC-560 had no effect on dexmedetomidine-induced contraction, whereas NS-398 attenuated contraction. SP600125, rauwolscine, quinacrine dihydrochloride, nordihydroguaiaretic acid, AA-861, phenidone and GF 109203X attenuated dexmedetomidine-induced JNK phosphorylation. 5-LOX and COX-2 were upregulated by dexmedetomidine. Thus, dexmedetomidine-induced alpha-2 adrenoceptor-mediated contraction is mediated mainly by 5-LOX and partially by COX-2, which leads to JNK phosphorylation.


Subject(s)
Aorta, Thoracic/drug effects , Arachidonate 5-Lipoxygenase/metabolism , Dexmedetomidine/pharmacology , Endothelium, Vascular/drug effects , JNK Mitogen-Activated Protein Kinases/metabolism , Phosphorylation/drug effects , Vasoconstriction/drug effects , Animals , Aorta, Thoracic/metabolism , Benzoquinones/pharmacology , Cyclooxygenase 2 Inhibitors/pharmacology , Endothelium, Vascular/metabolism , Male , Muscle Contraction/drug effects , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Nitrobenzenes/pharmacology , Pyrazoles/pharmacology , Rats , Rats, Sprague-Dawley , Sulfonamides/pharmacology
12.
Oncotarget ; 5(6): 1554-64, 2014 Mar 30.
Article in English | MEDLINE | ID: mdl-24721928

ABSTRACT

Rho GDP dissociation inhibitor 2 (RhoGDI2) expression correlates with tumor growth, metastasis, and chemoresistance in gastric cancer. Here, we show that RhoGDI2 functions in the epithelial-mesenchymal transition (EMT), which is responsible for invasiveness during tumor progression. This tumorigenic activity is associated with repression of E-cadherin by RhoGDI2 via upregulation of Snail. Overexpression of RhoGDI2 induced phenotypic changes consistent with EMT in gastric cancer cells, including abnormal epithelial cell morphology, fibroblast-like properties, and reduced intercellular adhesion. RhoGDI2 overexpression also resulted in decreased expression of the epithelial markers E-cadherin and ß-catenin and increased expression of the mesenchymal markers vimentin and fibronectin. Importantly, RhoGDI2 overexpression also stimulated the expression of Snail, a repressor of E-cadherin and inducer of EMT, but not other family members such as Slug or Twist. RNA interference-mediated knockdown of Snail expression suppressed RhoGDI2-induced EMT and invasion, confirming that the effect was Snail-specific. These results indicate that RhoGDI2 plays a critical role in tumor progression in gastric cancer through induction of EMT. Targeting RhoGDI2 may thus be a useful strategy to inhibit gastric cancer cell invasion and metastasis.


Subject(s)
Cadherins/metabolism , Cell Movement , Cell Proliferation , Epithelial-Mesenchymal Transition , Stomach Neoplasms/pathology , Transcription Factors/metabolism , rho Guanine Nucleotide Dissociation Inhibitor beta/metabolism , Antineoplastic Agents/pharmacology , Apoptosis , Blotting, Western , Cadherins/genetics , Drug Resistance, Neoplasm , Fluorescent Antibody Technique , Humans , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Snail Family Transcription Factors , Stomach Neoplasms/drug therapy , Stomach Neoplasms/metabolism , Transcription Factors/antagonists & inhibitors , Transcription Factors/genetics , Tumor Cells, Cultured , rho Guanine Nucleotide Dissociation Inhibitor beta/genetics
13.
Int J Cancer ; 135(7): 1553-63, 2014 Oct 01.
Article in English | MEDLINE | ID: mdl-24585459

ABSTRACT

Rho GDP dissociation inhibitor 2 (RhoGDI2) expression is correlated with tumor growth, metastasis and chemoresistance in gastric cancer. However, the mechanisms by which RhoGDI2 promotes tumor cell survival and metastasis remain unclear. In this study, we clearly demonstrate that RhoGDI2 upregulates VEGF-C expression and RhoGDI2 expression is positively correlated with VEGF-C expression in human gastric tumor tissues as well as parental gastric cancer cell lines. VEGF-C depletion suppressed RhoGDI2-induced gastric cancer metastasis and sensitized RhoGDI2-overexpressing cells to cisplatin-induced apoptosis in vitro and in vivo. Secreted VEGF-C enhanced gastric cancer cell invasion and conferred cisplatin resistance to RhoGDI2-overexpressing cells. We also show that RhoGDI2 positively regulates Rac1 activity in gastric cancer cells. Inhibition of Rac1 expression suppressed RhoGDI2-induced VEGF-C expression, and this inhibition was associated with decreased invasiveness and increased sensitivity to cisplatin in RhoGDI2-overexpressing cells. Our results indicate that RhoGDI2 might be a potential therapeutic target for simultaneously reducing metastasis risk and enhancing chemotherapy efficacy in gastric cancer.


