1
|
Lee JM, Lee WH, Cho SH, Park JW, Kwon HN, Kim JH, Lee SH, Yoon JH, Park S, Kim SC. DRG2 levels in prostate cancer cell lines predict response to PARP inhibitor during docetaxel treatment. Investig Clin Urol 2025; 66:56-66. [PMID: 39791585 PMCID: PMC11729229 DOI: 10.4111/icu.20240263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 11/25/2024] [Accepted: 12/02/2024] [Indexed: 01/30/2025] Open
Abstract
PURPOSE Developmentally regulated GTP-binding protein 2 (DRG2) regulates microtubule dynamics and G2/M arrest during docetaxel treatment. Poly ADP-ribose polymerase (PARP) acts as an important repair system for DNA damage caused by docetaxel treatment. This study investigated whether DRG2 expression affects response to PARP inhibitors (olaparib) using prostate cancer cell lines PC3, DU145, LNCaP-FGC, and LNCaP-LN3. MATERIALS AND METHODS The cell viability and DRG2 expression levels were assessed using colorimetric-based cell viability assay and western blot. Cells were transfected with DRG2 siRNA, and pcDNA6/V5-DRG2 was used to overexpress DRG2. Flow cytometry was applied for cell cycle assay and apoptosis analysis using the Annexing V cell death assay. RESULTS The expression of DRG2 was highest in LNCaP-LN3 and lowest in DU145 cells. Expressions of p53 in PC3, DU145, and the two LNCaP cell lines were null-type, high-expression, and medium-expression, respectively. In PC3 (DRG2 high, p53 null) cells, docetaxel increased G2/M arrest without apoptosis; however, subsequent treatment with olaparib promoted apoptosis. In DU145 and LNCaP-FGC (DRG2 low), docetaxel increased sub-G1 but not G2/M arrest and induced apoptosis, whereas olaparib had no additional effect. In LNCaP-LN3 (DRG2 high, p53 wild-type), docetaxel increased sub-G1 and G2/M arrest, furthermore olaparib enhanced cell death. Docetaxel and olaparib combination treatment had a slight effect on DRG2 knockdown PC3, but increased apoptosis in DRG2-overexpressed DU145 cells. CONCLUSIONS DRG2 and p53 expressions play an important role in prostate cancer cell lines treated with docetaxel, and DRG2 levels can predict the response to PARP inhibitors.
Collapse
Affiliation(s)
- Jeong Min Lee
- School of Biological Sciences, University of Ulsan, Ulsan, Korea
- Basic-Clinic Convergence Research Institute, University of Ulsan, Ulsan, Korea
| | - Won Hyeok Lee
- School of Biological Sciences, University of Ulsan, Ulsan, Korea
- Basic-Clinic Convergence Research Institute, University of Ulsan, Ulsan, Korea
| | - Seung Hyeon Cho
- School of Biological Sciences, University of Ulsan, Ulsan, Korea
- Basic-Clinic Convergence Research Institute, University of Ulsan, Ulsan, Korea
| | - Jeong Woo Park
- School of Biological Sciences, University of Ulsan, Ulsan, Korea
- Basic-Clinic Convergence Research Institute, University of Ulsan, Ulsan, Korea
| | - Hyuk Nam Kwon
- School of Biological Sciences, University of Ulsan, Ulsan, Korea
- Basic-Clinic Convergence Research Institute, University of Ulsan, Ulsan, Korea
| | - Ji Hye Kim
- Department of Pathology, Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan, Korea
| | - Sang Hun Lee
- Basic-Clinic Convergence Research Institute, University of Ulsan, Ulsan, Korea
- Department of Obstetrics & Gynecology, Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan, Korea
| | - Ji Hyung Yoon
- Basic-Clinic Convergence Research Institute, University of Ulsan, Ulsan, Korea
- Department of Urology, Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan, Korea
| | - Sungchan Park
- Basic-Clinic Convergence Research Institute, University of Ulsan, Ulsan, Korea
- Department of Urology, Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan, Korea
| | - Seong Cheol Kim
- Basic-Clinic Convergence Research Institute, University of Ulsan, Ulsan, Korea
- Department of Urology, Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan, Korea.
| |
Collapse
|
2
|
DRG2 Depletion Promotes Endothelial Cell Senescence and Vascular Endothelial Dysfunction. Int J Mol Sci 2022; 23:ijms23052877. [PMID: 35270019 PMCID: PMC8911374 DOI: 10.3390/ijms23052877] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Revised: 02/25/2022] [Accepted: 03/02/2022] [Indexed: 12/14/2022] Open
Abstract
Endothelial cell senescence is involved in endothelial dysfunction and vascular diseases. However, the detailed mechanisms of endothelial senescence are not fully understood. Here, we demonstrated that deficiency of developmentally regulated GTP-binding protein 2 (DRG2) induces senescence and dysfunction of endothelial cells. DRG2 knockout (KO) mice displayed reduced cerebral blood flow in the brain and lung blood vessel density. We also determined, by Matrigel plug assay, aorta ring assay, and in vitro tubule formation of primary lung endothelial cells, that deficiency in DRG2 reduced the angiogenic capability of endothelial cells. Endothelial cells from DRG2 KO mice showed a senescence phenotype with decreased cell growth and enhanced levels of p21 and phosphorylated p53, γH2AX, senescence-associated β-galactosidase (SA-β-gal) activity, and senescence-associated secretory phenotype (SASP) cytokines. DRG2 deficiency in endothelial cells upregulated arginase 2 (Arg2) and generation of reactive oxygen species. Induction of SA-β-gal activity was prevented by the antioxidant N-acetyl cysteine in endothelial cells from DRG2 KO mice. In conclusion, our results suggest that DRG2 is a key regulator of endothelial senescence, and its downregulation is probably involved in vascular dysfunction and diseases.
