Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Mol Ther Nucleic Acids ; 29: 803-822, 2022 Sep 13.
Article in English | MEDLINE | ID: mdl-36159587

ABSTRACT

Cancer stem-like cells (CSCs) have been suggested to be responsible for chemoresistance and tumor recurrence owing to their self-renewal capacity and differentiation potential. Although WEE1 is a strong candidate target for anticancer therapies, its role in ovarian CSCs is yet to be elucidated. Here, we show that WEE1 plays a key role in regulating CSC properties and tumor resistance to carboplatin via a microRNA-dependent mechanism. We found that WEE1 expression is upregulated in ovarian cancer spheroids because of the decreased expression of miR-424 and miR-503, which directly target WEE1. The overexpression of miR-424/503 suppressed CSC activity by inhibiting WEE1 expression, but this effect was reversed on the restoration of WEE1 expression. Furthermore, we demonstrated that NANOG modulates the miR-424/503-WEE1 axis that regulates the properties of CSCs. We also demonstrated the pharmacological restoration of the NANOG-miR-424/503-WEE1 axis and attenuation of ovarian CSC characteristics in response to atorvastatin treatment. Lastly, miR-424/503-mediated WEE1 inhibition re-sensitized chemoresistant ovarian cancer cells to carboplatin. Additionally, combined treatment with atorvastatin and carboplatin synergistically reduced tumor growth, chemoresistance, and peritoneal seeding in the intraperitoneal mouse models of ovarian cancer. We identified a novel NANOG-miR-424/503-WEE1 pathway for regulating ovarian CSCs, which has potential therapeutic utility in ovarian cancer treatment.

2.
Genes (Basel) ; 12(1)2020 12 23.
Article in English | MEDLINE | ID: mdl-33374832

ABSTRACT

Targeting the tumor vasculature is an attractive strategy for cancer treatment. However, the tumor vasculature is heterogeneous, and the mechanisms involved in the neovascularization of tumors are highly complex. Vasculogenic mimicry (VM) refers to the formation of vessel-like structures by tumor cells, which can contribute to tumor neovascularization, and is closely related to metastasis and a poor prognosis. Here, we report a novel function of AXL receptor tyrosine kinase (AXL) in the regulation of VM formation in breast cancer cells. MDA-MB-231 cells exhibited VM formation on Matrigel cultures, whereas MCF-7 cells did not. Moreover, AXL expression was positively correlated with VM formation. Pharmacological inhibition or AXL knockdown strongly suppressed VM formation in MDA-MB-231 cells, whereas the overexpression of AXL in MCF-7 cells promoted VM formation. In addition, AXL knockdown regulated epithelial-mesenchymal transition (EMT) features, increasing cell invasion and migration in MDA-MB-231 cells. Finally, the overexpression of microRNA-34a (miR-34a), which is a well-described EMT-inhibiting miRNA and targets AXL, inhibited VM formation, migration, and invasion in MDA-MB 231 cells. These results identify a miR-34a-AXL axis that is critical for the regulation of VM formation and may serve as a therapeutic target to inhibit tumor neovascularization.


Subject(s)
Breast Neoplasms/genetics , Gene Expression Regulation, Neoplastic , MicroRNAs/metabolism , Neovascularization, Pathologic/genetics , Proto-Oncogene Proteins/genetics , Receptor Protein-Tyrosine Kinases/genetics , Breast/blood supply , Breast/pathology , Breast Neoplasms/blood supply , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Movement/genetics , Epithelial-Mesenchymal Transition/genetics , Female , Humans , Neoplasm Invasiveness/genetics , Neovascularization, Pathologic/pathology , Axl Receptor Tyrosine Kinase
3.
BMB Rep ; 53(6): 291-298, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32438972

