1
|
Platko K, Gyulay G, Lebeau PF, MacDonald ME, Lynn EG, Byun JH, Igdoura SA, Holden RM, Roubtsova A, Seidah NG, Krepinsky JC, Austin RC. GDF10 is a negative regulator of vascular calcification. J Biol Chem 2024; 300:107805. [PMID: 39307303 DOI: 10.1016/j.jbc.2024.107805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 08/23/2024] [Accepted: 09/11/2024] [Indexed: 10/27/2024] Open
Abstract
Cardiovascular mortality is particularly high and increasing in patients with chronic kidney disease, with vascular calcification (VC) as a major pathophysiologic feature. VC is a highly regulated biological process similar to bone formation involving osteogenic transdifferentiation of vascular smooth muscle cells (VSMCs). We have previously demonstrated that loss of T-cell death-associated gene 51 (TDAG51) expression leads to an attenuation of medial VC. We now show a significant induction of circulating levels of growth differentiation factor 10 (GDF10) in TDAG51-/- mice, which was of interest due to its established role as an inhibitor of osteoblast differentiation. The objective of this study was to examine the role of GDF10 in the osteogenic transdifferentiation of VSMCs. Using primary mouse and human VSMCs, as well as ex vivo aortic ring cultures, we demonstrated that treatment with recombinant human (rh) GDF10 mitigated phosphate-mediated hydroxyapatite (HA) mineral deposition. Furthermore, ex vivo aortic rings from GDF10-/- mice exhibited increased HA deposition compared to C57BL/6J controls. To explain our observations, we identified that rhGDF10 treatment reduced protein expression of runt-related transcription factor 2, a key driver of osteogenic transdifferentiation of VSMCs and VC. In support of these findings, in vivo treatment with rhGDF10 attenuated VD3-induced VC. Furthermore, we demonstrated an increase in circulating GDF10 in patients with chronic kidney disease with clinically defined severe VC, as assessed by coronary artery calcium score. Thus, our studies identify GDF10 as a novel inhibitor of mineral deposition and as such, may represent a potential novel biomarker and therapeutic target for the detection and management of VC.
Collapse
Affiliation(s)
- Khrystyna Platko
- Department of Medicine, Division of Nephrology, McMaster University, and The Research Institute of St Joe's Hamilton, Hamilton, Ontario, Canada
| | - Gabriel Gyulay
- Department of Medicine, Division of Nephrology, McMaster University, and The Research Institute of St Joe's Hamilton, Hamilton, Ontario, Canada
| | - Paul F Lebeau
- Department of Medicine, Division of Nephrology, McMaster University, and The Research Institute of St Joe's Hamilton, Hamilton, Ontario, Canada
| | - Melissa E MacDonald
- Department of Medicine, Division of Nephrology, McMaster University, and The Research Institute of St Joe's Hamilton, Hamilton, Ontario, Canada
| | - Edward G Lynn
- Department of Medicine, Division of Nephrology, McMaster University, and The Research Institute of St Joe's Hamilton, Hamilton, Ontario, Canada
| | - Jae Hyun Byun
- Department of Medicine, Division of Nephrology, McMaster University, and The Research Institute of St Joe's Hamilton, Hamilton, Ontario, Canada
| | - Suleiman A Igdoura
- Department of Biology, McMaster University Medical Centre, Hamilton, Ontario, Canada; Department of Pathology and Molecular Medicine, Queen's University, Kingston, Ontario, Canada
| | - Rachel M Holden
- Department of Medicine, Queen's University, Kingston, Ontario, Canada
| | - Anna Roubtsova
- The Institut de Recherches Cliniques de Montréal (IRCM), Affiliated with Université de Montréal, Montréal, Quebec, Canada
| | - Nabil G Seidah
- The Institut de Recherches Cliniques de Montréal (IRCM), Affiliated with Université de Montréal, Montréal, Quebec, Canada
| | - Joan C Krepinsky
- Department of Medicine, Division of Nephrology, McMaster University, and The Research Institute of St Joe's Hamilton, Hamilton, Ontario, Canada.
| | - Richard C Austin
- Department of Medicine, Division of Nephrology, McMaster University, and The Research Institute of St Joe's Hamilton, Hamilton, Ontario, Canada.
| |
Collapse
|
2
|
Song X, Zhou L, Yang W, Li X, Ma J, Qi K, Liang R, Li M, Xie L, Su T, Huang D, Liang B. PHLDA1 is a P53 target gene involved in P53-mediated cell apoptosis. Mol Cell Biochem 2024; 479:653-664. [PMID: 37155089 DOI: 10.1007/s11010-023-04752-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Accepted: 04/26/2023] [Indexed: 05/10/2023]
Abstract
Pleckstrin homeolike domain, family A, member 1 (PHLDA1) is a multifunctional protein that plays diverse roles in A variety of biological processes, including cell death, and hence its altered expression has been found in different types of cancer. Although studies have shown a regulatory relationship between p53 and PHLDA1, the molecular mechanism is still unclear. Especially, the role of PHLDA1 in the process of apoptosis is still controversial. In this study, we found that the expression of PHLDA1 in human cervical cancer cell lines was correlated with the up-expression of p53 after treatment with apoptosis-inducing factors. Subsequently, the binding site and the binding effect of p53 on the promoter region of PHLDA1 were verified by our bioinformatics data analysis and luciferase reporter assay. Indeed, we used CRISPR-Cas9 to knockout the p53 gene in HeLa cells and further confirmed that p53 can bind to the promoter region of PHLDA1 gene, and then directly regulate the expression of PHLDA1 by recruiting P300 and CBP to change the acetylation and methylation levels in the promoter region. Finally, a series of gain-of-function experiments further confirmed that p53 re-expression in HeLap53-/- cell can up-regulate the reduction of PHLDA1 caused by p53 knockout, and affect cell apoptosis and proliferation. Our study is the first to explore the regulatory mechanism of p53 on PHLDA1 by using the p53 gene knockout cell model, which further proves that PHLDA1 is a target-gene in p53-mediated apoptosis, and reveals the important role of PHLDA1 in cell fate determination.
Collapse
Affiliation(s)
- Xuhong Song
- Center for Cancer Research, Shantou University Medical College, Shantou, Guangdong, China
- Section of Cell Biology and Genetics, Shantou University Medical College, Shantou, Guangdong, China
| | - Lulu Zhou
- Section of Cell Biology and Genetics, Shantou University Medical College, Shantou, Guangdong, China
| | - Wenrui Yang
- Section of Cell Biology and Genetics, Shantou University Medical College, Shantou, Guangdong, China
| | - Xinyan Li
- Section of Cell Biology and Genetics, Shantou University Medical College, Shantou, Guangdong, China
| | - Jiazi Ma
- Section of Cell Biology and Genetics, Shantou University Medical College, Shantou, Guangdong, China
| | - Kun Qi
- Section of Cell Biology and Genetics, Shantou University Medical College, Shantou, Guangdong, China
| | - Rui Liang
- Section of Cell Biology and Genetics, Shantou University Medical College, Shantou, Guangdong, China
| | - Meijing Li
- Section of Cell Biology and Genetics, Shantou University Medical College, Shantou, Guangdong, China
| | - Lingzhu Xie
- Section of Cell Biology and Genetics, Shantou University Medical College, Shantou, Guangdong, China
| | - Tin Su
- Section of Cell Biology and Genetics, Shantou University Medical College, Shantou, Guangdong, China
| | - Dongyang Huang
- Section of Cell Biology and Genetics, Shantou University Medical College, Shantou, Guangdong, China.
| | - Bin Liang
- Section of Cell Biology and Genetics, Shantou University Medical College, Shantou, Guangdong, China.
- Biomedical Research Center, Shantou University Medical College, Shantou, Guangdong, China.
| |
Collapse
|
3
|
Yousof TR, Bouchard CC, Alb M, Lynn EG, Lhoták S, Jiang H, MacDonald M, Li H, Byun JH, Makda Y, Athanasopoulos M, Maclean KN, Cherrington NJ, Naqvi A, Igdoura SA, Krepinsky JC, Steinberg GR, Austin RC. Restoration of the ER stress response protein TDAG51 in hepatocytes mitigates NAFLD in mice. J Biol Chem 2024; 300:105655. [PMID: 38237682 PMCID: PMC10875272 DOI: 10.1016/j.jbc.2024.105655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 12/24/2023] [Accepted: 12/27/2023] [Indexed: 02/16/2024] Open
Abstract
Endoplasmic reticulum stress is associated with insulin resistance and the development of nonalcoholic fatty liver disease. Deficiency of the endoplasmic reticulum stress response T-cell death-associated gene 51 (TDAG51) (TDAG51-/-) in mice promotes the development of high-fat diet (HFD)-induced obesity, fatty liver, and hepatic insulin resistance. However, whether this effect is due specifically to hepatic TDAG51 deficiency is unknown. Here, we report that hepatic TDAG51 protein levels are consistently reduced in multiple mouse models of liver steatosis and injury as well as in liver biopsies from patients with liver disease compared to normal controls. Delivery of a liver-specific adeno-associated virus (AAV) increased hepatic expression of a TDAG51-GFP fusion protein in WT, TDAG51-/-, and leptin-deficient (ob/ob) mice. Restoration of hepatic TDAG51 protein was sufficient to increase insulin sensitivity while reducing body weight and fatty liver in HFD fed TDAG51-/- mice and in ob/ob mice. TDAG51-/- mice expressing ectopic TDAG51 display improved Akt (Ser473) phosphorylation, post-insulin stimulation. HFD-fed TDAG51-/- mice treated with AAV-TDAG51-GFP displayed reduced lipogenic gene expression, increased beta-oxidation and lowered hepatic and serum triglycerides, findings consistent with reduced liver weight. Further, AAV-TDAG51-GFP-treated TDAG51-/- mice exhibited reduced hepatic precursor and cleaved sterol regulatory-element binding proteins (SREBP-1 and SREBP-2). In vitro studies confirmed the lipid-lowering effect of TDAG51 overexpression in oleic acid-treated Huh7 cells. These studies suggest that maintaining hepatic TDAG51 protein levels represents a viable therapeutic approach for the treatment of obesity and insulin resistance associated with nonalcoholic fatty liver disease.