Collapse
|
3
|
ZC3H15 Correlates with a Poor Prognosis and Tumor Progression in Melanoma. BIOMED RESEARCH INTERNATIONAL 2022; 2021:8305299. [PMID: 34988227 PMCID: PMC8723872 DOI: 10.1155/2021/8305299] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Accepted: 10/25/2021] [Indexed: 11/28/2022]
Abstract
Zinc figure CCCH-type containing 15 (ZC3H15), also called developmentally regulated GTP-binding protein 1 (DRG1) family regulatory protein 1 (DFRP1), is a zinc finger containing protein. Despite playing a role in cellular signaling, it is found overexpressed in acute myeloid leukemia and also an independent prognostic marker in hepatocellular carcinoma patients. However, the biological effect of ZC3H15 in malignant melanoma (MM) remains unexplored. The expression of ZC3H15 in patients was analyzed using the R2: Genomics Analysis and Visualization Platform database. Immunohistochemical analysis, western blot, and qRT-PCR were used to detect ZC3H15 expression in melanoma tissues and cell lines. MTT, BrdU, flow cytometry assay, transwell, and western blot were performed to explore the proliferation, cell cycle, invasion, and migration of melanoma cells. We undertaken colony formation assay in vitro and tumor xenograft in vivo to detect the tumorigenicity of melanoma cells. In the present study, ZC3H15 was demonstrated highly expressed in melanoma tissues and cells. Elevated ZC3H15 impairs the survival of melanoma patients. Meanwhile, attenuation of ZC3H15 in melanoma cells inhibited cell proliferation and induced cycle arrest at G0/G1 phase. Consistently, the expression of cell cycle-related proteins cyclin dependent kinase 4 (CDK4), CDK6, and cyclin D1 (CCND1) was decreased while p21 was upregulated. Furthermore, we found the migration and invasion abilities were inhibited in ZC3H15-knockdown melanoma cells. In addition, downregulation of ZC3H15 resulted in inhibition of colony formation abilities in vitro and tumorigenesis in vivo. ZC3H15 promotes proliferation, migration/invasion, and tumorigenicity of melanoma cells. As a promising biomarker and therapeutic target in MM, ZC3H15 is worthy of further exploration.
Collapse
|
4
|
Westrip CAE, Zhuang Q, Hall C, Eaton CD, Coleman ML. Developmentally regulated GTPases: structure, function and roles in disease. Cell Mol Life Sci 2021; 78:7219-7235. [PMID: 34664086 PMCID: PMC8629797 DOI: 10.1007/s00018-021-03961-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 08/13/2021] [Accepted: 09/28/2021] [Indexed: 01/01/2023]
Abstract
GTPases are a large superfamily of evolutionarily conserved proteins involved in a variety of fundamental cellular processes. The developmentally regulated GTP-binding protein (DRG) subfamily of GTPases consists of two highly conserved paralogs, DRG1 and DRG2, both of which have been implicated in the regulation of cell proliferation, translation and microtubules. Furthermore, DRG1 and 2 proteins both have a conserved binding partner, DRG family regulatory protein 1 and 2 (DFRP1 and DFRP2), respectively, that prevents them from being degraded. Similar to DRGs, the DFRP proteins have also been studied in the context of cell growth control and translation. Despite these proteins having been implicated in several fundamental cellular processes they remain relatively poorly characterized, however. In this review, we provide an overview of the structural biology and biochemistry of DRG GTPases and discuss current understanding of DRGs and DFRPs in normal physiology, as well as their emerging roles in diseases such as cancer.
Collapse
Affiliation(s)
- Christian A E Westrip
- Tumour Oxygenase Group, Institute of Cancer and Genomic Sciences, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - Qinqin Zhuang
- Tumour Oxygenase Group, Institute of Cancer and Genomic Sciences, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
- Institute of Inflammation and Ageing, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - Charlotte Hall
- Tumour Oxygenase Group, Institute of Cancer and Genomic Sciences, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - Charlotte D Eaton
- Tumour Oxygenase Group, Institute of Cancer and Genomic Sciences, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
- Neurological Surgery, School of Medicine, University of California, 1450 Third St, San Francisco, CA, 94158, USA
| | - Mathew L Coleman
- Tumour Oxygenase Group, Institute of Cancer and Genomic Sciences, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK.
| |
Collapse
|
5
|
Park BS, Im HL, Yoon NA, Tu TH, Park JW, Kim JG, Lee BJ. Developmentally regulated GTP-binding protein-2 regulates adipocyte differentiation. Biochem Biophys Res Commun 2021; 578:1-6. [PMID: 34520979 DOI: 10.1016/j.bbrc.2021.08.081] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Accepted: 08/27/2021] [Indexed: 10/20/2022]
Abstract
Developmentally regulated GTP-binding protein 2 (DRG2) participates in the regulation of proliferation and differentiation of multiple cells. However, whether DRG2 regulates adipocyte differentiation and related metabolic control remains elusive. This study revealed increases in body weight and adiposity in DRG2 transgenic (Tg) mice overexpressing DRG2. Consistent with these results, DRG2 Tg mice showed increased expression of genes involved in adipogenesis and lipid metabolism in the white adipose tissue. DRG2 was also identified to control adipogenesis by cooperating with peroxisome proliferator activated receptor-γ (PPAR-γ) in cultured adipocytes. Overall, the findings of the current study suggest that DRG2 plays an active role in regulating adipocyte differentiation, and thus participates in the development of obesity during exposure to a fat-rich diet.
Collapse
Affiliation(s)
- Byong Seo Park
- Department of Biological Science, University of Ulsan, Ulsan, 44610, South Korea; Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon, 22012, South Korea
| | - Hye Li Im
- Department of Biological Science, University of Ulsan, Ulsan, 44610, South Korea
| | - Nal Ae Yoon
- Department of Biological Science, University of Ulsan, Ulsan, 44610, South Korea
| | - Thai Hien Tu
- Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon, 22012, South Korea
| | - Jeong Woo Park
- Department of Biological Science, University of Ulsan, Ulsan, 44610, South Korea
| | - Jae Geun Kim
- Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon, 22012, South Korea.
| | - Byung Ju Lee
- Department of Biological Science, University of Ulsan, Ulsan, 44610, South Korea.
| |
Collapse
|
6
|
Li BS, Jin AL, Zhou Z, Seo JH, Choi BM. DRG2 Accelerates Senescence via Negative Regulation of SIRT1 in Human Diploid Fibroblasts. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:7301373. [PMID: 34777693 PMCID: PMC8580627 DOI: 10.1155/2021/7301373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Accepted: 10/04/2021] [Indexed: 11/21/2022]
Abstract
Accumulating evidence suggests that developmentally regulated GTP-binding protein 2 (DRG2), an evolutionarily conserved GTP-binding protein, plays an important role in regulating cell growth, inflammation, and mitochondria dynamics. However, the effect of DRG2 in aging remains unclear. In this study, we found that endogenous DRG2 protein expression is upregulated in oxidative stress-induced premature senescence models and tissues of aged mice. Ectopic expression of DRG2 significantly promoted senescence-associated β-galactosidase (SA-β-gal) activity and inhibited cell growth, concomitant with increase in levels of acetyl (ac)-p53 (Lys382), ac-nuclear factor-kB (NF-κB) p65 (Lys310), p21 Waf1/Cip1 , and p16 Ink4a and a decrease in cyclin D1. In this process, reactive oxygen species (ROS) and phosphorylation of H2A histone family member X (H2A.X), forming γ-H2A.X, were enhanced. Mechanistically, ectopic expression of DRG2 downregulated Sirtuin-1 (SIRT1), resulting in augmented acetylation of p53 and NF-κB p65. Additionally, DRG2 knockdown significantly abolished oxidative stress-induced premature senescence. Our results provide a possible molecular mechanism for investigation of cellular senescence and aging regulated by DRG2.