ABSTRACT

Tumor angiogenesis is an essential process for growth and metastasis of cancer cells as it supplies tumors with oxygen and nutrients. During tumor angiogenesis, many pro-angiogenic factors are secreted by tumor cells to induce their own vascularization via activation of pre-existing host endothelium. However, accumulating evidence suggests that vasculogenic mimicry (VM) is a key alternative mechanism for tumor vascularization when tumors are faced with insufficient supply of oxygen and nutrients. VM is a tumor vascularization mechanism in which tumors create a blood supply system, in contrast to tumor angiogenesis mechanisms that depend on pre-existing host endothelium. VM is closely associated with tumor progression and poor prognosis in many cancers. Therefore, inhibition of VM may be a promising therapeutic strategy and may overcome the limitations of anti-angiogenesis therapy for cancer patients. In this review, we provide an overview of the current anti-angiogenic therapies for ovarian cancer and the current state of knowledge regarding the links between microRNAs and the VM process, with a focus on the mechanism that regulates associated signaling pathways in ovarian cancer. Moreover, we discuss the potential for VM as a therapeutic strategy against ovarian cancer. [BMB Reports 2020; 53(6): 291-298].


Subject(s)
Antineoplastic Agents/pharmacology , Neovascularization, Pathologic/drug therapy , Ovarian Neoplasms/drug therapy , Female , Humans , MicroRNAs/drug effects , MicroRNAs/metabolism , Neovascularization, Pathologic/metabolism , Ovarian Neoplasms/metabolism , Signal Transduction/drug effects
4.
Genes (Basel) ; 12(1)2020 12 30.
Article in English | MEDLINE | ID: mdl-33396906

ABSTRACT

Cancer metastasis is the primary cause of cancer-related death and metastatic cancer has circulating-tumor cells (CTCs), which circulate in the bloodstream before invading other organs. Thus, understanding the precise role of CTCs may provide new insights into the metastasis process and reduce cancer mortality. However, the molecular characteristics of CTCs are not well understood due to a lack of number of CTCs. Therefore, suspension cells were generated from MDA-MB-468 cells to mimic CTCs, and we investigate the microRNA (miRNA)-dependent molecular networks and their role in suspension cells. Here, we present an integrated analysis of mRNA and miRNA sequencing data for suspension cell lines, through comparison with adherent cells. Among the differentially regulated miRNA-mRNAs axes, we focus on the miR-146a-Neuropilin2 (NRP2) axis, which is known to influence tumor aggressiveness. We show that miR-146a directly regulates NRP2 expression and inhibits Semaphorin3C (SEMA3C) signaling. Functional studies reveal that miR-146a represses SEMA3C-induced invasion and proliferation by targeting NRP2. Finally, high-NRP2 is shown to be associated with poor outcomes in breast cancer patients. This study identifies the key role of the miR-146a-NRP2 signaling axis that is critical for the regulation of migration and invasion in CTC-mimicking cells.


Subject(s)
Breast Neoplasms/genetics , MicroRNAs/genetics , Neoplastic Cells, Circulating/metabolism , Neuropilin-2/genetics , Semaphorins/genetics , Base Pairing , Base Sequence , Breast Neoplasms/metabolism , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Movement , Cell Proliferation , Female , Gene Expression Regulation, Neoplastic , Genes, Reporter , Humans , Luciferases/genetics , Luciferases/metabolism , MicroRNAs/metabolism , Models, Biological , Neoplastic Cells, Circulating/pathology , Neuropilin-2/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Semaphorins/metabolism , Signal Transduction , Survival Analysis
5.
Int J Mol Sci ; 20(7)2019 Apr 09.
Article in English | MEDLINE | ID: mdl-30970566