Collapse
Affiliation(s)
- Tamana R Yousof
- Division of Nephrology, Department of Medicine, The Research Institute of St. Joe's Hamilton and the Hamilton Centre for Kidney Research, McMaster University, Hamilton, Ontario, Canada
| | - Celeste C Bouchard
- Division of Nephrology, Department of Medicine, The Research Institute of St. Joe's Hamilton and the Hamilton Centre for Kidney Research, McMaster University, Hamilton, Ontario, Canada
| | - Mihnea Alb
- Division of Nephrology, Department of Medicine, The Research Institute of St. Joe's Hamilton and the Hamilton Centre for Kidney Research, McMaster University, Hamilton, Ontario, Canada
| | - Edward G Lynn
- Division of Nephrology, Department of Medicine, The Research Institute of St. Joe's Hamilton and the Hamilton Centre for Kidney Research, McMaster University, Hamilton, Ontario, Canada
| | - Sárka Lhoták
- Division of Nephrology, Department of Medicine, The Research Institute of St. Joe's Hamilton and the Hamilton Centre for Kidney Research, McMaster University, Hamilton, Ontario, Canada
| | - Hua Jiang
- Department of Pediatrics, School of Medicine, University of Colorado Health Sciences Center, Aurora, Colorado, USA
| | - Melissa MacDonald
- Division of Nephrology, Department of Medicine, The Research Institute of St. Joe's Hamilton and the Hamilton Centre for Kidney Research, McMaster University, Hamilton, Ontario, Canada
| | - Hui Li
- Department of Pharmacology and Toxicology, University of Arizona, Tucson, Arizona, USA
| | - Jae H Byun
- Division of Nephrology, Department of Medicine, The Research Institute of St. Joe's Hamilton and the Hamilton Centre for Kidney Research, McMaster University, Hamilton, Ontario, Canada
| | - Yumna Makda
- Division of Nephrology, Department of Medicine, The Research Institute of St. Joe's Hamilton and the Hamilton Centre for Kidney Research, McMaster University, Hamilton, Ontario, Canada
| | | | - Kenneth N Maclean
- Department of Pediatrics, School of Medicine, University of Colorado Health Sciences Center, Aurora, Colorado, USA
| | - Nathan J Cherrington
- Department of Pharmacology and Toxicology, University of Arizona, Tucson, Arizona, USA
| | - Asghar Naqvi
- Department of Pathology and Molecular Medicine, St. Joseph's Healthcare Hamilton, McMaster University, Hamilton, Ontario, Canada
| | - Suleiman A Igdoura
- Department of Biology, McMaster University, Hamilton, Ontario, Canada; Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Joan C Krepinsky
- Division of Nephrology, Department of Medicine, The Research Institute of St. Joe's Hamilton and the Hamilton Centre for Kidney Research, McMaster University, Hamilton, Ontario, Canada; Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, Ontario, Canada
| | - Gregory R Steinberg
- Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, Ontario, Canada; Division of Endocrinology, Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Richard C Austin
- Division of Nephrology, Department of Medicine, The Research Institute of St. Joe's Hamilton and the Hamilton Centre for Kidney Research, McMaster University, Hamilton, Ontario, Canada; Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, Ontario, Canada.
| |
Collapse
|
4
|
Park E, Jeon H, Lee N, Yu J, Park H, Satoh T, Akira S, Furuyama T, Lee C, Choi J, Rho J. TDAG51 promotes transcription factor FoxO1 activity during LPS-induced inflammatory responses. EMBO J 2023; 42:e111867. [PMID: 37203866 PMCID: PMC10308371 DOI: 10.15252/embj.2022111867] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 04/28/2023] [Accepted: 05/03/2023] [Indexed: 05/20/2023] Open
Abstract
Tight regulation of Toll-like receptor (TLR)-mediated inflammatory responses is important for innate immunity. Here, we show that T-cell death-associated gene 51 (TDAG51/PHLDA1) is a novel regulator of the transcription factor FoxO1, regulating inflammatory mediator production in the lipopolysaccharide (LPS)-induced inflammatory response. TDAG51 induction by LPS stimulation was mediated by the TLR2/4 signaling pathway in bone marrow-derived macrophages (BMMs). LPS-induced inflammatory mediator production was significantly decreased in TDAG51-deficient BMMs. In TDAG51-deficient mice, LPS- or pathogenic Escherichia coli infection-induced lethal shock was reduced by decreasing serum proinflammatory cytokine levels. The recruitment of 14-3-3ζ to FoxO1 was competitively inhibited by the TDAG51-FoxO1 interaction, leading to blockade of FoxO1 cytoplasmic translocation and thereby strengthening FoxO1 nuclear accumulation. TDAG51/FoxO1 double-deficient BMMs showed significantly reduced inflammatory mediator production compared with TDAG51- or FoxO1-deficient BMMs. TDAG51/FoxO1 double deficiency protected mice against LPS- or pathogenic E. coli infection-induced lethal shock by weakening the systemic inflammatory response. Thus, these results indicate that TDAG51 acts as a regulator of the transcription factor FoxO1, leading to strengthened FoxO1 activity in the LPS-induced inflammatory response.
Collapse
Affiliation(s)
- Eui‐Soon Park
- Department of Microbiology and Molecular BiologyChungnam National UniversityDaejeonKorea
| | - Hyoeun Jeon
- Department of Microbiology and Molecular BiologyChungnam National UniversityDaejeonKorea
| | - Nari Lee
- Department of Microbiology and Molecular BiologyChungnam National UniversityDaejeonKorea
| | - Jiyeon Yu
- Department of Microbiology and Molecular BiologyChungnam National UniversityDaejeonKorea
| | - Hye‐Won Park
- Department of Microbiology and Molecular BiologyChungnam National UniversityDaejeonKorea
| | - Takashi Satoh
- Department of Immune Regulation, Graduate School of Medical and Dental SciencesTokyo Medical and Dental UniversityTokyoJapan
| | - Shizuo Akira
- Laboratory of Host Defense, WPI Immunology Frontier Research CenterOsaka UniversityOsakaJapan
| | - Tatsuo Furuyama
- Department of Clinical ExaminationKagawa Prefectural University of Health SciencesKagawaJapan
| | - Chul‐Ho Lee
- Laboratory Animal CenterKorea Research Institute of Bioscience & Biotechnology (KRIBB)DaejeonKorea
| | - Jong‐Soon Choi
- Division of Life ScienceKorea Basic Science Institute (KBSI)DaejeonKorea
| | - Jaerang Rho
- Department of Microbiology and Molecular BiologyChungnam National UniversityDaejeonKorea
| |
Collapse
|
5
|
Jeon H, Amarasekara DS, Lee N, Park HW, Yu J, Rho J. TDAG51 deficiency attenuates dextran sulfate sodium-induced colitis in mice. Sci Rep 2022; 12:20619. [PMID: 36450854 PMCID: PMC9712416 DOI: 10.1038/s41598-022-24873-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 11/22/2022] [Indexed: 12/12/2022] Open
Abstract
Inflammatory bowel disease (IBD), including ulcerative colitis and Crohn's disease, is a group of chronic inflammatory diseases of the gastrointestinal tract. Although the multifactorial etiology of IBD pathogenesis is relatively well documented, the regulatory factors that confer a risk of IBD pathogenesis remain less explored. In this study, we report that T-cell death-associated gene 51 (TDAG51/PHLDA1) is a novel regulator of the development of dextran sulfate sodium (DSS)-induced colitis in mice. TDAG51 expression was elevated in the colon tissues of DSS-induced experimental colitis mice. TDAG51 deficiency protected mice against acute DSS-induced lethality and body weight changes and disease severity. DSS-induced structural damage and mucus secretion in colon tissues were significantly reduced in TDAG51-deficient mice compared with wild-type mice. We observed similar results in a DSS-induced chronic colitis mouse model. Finally, we showed that the production of inflammatory mediators, including proinflammatory enzymes, molecules and cytokines, was decreased in DSS-treated TDAG51-deficient mice compared with DSS-treated wild-type mice. Thus, we demonstrated that TDAG51 deficiency plays a protective role against DSS-induced colitis by decreasing the production of inflammatory mediators in mice. These findings suggest that TDAG51 is a novel regulator of the development of DSS-induced colitis and is a potential therapeutic target for IBD.