Collapse
Affiliation(s)
- Bing Si Li
- Department of Biochemistry, Wonkwang University School of Medicine, Iksan, Jeonbuk 54538, Republic of Korea
| | - Ai Lin Jin
- Department of Biochemistry, Wonkwang University School of Medicine, Iksan, Jeonbuk 54538, Republic of Korea
| | - ZiQi Zhou
- Department of Herbology, Wonkwang University School of Korean Medicine, Iksan, Jeonbuk 54538, Republic of Korea
| | - Jae Ho Seo
- Department of Biochemistry, Wonkwang University School of Medicine, Iksan, Jeonbuk 54538, Republic of Korea
- Sarcopenia Total Solution Center, Wonkwang University School of Medicine, Iksan, Jeonbuk 54538, Republic of Korea
| | - Byung-Min Choi
- Department of Biochemistry, Wonkwang University School of Medicine, Iksan, Jeonbuk 54538, Republic of Korea
| |
Collapse
|
7
|
Kim SC, Lee WH, Kim SH, Abdulkhayevich AA, Park JW, Kim YM, Moon KH, Lee SH, Park S. Developmentally regulated GTP-binding protein 2 levels in prostate cancer cell lines impact docetaxel-induced apoptosis. Investig Clin Urol 2021; 62:485-495. [PMID: 34190439 PMCID: PMC8246011 DOI: 10.4111/icu.20200574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 02/18/2021] [Accepted: 04/01/2021] [Indexed: 11/30/2022] Open
Abstract
Purpose This study aimed to confirm the association between developmentally regulated GTP-binding protein 2 (DRG2) expression and docetaxel-induced apoptosis and to determine whether prostate cancer responses to docetaxel treatment differ with DRG2 expression. Materials and Methods PC3, DU145, and LNCaP prostate cancer cell lines were used. The MTT assay was used to determine cell viability. Western blotting analysis was performed using anti-DRG2 antibodies. Cells were transfected with 50 nmol DRG2 siRNA using an siRNA transfection reagent for DRG2 knockdown. The cell cycle was analyzed by using flow cytometry, and apoptosis was detected by using the Annexin V cell death assay. Results DRG2 expression differed in each prostate cancer cell line. Docetaxel reduced DRG2 expression in a dose-dependent manner. Upon DRG2 knockdown in prostate cancer cells, an increase in the sub-G1 phase was observed without a change in the G1 or G2/M phases. When 4 nM docetaxel was administered to DRG2 knockdown prostate cancer cell lines, an increase in the sub-G1 phase was observed without increasing the G2/M phase, which was similar to that in DU145 cells before DRG2 knockdown. In PC3 and DU145 cell lines, DRG2 knockdown increased docetaxel-induced Annexin V (+) apoptosis by 8.7 and 2.7 times, respectively. Conclusions In prostate cancer cells, DRG2 regulates G2/M arrest after docetaxel treatment. In prostate cancer cells with DRG2 knockdown, apoptosis increases without G2/M arrest in response to docetaxel treatment. These results show that inhibition of DRG2 expression can be useful to enhance docetaxel-induced apoptosis despite low-dose administration in castration-resistant prostate cancer.
Collapse
Affiliation(s)
- Seong Cheol Kim
- Department of Urology, Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan, Korea
| | - Won Hyeok Lee
- Biomedical Research Center, Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan, Korea
| | - Song Hee Kim
- Biomedical Research Center, Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan, Korea
| | | | - Jeong Woo Park
- Biomedical Research Center, Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan, Korea
| | - Young Min Kim
- Department of Pathology, Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan, Korea
| | - Kyung Hyun Moon
- Department of Urology, Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan, Korea
| | - Sang Hun Lee
- Department of Obstetrics and Gynecology, Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan, Korea
| | - Sungchan Park
- Department of Urology, Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan, Korea.
| |
Collapse
|
8
|
Yoon NA, Jung SJ, Choi SH, Ryu JH, Mani M, Lee UH, Vo MT, Jeon DY, Chung SW, Ju Lee B, Koh YW, Park SE, Shin YJ, Kang SS, Cho WJ, Cha HJ, Park JW. DRG2 supports the growth of primary tumors and metastases of melanoma by enhancing VEGF-A expression. FEBS J 2019; 287:2070-2086. [PMID: 31693298 DOI: 10.1111/febs.15125] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Revised: 08/20/2019] [Accepted: 11/04/2019] [Indexed: 12/15/2022]
Abstract
Malignant metastatic melanoma (MM) is the most lethal of all skin cancers, but detailed mechanisms for regulation of melanoma metastasis are not fully understood. Here, we demonstrated that developmentally regulated GTP-binding protein 2 (DRG2) is required for the growth of primary tumors and for metastasis. DRG2 expression was significantly increased in MM compared with primary melanoma (PM) and dysplastic nevi. A correlation between DRG2 expression and poor disease-specific survival in melanoma patients was also identified. Furthermore, inhibition of DRG2 suppressed the binding of Hypoxia-inducible factor 1α to the VEGF-A promoter region, expression of vascular endothelial growth factor (VEGF)-A, and formation of endothelial cell tubes. In experimental mice, DRG2 depletion inhibited the growth of PM and lung metastases and increased survival. These results identify DRG2 as a critical regulator of VEGF-A expression and of growth of PMs and lung metastases.