ABSTRACT

The mammalian mediator complex subunit 28 (MED28) is overexpressed in a variety of cancers and it regulates cell migration/invasion and epithelial-mesenchymal transition. However, transcription factors that increase MED28 expression have not yet been identified. In this study, we performed a luciferase reporter assay to identify and characterize the prospective transcription factors, namely E2F transcription factor 1, nuclear respiratory factor 1, E-26 transforming sequence 1, and CCAAT/enhancer-binding protein ß, which increased MED28 expression. In addition, the release from the arrest at the G1-S or G2-M phase transition after cell cycle synchronization using thymidine or nocodazole, respectively, showed enhanced MED28 expression at the G1-S transition and mitosis. Furthermore, the overexpression of MED28 significantly decreased the duration of interphase and mitosis. Conversely, a knockdown of MED28 using si-RNA increased the duration of interphase and mitosis. Of note, the overexpression of MED28 significantly increased micronucleus and nuclear budding in HeLa cells. In addition, flow cytometry and fluorescence microscopy analyses showed that the overexpression of MED28 significantly increased aneuploid cells. Taken together, these results suggest that MED28 expression is increased by oncogenic transcription factors and its overexpression disturbs the cell cycle, which results in genomic instability and aneuploidy.


Subject(s)
Genomic Instability , Mediator Complex/genetics , Mediator Complex/metabolism , Transcription Factors/metabolism , Aneuploidy , Cell Cycle/drug effects , Genomic Instability/drug effects , HEK293 Cells , HeLa Cells , Humans , MCF-7 Cells , Nocodazole/pharmacology , Promoter Regions, Genetic , Thymidine/pharmacology , Up-Regulation
6.
Front Immunol ; 9: 294, 2018.
Article in English | MEDLINE | ID: mdl-29515588

ABSTRACT

Endothelial cells that line the inner walls of blood vessels are in direct contact with blood and display remarkable heterogeneity in their response to exogenous stimuli. These ECs have unique location-dependent properties determined by the corresponding vascular beds and play an important role in regulating the homeostasis of the vascular system. Evidence suggests that vascular endothelial cells exposed to various environments undergo dynamic phenotypic switching, a key biological program in the context of endothelial heterogeneity, but that might result in EC dysfunction and, in turn, cause a variety of human diseases. Emerging studies show the importance of endothelial to mesenchymal transition (EndMT) in endothelial dysfunction during inflammation. EndMT is a complex biological process in which ECs lose their endothelial characteristics, acquire mesenchymal phenotypes, and express mesenchymal cell markers, such as alpha smooth muscle actin and fibroblast-specific protein 1. EndMT is induced by inflammatory responses, leading to pathological states, including tissue fibrosis, pulmonary arterial hypertension, and atherosclerosis, via dysfunction of the vascular system. Although the mechanisms associated with inflammation-induced EndMT have been identified, unraveling the specific role of this phenotypic switching in vascular dysfunction remains a challenge. Here, we review the current understanding on the interactions between inflammatory processes, EndMT, and endothelial dysfunction, with a focus on the mechanisms that regulate essential signaling pathways. Identification of such mechanisms will guide future research and could provide novel therapeutic targets for the treatment of vascular diseases.


Subject(s)
Cell Transdifferentiation/physiology , Endothelial Cells/pathology , Inflammation/pathology , Humans , Vascular Diseases/pathology
7.
Oncol Lett ; 15(3): 3147-3154, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29435049

ABSTRACT

The regulation of gene expression by transcription factors serves a critical function in cell proliferation. Zinc-finger protein 224 (ZNF224), a Krüppel-associated-box-containing zinc finger protein, is known to serve a crucial function in integrating the transcriptional co-factors that activate transcriptional regulation pathways in the cell. A previous study demonstrated that ZNF224 enhances cell proliferation by downregulating the expression of p21 and p53. The present study identified mediator complex subunit 28 (MED28) as a potential binding partner for ZNF224; this was confirmed by co-immunoprecipitation and a surface plasmon resonance assay. Additionally, the KRAB domain at the N-terminal of ZNF224 interacts with the MED domain of MED28. Bimolecular fluorescence complementation analysis revealed that ZNF224 associates with MED28 in the nucleus. In addition, ZNF224 was rapidly degraded upon treatment with the DNA-damaging agent camptothecin (CPT). Transient overexpression of MED28 inhibited the CPT-mediated degradation of ZNF224, resulting in increased colony formation by MCF-7 cells. The molecular mechanisms that underlie the biological outcomes of MED28 expression have not yet been fully elucidated. The present study provides molecular evidence for the function of ZNF224 and MED28 in the DNA-damage response.