Subject(s)
Cisplatin/pharmacology , Drug Resistance, Neoplasm , Lung Neoplasms/secondary , Stomach Neoplasms/pathology , Vascular Endothelial Growth Factor C/metabolism , rho Guanine Nucleotide Dissociation Inhibitor beta/metabolism , Animals , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Blotting, Western , Cell Proliferation/drug effects , Chromatin Immunoprecipitation , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Humans , Immunoenzyme Techniques , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Nude , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Stomach Neoplasms/drug therapy , Stomach Neoplasms/metabolism , Tissue Array Analysis , Tumor Cells, Cultured , Vascular Endothelial Growth Factor C/genetics , Xenograft Model Antitumor Assays , rho Guanine Nucleotide Dissociation Inhibitor beta/antagonists & inhibitors , rho Guanine Nucleotide Dissociation Inhibitor beta/genetics
14.
Oncotarget ; 4(11): 2045-56, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24185104

ABSTRACT

Rho GDP dissociation inhibitor 2 (RhoGDI2) promotes tumor growth and malignant progression and enhances chemoresistance of gastric cancer. Recently, we noted an inverse correlation between RhoGDI2 and 14-3-3σ expression, which suggests that 14-3-3σ is a target of gastric cancer metastasis and the chemoresistance-promoting effect of RhoGDI2. Herein, we evaluated whether 14-3-3σ is regulated by RhoGDI2 and is functionally important for the RhoGDI2-induced cisplatin resistance of gastric cancer cells. We used highly metastatic and cisplatin-resistant RhoGDI2-overexpressing SNU-484 cells and observed decreased 14-3-3σ mRNA and protein expression. Depletion of 14-3-3σ in SNU-484 control cells enhanced cisplatin resistance, whereas restoration of 14-3-3σ in RhoGDI2-overexpressing SNU-484 cells impaired cisplatin resistance in vitro and in vivo. We also found that the phosphorylation levels of Erk and p38 kinases significantly decreased in RhoGDI2-overexpressing SNU-484 cells and recovered after 14-3-3σ expression, and that decreased activities of these kinases were critical for RhoGDI2-induced cisplatin resistance. In conclusion, 14-3-3σ is a RhoGDI2-regulated gene that appears to be important for suppressing the chemoresistance of gastric cancer cells.


Subject(s)
14-3-3 Proteins/metabolism , Biomarkers, Tumor/metabolism , Cisplatin/pharmacology , Exoribonucleases/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Stomach Neoplasms/drug therapy , Stomach Neoplasms/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , rho Guanine Nucleotide Dissociation Inhibitor beta/metabolism , 14-3-3 Proteins/biosynthesis , 14-3-3 Proteins/genetics , Apoptosis/physiology , Biomarkers, Tumor/biosynthesis , Biomarkers, Tumor/genetics , Cell Line, Tumor , Cell Movement/physiology , Disease Progression , Down-Regulation , Drug Resistance, Neoplasm , Enzyme Activation , Exoribonucleases/biosynthesis , Exoribonucleases/genetics , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , HeLa Cells , Humans , MAP Kinase Signaling System , MCF-7 Cells , Neoplasm Metastasis , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/genetics , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Transfection , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , rho Guanine Nucleotide Dissociation Inhibitor beta/genetics
15.
J Proteome Res ; 11(4): 2355-64, 2012 Apr 06.
Article in English | MEDLINE | ID: mdl-22364609

ABSTRACT

Rho GDP dissociation inhibitor 2 (RhoGDI2) was initially identified as a regulator of the Rho family of GTPases. Our recent works suggest that RhoGDI2 promotes tumor growth and malignant progression, as well as enhances chemoresistance in gastric cancer. Here, we delineate the mechanism by which RhoGDI2 promotes gastric cancer cell invasion and chemoresistance using two-dimensional gel electrophoresis (2-DE) on proteins derived from a RhoGDI2-overexpressing SNU-484 human gastric cancer cell line and control cells. Differentially expressed proteins were identified using matrix-assisted laser desorption ionization-time-of-flight mass spectrometry (MALDI-TOF-MS). In total, 47 differential protein spots were identified; 33 were upregulated, and 14 were downregulated by RhoGDI2 overexpression. Upregulation of SAE1, Cathepsin D, Cofilin1, CIAPIN1, and PAK2 proteins was validated by Western blot analysis. Loss-of-function analysis using small interference RNA (siRNA) directed against candidate genes reveals the need for CIAPIN1 and PAK2 in RhoGDI2-induced cancer cell invasion and Cathepsin D and PAK2 in RhoGDI2-mediated chemoresistance in gastric cancer cells. These data extend our understanding of the genes that act downstream of RhoGDI2 during the progression of gastric cancer and the acquisition of chemoresistance.