Collapse
Affiliation(s)
- Nal Ae Yoon
- Department of Biological Sciences, University of Ulsan, Korea
| | - Se Jin Jung
- Department of Pathology, Ulsan University Hospital, University of Ulsan College of Medicine, Korea
| | - Seong Hee Choi
- Department of Biological Sciences, University of Ulsan, Korea
| | - Jin Hyun Ryu
- Department of Anatomy and Convergence Medical Science, Institute of Health Sciences, School of Medicine, Gyeongsang National University, Jinju, Korea
| | | | - Unn Hwa Lee
- Department of Biological Sciences, University of Ulsan, Korea
| | - Mai-Tram Vo
- Department of Biological Sciences, University of Ulsan, Korea
| | - Do Yong Jeon
- Department of Biological Sciences, University of Ulsan, Korea
| | - Su Wol Chung
- Department of Biological Sciences, University of Ulsan, Korea
| | - Byung Ju Lee
- Department of Biological Sciences, University of Ulsan, Korea
| | - Young Wha Koh
- Department of Pathology, Ajou University School of Medicine, Suwon, Korea
| | - Soon Eun Park
- Department of Anesthesiology, Ulsan University Hospital, University of Ulsan College of Medicine, Korea
| | - Yong Joon Shin
- Department of Anesthesiology, Ulsan University Hospital, University of Ulsan College of Medicine, Korea
| | - Sang Soo Kang
- Department of Anatomy and Convergence Medical Science, Institute of Health Sciences, School of Medicine, Gyeongsang National University, Jinju, Korea
| | - Wha Ja Cho
- Department of Biological Sciences, University of Ulsan, Korea
| | - Hee Jeong Cha
- Department of Pathology, Ulsan University Hospital, University of Ulsan College of Medicine, Korea
| | - Jeong Woo Park
- Department of Biological Sciences, University of Ulsan, Korea
| |
Collapse
|
9
|
Mani M, Thao DT, Kim BC, Lee UH, Kim DJ, Jang SH, Back SH, Lee BJ, Cho WJ, Han IS, Park JW. DRG2 knockdown induces Golgi fragmentation via GSK3β phosphorylation and microtubule stabilization. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2019; 1866:1463-1474. [PMID: 31199931 DOI: 10.1016/j.bbamcr.2019.06.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/09/2019] [Revised: 05/19/2019] [Accepted: 06/04/2019] [Indexed: 01/01/2023]
Abstract
The perinuclear stacks of the Golgi apparatus maintained by dynamic microtubules are essential for cell migration. Activation of Akt (protein kinase B, PKB) negatively regulates glycogen synthase kinase 3β (GSK3β)-mediated tau phosphorylation, which enhances tau binding to microtubules and microtubule stability. In this study, experiments were performed on developmentally regulated GTP-binding protein 2 (DRG2)-stably knockdown HeLa cells to determine whether knockdown of DRG2 in HeLa cells treated with epidermal growth factor (EGF) affects microtubule dynamics, perinuclear Golgi stacking, and cell migration. Here, we show that DRG2 plays a key role in regulating microtubule stability, perinuclear Golgi stack formation, and cell migration. DRG2 knockdown prolonged the EGF receptor (EGFR) localization in endosome, enhanced Akt activity and inhibitory phosphorylation of GSK3β. Tau, a target of GSK3β, was hypo-phosphorylated in DRG2-knockdown cells and showed greater association with microtubules, resulting in microtubule stabilization. DRG2-knockdown cells showed defects in microtubule growth and microtubule organizing centers (MTOC), Golgi fragmentation, and loss of directional cell migration. These results reveal a previously unappreciated role for DRG2 in the regulation of perinuclear Golgi stacking and cell migration via its effects on GSK3β phosphorylation, and microtubule stability.
Collapse
Affiliation(s)
- Muralidharan Mani
- Department of Biological Sciences, University of Ulsan, Ulsan 680-749, Republic of Korea
| | - Dang Thi Thao
- Department of Biological Sciences, University of Ulsan, Ulsan 680-749, Republic of Korea
| | - Beom Chang Kim
- Department of Biological Sciences, University of Ulsan, Ulsan 680-749, Republic of Korea
| | - Unn Hwa Lee
- Department of Biological Sciences, University of Ulsan, Ulsan 680-749, Republic of Korea
| | - Dong Jun Kim
- Department of Biological Sciences, University of Ulsan, Ulsan 680-749, Republic of Korea
| | - Soo Hwa Jang
- Department of Biological Sciences, University of Ulsan, Ulsan 680-749, Republic of Korea
| | - Sung Hoon Back
- Department of Biological Sciences, University of Ulsan, Ulsan 680-749, Republic of Korea
| | - Byung Ju Lee
- Department of Biological Sciences, University of Ulsan, Ulsan 680-749, Republic of Korea
| | - Wha Ja Cho
- Metainflammation Research Center, University of Ulsan, Ulsan 680-749, Republic of Korea
| | - In-Seob Han
- Department of Biological Sciences, University of Ulsan, Ulsan 680-749, Republic of Korea.
| | - Jeong Woo Park
- Department of Biological Sciences, University of Ulsan, Ulsan 680-749, Republic of Korea.
| |
Collapse
|
10
|
Dang T, Jang SH, Back SH, Park JW, Han IS. DRG2 Deficiency Causes Impaired Microtubule Dynamics in HeLa Cells. Mol Cells 2018; 41:1045-1051. [PMID: 30453731 PMCID: PMC6315320 DOI: 10.14348/molcells.2018.0129] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Revised: 09/17/2018] [Accepted: 09/27/2018] [Indexed: 01/03/2023] Open
Abstract
The developmentally regulated GTP binding protein 2 (DRG2) is involved in the control of cell growth and differentiation. Here, we demonstrate that DRG2 regulates microtubule dynamics in HeLa cells. Analysis of live imaging of the plus-ends of microtubules with EB1-EGFP showed that DRG2 deficiency (shDRG2) significantly reduced the growth rate of HeLa cells. Depletion of DRG2 increased 'slow and long-lived' subpopulations, but decreased 'fast and short-lived' subpopulations of microtubules. Microtubule polymerization inhibitor exhibited a reduced response in shDRG2 cells. Using immunoprecipitation, we show that DRG2 interacts with tau, which regulates microtubule polymerization. Collectively, these data demonstrate that DRG2 may aid in affecting microtubule dynamics in HeLa cells.
Collapse
Affiliation(s)
- Thao Dang
- School of Biological Sciences, University of Ulsan, Ulsan 44610,
Korea
| | - Soo Hwa Jang
- School of Biological Sciences, University of Ulsan, Ulsan 44610,
Korea
| | - Sung Hoon Back
- School of Biological Sciences, University of Ulsan, Ulsan 44610,
Korea
| | - Jeong Woo Park
- School of Biological Sciences, University of Ulsan, Ulsan 44610,
Korea
| | - In-Seob Han
- School of Biological Sciences, University of Ulsan, Ulsan 44610,
Korea
| |
Collapse
|
11
|
Jiang BG, Wan ZH, Huang J, Li LM, Liu H, Fu SY, Yang Y, Zhang J, Yuan SX, Wang RY, Yang Y, Gu FM, Dong LW, Pan ZY, Zhou WP. Elevated ZC3H15 increases HCC growth and predicts poor survival after surgical resection. Oncotarget 2018; 7:37238-37249. [PMID: 27191988 PMCID: PMC5095072 DOI: 10.18632/oncotarget.9361] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Accepted: 04/24/2016] [Indexed: 02/07/2023] Open
Abstract
Zinc finger CCCH-type containing 15 (ZC3H15), also known as DRG family regulatory protein 1 (DFRP1), is a highly conserved eukaryotic protein that associates with active translation machinery. The aim of our study was to explore the clinical relevance and intrinsic functions of ZC3H15 in hepatocellular carcinoma (HCC). We constructed a cohort with 261 tumor and matched normal tissues from HCC patients. ZC3H15 protein and mRNA levels were determined using immunohistochemistry, western blot analysis, and quantitative polymerase chain reaction. ZC3H15 was highly expressed in the majority of HCC cases, and high ZC3H15 levels were significantly associated with high serum a-fetoprotein (AFP) levels (>20 ng/mL) and vascular invasion. Kaplan-Meier and Cox regression data indicated that elevated ZC3H15 was an independent predictor for HCC-specific disease-free survival (hazards ratio [HR], 1.789; 95% confidence interval [95% CI], 1.298-2.466 [P=0.0004]) and overall survival (HR, 1.613; 95% CI, 1.120-2.322 [P=0.0101]). Interaction of ZC3H15 with TRAF2 increased activation of NFκB signaling. These results suggest ZC3H15 is an independent prognostic marker in HCC patients that is clinicopathologically associated with tumor invasion and serum AFP levels.