8.
Biochem Biophys Res Commun ; 482(1): 28-34, 2017 Jan 01.
Article in English | MEDLINE | ID: mdl-27836539

ABSTRACT

Infection with pathogens activates the endothelial cell and its sustained activation may result in impaired endothelial function. Endothelial dysfunction contributes to the pathologic angiogenesis that is characteristic of infection-induced inflammatory pathway activation. Nucleotide-binding oligomerization domain-containing protein 1 (NOD1) is a protein receptor which recognizes bacterial molecules and stimulates an immune reaction in various cells; however, the underlying molecular mechanisms in the regulation of inflammation-triggered angiogenesis are not fully understood. Here we report that peroxisome proliferator-activated receptor gamma (PPARγ)-mediated miR-125a serves as an important regulator of NOD1 agonist-mediated angiogenesis in endothelial cells by directly targeting NOD1. Treatment of human umbilical vein endothelial cells with natural PPARγ ligand, 15-Deoxy-Delta12,14-prostaglandin J2, led to inhibition of NOD1 expression; contrarily, protein levels of NOD1 were significantly increased by PPARγ knockdown. We report that PPARγ regulation of NOD1 expression is a novel microRNA-mediated regulation in endothelial cells. MiR-125a expression was markedly decreased in human umbilical vein endothelial cells subjected to PPARγ knockdown while 15-Deoxy-Delta12,14-prostaglandin J2 treatment increased the level of miR-125a. In addition, NOD1 is closely regulated by miR-125a, which directly targets the 3' untranslated region of NOD1. Moreover, both overexpression of miR-125a and PPARγ activation led to inhibition of NOD1 agonist-induced tube formation in endothelial cells. Finally, NOD1 agonist increased the formation of cranial and subintestinal vessel plexus in zebrafish, and this effect was abrogated by concurrent PPARγ activation. Overall, these findings identify a PPARγ-miR-125a-NOD1 signaling axis in endothelial cells that is critical in the regulation of inflammation-mediated angiogenesis.


Subject(s)
Endothelial Cells/metabolism , MicroRNAs/metabolism , Neovascularization, Pathologic/metabolism , Nod1 Signaling Adaptor Protein/metabolism , PPAR gamma/metabolism , Vasculitis/metabolism , Animals , Cells, Cultured , Down-Regulation , Endothelial Cells/pathology , Humans , Neovascularization, Pathologic/pathology , Vasculitis/pathology , Zebrafish
9.
Biochem Biophys Res Commun ; 478(4): 1682-7, 2016 09 30.
Article in English | MEDLINE | ID: mdl-27596970

ABSTRACT

Tauroursodeoxycholic acid (TUDCA) is known to prevent apoptosis through the Bax pathway and to promote neovascularization by enhancing the mobilization of stem cells, their differentiation. This study was performed to investigate the effect of TUDCA on erythropoiesis in hematopoietic stem cells (HSCs). Since erythropoiesis of CD34(+) HSCs is divided into four phases, the total cell number, viable cell number, cell viability, cell morphology, and expressed erythroid markers in each phase were examined. The number of viable control cells and their viability did not differ from those of the TUDCA-treated cells in phase I and II. However, TUDCA increased cell viability compared to the control in phases III and IV. Cell distribution differed that the immature erythroid cell number was higher for the TUDCA-treated cells than for the control cells until phase III, but all developed into RBCs in the last. The final RBC number and viability was significantly higher in TUDCA-treated cells compared to the control cells. Taken together, we suggest that TUDCA addition to cell cultures for artificial RBC production could be used as a new protocol for improving the viability of RBCs.