Subject(s)
Guanine Nucleotide Dissociation Inhibitors/metabolism , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology , Cell Line, Tumor , Drug Resistance, Neoplasm , Electrophoresis, Gel, Two-Dimensional , Humans , Intracellular Signaling Peptides and Proteins/analysis , Intracellular Signaling Peptides and Proteins/metabolism , Neoplasm Metastasis , Proteome/analysis , Proteome/metabolism , Proteomics/methods , Reproducibility of Results , Stomach Neoplasms/drug therapy , Up-Regulation , p21-Activated Kinases/analysis , p21-Activated Kinases/metabolism , rho Guanine Nucleotide Dissociation Inhibitor beta , rho-Specific Guanine Nucleotide Dissociation Inhibitors
16.
Biochem Biophys Res Commun ; 414(3): 575-80, 2011 Oct 28.
Article in English | MEDLINE | ID: mdl-21986528

ABSTRACT

Rho GDP dissociation inhibitor 2 (RhoGDI2) is a regulator of the Rho family GTPases. Recent work from our laboratory suggests that RhoGDI2 expression potentially enhances resistance to cisplatin as well as promotes tumor growth and malignant progression in gastric cancer. In this study, we demonstrate that phospholipase C-gamma (PLCγ) is required for RhoGDI2-mediated cisplatin resistance and cancer cell invasion in gastric cancer. The levels of phosphorylated PLCγ are markedly enhanced in RhoGDI2-overexpressing SNU-484 cells and, by contrast, repressed in RhoGDI2-depleted MKN-28 cells. Depletion of PLCγ expression or inhibition of its activity not only significantly increases cisplatin-induced apoptosis but also suppresses the invasive ability of RhoGDI2-overexpressing SNU-484 cells. Taken together, our results suggest that PLCγ plays a key role in RhoGDI2-mediated cisplatin resistance and cell invasion in gastric cancer cells.


Subject(s)
Antineoplastic Agents/therapeutic use , Cisplatin/therapeutic use , Drug Resistance, Neoplasm/genetics , Guanine Nucleotide Dissociation Inhibitors/metabolism , Phospholipase C gamma/physiology , Stomach Neoplasms/drug therapy , Tumor Suppressor Proteins/metabolism , Cell Line, Tumor , Enzyme Activation , Guanine Nucleotide Dissociation Inhibitors/genetics , Humans , Neoplasm Invasiveness , Phospholipase C gamma/genetics , Phosphorylation , Stomach Neoplasms/pathology , Tumor Suppressor Proteins/genetics , rho Guanine Nucleotide Dissociation Inhibitor beta , rho-Specific Guanine Nucleotide Dissociation Inhibitors
17.
Cancer Lett ; 311(1): 48-56, 2011 Dec 01.
Article in English | MEDLINE | ID: mdl-21752536

ABSTRACT

Rho GDP dissociation inhibitor (RhoGDI)2 has been identified as a regulator of Rho family GTPase. Recently, we suggested that RhoGDI2 could promote tumor growth and malignant progression in gastric cancer. In this study, we demonstrate that RhoGDI2 contributes to another important feature of aggressive cancers, i.e., resistance to chemotherapeutic agents such as cisplatin. Forced expression of RhoGDI2 attenuated cisplatin-induced apoptosis, whereas RhoGDI2 depletion showed opposite effects in vitro. Moreover, the increased anti-apoptotic effect of RhoGDI2 on cisplatin was further validated in RhoGDI2-overexpressing SNU-484 xenograft model in nude mice. Furthermore, we identified Bcl-2 as a major determinant of RhoGDI2-mediated cisplatin resistance in gastric cancer cells. Depletion of Bcl-2 expression significantly increased cisplatin-induced apoptosis in RhoGDI2-overexpressing gastric cancer cells, whereas overexpression of Bcl-2 blocked cisplatin-induced apoptosis in RhoGDI2-depleted gastric cancer cells. Overall, these findings establish RhoGDI2 as an important therapeutic target for simultaneously enhancing chemotherapy efficacy and reducing metastasis risk in gastric cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cisplatin/pharmacology , Guanine Nucleotide Dissociation Inhibitors/biosynthesis , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Stomach Neoplasms/drug therapy , Stomach Neoplasms/metabolism , Tumor Suppressor Proteins/biosynthesis , Animals , Apoptosis/physiology , Cell Line, Tumor , Drug Resistance, Neoplasm , Etoposide/pharmacology , Guanine Nucleotide Dissociation Inhibitors/deficiency , Guanine Nucleotide Dissociation Inhibitors/genetics , Guanine Nucleotide Dissociation Inhibitors/metabolism , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Proto-Oncogene Proteins c-bcl-2/genetics , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/genetics , Staurosporine/pharmacology , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Transfection , Tumor Suppressor Proteins/deficiency , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Up-Regulation , rho Guanine Nucleotide Dissociation Inhibitor alpha , rho Guanine Nucleotide Dissociation Inhibitor beta , rho-Specific Guanine Nucleotide Dissociation Inhibitors
18.
J Biol Chem ; 285(33): 25500-5, 2010 Aug 13.
Article in English | MEDLINE | ID: mdl-20558744