Collapse
Affiliation(s)
- Bei-Ge Jiang
- Hepatic Surgery Department III, Eastern Hepatobiliary Surgery Hospital, The Second Military Medical University, National Innovation Alliance for Hepatitis & Liver Cancer, Shanghai, P. R. China
| | - Zheng-Hua Wan
- International Cooperation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Institute, The Second Military Medical University, Shanghai, P. R. China
| | - Jian Huang
- Hepatic Surgery Department III, Eastern Hepatobiliary Surgery Hospital, The Second Military Medical University, National Innovation Alliance for Hepatitis & Liver Cancer, Shanghai, P. R. China
| | - Li-Mei Li
- State Key Laboratory of Cell Biology, Institute of Biochemistry and Cell biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, P. R. China
| | - Hui Liu
- Hepatic Surgery Department III, Eastern Hepatobiliary Surgery Hospital, The Second Military Medical University, National Innovation Alliance for Hepatitis & Liver Cancer, Shanghai, P. R. China
| | - Si-Yuan Fu
- Hepatic Surgery Department III, Eastern Hepatobiliary Surgery Hospital, The Second Military Medical University, National Innovation Alliance for Hepatitis & Liver Cancer, Shanghai, P. R. China
| | - Yuan Yang
- Hepatic Surgery Department III, Eastern Hepatobiliary Surgery Hospital, The Second Military Medical University, National Innovation Alliance for Hepatitis & Liver Cancer, Shanghai, P. R. China
| | - Jin Zhang
- Hepatic Surgery Department III, Eastern Hepatobiliary Surgery Hospital, The Second Military Medical University, National Innovation Alliance for Hepatitis & Liver Cancer, Shanghai, P. R. China
| | - Shen-Xian Yuan
- Hepatic Surgery Department III, Eastern Hepatobiliary Surgery Hospital, The Second Military Medical University, National Innovation Alliance for Hepatitis & Liver Cancer, Shanghai, P. R. China
| | - Ruo-Yu Wang
- Hepatic Surgery Department III, Eastern Hepatobiliary Surgery Hospital, The Second Military Medical University, National Innovation Alliance for Hepatitis & Liver Cancer, Shanghai, P. R. China
| | - Yun Yang
- Hepatic Surgery Department III, Eastern Hepatobiliary Surgery Hospital, The Second Military Medical University, National Innovation Alliance for Hepatitis & Liver Cancer, Shanghai, P. R. China
| | - Fang-Ming Gu
- Hepatic Surgery Department III, Eastern Hepatobiliary Surgery Hospital, The Second Military Medical University, National Innovation Alliance for Hepatitis & Liver Cancer, Shanghai, P. R. China
| | - Li-Wei Dong
- International Cooperation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Institute, The Second Military Medical University, Shanghai, P. R. China
| | - Ze-Ya Pan
- Hepatic Surgery Department III, Eastern Hepatobiliary Surgery Hospital, The Second Military Medical University, National Innovation Alliance for Hepatitis & Liver Cancer, Shanghai, P. R. China
| | - Wei-Ping Zhou
- Hepatic Surgery Department III, Eastern Hepatobiliary Surgery Hospital, The Second Military Medical University, National Innovation Alliance for Hepatitis & Liver Cancer, Shanghai, P. R. China
| |
Collapse
|
12
|
Li Y, Kikuchi M, Li X, Gao Q, Xiong Z, Ren Y, Zhao R, Mao B, Kondo M, Irie N, Wang W. Weighted gene co-expression network analysis reveals potential genes involved in early metamorphosis process in sea cucumber Apostichopus japonicus. Biochem Biophys Res Commun 2017; 495:1395-1402. [PMID: 29180012 DOI: 10.1016/j.bbrc.2017.11.154] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Accepted: 11/22/2017] [Indexed: 11/15/2022]
Abstract
Sea cucumbers, one main class of Echinoderms, have a very fast and drastic metamorphosis process during their development. However, the molecular basis under this process remains largely unknown. Here we systematically examined the gene expression profiles of Japanese common sea cucumber (Apostichopus japonicus) for the first time by RNA sequencing across 16 developmental time points from fertilized egg to juvenile stage. Based on the weighted gene co-expression network analysis (WGCNA), we identified 21 modules. Among them, MEdarkmagenta was highly expressed and correlated with the early metamorphosis process from late auricularia to doliolaria larva. Furthermore, gene enrichment and differentially expressed gene analysis identified several genes in the module that may play key roles in the metamorphosis process. Our results not only provide a molecular basis for experimentally studying the development and morphological complexity of sea cucumber, but also lay a foundation for improving its emergence rate.
Collapse
Affiliation(s)
- Yongxin Li
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China; Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming 650203, China
| | - Mani Kikuchi
- Department of Cell Biological Science, Faculty of Advanced Life Science, Graduate School of Life Science, Hokkaido University, Sapporo, Hokkaido 001-0021, Japan
| | - Xueyan Li
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China
| | - Qionghua Gao
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China
| | - Zijun Xiong
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China; College of Forensic Medicine, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China; China National GeneBank, BGI-Shenzhen, Shenzhen, 518083, China
| | - Yandong Ren
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China; Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming 650203, China
| | - Ruoping Zhao
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China
| | - Bingyu Mao
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China
| | - Mariko Kondo
- Misaki Marine Biological Station, Graduate School of Science, The University of Tokyo, Kanagawa 238-0225, Japan
| | - Naoki Irie
- Department of Biological Sciences, Graduate School of Science, University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Wen Wang
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China.