Subject(s)
Cell Differentiation/drug effects , Erythrocytes/drug effects , Erythroid Cells/drug effects , Hematopoietic Stem Cells/drug effects , Taurochenodeoxycholic Acid/pharmacology , Adult , Aged , Antigens, CD34/metabolism , Biomarkers/metabolism , Cell Survival/drug effects , Cells, Cultured , Cholagogues and Choleretics/pharmacology , Erythrocyte Count , Erythrocytes/cytology , Erythrocytes/metabolism , Erythroid Cells/cytology , Erythroid Cells/metabolism , Erythropoiesis/drug effects , Female , Flow Cytometry , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Humans , Immunophenotyping , Male , Middle Aged , Time Factors
10.
Oncotarget ; 7(21): 31177-90, 2016 May 24.
Article in English | MEDLINE | ID: mdl-27105517

ABSTRACT

ZNF224 is a Krüppel-associated box-containing zinc-finger protein which represses gene transcription by interacting with various co-repressors. However, its consensus DNA sequences and target genes are not fully identified. In this study, we identified and characterized consensus DNA sequences containing 5'-CAGC-3' recognized by ZNF224 through ChIP-sequencing, which further confirmed by ELISA, SPR, qPCR, and luciferase activity assay. ZNF224 increased miR-663a transcription by binding to miR-663a promoter, which in turn binds to 3' UTR of p53 and p21 to decrease their expression. miR-663a antagonist abolished ZNF224-mediated suppression of p21 and p53, resulting in the enhanced apoptosis by CPT. The analyses using human breast ductal carcinoma tissues exhibited that the expression of ZNF224 and miR-663a was increased in cancer compared to non-cancer region. Consequently, ZNF224 increases cell survival and decreases apoptosis by decreasing the expression of p53 and p21 via miR-663a as a transcriptional activator. Taken together, we identified and characterized DNA binding element of ZNF224, and its target genes, miR-663a, which provides a novel insight in the down-regulation of p21 and p53 via miR-663a by ZNF224 in breast cancer.


Subject(s)
Breast Neoplasms/metabolism , Carcinoma, Ductal, Breast/metabolism , Cyclin-Dependent Kinase Inhibitor p21/metabolism , MicroRNAs/metabolism , Repressor Proteins/metabolism , Tumor Suppressor Protein p53/metabolism , Apoptosis/physiology , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Carcinoma, Ductal, Breast/genetics , Carcinoma, Ductal, Breast/pathology , Cell Cycle/physiology , Cell Line, Tumor , Cell Proliferation/physiology , Cyclin-Dependent Kinase Inhibitor p21/genetics , Down-Regulation , Genes, p53 , HEK293 Cells , Humans , MCF-7 Cells , MicroRNAs/genetics , Repressor Proteins/genetics , Transfection , Tumor Suppressor Protein p53/genetics
11.
Mol Med Rep ; 13(3): 2078-86, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26781331

ABSTRACT

Interruptin B has been isolated from Cyclosorus terminans, however, its pharamcological effect has not been fully identified. In the present study, the effects of interruptin B, from C. terminans, on brown adipocyte differentiation and glucose uptake in adipose­derived stem cells (ASCs) were investigated. The results revealed that interruptin B dose­dependently enhanced the adipogenic differentiation of ASCs, with an induction in the mRNA expression levels of peroxisome proliferator­activated receptor (PPAR)­α and PPAR­Î³. In addition, interruptin B efficiently increased the number and the membrane potential of mitochondria and upregulated the mRNA expression levels of uncoupling protein (UCP)­1 and cyclooxygenase (COX)­2, which are all predominantly expressed in brown adipocytes. Interruptin B increased glucose consumption in differentiated ASCs, accompanied by the upregulation in the mRNA expression levels of glucose transporter (GLUT)­1 and GLUT­4. The computational analysis of molecular docking, a luciferase reporter assay and surface plasmon resonance confirmed the marked binding affinity of interruptin B to PPAR­α and PPAR­Î³ (K(D) values of 5.32 and 0.10 µm, respectively). To the best of our knowledge, the present study is the first report to show the stimulatory effects of interruptin B on brown adipocyte differentiation and glucose uptake in ASCs, through its role as a dual PPAR­α and PPAR­Î³ ligand. Therefore, interruptin B could be further developed as a therapeutic agent for the treatment of diabetes.