Collapse
Affiliation(s)
- Hyoeun Jeon
- Department of Microbiology and Molecular Biology, College of Bioscience and Biotechnology, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon, 34134, Korea
| | - Dulshara Sachini Amarasekara
- Department of Microbiology and Molecular Biology, College of Bioscience and Biotechnology, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon, 34134, Korea
| | - Nari Lee
- Department of Microbiology and Molecular Biology, College of Bioscience and Biotechnology, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon, 34134, Korea
| | - Hye-Won Park
- Department of Microbiology and Molecular Biology, College of Bioscience and Biotechnology, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon, 34134, Korea
| | - Jiyeon Yu
- Department of Microbiology and Molecular Biology, College of Bioscience and Biotechnology, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon, 34134, Korea
| | - Jaerang Rho
- Department of Microbiology and Molecular Biology, College of Bioscience and Biotechnology, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon, 34134, Korea.
| |
Collapse
|
6
|
Xu J, Bi G, Luo Q, Liu Y, Liu T, Li L, Zeng Q, Wang Q, Wang Y, Yu J, Yi P. PHLDA1 Modulates the Endoplasmic Reticulum Stress Response and is required for Resistance to Oxidative Stress-induced Cell Death in Human Ovarian Cancer Cells. J Cancer 2021; 12:5486-5493. [PMID: 34405011 PMCID: PMC8364641 DOI: 10.7150/jca.45262] [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: 02/24/2020] [Accepted: 04/21/2021] [Indexed: 11/10/2022] Open
Abstract
Objective: Pleckstrin homology-like domain family A member 1 (PHLDA1) has been implicated in the regulation of apoptosis in a variety of normal cell types and cancers. However, its precise pathophysiological functions remain unclear. Here, we examined the expression of PHLDA1 in human ovarian cancer (OvCa), the most lethal gynecologic malignancy, and investigated its functions in vitro. Materials and Methods: The expression of PHLDA1 was detected by reverse-transcription quantitative PCR (RT-qPCR), immunohistochemical analysis, or western blotting, silencing of PHLDA was achieved by shRNA, cell proliferation was detected by MTT assay, apoptosis was detected by flow cytometric analysis, PHLDA1 transcriptional activity was detected by dual luciferase reporter assay. Results: PHLDA1 mRNA levels were significantly higher in serous OvCa specimens compared with normal ovarian tissue, confirmed by immunohistochemical staining of PHLDA1 protein, which also indicated the expression was predominantly cytoplasmic. Bioinformatics analysis of publicly available datasets indicated that PHLDA1 expression in clinical specimens was significantly associated with disease stage, progression-free survival, and overall survival. In human OvCa cell lines, shRNA-mediated silencing of PHLDA1 expression enhanced apoptosis after exposure to oxidative stress- and endoplasmic reticulum stress-inducing agents. PHLDA1 silencing increased not the expression of anti-apoptotic or autophagy-related proteins, but the expression of ER stress response-associated proteins. Conclusion: PHLDA1 modulates the susceptibility of human OvCa cells to apoptosis via the endoplasmic reticulum stress response pathway.
Collapse
Affiliation(s)
- Jing Xu
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing 401120, PR China
| | - Gang Bi
- Department of Urology, Daping Hospital, Army Medical University, Chongqing 400042, PR China
| | - Qingya Luo
- Department of Obstetrics and Gynecology, Daping Hospital, Army Medical University, Chongqing 400042, PR China
| | - Yi Liu
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing 401120, PR China.,Department of Obstetrics and Gynecology, Daping Hospital, Army Medical University, Chongqing 400042, PR China
| | - Tao Liu
- Department of Obstetrics and Gynecology, Daping Hospital, Army Medical University, Chongqing 400042, PR China
| | - Lanfang Li
- Department of Obstetrics and Gynecology, Daping Hospital, Army Medical University, Chongqing 400042, PR China
| | - Qi Zeng
- Department of Obstetrics and Gynecology, Daping Hospital, Army Medical University, Chongqing 400042, PR China
| | - Qien Wang
- The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA
| | - Yufeng Wang
- Cancer Research Institute of Jilin University, The First Hospital of Jilin University, Changchun 130021, PR China
| | - Jianhua Yu
- The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA.,Department of Hematology and Hematopoietic Cell Transplantation, Comprehensive Cancer Center, City of Hope National Medical Center, Duarte, California 91010, USA
| | - Ping Yi
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing 401120, PR China.,Department of Obstetrics and Gynecology, Daping Hospital, Army Medical University, Chongqing 400042, PR China
| |
Collapse
|
7
|
Han C, Yan P, He T, Cheng J, Zheng W, Zheng LT, Zhen X. PHLDA1 promotes microglia-mediated neuroinflammation via regulating K63-linked ubiquitination of TRAF6. Brain Behav Immun 2020; 88:640-653. [PMID: 32353516 DOI: 10.1016/j.bbi.2020.04.064] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 04/24/2020] [Accepted: 04/25/2020] [Indexed: 12/22/2022] Open
Abstract
Microglia-mediated neuroinflammation plays an important role in the progression of neurodegenerative diseases including Parkinson's disease (PD). Pleckstrin homology-like domain family A member 1 (PHLDA1) plays an important role in immunological regulation, particularly in the Toll-like receptor-mediated immune response. Here, we explored the potential roles of PHLDA1 in microglia-mediated inflammation and neuronal protection. We found that PHLDA1 expression was rapidly increased in response to inflammatory stimuli in microglia cells in vivo or in vitro. Knockdown of PHLDA1 using adeno-associated virus serotype (AAV) ameliorated MPTP-induced motor deficits and inhibited neuroinflammation in mice. In support of this observation in vivo, we found that LPS-induced proinflammatory gene expression, including TNF-α, IL-1β, iNOS, and COX-2, was decreased in PHLDA1-deficient microglial cells. Mechanistic studies demonstrated that increased expression of PHLDA1, upon LPS stimulation in microglia, led to direct interaction with TRAF6 and enhanced its K63-linked ubiquitination-mediated NF-κB signaling activation. PHLDA1 deficiency interfered with TRAF6 K63-linked ubiquitination and inhibited microglial inflammatory responses. These findings reveal the first evidence that PHLDA1 is an important modulator of microglial function that is associated with microglia-mediated dopaminergic neurotoxicity. The data therefore provided the first evidence that PHLDA1 may be a potent modulator for neuroinflammation, and PHLDA1 may be a novel drug target for treatment of neuroinflammation-related diseases such as PD.
Collapse
Affiliation(s)
- Chaojun Han
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu 215123, China; College of Pharmaceutical Sciences and the Collaborative Innovation Center for Brain Science, Soochow University, Suzhou, China
| | - Pengju Yan
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu 215123, China; College of Pharmaceutical Sciences and the Collaborative Innovation Center for Brain Science, Soochow University, Suzhou, China
| | - Tao He
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu 215123, China; College of Pharmaceutical Sciences and the Collaborative Innovation Center for Brain Science, Soochow University, Suzhou, China
| | - Junjie Cheng
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu 215123, China; College of Pharmaceutical Sciences and the Collaborative Innovation Center for Brain Science, Soochow University, Suzhou, China
| | - Wenhua Zheng
- Center of Reproduction, Development and Aging, Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Macau SAR, China
| | - Long-Tai Zheng
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu 215123, China; College of Pharmaceutical Sciences and the Collaborative Innovation Center for Brain Science, Soochow University, Suzhou, China.
| | - Xuechu Zhen
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu 215123, China; College of Pharmaceutical Sciences and the Collaborative Innovation Center for Brain Science, Soochow University, Suzhou, China.
| |
Collapse
|
8
|
Platko K, Lebeau PF, Gyulay G, Lhoták Š, MacDonald ME, Pacher G, Hyun Byun J, Boivin FJ, Igdoura SA, Cutz JC, Bridgewater D, Ingram AJ, Krepinsky JC, Austin RC. TDAG51 (T-Cell Death-Associated Gene 51) Is a Key Modulator of Vascular Calcification and Osteogenic Transdifferentiation of Arterial Smooth Muscle Cells. Arterioscler Thromb Vasc Biol 2020; 40:1664-1679. [PMID: 32434409 DOI: 10.1161/atvbaha.119.313779] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
OBJECTIVE Cardiovascular disease is the primary cause of mortality in patients with chronic kidney disease. Vascular calcification (VC) in the medial layer of the vessel wall is a unique and prominent feature in patients with advanced chronic kidney disease and is now recognized as an important predictor and independent risk factor for cardiovascular and all-cause mortality in these patients. VC in chronic kidney disease is triggered by the transformation of vascular smooth muscle cells (VSMCs) into osteoblasts as a consequence of elevated circulating inorganic phosphate (Pi) levels, due to poor kidney function. The objective of our study was to investigate the role of TDAG51 (T-cell death-associated gene 51) in the development of medial VC. METHODS AND RESULTS Using primary mouse and human VSMCs, we found that TDAG51 is induced in VSMCs by Pi and is expressed in the medial layer of calcified human vessels. Furthermore, the transcriptional activity of RUNX2 (Runt-related transcription factor 2), a well-established driver of Pi-mediated VC, is reduced in TDAG51-/- VSMCs. To explain these observations, we identified that TDAG51-/- VSMCs express reduced levels of the type III sodium-dependent Pi transporter, Pit-1, a solute transporter, a solute transporter, a solute transporter responsible for cellular Pi uptake. Significantly, in response to hyperphosphatemia induced by vitamin D3, medial VC was attenuated in TDAG51-/- mice. CONCLUSIONS Our studies highlight TDAG51 as an important mediator of Pi-induced VC in VSMCs through the downregulation of Pit-1. As such, TDAG51 may represent a therapeutic target for the prevention of VC and cardiovascular disease in patients with chronic kidney disease.