ABSTRACT

Gadd45b has been known as a positive mediator of apoptosis induced by certain cytokines and oncogenes. Here, we identified Gadd45b as an effector of Fas-induced apoptosis and found that p38-mediated Rb hyperphosphorylation is one of the mechanisms of Fas-induced apoptosis in murine hepatocyte AML12 cells. Gadd45b has been shown to activate p38 through its physical interaction with MTK1 and induce apoptosis. However, in this study, we have showed that the function of Gadd45b during Fas-induced apoptosis in AML12 cells is different from that reported in previous studies. Depletion of Gadd45b expression did not inhibit the phosphorylation of p38, but it suppressed p38-mediated Rb phosphorylation and apoptosis in response to Fas stimulation by reducing the interaction between p38 and Rb. Ectopic expression of Gadd45b was sufficient to enhance this interaction. These findings suggest that Gadd45b mediates p38-induced Rb phosphorylation by enhancing the interaction between p38 and Rb during Fas-induced apoptosis in murine hepatocytes.


Subject(s)
Antigens, Differentiation/metabolism , Retinoblastoma Protein/metabolism , fas Receptor/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Apoptosis/drug effects , Cell Line , Cell Line, Tumor , Enzyme Inhibitors/pharmacology , HeLa Cells , Humans , Imidazoles/pharmacology , Immunoprecipitation , In Situ Nick-End Labeling , Mice , Protein Binding/genetics , Protein Binding/physiology , Pyridines/pharmacology , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , fas Receptor/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors
19.
Clin Cancer Res ; 15(8): 2612-9, 2009 Apr 15.
Article in English | MEDLINE | ID: mdl-19351766

ABSTRACT

PURPOSE: Rho GDP dissociation inhibitor 2 (RhoGDI2) has been identified as a regulator of Rho family GTPase. However, there is currently no direct evidence suggesting whether RhoGDI2 activates or inhibits Rho family GTPase in vivo (and which type), and the role of RhoGDI2 in tumor remains controversial. Here, we assessed the effects of RhoGDI2 expression on gastric tumor growth and metastasis progression. EXPERIMENTAL DESIGN: Proteomic analysis was done to investigate the tumor-specific protein expression in gastric cancer and RhoGDI2 was selected for further study. Immunohistochemistry was used to detect RhoGDI2 expression in clinical samples of primary gastric tumor tissues which have different pathologic stages. Gain-of-function and loss-of-function approaches were done to examine the malignant phenotypes of the RhoGDI2-expressing or RhoGDI2-depleting cells. RESULTS: RhoGDI2 expression was correlated positively with tumor progression and metastasis potential in human gastric tumor tissues, as well as cell lines. The forced expression of RhoGDI2 caused a significant increase in gastric cancer cell invasion in vitro, and tumor growth, angiogenesis, and metastasis in vivo, whereas RhoGDI2 depletion evidenced opposite effects. CONCLUSION: Our findings indicate that RhoGDI2 is involved in gastric tumor growth and metastasis, and that RhoGDI2 may be a useful marker for tumor progression of human gastric cancer.


Subject(s)
Guanine Nucleotide Dissociation Inhibitors/biosynthesis , Stomach Neoplasms/pathology , Tumor Suppressor Proteins/biosynthesis , Animals , Cell Line, Tumor , Disease Progression , Gene Knockdown Techniques , Guanine Nucleotide Dissociation Inhibitors/genetics , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Metastasis , Proteomics , RNA, Small Interfering/metabolism , Stomach Neoplasms/metabolism , Transplantation, Heterologous , Tumor Suppressor Proteins/genetics , rho Guanine Nucleotide Dissociation Inhibitor beta , rho-Specific Guanine Nucleotide Dissociation Inhibitors
SELECTION OF CITATIONS
SEARCH DETAIL
...