Subject(s)
Adipocytes, Brown/cytology , Adipose Tissue/cytology , Cell Differentiation/drug effects , Chalcones/pharmacology , Coumarins/pharmacology , Glucose/metabolism , Stem Cells/cytology , Stem Cells/metabolism , Chalcones/chemistry , Computer Simulation , Coumarins/chemistry , Hep G2 Cells , Humans , Ligands , Molecular Docking Simulation , PPAR alpha/antagonists & inhibitors , PPAR alpha/metabolism , PPAR gamma/antagonists & inhibitors , PPAR gamma/metabolism , Stem Cells/drug effects , Surface Plasmon Resonance
12.
Article in English | MEDLINE | ID: mdl-25723281

ABSTRACT

Med28 plays a role in transcription, signal transduction, and cell proliferation. The overexpression of med28 is associated with tumor progression in in vitro and in vivo models. Recently it has been reported that the elevated expression of med28 is associated with poor outcome in women with breast cancer. The expression level of med28 in in vitro and in vivo was examined by using anti-rabbit polyclonal antibody in previous reports. In this study, we report for the first time the generation and characterization of four monoclonal antibodies against med28 through immunoblotting, immunofluorescence microscopy, immunoprecipitation, and immunohistochemical analyses. These antibodies will be useful in detecting med28 in in vitro and in vivo.


Subject(s)
Antibodies, Monoclonal/biosynthesis , Antigens, Neoplasm/analysis , Breast Neoplasms/diagnosis , Mediator Complex/analysis , Animals , Antibodies, Monoclonal/chemistry , Antibody Specificity , Antigens, Neoplasm/immunology , Blotting, Western , Breast Neoplasms/immunology , Breast Neoplasms/pathology , Enzyme-Linked Immunosorbent Assay , Escherichia coli/genetics , Escherichia coli/metabolism , Female , Freund's Adjuvant/administration & dosage , Freund's Adjuvant/immunology , Gene Expression , HeLa Cells , Humans , Hybridomas/immunology , Immunization , Immunohistochemistry , MCF-7 Cells , Mediator Complex/immunology , Mice , Mice, Inbred BALB C , Recombinant Proteins/administration & dosage , Recombinant Proteins/biosynthesis , Recombinant Proteins/immunology
13.
Biomaterials ; 44: 45-54, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25617125

ABSTRACT

ARS-interacting multifunctional protein 1 (AIMP1) induces production of inflammatory cytokines from immune cells. Since osteoclastogenesis is promoted by positive regulation of inflammatory cytokines, whether AIMP1 could promote osteoclastogenesis was investigated. AIMP1 induced osteoclastogenesis and acted synergistically with RANKL to promote osteoclastogenesis. Down-regulation of CD23, an AIMP1 receptor, abolished AIMP1-mediated osteoclastogenesis. Enzyme-linked immunosorbent assays showed that the AIMP1 level was significantly higher in the peripheral blood (PB) and synovial fluid of rheumatoid arthritis patients than in normal PB. A monoclonal antibody (clone 15B3AF) that blocked the cytokine activity of AIMP1 inhibited the AIMP1-mediated production of inflammatory cytokines. Clone 15B3AF inhibited the AIMP1-mediated osteoclastogenesis in vitro. We then cloned the complementary determining regions of clone 15B3AF and generated a chimeric antibody (atliximab). In a collagen-induced arthritis mouse model (CIA), atliximab administration significantly attenuated disease severity and improved various histopathological parameters. Three-dimensional micro-computed tomography scanning confirmed that atliximab enhanced the joint structures in CIA mice. Furthermore, atliximab decreased the expression of inflammatory cytokines in the serum and inflamed joints of CIA mice. Taken together, our findings suggest that AIMP1 exacerbates RA by promoting inflammation and osteoclastogenesis and that atliximab could be developed as a therapeutic antibody to target inflammatory diseases, including RA.