Collapse
Affiliation(s)
- Khrystyna Platko
- From the Division of Nephrology, Department of Medicine (K.P., P.F.L., G.G., Š.L., M.E.M., G.P., J.H.B., A.J.I., J.C.K., R.C.A.), McMaster University, and The Research Institute of St. Joseph's Hamilton, ON, Canada
| | - Paul F Lebeau
- From the Division of Nephrology, Department of Medicine (K.P., P.F.L., G.G., Š.L., M.E.M., G.P., J.H.B., A.J.I., J.C.K., R.C.A.), McMaster University, and The Research Institute of St. Joseph's Hamilton, ON, Canada
| | - Gabriel Gyulay
- From the Division of Nephrology, Department of Medicine (K.P., P.F.L., G.G., Š.L., M.E.M., G.P., J.H.B., A.J.I., J.C.K., R.C.A.), McMaster University, and The Research Institute of St. Joseph's Hamilton, ON, Canada
| | - Šárka Lhoták
- From the Division of Nephrology, Department of Medicine (K.P., P.F.L., G.G., Š.L., M.E.M., G.P., J.H.B., A.J.I., J.C.K., R.C.A.), McMaster University, and The Research Institute of St. Joseph's Hamilton, ON, Canada
| | - Melissa E MacDonald
- From the Division of Nephrology, Department of Medicine (K.P., P.F.L., G.G., Š.L., M.E.M., G.P., J.H.B., A.J.I., J.C.K., R.C.A.), McMaster University, and The Research Institute of St. Joseph's Hamilton, ON, Canada
| | - Giusepina Pacher
- From the Division of Nephrology, Department of Medicine (K.P., P.F.L., G.G., Š.L., M.E.M., G.P., J.H.B., A.J.I., J.C.K., R.C.A.), McMaster University, and The Research Institute of St. Joseph's Hamilton, ON, Canada
| | - Jae Hyun Byun
- From the Division of Nephrology, Department of Medicine (K.P., P.F.L., G.G., Š.L., M.E.M., G.P., J.H.B., A.J.I., J.C.K., R.C.A.), McMaster University, and The Research Institute of St. Joseph's Hamilton, ON, Canada
| | - Felix J Boivin
- Department of Pathology and Molecular Medicine (F.J.B., S.A.I., D.B.), McMaster University Medical Centre, Hamilton, ON, Canada
| | - Suleiman A Igdoura
- Department of Pathology and Molecular Medicine (F.J.B., S.A.I., D.B.), McMaster University Medical Centre, Hamilton, ON, Canada.,Department of Biology (S.A.I.), McMaster University Medical Centre, Hamilton, ON, Canada
| | - Jean-Claude Cutz
- Department of Pathology and Molecular Medicine (J.-C.C.), McMaster University, and The Research Institute of St. Joseph's Hamilton, ON, Canada
| | - Darren Bridgewater
- Department of Pathology and Molecular Medicine (F.J.B., S.A.I., D.B.), McMaster University Medical Centre, Hamilton, ON, Canada
| | - Alistair J Ingram
- From the Division of Nephrology, Department of Medicine (K.P., P.F.L., G.G., Š.L., M.E.M., G.P., J.H.B., A.J.I., J.C.K., R.C.A.), McMaster University, and The Research Institute of St. Joseph's Hamilton, ON, Canada
| | - Joan C Krepinsky
- From the Division of Nephrology, Department of Medicine (K.P., P.F.L., G.G., Š.L., M.E.M., G.P., J.H.B., A.J.I., J.C.K., R.C.A.), McMaster University, and The Research Institute of St. Joseph's Hamilton, ON, Canada
| | - Richard C Austin
- From the Division of Nephrology, Department of Medicine (K.P., P.F.L., G.G., Š.L., M.E.M., G.P., J.H.B., A.J.I., J.C.K., R.C.A.), McMaster University, and The Research Institute of St. Joseph's Hamilton, ON, Canada
| |
Collapse
|
9
|
PHLDA1 Does Not Contribute Directly to Heat Shock-Induced Apoptosis of Spermatocytes. Int J Mol Sci 2019; 21:ijms21010267. [PMID: 31906015 PMCID: PMC6982182 DOI: 10.3390/ijms21010267] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 12/25/2019] [Accepted: 12/27/2019] [Indexed: 01/09/2023] Open
Abstract
Spermatocytes are among the most heat-sensitive cells and the exposure of testes to heat shock results in their Heat Shock Factor 1 (HSF1)-mediated apoptosis. Several lines of evidence suggest that pleckstrin-homology-like domain family A, member 1 (PHLDA1) plays a role in promoting heat shock-induced cell death in spermatogenic cells, yet its precise physiological role is not well understood. Aiming to elucidate the hypothetical role of PHLDA1 in HSF1-mediated apoptosis of spermatogenic cells we characterized its expression in mouse testes during normal development and after heat shock. We stated that transcription of Phlda1 is upregulated by heat shock in many adult mouse organs including the testes. Analyzes of the Phlda1 expression during postnatal development indicate that it is expressed in pre-meiotic or somatic cells of the testis. It starts to be transcribed much earlier than spermatocytes are fully developed and its transcripts and protein products do not accumulate further in the later stages. Moreover, neither heat shock nor expression of constitutively active HSF1 results in the accumulation of PHLDA1 protein in meiotic and post-meiotic cells although both conditions induce massive apoptosis of spermatocytes. Furthermore, the overexpression of PHLDA1 in NIH3T3 cells leads to cell detachment, yet classical apoptosis is not observed. Therefore, our findings indicate that PHLDA1 cannot directly contribute to the heat-induced apoptosis of spermatocytes. Instead, PHLDA1 could hypothetically participate in death of spermatocytes indirectly via activation of changes in the somatic or pre-meiotic cells present in the testes.
Collapse
|
10
|
Yun H, Park ES, Choi S, Shin B, Yu J, Yu J, Amarasekara DS, Kim S, Lee N, Choi JS, Choi Y, Rho J. TDAG51 is a crucial regulator of maternal care and depressive-like behavior after parturition. PLoS Genet 2019; 15:e1008214. [PMID: 31251738 PMCID: PMC6599150 DOI: 10.1371/journal.pgen.1008214] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2018] [Accepted: 05/27/2019] [Indexed: 12/11/2022] Open
Abstract
Postpartum depression is a severe emotional and mental disorder that involves maternal care defects and psychiatric illness. Postpartum depression is closely associated with a combination of physical changes and physiological stress during pregnancy or after parturition in stress-sensitive women. Although postpartum depression is relatively well known to have deleterious effects on the developing fetus, the influence of genetic risk factors on the development of postpartum depression remains unclear. In this study, we discovered a novel function of T cell death-associated gene 51 (TDAG51/PHLDA1) in the regulation of maternal and depressive-like behavior. After parturition, TDAG51-deficient dams showed impaired maternal behavior in pup retrieving, nursing and nest building tests. In contrast to the normal dams, the TDAG51-deficient dams also exhibited more sensitive depressive-like behaviors after parturition. Furthermore, changes in the expression levels of various maternal and depressive-like behavior-associated genes regulating neuroendocrine factor and monoamine neurotransmitter levels were observed in TDAG51-deficient postpartum brain tissues. These findings indicate that TDAG51 plays a protective role against maternal care defects and depressive-like behavior after parturition. Thus, TDAG51 is a maternal care-associated gene that functions as a crucial regulator of maternal and depressive-like behavior after parturition. Postpartum depression is a severe emotional and mental disease that can affect women typically after parturition. However, the genetic risk factors associated with the development of postpartum depression are still largely unknown. We discovered a novel function of T cell death-associated gene 51 (TDAG51) in the regulation of maternal behavior and postpartum depression. We report that TDAG51 deficiency induces depressive-like and abnormal maternal behavior after parturition. The loss of TDAG51 in postpartum brain tissues induces changes in the expression levels of various maternal and depressive-like behavior-associated genes that regulate the levels of neuroendocrine factors and monoamine neurotransmitters. TDAG51 is a maternal care-associated gene that functions as a crucial regulator of maternal and depressive-like behavior after parturition.