| |
Collapse
|
13
|
Vo MT, Ko MS, Lee UH, Yoon EH, Lee BJ, Cho WJ, Ha CM, Kim K, Park JW. Developmentally regulated GTP-binding protein 2 depletion leads to mitochondrial dysfunction through downregulation of dynamin-related protein 1. Biochem Biophys Res Commun 2017; 486:1014-1020. [PMID: 28363867 DOI: 10.1016/j.bbrc.2017.03.154] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Accepted: 03/27/2017] [Indexed: 12/20/2022]
Abstract
Mitochondrial dynamics, including constant fusion and fission, play critical roles in maintaining mitochondrial morphology and function. Here, we report that developmentally regulated GTP-binding protein 2 (DRG2) regulates mitochondrial morphology by modulating the expression of the mitochondrial fission gene dynamin-related protein 1 (Drp1). shRNA-mediated silencing of DRG2 induced mitochondrial swelling, whereas expression of an shRNA-resistant version of DRG2 decreased mitochondrial swelling in DRG2-depleted cells. Analysis of the expression levels of genes involved in mitochondrial fusion and fission revealed that DRG2 depletion significantly decreased the level of Drp1. Overexpression of Drp1 rescued the defect in mitochondrial morphology induced by DRG2 depletion. DRG2 depletion reduced the mitochondrial membrane potential, oxygen consumption rate (OCR), and amount of mitochondrial DNA (mtDNA), whereas it increased reactive oxygen species (ROS) production and apoptosis. Taken together, our data demonstrate that DRG2 acts as a regulator of mitochondrial fission by controlling the expression of Drp1.
Collapse
Affiliation(s)
- Mai-Tram Vo
- Department of Biological Sciences, University of Ulsan, Ulsan 680-749, South Korea
| | - Myoung Seok Ko
- Asan Medical Center, 88 Olympic-ro 43-Gil, Songpa-gu, Seoul 05505, South Korea
| | - Unn Hwa Lee
- Department of Biological Sciences, University of Ulsan, Ulsan 680-749, South Korea
| | - Eun Hye Yoon
- Department of Biological Sciences, University of Ulsan, Ulsan 680-749, South Korea
| | - Byung Ju Lee
- Department of Biological Sciences, University of Ulsan, Ulsan 680-749, South Korea
| | - Wha Ja Cho
- Department of Biological Sciences, University of Ulsan, Ulsan 680-749, South Korea
| | - Chang Man Ha
- Research Division, Korea Brain Research Institute, Daegu 41068, South Korea
| | - Kyungjin Kim
- Research Division, Korea Brain Research Institute, Daegu 41068, South Korea; Department of Brain and Cognitive Science, DGIST, Daegu 711-873, South Korea
| | - Jeong Woo Park
- Department of Biological Sciences, University of Ulsan, Ulsan 680-749, South Korea.
| |
Collapse
|
14
|
Jang SH, Kim AR, Park NH, Park JW, Han IS. DRG2 Regulates G2/M Progression via the Cyclin B1-Cdk1 Complex. Mol Cells 2016; 39:699-704. [PMID: 27669826 PMCID: PMC5050535 DOI: 10.14348/molcells.2016.0149] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Revised: 08/25/2016] [Accepted: 08/25/2016] [Indexed: 02/01/2023] Open
Abstract
Developmentally regulated GTP-binding protein 2 (DRG2) plays an important role in cell growth. Here we explored the linkage between DRG2 and G2/M phase checkpoint function in cell cycle progression. We observed that knockdown of DRG2 in HeLa cells affected growth in a wound-healing assay, and tumorigenicity in nude mice xenografts. Flow cytometry assays and [(3)H] incorporation assays indicated that G2/M phase arrest was responsible for the decreased proliferation of these cells. Knockdown of DRG2 elicited down-regulation of the major mitotic promoting factor, the cyclin B1/Cdk1 complex, but up-regulation of the cell cycle arresting proteins, Wee1, Myt1, and p21. These findings identify a novel role of DRG2 in G2/M progression.
Collapse
Affiliation(s)
- Soo Hwa Jang
- School of Biological Sciences, University of Ulsan, Ulsan 44610,
Korea
| | - Ah-Ram Kim
- School of Biological Sciences, University of Ulsan, Ulsan 44610,
Korea
| | - Neung-Hwa Park
- Department of Internal Medicine, University of Ulsan College of Medicine, Ulsan University Hospital, Ulsan 44033,
Korea
| | - Jeong Woo Park
- School of Biological Sciences, University of Ulsan, Ulsan 44610,
Korea
| | - In-Seob Han
- School of Biological Sciences, University of Ulsan, Ulsan 44610,
Korea
| |
Collapse
|
15
|
Mani M, Lee UH, Yoon NA, Kim HJ, Ko MS, Seol W, Joe Y, Chung HT, Lee BJ, Moon CH, Cho WJ, Park JW. Developmentally regulated GTP-binding protein 2 coordinates Rab5 activity and transferrin recycling. Mol Biol Cell 2015; 27:334-48. [PMID: 26582392 PMCID: PMC4713135 DOI: 10.1091/mbc.e15-08-0558] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2015] [Accepted: 11/12/2015] [Indexed: 01/26/2023] Open
Abstract
The small GTPase Rab5 regulates the early endocytic pathway of transferrin (Tfn), and Rab5 deactivation is required for Tfn recycling. Developmentally regulated GTP-binding protein 2 is required for interaction between Rab5 and RabGAP5 on endosomes and acts as a key regulator for Rab5 deactivation and Tfn recycling. The small GTPase Rab5 regulates the early endocytic pathway of transferrin (Tfn), and Rab5 deactivation is required for Tfn recycling. Rab5 deactivation is achieved by RabGAP5, a GTPase-activating protein, on the endosomes. Here we report that recruitment of RabGAP5 is insufficient to deactivate Rab5 and that developmentally regulated GTP-binding protein 2 (DRG2) is required for Rab5 deactivation and Tfn recycling. DRG2 was associated with phosphatidylinositol 3-phosphate–containing endosomes. It colocalized and interacted with EEA1 and Rab5 on endosomes in a phosphatidylinositol 3-kinase–dependent manner. DRG2 depletion did not affect Tfn uptake and recruitment of RabGAP5 and Rac1 to Rab5 endosomes. However, it resulted in impairment of interaction between Rab5 and RabGAP5, Rab5 deactivation on endosomes, and Tfn recycling. Ectopic expression of shRNA-resistant DRG2 rescued Tfn recycling in DRG2-depleted cells. Our results demonstrate that DRG2 is an endosomal protein and a key regulator of Rab5 deactivation and Tfn recycling.