Subject(s)
Antibodies, Neutralizing/pharmacology , Antibodies/pharmacology , Arthritis, Experimental/pathology , Cytokines/metabolism , Inflammation Mediators/metabolism , Neoplasm Proteins/metabolism , Osteoclasts/pathology , Osteogenesis/drug effects , RNA-Binding Proteins/metabolism , Recombinant Fusion Proteins/pharmacology , Animals , Arthritis, Experimental/diagnostic imaging , Cell Line , Disease Models, Animal , Humans , Mice , Osteoclasts/drug effects , RANK Ligand/pharmacology , X-Ray Microtomography
14.
Stem Cells ; 33(3): 792-805, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25407160

ABSTRACT

Although serum bile acid concentrations are approximately 10 µM in healthy subjects, the crosstalk between the biliary system and vascular repair has never been investigated. In this study, tauroursodeoxycholic acid (TUDCA) induced dissociation of CD34(+) hematopoietic stem cells (HSCs) from stromal cells by reducing adhesion molecule expression. TUDCA increased CD34(+) /Sca1(+) progenitors in mice peripheral blood (PB), and CD34(+) , CD31(+) , and c-kit(+) progenitors in human PB. In addition, TUDCA increased differentiation of CD34(+) HSCs into EPC lineage cells via Akt activation. EPC invasion was increased by TUDCA, which was mediated by fibroblast activating protein via Akt activation. Interestingly, TUDCA induced integration of EPCs into human aortic endothelial cells (HAECs) by increasing adhesion molecule expression. In the mouse hind limb ischemia model, TUDCA promoted blood perfusion by enhancing angiogenesis through recruitment of Flk-1(+) /CD34(+) and Sca-1(+) /c-kit(+) progenitors into damaged tissue. In GFP(+) bone marrow-transplanted hind limb ischemia, TUDCA induced recruitment of GFP(+) /c-kit(+) progenitors to the ischemic area, resulting in an increased blood perfusion ratio. Histological analysis suggested that GFP(+) progenitors mobilized from bone marrow, integrated into blood vessels, and differentiated into VEGFR(+) cells. In addition, TUDCA decreased cellular senescence by reducing levels of p53, p21, and reactive oxygen species and increased nitric oxide. Transplantation of TUDCA-primed senescent EPCs in hind limb ischemia significantly improved blood vessel regeneration, as compared with senescent EPCs. Our results suggested that TUDCA promoted neovascularization by enhancing the mobilization of stem/progenitor cells from bone marrow, their differentiation into EPCs, and their integration with preexisting endothelial cells.


Subject(s)
Blood Vessels/cytology , Blood Vessels/drug effects , Hematopoietic Stem Cells/cytology , Stem Cells/cytology , Stem Cells/drug effects , Taurochenodeoxycholic Acid/pharmacology , Adult , Animals , Cell Differentiation/physiology , Endothelial Cells/cytology , Endothelial Cells/metabolism , Female , Hematopoietic Stem Cells/metabolism , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Nude , Neovascularization, Pathologic/pathology , Stem Cells/metabolism
15.
J Biol Chem ; 288(46): 32886-96, 2013 Nov 15.
Article in English | MEDLINE | ID: mdl-24072712