Collapse
Affiliation(s)
- Hyeongseok Yun
- Department of Microbiology and Molecular Biology, Chungnam National University, Daejeon, Korea
| | - Eui-Soon Park
- Department of Microbiology and Molecular Biology, Chungnam National University, Daejeon, Korea
| | - Seunga Choi
- Department of Microbiology and Molecular Biology, Chungnam National University, Daejeon, Korea
| | - Bongjin Shin
- Department of Microbiology and Molecular Biology, Chungnam National University, Daejeon, Korea
| | - Jungeun Yu
- Department of Microbiology and Molecular Biology, Chungnam National University, Daejeon, Korea
| | - Jiyeon Yu
- Department of Microbiology and Molecular Biology, Chungnam National University, Daejeon, Korea
| | | | - Sumi Kim
- Department of Microbiology and Molecular Biology, Chungnam National University, Daejeon, Korea
| | - Nari Lee
- Department of Microbiology and Molecular Biology, Chungnam National University, Daejeon, Korea
| | - Jong-Soon Choi
- Division of Life Science, Korea Basic Science Institute, Daejeon, Korea
| | - Yongwon Choi
- Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Jaerang Rho
- Department of Microbiology and Molecular Biology, Chungnam National University, Daejeon, Korea
- * E-mail:
| |
Collapse
|
11
|
Coleman SK, Cao AW, Rebalka IA, Gyulay G, Chambers PJ, Tupling AR, Austin RC, Hawke TJ. The Pleckstrin homology like domain family member, TDAG51, is temporally regulated during skeletal muscle regeneration. Biochem Biophys Res Commun 2017; 495:499-505. [PMID: 29127005 DOI: 10.1016/j.bbrc.2017.11.019] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Accepted: 11/02/2017] [Indexed: 01/23/2023]
Abstract
The capacity for skeletal muscle to repair from daily insults as well as larger injuries is a vital component to maintaining muscle health over our lifetime. Given the importance of skeletal muscle for our physical and metabolic well-being, identifying novel factors mediating the growth and repair of skeletal muscle will thus build our foundational knowledge and help lead to potential therapeutic avenues for muscle wasting disorders. To that end, we investigated the expression of T-cell death associated gene 51 (TDAG51) during skeletal muscle repair and studied the response of TDAG51 deficient (TDAG51-/-) mice to chemically-induced muscle damage. TDAG51 mRNA and protein expression within uninjured skeletal muscle is almost undetectable but, in response to chemically-induced muscle damage, protein levels increase by 5 days post-injury and remain elevated for up to 10 days of regeneration. To determine the impact of TDAG51 deletion on skeletal muscle form and function, we compared adult male TDAG51-/- mice with age-matched wild-type (WT) mice. Body and muscle mass were not different between the two groups, however, in situ muscle testing demonstrated a significant reduction in force production both before and after fatiguing contractions in TDAG51-/- mice. During the early phases of the regenerative process (5 days post-injury), TDAG51-/- muscles display a significantly larger area of degenerating muscle tissue concomitant with significantly less regenerating area compared to WT (as demonstrated by embryonic myosin heavy chain expression). Despite these early deficits in regeneration, TDAG51-/- muscles displayed no morphological deficits by 10 days post injury compared to WT mice. Taken together, the data presented herein demonstrate TDAG51 expression to be upregulated in damaged skeletal muscle and its absence attenuates the early phases of muscle regeneration.
Collapse
Affiliation(s)
- Samantha K Coleman
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, L8S 4L8, Canada
| | - Andrew W Cao
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, L8S 4L8, Canada
| | - Irena A Rebalka
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, L8S 4L8, Canada
| | - Gabriel Gyulay
- Department of Medicine, Division of Nephrology, McMaster University, Hamilton, ON, L8S 4L8, Canada
| | - Paige J Chambers
- Department of Kinesiology, University of Waterloo, Waterloo, ON, N2L 3G1, Canada
| | - A Russell Tupling
- Department of Kinesiology, University of Waterloo, Waterloo, ON, N2L 3G1, Canada
| | - Richard C Austin
- Department of Medicine, Division of Nephrology, McMaster University, Hamilton, ON, L8S 4L8, Canada
| | - Thomas J Hawke
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, L8S 4L8, Canada.
| |
Collapse
|
12
|
Jiao HW, Jia XX, Zhao TJ, Rong H, Zhang JN, Cheng Y, Zhu HP, Xu KL, Guo SY, Shi QY, Zhang H, Wang FY, Chen CF, Du L. Up-regulation of TDAG51 is a dependent factor of LPS-induced RAW264.7 macrophages proliferation and cell cycle progression. Immunopharmacol Immunotoxicol 2016; 38:124-30. [PMID: 26873343 DOI: 10.3109/08923973.2016.1138968] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
CONTEXT As a component of the outer membrane in Gram-negative bacteria, lipopolysaccharide (LPS)-induced proliferation and cell cycle progression of monocytes/macrophages. It has been suggested that the proapoptotic T-cell death-associated gene 51 (TDAG51) might be associated with cell proliferation and cell cycle progression; however, its role in the interaction between LPS and macrophages remains unclear. OBJECTIVE We attempted to elucidate the role(s) of TDAG51 played in the interaction between LPS and macrophages. MATERIALS AND METHODS We investigated TDAG51 expression in RAW264.7 cells stimulated with LPS and examined the effects of RNA interference-mediated TDAG51 down-regulation. We used CCK-8 assay and flow cytometry analysis to evaluate the interaction between TDAG51 and LPS-induced proliferation and cell cycle progression in RAW264.7 cells. RESULTS Our findings indicate that TDAG51 is up-regulated in LPS-stimulated RAW264.7 cells, the TDAG51 siRNA effectively reduced TDAG51 protein up-regulation following LPS stimulation in RAW264.7 cells, the significant changes of the proliferation and cell cycle progression of RAW264.7 cells in TDAG51 Knockdown RAW264.7 cells treated with LPS were observed. CONCLUSION These findings suggested that TDAG51 up-regulation is a dependent event during LPS-mediated proliferation and cell cycle progression, and which increase our understanding of the interaction mechanism between LPS and macrophages.
Collapse
Affiliation(s)
- Han-Wei Jiao
- a College of Agriculture, Hainan University, Hainan Key Lab of Tropical Animal Reproduction & Breeding and Epidemic Disease Research, Animal Genetic Engineering Key Lab of Haikou , Haidian Island , Haikou , People's Republic of China and
| | - Xiao-Xiao Jia
- a College of Agriculture, Hainan University, Hainan Key Lab of Tropical Animal Reproduction & Breeding and Epidemic Disease Research, Animal Genetic Engineering Key Lab of Haikou , Haidian Island , Haikou , People's Republic of China and
| | - Tian-Jing Zhao
- a College of Agriculture, Hainan University, Hainan Key Lab of Tropical Animal Reproduction & Breeding and Epidemic Disease Research, Animal Genetic Engineering Key Lab of Haikou , Haidian Island , Haikou , People's Republic of China and
| | - Hui Rong
- a College of Agriculture, Hainan University, Hainan Key Lab of Tropical Animal Reproduction & Breeding and Epidemic Disease Research, Animal Genetic Engineering Key Lab of Haikou , Haidian Island , Haikou , People's Republic of China and
| | - Jia-Ning Zhang
- a College of Agriculture, Hainan University, Hainan Key Lab of Tropical Animal Reproduction & Breeding and Epidemic Disease Research, Animal Genetic Engineering Key Lab of Haikou , Haidian Island , Haikou , People's Republic of China and
| | - Ying Cheng
- a College of Agriculture, Hainan University, Hainan Key Lab of Tropical Animal Reproduction & Breeding and Epidemic Disease Research, Animal Genetic Engineering Key Lab of Haikou , Haidian Island , Haikou , People's Republic of China and
| | - Hua-Pei Zhu
- a College of Agriculture, Hainan University, Hainan Key Lab of Tropical Animal Reproduction & Breeding and Epidemic Disease Research, Animal Genetic Engineering Key Lab of Haikou , Haidian Island , Haikou , People's Republic of China and
| | - Kai-Lian Xu
- a College of Agriculture, Hainan University, Hainan Key Lab of Tropical Animal Reproduction & Breeding and Epidemic Disease Research, Animal Genetic Engineering Key Lab of Haikou , Haidian Island , Haikou , People's Republic of China and
| | - Shi-Yu Guo
- a College of Agriculture, Hainan University, Hainan Key Lab of Tropical Animal Reproduction & Breeding and Epidemic Disease Research, Animal Genetic Engineering Key Lab of Haikou , Haidian Island , Haikou , People's Republic of China and
| | - Qiao-Yun Shi
- a College of Agriculture, Hainan University, Hainan Key Lab of Tropical Animal Reproduction & Breeding and Epidemic Disease Research, Animal Genetic Engineering Key Lab of Haikou , Haidian Island , Haikou , People's Republic of China and
| | - Hui Zhang
- b College of Animal Science and Technology, Shihezi University , North 4th Road, Shihezi , People's Republic of China
| | - Feng-Yang Wang
- a College of Agriculture, Hainan University, Hainan Key Lab of Tropical Animal Reproduction & Breeding and Epidemic Disease Research, Animal Genetic Engineering Key Lab of Haikou , Haidian Island , Haikou , People's Republic of China and
| | - Chuang-Fu Chen
- b College of Animal Science and Technology, Shihezi University , North 4th Road, Shihezi , People's Republic of China
| | - Li Du
- a College of Agriculture, Hainan University, Hainan Key Lab of Tropical Animal Reproduction & Breeding and Epidemic Disease Research, Animal Genetic Engineering Key Lab of Haikou , Haidian Island , Haikou , People's Republic of China and
| |
Collapse
|
13
|
Nagai MA. Pleckstrin homology-like domain, family A, member 1 ( PHLDA1) and cancer. Biomed Rep 2016; 4:275-281. [PMID: 26998263 DOI: 10.3892/br.2016.580] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Accepted: 01/19/2016] [Indexed: 12/12/2022] Open
Abstract
Pleckstrin homology-like domain, family A, member 1 (PHLDA1) encodes a member of an evolutionarily conserved pleckstrin homology-related domain protein family. It was first identified as a potential transcription factor required for Fas expression and activation-induced apoptosis in mouse T cell hybridomas. The exact molecular and biological functions of PHLDA1 remain to be elucidated. However, its expression is induced by a variety of external stimuli and there is evidence that it may function as a transcriptional activator that acts as a mediator of apoptosis, proliferation, differentiation and cell migration dependent on the cellular type and context. Recently, PHLDA1 has received attention due to its association with cancer. In the present review, the current knowledge of PHLDA1 protein structure, expression regulation and function is summarized. In addition, the current data in the literature is reviewed with regards to the role of PHLDA1 in cancer pathogenesis.