Collapse
Affiliation(s)
- Muralidharan Mani
- Department of Biological Sciences, University of Ulsan, Ulsan 680-749, Korea
| | - Unn Hwa Lee
- Department of Biological Sciences, University of Ulsan, Ulsan 680-749, Korea
| | - Nal Ae Yoon
- Department of Biological Sciences, University of Ulsan, Ulsan 680-749, Korea
| | - Hyo Jeong Kim
- Department of Biological Sciences, University of Ulsan, Ulsan 680-749, Korea
| | - Myoung Seok Ko
- Department of Medical Science, University of Ulsan College of Medicine, Seoul 138-736, Korea
| | - Wongi Seol
- Inam Institute for Brain Science, Wonkwang University Sanbon Hospital, Gunpo 435-040, Korea
| | - Yeonsoo Joe
- Department of Biological Sciences, University of Ulsan, Ulsan 680-749, Korea
| | - Hun Taeg Chung
- Department of Biological Sciences, University of Ulsan, Ulsan 680-749, Korea
| | - Byung Ju Lee
- Department of Biological Sciences, University of Ulsan, Ulsan 680-749, Korea
| | - Chang Hoon Moon
- Biomedical Research Center, Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan 682-060, Korea
| | - Wha Ja Cho
- Biomedical Research Center, Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan 682-060, Korea
| | - Jeong Woo Park
- Department of Biological Sciences, University of Ulsan, Ulsan 680-749, Korea
| |
Collapse
|
16
|
Xu C, Li H, Zhang L, Jia T, Duan L, Lu C. MicroRNA‑1915‑3p prevents the apoptosis of lung cancer cells by downregulating DRG2 and PBX2. Mol Med Rep 2015; 13:505-12. [PMID: 26572100 DOI: 10.3892/mmr.2015.4565] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2015] [Accepted: 10/29/2015] [Indexed: 11/05/2022] Open
Abstract
Micro (mi)RNAs are short non‑coding RNA molecules, which post‑transcriptionally regulate gene expression and exert key roles in cell growth, differentiation and apoptosis. In the present study, the mechanism and the function of miR‑1915‑3p in the apoptotic regulation of lung cancer cell lines (NCI‑H441 and NCI‑H1650) were investigated. The expression analysis confirmed that the expression of miR‑1915‑3p was markedly decreased in the apoptotic cells. The overexpression of miR‑1915‑3p in the lung cancer cells prevented apoptosis induced by etoposide. Developmentally regulated GTP‑binding protein 2 (DRG2) and pre‑B cell leukemia homeobox 2 (PBX2) were identified as downstream targets of miR‑1915‑3p, which was shown to bind directly to the 3'‑untranslated region of DRG2 and PBX2, subsequently lowering their mRNA and protein expression levels. Co‑expression of miR‑1915‑3p and DRG2/PBX2 in the NCI‑H441 and NCI‑H1650 cells partly circumvented the effect of miR‑1915‑3p on apoptosis. The results in the present study revealed that miR‑1915‑3p functions as a silencer of apoptosis, which regulates lung cancer apoptosis via targeting DRG2/PBX2, and consequently this miRNA may be a putative therapeutic target in lung cancer.
Collapse
Affiliation(s)
- Chengshan Xu
- Aviation Medicine Research Laboratory, Air Force General Hospital, PLA, Beijing 100142, P.R. China
| | - Hengheng Li
- Graduate Student Ministry of Education, Hebei North University, Zhangjiakou, Hebei 075000, P.R. China
| | - Ling Zhang
- Aviation Medicine Research Laboratory, Air Force General Hospital, PLA, Beijing 100142, P.R. China
| | - Tianjun Jia
- College of Medical Laboratory, Hebei North University, Zhangjiakou, Hebei 075000, P.R. China
| | - Lianning Duan
- Aviation Medicine Research Laboratory, Air Force General Hospital, PLA, Beijing 100142, P.R. China
| | - Chengrong Lu
- Aviation Medicine Research Laboratory, Air Force General Hospital, PLA, Beijing 100142, P.R. China
| |
Collapse
|
17
|
Developmentally regulated GTP-binding protein 2 ameliorates EAE by suppressing the development of TH17 cells. Clin Immunol 2013; 150:225-35. [PMID: 24463315 DOI: 10.1016/j.clim.2013.12.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2013] [Revised: 11/27/2013] [Accepted: 12/05/2013] [Indexed: 01/18/2023]
Abstract
Developmentally regulated GTP-binding protein 2 (DRG2) represents a novel subclass of GTP-binding proteins. We here report that transgenic overexpression of DRG2 in mice ameliorates experimental autoimmune encephalomyelitis (EAE), a murine model of multiple sclerosis (MS). The protective effect of DRG2 in EAE was mediated by the inhibition of the development of T(H)17 cells. DRG2 enhanced the activity of PPARγ, which led to an inhibition of the nuclear factor kappa B (NF-κB) activity and IL-6 production in antigen presenting cells and an inhibition of the development of T(H)17 cells. Our results demonstrate that DRG2 is an essential modulator of EAE.
Collapse
|
18
|
Ke K, Sul OJ, Kim WK, Lee MH, Ko MS, Suh JH, Kim HJ, Kim SY, Park JW, Choi HS. Overexpression of developmentally regulated GTP-binding protein-2 increases bone loss. Am J Physiol Endocrinol Metab 2013; 304:E703-10. [PMID: 23360825 DOI: 10.1152/ajpendo.00517.2012] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
The developmentally regulated GTP-binding protein-2 (DRG2) is a novel subclass of GTP-binding proteins. Many functional characteristics of osteoclasts (OC) are associated with small GTPases. We hypothesized that DRG2 affects bone mass via modulating OC activity. Using DRG2 transgenic mice, we investigated the role of DRG2 in bone remodeling. DRG2 overexpression caused a decrease in bone mass and an increase in the number and activity of OC in vivo. DRG2 overexpression increased fusion, spreading, survival, and resorption activity of OC in vitro. Downregulation of DRG2 by siRNA decreased fusion, spreading, and survival of OC, supporting the observations found in DRG2 transgenic OC. Transgenic mature OCs were larger, with actin rings and higher ERK, Akt, Rac1 and Rho activities than wild-type OCs. Inhibition of these proteins abolished the effects of DRG2 on formation of large OCs with actin rings, implying that DRG2 affects cytoskeleton reorganization in a Rac1/Rho/ERK/Akt-dependent manner. In summary, DRG2 is associated with survival and cytoskeleton organization of OC under influence of macrophage colony-stimulating factor, and its overexpression leads to elevated bone resorptive activity of OC, resulting in bone loss.