ABSTRACT

The peroxisome proliferator-activated receptor γ (PPARγ) is a central regulator of adipogenesis and modulates glucose and lipid metabolism. In this study, herpesvirus-associated ubiquitin-specific protease (HAUSP) was isolated as a binding partner of PPARγ. Both endogenous and exogenous PPARγ associated with HAUSP in co-immunoprecipitation analysis. HAUSP, but not the catalytically inactive HAUSP C223S mutant, increased the stability of both endogenous and exogenous PPARγ through its deubiquitinating activity. Site-directed mutagenesis experiments showed that the Lys(462) residue of PPARγ is critical for ubiquitination. HBX 41,108, a specific inhibitor of HAUSP, abolished the increase in PPARγ stability induced by HAUSP. In addition, knockdown of endogenous HAUSP using siRNA decreased PPARγ protein levels. HAUSP enhanced the transcriptional activity of both exogenous and endogenous PPARγ in luciferase activity assays. Quantitative RT-PCR analysis showed that HAUSP increased the transcript levels of PPARγ target genes in HepG2 cells, resulting in the enhanced uptake of glucose and fatty acids, and vice versa, upon siRNA knockdown of HAUSP. In vivo analysis using adenoviruses confirmed that HAUSP, but not the HAUSP C223S mutant, decreased blood glucose and triglyceride levels, which are associated with the increased expression of endogenous PPARγ and lipid accumulation in the liver. Our results demonstrate that the stability and activity of PPARγ are modulated by the deubiquitinating activity of HAUSP, which may be a target for the development of anti-diabetic drugs.


Subject(s)
PPAR gamma/metabolism , Transcription, Genetic/physiology , Ubiquitin Thiolesterase/metabolism , Ubiquitin-Specific Proteases/metabolism , Ubiquitination/physiology , Adenoviridae , Amino Acid Substitution , Animals , Biological Transport, Active/drug effects , Biological Transport, Active/physiology , Blood Glucose/genetics , Blood Glucose/metabolism , COS Cells , Chlorocebus aethiops , Fatty Acids/blood , Fatty Acids/genetics , Gene Knockdown Techniques , HeLa Cells , Hep G2 Cells , Humans , Indenes/pharmacology , Male , Mice , Mutagenesis, Site-Directed , Mutation, Missense , PPAR gamma/genetics , Protein Stability , Pyrazines/pharmacology , Transcription, Genetic/drug effects , Transduction, Genetic , Ubiquitin Thiolesterase/antagonists & inhibitors , Ubiquitin Thiolesterase/genetics , Ubiquitin-Specific Peptidase 7 , Ubiquitin-Specific Proteases/genetics , Ubiquitination/drug effects
16.
Hybridoma (Larchmt) ; 31(5): 358-63, 2012 Oct.
Article in English | MEDLINE | ID: mdl-23098303

ABSTRACT

The foot and mouth disease virus (FMDV) is an RNA virus composed of single stranded positive sense RNA. FMDV has been known to infect cloven-hoofed animals, including pigs, cattle, and sheep. FMDV is rapidly spreading outward to neighboring regions, often leading to a high mortality rate. Thus, early diagnosis of FMDV is critical to suppress propagation of FMDV and minimize economic losses. In this study, we report the generation and characterization of polyclonal and six monoclonal antibodies against VP1 through immunoblotting and immunofluorescence microscopy analyses. These VP1 antibodies will be useful as tools to detect serotypes A and O of FMDVs for diagnostic usage.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Viral/immunology , Antibody Specificity , Antigens, Viral/immunology , Capsid Proteins/immunology , Foot-and-Mouth Disease Virus/immunology , Animals , Antibodies, Monoclonal/biosynthesis , Antibodies, Viral/biosynthesis , Antigens, Viral/administration & dosage , Antigens, Viral/genetics , Binding Sites, Antibody , Blotting, Western , Capsid Proteins/administration & dosage , Capsid Proteins/genetics , Escherichia coli/genetics , Female , Foot-and-Mouth Disease Virus/chemistry , Gene Expression , Hybridomas/immunology , Immunization , Mice , Mice, Inbred BALB C , Microscopy, Fluorescence , Rabbits , Recombinant Proteins/administration & dosage , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Serotyping
SELECTION OF CITATIONS
SEARCH DETAIL
...