Collapse
Affiliation(s)
- Maria Aparecida Nagai
- Discipline of Oncology, Department of Radiology, Faculty of Medicine, University of São Paulo, Laboratory of Molecular Genetics, Center for Translational Research in Oncology, Cancer Institute of São Paulo, São Paulo, SP 01246-000, Brazil
| |
Collapse
|
14
|
Shin B, Yu J, Park ES, Choi S, Yu J, Hwang JM, Yun H, Chung YH, Hong KS, Choi JS, Takami M, Rho J. Secretion of a truncated osteopetrosis-associated transmembrane protein 1 (OSTM1) mutant inhibits osteoclastogenesis through down-regulation of the B lymphocyte-induced maturation protein 1 (BLIMP1)-nuclear factor of activated T cells c1 (NFATc1) axis. J Biol Chem 2014; 289:35868-81. [PMID: 25359771 DOI: 10.1074/jbc.m114.589614] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Genetic mutations in osteoclastogenic genes are closely associated with osteopetrotic bone diseases. Genetic defects in OSTM1 (osteopetrosis-associated transmembrane protein 1) cause autosomal recessive osteopetrosis in humans. In particular, OSTM1 mutations that exclude the transmembrane domain might lead to the production of a secreted form of truncated OSTM1. However, the precise role of the secreted form of truncated OSTM1 remains unknown. In this study, we analyzed the functional role of truncated OSTM1 in osteoclastogenesis. Here, we showed that a secreted form of truncated OSTM1 binds to the cell surface of osteoclast (OC) precursors and inhibits the formation of multinucleated OCs through the reduction of cell fusion and survival. Truncated OSTM1 significantly inhibited the expression of OC marker genes through the down-regulation of the BLIMP1 (B lymphocyte-induced maturation protein 1)-NFATc1 (nuclear factor of activated T cells c1) axis. Finally, we demonstrated that truncated OSTM1 reduces lipopolysaccharide-induced bone destruction in vivo. Thus, these findings suggest that autosomal recessive osteopetrosis patients with an OSTM1 gene mutation lacking the transmembrane domain produce a secreted form of truncated OSTM1 that inhibits osteoclastogenesis.
Collapse
Affiliation(s)
- Bongjin Shin
- From the Department of Microbiology and Molecular Biology and
| | - Jungeun Yu
- From the Department of Microbiology and Molecular Biology and
| | - Eui-Soon Park
- From the Department of Microbiology and Molecular Biology and
| | - Seunga Choi
- From the Department of Microbiology and Molecular Biology and
| | - Jiyeon Yu
- From the Department of Microbiology and Molecular Biology and
| | - Jung Me Hwang
- From the Department of Microbiology and Molecular Biology and
| | - Hyeongseok Yun
- From the Department of Microbiology and Molecular Biology and
| | - Young-Ho Chung
- the Division of Life Science, Korea Basic Science Institute, 169-148 Gwahak-ro, Yuseong-gu, Daejeon 305-806, Korea, and
| | - Kwan Soo Hong
- the Division of Life Science, Korea Basic Science Institute, 169-148 Gwahak-ro, Yuseong-gu, Daejeon 305-806, Korea, and the Graduate School of Analytical Science and Technology (GRAST), Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon 305-764, Korea
| | - Jong-Soon Choi
- the Division of Life Science, Korea Basic Science Institute, 169-148 Gwahak-ro, Yuseong-gu, Daejeon 305-806, Korea, and the Graduate School of Analytical Science and Technology (GRAST), Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon 305-764, Korea
| | - Masamichi Takami
- the Department of Biochemistry, School of Dentistry, Showa University, 1-5-8 Hatanodai, Shinagawaku 142-8555, Japan
| | - Jaerang Rho
- From the Department of Microbiology and Molecular Biology and the Graduate School of Analytical Science and Technology (GRAST), Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon 305-764, Korea,
| |
Collapse
|
15
|
Murata T, Sato T, Kamoda T, Moriyama H, Kumazawa Y, Hanada N. Differential susceptibility to hydrogen sulfide-induced apoptosis between PHLDA1-overexpressing oral cancer cell lines and oral keratinocytes: Role of PHLDA1 as an apoptosis suppressor. Exp Cell Res 2014; 320:247-57. [DOI: 10.1016/j.yexcr.2013.10.023] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2013] [Revised: 10/28/2013] [Accepted: 10/30/2013] [Indexed: 11/16/2022]
|
16
|
Park ES, Kim J, Ha TU, Choi JS, Soo Hong K, Rho J. TDAG51 deficiency promotes oxidative stress-induced apoptosis through the generation of reactive oxygen species in mouse embryonic fibroblasts. Exp Mol Med 2013; 45:e35. [PMID: 23928855 PMCID: PMC3789259 DOI: 10.1038/emm.2013.67] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2012] [Revised: 05/30/2013] [Accepted: 06/11/2013] [Indexed: 01/05/2023] Open
Abstract
Apoptosis has an important role in maintaining tissue homeostasis in cellular stress responses such as inflammation, endoplasmic reticulum stress, and oxidative stress. T-cell death-associated gene 51 (TDAG51) is a member of the pleckstrin homology-like domain family and was first identified as a pro-apoptotic gene in T-cell receptor-mediated cell death. However, its pro-apoptotic function remains controversial. In this study, we investigated the role of TDAG51 in oxidative stress-induced apoptotic cell death in mouse embryonic fibroblasts (MEFs). TDAG51 expression was highly increased by oxidative stress responses. In response to oxidative stress, the production of intracellular reactive oxygen species was significantly enhanced in TDAG51-deficient MEFs, resulting in the activation of caspase-3. Thus, TDAG51 deficiency promotes apoptotic cell death in MEFs, and these results indicate that TDAG51 has a protective role in oxidative stress-induced cell death in MEFs.
Collapse
Affiliation(s)
- Eui-Soon Park
- Department of Microbiology and Molecular Biology, College of Bioscience and Biotechnology, Chungnam National University, Daejeon, Korea
| | | | | | | | | | | |
Collapse
|
17
|
Hossain GS, Lynn EG, Maclean KN, Zhou J, Dickhout JG, Lhoták S, Trigatti B, Capone J, Rho J, Tang D, McCulloch CA, Al-Bondokji I, Malloy MJ, Pullinger CR, Kane JP, Li Y, Shiffman D, Austin RC. Deficiency of TDAG51 protects against atherosclerosis by modulating apoptosis, cholesterol efflux, and peroxiredoxin-1 expression. J Am Heart Assoc 2013; 2:e000134. [PMID: 23686369 PMCID: PMC3698773 DOI: 10.1161/jaha.113.000134] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
BACKGROUND Apoptosis caused by endoplasmic reticulum (ER) stress contributes to atherothrombosis, the underlying cause of cardiovascular disease (CVD). T-cell death-associated gene 51 (TDAG51), a member of the pleckstrin homology-like domain gene family, is induced by ER stress, causes apoptosis when overexpressed, and is present in lesion-resident macrophages and endothelial cells. METHODS AND RESULTS To study the role of TDAG51 in atherosclerosis, male mice deficient in TDAG51 and apolipoprotein E (TDAG51(-/-)/ApoE(-/-)) were generated and showed reduced atherosclerotic lesion growth (56 ± 5% reduction at 40 weeks, relative to ApoE(-/-) controls, P<0.005) and necrosis (41 ± 4% versus 63 ± 8% lesion area in TDAG51(-/-)/ApoE(-/-) and ApoE(-/-), respectively; P<0.05) without changes in plasma levels of lipids, glucose, and inflammatory cytokines. TDAG51 deficiency caused several phenotypic changes in macrophages and endothelial cells that increase cytoprotection against oxidative and ER stress, enhance PPARγ-dependent reverse cholesterol transport, and upregulate peroxiredoxin-1 (Prdx-1), an antioxidant enzyme with antiatherogenic properties (1.8 ± 0.1-fold increase in Prdx-1 protein expression, relative to control macrophages; P<0.005). Two independent case-control studies found that a genetic variant in the human TDAG51 gene region (rs2367446) is associated with CVD (OR, 1.15; 95% CI, 1.07 to 1.24; P=0.0003). CONCLUSIONS These findings provide evidence that TDAG51 affects specific cellular pathways known to reduce atherogenesis, suggesting that modulation of TDAG51 expression or its activity may have therapeutic benefit for the treatment of CVD.