Collapse
Affiliation(s)
- Ke Ke
- Department of Biological Sciences (BK21 Program) and the Immunomodulation Research Center, University of Ulsan, Ulsan, Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Kim HJ, Ko MS, Kim HK, Cho WJ, Lee SH, Lee BJ, Park JW. Transcription factor Sp1 regulates basal transcription of the human DRG2 gene. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2011; 1809:184-90. [PMID: 21296692 DOI: 10.1016/j.bbagrm.2011.01.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/04/2010] [Revised: 01/18/2011] [Accepted: 01/20/2011] [Indexed: 11/30/2022]
Abstract
Developmentally regulated GTP-binding protein 2 (DRG2) is an evolutionarily conserved GTP-binding protein. DRG2 mRNA expression has been confirmed in many animal and human tissues. DRG2 is thought to play an essential role in the control of cell growth and differentiation. However, transcriptional regulation of DRG2 is largely unknown. To investigate the mechanisms controlling DRG2 expression, we cloned 1509bp of the 5'-flanking sequence of this gene. Deletion analysis showed that the region between -113 and -70 is essential for the basal level expression of the DRG2 gene in K562 human erythroleukemic cells. Mutation of a putative stimulating protein 1 (Sp1) regulatory site located at position -108 resulted in a significant decline in DRG2 promoter activity. Electrophoretic mobility shift assay and chromatin immunoprecipitation analysis revealed that Sp1 binds to this site. Knockdown of Sp1 expression using siRNA inhibited the promoter activation as well as the endogenous DRG2 transcriptional level. Taken together, these results demonstrate that basal expression level of DRG2 is regulated by the Sp1 transcription factor.
Collapse
Affiliation(s)
- Hyo Jeong Kim
- Department of Biological Sciences, University of Ulsan, Ulsan 680-749, Korea
| | | | | | | | | | | | | |
Collapse
|
20
|
Daugeron MC, Prouteau M, Lacroute F, Séraphin B. The highly conserved eukaryotic DRG factors are required for efficient translation in a manner redundant with the putative RNA helicase Slh1. Nucleic Acids Res 2010; 39:2221-33. [PMID: 21076151 PMCID: PMC3064805 DOI: 10.1093/nar/gkq898] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Eukaryotic and archaeal DRG factors are highly conserved proteins with characteristic GTPase motifs. This suggests their implication in a central biological process, which has so far escaped detection. We show here that the two Saccharomyces cerevisiae DRGs form distinct complexes, RBG1 and RBG2, and that the former co-fractionate with translating ribosomes. A genetic screen for triple synthetic interaction demonstrates that yeast DRGs have redundant function with Slh1, a putative RNA helicase also associating with translating ribosomes. Translation and cell growth are severely impaired in a triple mutant lacking both yeast DRGs and Slh1, but not in double mutants. This new genetic assay allowed us to characterize the roles of conserved motifs present in these proteins for efficient translation and/or association with ribosomes. Altogether, our results demonstrate for the first time a direct role of the highly conserved DRG factors in translation and indicate that this function is redundantly shared by three factors. Furthermore, our data suggest that important cellular processes are highly buffered against external perturbation and, consequently, that redundantly acting factors may escape detection in current high-throughput binary genetic interaction screens.
Collapse
Affiliation(s)
- Marie-Claire Daugeron
- Equipe Labellisée La Ligue, CGM, CNRS FRE3144, 1 Avenue de la Terrasse, 91198 Gif sur Yvette Cedex, Univ Paris-Sud, Orsay F-91405, France
| | | | | | | |
Collapse
|
21
|
Ishikawa K, Akiyama T, Ito K, Semba K, Inoue JI. Independent stabilizations of polysomal Drg1/Dfrp1 complex and non-polysomal Drg2/Dfrp2 complex in mammalian cells. Biochem Biophys Res Commun 2009; 390:552-6. [DOI: 10.1016/j.bbrc.2009.10.003] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2009] [Accepted: 10/02/2009] [Indexed: 11/16/2022]
|
22
|
Eswaran J, Bernad A, Ligos JM, Guinea B, Debreczeni JE, Sobott F, Parker SA, Najmanovich R, Turk BE, Knapp S. Structure of the human protein kinase MPSK1 reveals an atypical activation loop architecture. Structure 2008; 16:115-24. [PMID: 18184589 PMCID: PMC2194165 DOI: 10.1016/j.str.2007.10.026] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2007] [Revised: 10/11/2007] [Accepted: 10/18/2007] [Indexed: 11/30/2022]
Abstract
The activation segment of protein kinases is structurally highly conserved and central to regulation of kinase activation. Here we report an atypical activation segment architecture in human MPSK1 comprising a β sheet and a large α-helical insertion. Sequence comparisons suggested that similar activation segments exist in all members of the MPSK1 family and in MAST kinases. The consequence of this nonclassical activation segment on substrate recognition was studied using peptide library screens that revealed a preferred substrate sequence of X-X-P/V/I-ϕ-H/Y-T∗-N/G-X-X-X (ϕ is an aliphatic residue). In addition, we identified the GTPase DRG1 as an MPSK1 interaction partner and specific substrate. The interaction domain in DRG1 was mapped to the N terminus, leading to recruitment and phosphorylation at Thr100 within the GTPase domain. The presented data reveal an atypical kinase structural motif and suggest a role of MPSK1 regulating DRG1, a GTPase involved in regulation of cellular growth.
Collapse
Affiliation(s)
- Jeyanthy Eswaran
- Structural Genomics Consortium, Botnar Research Centre, University of Oxford, Oxford, United Kingdom
| | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Ishikawa K, Azuma S, Ikawa S, Semba K, Inoue JI. Identification of DRG family regulatory proteins (DFRPs): specific regulation of DRG1 and DRG2. Genes Cells 2005; 10:139-50. [PMID: 15676025 DOI: 10.1111/j.1365-2443.2005.00825.x] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
DRG1 and DRG2 comprise a highly conserved subfamily of GTP-binding proteins and are thought to act as critical regulators of cell growth. Their abnormal expressions may trigger cell transformation or cell cycle arrest. Our aim is to clarify their physiological functions and regulatory mechanisms. Here we report identification of novel proteins, DRG family regulatory protein (DFRP) 1 and DFRP2, which regulate expression of DRG proteins through specific binding. In transient transfection experiments, DFRP1 specifically binds DRG1, and DFRP2 preferentially binds DRG2. DFRPs provide stability to the target DRG proteins through physical association, possibly by blocking the poly-ubiquitination that would precede proteolysis of DRG proteins. DFRPs are highly conserved in eucaryotes, and the expression patterns of dfrp1 and drg1 transcripts in Xenopus embryos and tissues are similar, indicating that these genes work cooperatively in various types of eukaryotic cells. Immunofluorescence experiments have revealed that the interaction between DRG1 and DFRP1 may occur in the cytoplasm. We generated dfrp1- knockout cells and found that endogenous expression of DRG1 is regulated by DFRP1, confirming that DFRP1 is a specific up-regulator of DRG1 in vivo. On the basis of these results, we propose that DRG1 and DRG2 are regulated differently despite their structural similarities.
Collapse
Affiliation(s)
- Kosuke Ishikawa
- Division of Cellular and Molecular Biology, Department of Cancer Biology, Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minatoku, Tokyo 108-8639, Japan
| | | | | | | | | |
Collapse
|