Collapse
Affiliation(s)
- Gazi S Hossain
- Division of Nephrology, Department of Medicine, McMaster University and St. Joseph's Healthcare Hamilton, Ontario, Canada
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Basseri S, Lhoták Š, Fullerton MD, Palanivel R, Jiang H, Lynn EG, Ford RJ, Maclean KN, Steinberg GR, Austin RC. Loss of TDAG51 results in mature-onset obesity, hepatic steatosis, and insulin resistance by regulating lipogenesis. Diabetes 2013; 62:158-69. [PMID: 22961087 PMCID: PMC3526025 DOI: 10.2337/db12-0256] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Regulation of energy metabolism is critical for the prevention of obesity, diabetes, and hepatic steatosis. Here, we report an important role for the pleckstrin homology-related domain family member, T-cell death-associated gene 51 (TDAG51), in the regulation of energy metabolism. TDAG51 expression was examined during adipocyte differentiation. Adipogenic potential of preadipocytes with knockdown or absence of TDAG51 was assessed. Weight gain, insulin sensitivity, metabolic rate, and liver lipid content were also compared between TDAG51-deficient (TDAG51(-/-)) and wild-type mice. In addition to its relatively high expression in liver, TDAG51 was also present in white adipose tissue (WAT). TDAG51 was downregulated during adipogenesis, and TDAG51(-/-) preadipocytes exhibited greater lipogenic potential. TDAG51(-/-) mice fed a chow diet exhibited greater body and WAT mass, had reduced energy expenditure, displayed mature-onset insulin resistance (IR), and were predisposed to hepatic steatosis. TDAG51(-/-) mice had increased hepatic triglycerides and SREBP-1 target gene expression. Furthermore, TDAG51 expression was inversely correlated with fatty liver in multiple mouse models of hepatic steatosis. Taken together, our findings suggest that TDAG51 is involved in energy homeostasis at least in part by regulating lipogenesis in liver and WAT, and hence, may constitute a novel therapeutic target for the treatment of obesity and IR.
Collapse
Affiliation(s)
- Sana Basseri
- Division of Nephrology, Department of Medicine, McMaster University, Hamilton, Ontario, Canada
- Hamilton Centre for Kidney Research, St. Joseph’s Healthcare Hamilton, Hamilton, Ontario, Canada
| | - Šárka Lhoták
- Hamilton Centre for Kidney Research, St. Joseph’s Healthcare Hamilton, Hamilton, Ontario, Canada
| | - Morgan D. Fullerton
- Division of Endocrinology and Metabolism, Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Rengasamy Palanivel
- Division of Endocrinology and Metabolism, Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Hua Jiang
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, Colorado
| | - Edward G. Lynn
- Division of Nephrology, Department of Medicine, McMaster University, Hamilton, Ontario, Canada
- Hamilton Centre for Kidney Research, St. Joseph’s Healthcare Hamilton, Hamilton, Ontario, Canada
| | - Rebecca J. Ford
- Division of Endocrinology and Metabolism, Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Kenneth N. Maclean
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, Colorado
| | - Gregory R. Steinberg
- Division of Endocrinology and Metabolism, Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Richard C. Austin
- Division of Nephrology, Department of Medicine, McMaster University, Hamilton, Ontario, Canada
- Hamilton Centre for Kidney Research, St. Joseph’s Healthcare Hamilton, Hamilton, Ontario, Canada
- Corresponding author: Richard C. Austin,
| |
Collapse
|
19
|
Carlisle RE, Heffernan A, Brimble E, Liu L, Jerome D, Collins CA, Mohammed-Ali Z, Margetts PJ, Austin RC, Dickhout JG. TDAG51 mediates epithelial-to-mesenchymal transition in human proximal tubular epithelium. Am J Physiol Renal Physiol 2012; 303:F467-81. [DOI: 10.1152/ajprenal.00481.2011] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Epithelial-to-mesenchymal transition (EMT) contributes to renal fibrosis in chronic kidney disease. Endoplasmic reticulum (ER) stress, a feature of many forms of kidney disease, results from the accumulation of misfolded proteins in the ER and leads to the unfolded protein response (UPR). We hypothesized that ER stress mediates EMT in human renal proximal tubules. ER stress is induced by a variety of stressors differing in their mechanism of action, including tunicamycin, thapsigargin, and the calcineurin inhibitor cyclosporine A. These ER stressors increased the UPR markers GRP78, GRP94, and phospho-eIF2α in human proximal tubular cells. Thapsigargin and cyclosporine A also increased cytosolic Ca2+ concentration and T cell death-associated gene 51 (TDAG51) expression, whereas tunicamycin did not. Thapsigargin was also shown to increase levels of active transforming growth factor (TGF)-β1 in the media of cultured human proximal tubular cells. Thapsigargin induced cytoskeletal rearrangement, β-catenin nuclear translocation, and α-smooth muscle actin and vinculin expression in proximal tubular cells, indicating an EMT response. Subconfluent primary human proximal tubular cells were induced to undergo EMT by TGF-β1 treatment. In contrast, tunicamycin treatment did not produce an EMT response. Plasmid-mediated overexpression of TDAG51 resulted in cell shape change and β-catenin nuclear translocation. These results allowed us to develop a two-hit model of ER stress-induced EMT, where Ca2+ dysregulation-mediated TDAG51 upregulation primes the cell for mesenchymal transformation via Wnt signaling and then TGF-β1 activation leads to a complete EMT response. Thus the release of Ca2+ from ER stores mediates EMT in human proximal tubular epithelium via the induction of TDAG51.
Collapse
Affiliation(s)
- Rachel E. Carlisle
- Department of Medicine, Division of Nephrology, McMaster University and St. Joseph's Healthcare Hamilton, Hamilton, Ontario, Canada
| | - Alana Heffernan
- Department of Medicine, Division of Nephrology, McMaster University and St. Joseph's Healthcare Hamilton, Hamilton, Ontario, Canada
| | - Elise Brimble
- Department of Medicine, Division of Nephrology, McMaster University and St. Joseph's Healthcare Hamilton, Hamilton, Ontario, Canada
| | - Limin Liu
- Department of Medicine, Division of Nephrology, McMaster University and St. Joseph's Healthcare Hamilton, Hamilton, Ontario, Canada
| | - Danielle Jerome
- Department of Medicine, Division of Nephrology, McMaster University and St. Joseph's Healthcare Hamilton, Hamilton, Ontario, Canada
| | - Celeste A. Collins
- Department of Medicine, Division of Nephrology, McMaster University and St. Joseph's Healthcare Hamilton, Hamilton, Ontario, Canada
| | - Zahraa Mohammed-Ali
- Department of Medicine, Division of Nephrology, McMaster University and St. Joseph's Healthcare Hamilton, Hamilton, Ontario, Canada
| | - Peter J. Margetts
- Department of Medicine, Division of Nephrology, McMaster University and St. Joseph's Healthcare Hamilton, Hamilton, Ontario, Canada
| | - Richard C. Austin
- Department of Medicine, Division of Nephrology, McMaster University and St. Joseph's Healthcare Hamilton, Hamilton, Ontario, Canada
| | - Jeffrey G. Dickhout
- Department of Medicine, Division of Nephrology, McMaster University and St. Joseph's Healthcare Hamilton, Hamilton, Ontario, Canada
| |
Collapse
|
20
|
Champier J, Claustrat F, Nazaret N, Montange MF, Claustrat B. Folate depletion changes gene expression of fatty acid metabolism, DNA synthesis, and circadian cycle in male mice. Nutr Res 2012; 32:124-32. [DOI: 10.1016/j.nutres.2011.12.012] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2011] [Revised: 12/20/2011] [Accepted: 12/28/2011] [Indexed: 11/16/2022]
|
21
|
Johnson EO, Chang KH, de Pablo Y, Ghosh S, Mehta R, Badve S, Shah K. PHLDA1 is a crucial negative regulator and effector of Aurora A kinase in breast cancer. J Cell Sci 2011; 124:2711-22. [PMID: 21807936 DOI: 10.1242/jcs.084970] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Aurora A kinase is overexpressed in the majority of breast carcinomas. A chemical genetic approach was used to identify the malignant targets of Aurora A, which revealed pleckstrin-homology-like domain protein PHLDA1 as an Aurora A substrate. PHLDA1 downregulation is a powerful prognostic predictor for breast carcinoma, which was confirmed in our study. We further show that downregulation of PHLDA1 is associated with estrogen receptor (ER) expression in breast carcinoma. Aurora A directly phosphorylates PHLDA1 leading to its degradation. PHLDA1 also negatively regulates Aurora A, thereby triggering a feedback loop. We demonstrate the underlying mechanisms by which PHLDA1 upregulation strongly antagonizes Aurora-A-mediated oncogenic pathways, thereby revealing PHLDA1 degradation as a key mechanism by which Aurora A promotes breast malignancy. Thus, not surprisingly, PHLDA1 upregulation acts synergistically with Aurora A inhibition in promoting cell death. PHLDA1 overexpression might therefore be an alternative method to modulate Aurora A deregulation in breast carcinoma. Finally, this study led to the discovery of a mutation in the Aurora A active site that renders it amenable to the chemical genetic approach. Similar mutations are required for Aurora B, suggesting that this modified approach can be extended to other kinases that have hitherto not been amenable to this methodology.
Collapse
Affiliation(s)
- Emmanuel O Johnson
- Department of Chemistry and Purdue University Center for Cancer Research, Purdue University, West Lafayette, IN 47907, USA
| | | | | | | | | | | | | |
Collapse
|
22
|
Dai H, Huang Y, Li Y, Meng G, Wang Y, Guo QN. TSSC3 overexpression associates with growth inhibition, apoptosis induction and enhances chemotherapeutic effects in human osteosarcoma. Carcinogenesis 2011; 33:30-40. [PMID: 22021909 DOI: 10.1093/carcin/bgr232] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Loss of expression of TSSC3, an apoptosis-related imprinted gene, has been reported in several cases of malignant tumors. However, the roles and mechanisms of TSSC3 in human osteosarcoma remain to be defined. In this study, we found TSSC3 to be downregulated during osteosarcoma transformation and progression in osteosarcoma cell lines and tissues. The SaOS2 cell line was used to further evaluate the precise role of TSSC3 in osteosarcoma development. Overexpression of TSSC3 markedly reduced cell vitality and growth, colony formation, Ki67 expression as well as cell cycle arrest in the G(0)/G(1) phase. Consistently, TSSC3 overexpression was associated with increased apoptosis assayed by annexin V/propidium iodide and terminal deoxynucleotidyl transferase-mediated dUTP-biotin nick end labeling staining. Subcutaneous injection of TSSC3 overexpressing SaOS2 cells into athymic nude mice showed that TSSC3 also inhibited tumorigenesis through growth inhibition and apoptosis induction in vivo. Further mechanistic studies revealed that the mitochondrial apoptosis pathway was required for TSSC3-mediated cell apoptosis. These findings support a suppressor role for TSSC3 in osteosarcoma development by regulating apoptosis. In addition, constitutive TSSC3 expression greatly enhanced the sensitivity of human osteosarcoma cells to the chemotherapeutic drugs cisplatin and epirubicin. Conversely, TSSC3 knockdown increased SaOS2 cell growth and decreased apoptosis in vitro and in vivo and reduced sensitivity of the cells to chemotherapy. This is the first study to demonstrate that TSSC3 has a potent tumor suppressor role in osteosarcoma, probably by inhibition of growth and induction of apoptosis via the mitochondrial apoptosis pathway.
Collapse
Affiliation(s)
- Huanzi Dai
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | | | | | | | | | | |
Collapse
|
23
|
Barrow AD, Raynal N, Andersen TL, Slatter DA, Bihan D, Pugh N, Cella M, Kim T, Rho J, Negishi-Koga T, Delaisse JM, Takayanagi H, Lorenzo J, Colonna M, Farndale RW, Choi Y, Trowsdale J. OSCAR is a collagen receptor that costimulates osteoclastogenesis in DAP12-deficient humans and mice. J Clin Invest 2011; 121:3505-16. [PMID: 21841309 PMCID: PMC3163954 DOI: 10.1172/jci45913] [Citation(s) in RCA: 150] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2010] [Accepted: 07/01/2011] [Indexed: 12/14/2022] Open
Abstract
Osteoclasts are terminally differentiated leukocytes that erode the mineralized bone matrix. Osteoclastogenesis requires costimulatory receptor signaling through adaptors containing immunoreceptor tyrosine-based activation motifs (ITAMs), such as Fc receptor common γ (FcRγ) and DNAX-activating protein of 12 kDa. Identification of these ITAM-containing receptors and their ligands remains a high research priority, since the stimuli for osteoclastogenesis are only partly defined. Osteoclast-associated receptor (OSCAR) was proposed to be a potent FcRγ-associated costimulatory receptor expressed by preosteoclasts in vitro, but OSCAR lacks a cognate ligand and its role in vivo has been unclear. Using samples from mice and patients deficient in various ITAM signaling pathways, we show here that OSCAR costimulates one of the major FcRγ-associated pathways required for osteoclastogenesis in vivo. Furthermore, we found that OSCAR binds to specific motifs within fibrillar collagens in the ECM that become revealed on nonquiescent bone surfaces in which osteoclasts undergo maturation and terminal differentiation in vivo. OSCAR promoted osteoclastogenesis in vivo, and OSCAR binding to its collagen motif led to signaling that increased numbers of osteoclasts in culture. Thus, our results suggest that ITAM-containing receptors can respond to exposed ligands in collagen, leading to the functional differentiation of leukocytes, which provides what we believe to be a new concept for ITAM regulation of cytokine receptors in different tissue microenvironments.
Collapse
Affiliation(s)
- Alexander David Barrow
- Department of Pathology, University of Cambridge, Cambridge, United Kingdom.
Washington University School of Medicine, Department of Pathology and Immunology, St. Louis, Missouri, USA.
Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom.
Department of Clinical Cell Biology, University of Southern Denmark, Vejle/Lillebælt Hospital, Institute of Regional Health Services Research, Vejle, Denmark.
Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Department of Microbiology, BK21 BioBC, and Graduate of Analytical Science and Technology, Chungnam National University, Yuseong-gu, Daejon, Republic of Korea.
Department of Cell Signaling, Tokyo Medical and Dental University, Tokyo, Japan.
Division of Endocrinology, Department of Medicine, University of Connecticut Health Center, Farmington, Connecticut, USA
| | - Nicolas Raynal
- Department of Pathology, University of Cambridge, Cambridge, United Kingdom.
Washington University School of Medicine, Department of Pathology and Immunology, St. Louis, Missouri, USA.
Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom.
Department of Clinical Cell Biology, University of Southern Denmark, Vejle/Lillebælt Hospital, Institute of Regional Health Services Research, Vejle, Denmark.
Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Department of Microbiology, BK21 BioBC, and Graduate of Analytical Science and Technology, Chungnam National University, Yuseong-gu, Daejon, Republic of Korea.
Department of Cell Signaling, Tokyo Medical and Dental University, Tokyo, Japan.
Division of Endocrinology, Department of Medicine, University of Connecticut Health Center, Farmington, Connecticut, USA
| | - Thomas Levin Andersen
- Department of Pathology, University of Cambridge, Cambridge, United Kingdom.
Washington University School of Medicine, Department of Pathology and Immunology, St. Louis, Missouri, USA.
Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom.
Department of Clinical Cell Biology, University of Southern Denmark, Vejle/Lillebælt Hospital, Institute of Regional Health Services Research, Vejle, Denmark.
Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Department of Microbiology, BK21 BioBC, and Graduate of Analytical Science and Technology, Chungnam National University, Yuseong-gu, Daejon, Republic of Korea.
Department of Cell Signaling, Tokyo Medical and Dental University, Tokyo, Japan.
Division of Endocrinology, Department of Medicine, University of Connecticut Health Center, Farmington, Connecticut, USA
| | - David A. Slatter
- Department of Pathology, University of Cambridge, Cambridge, United Kingdom.
Washington University School of Medicine, Department of Pathology and Immunology, St. Louis, Missouri, USA.
Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom.
Department of Clinical Cell Biology, University of Southern Denmark, Vejle/Lillebælt Hospital, Institute of Regional Health Services Research, Vejle, Denmark.
Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Department of Microbiology, BK21 BioBC, and Graduate of Analytical Science and Technology, Chungnam National University, Yuseong-gu, Daejon, Republic of Korea.
Department of Cell Signaling, Tokyo Medical and Dental University, Tokyo, Japan.
Division of Endocrinology, Department of Medicine, University of Connecticut Health Center, Farmington, Connecticut, USA
| | - Dominique Bihan
- Department of Pathology, University of Cambridge, Cambridge, United Kingdom.
Washington University School of Medicine, Department of Pathology and Immunology, St. Louis, Missouri, USA.
Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom.
Department of Clinical Cell Biology, University of Southern Denmark, Vejle/Lillebælt Hospital, Institute of Regional Health Services Research, Vejle, Denmark.
Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Department of Microbiology, BK21 BioBC, and Graduate of Analytical Science and Technology, Chungnam National University, Yuseong-gu, Daejon, Republic of Korea.
Department of Cell Signaling, Tokyo Medical and Dental University, Tokyo, Japan.
Division of Endocrinology, Department of Medicine, University of Connecticut Health Center, Farmington, Connecticut, USA
| | - Nicholas Pugh
- Department of Pathology, University of Cambridge, Cambridge, United Kingdom.
Washington University School of Medicine, Department of Pathology and Immunology, St. Louis, Missouri, USA.
Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom.
Department of Clinical Cell Biology, University of Southern Denmark, Vejle/Lillebælt Hospital, Institute of Regional Health Services Research, Vejle, Denmark.
Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Department of Microbiology, BK21 BioBC, and Graduate of Analytical Science and Technology, Chungnam National University, Yuseong-gu, Daejon, Republic of Korea.
Department of Cell Signaling, Tokyo Medical and Dental University, Tokyo, Japan.
Division of Endocrinology, Department of Medicine, University of Connecticut Health Center, Farmington, Connecticut, USA
| | - Marina Cella
- Department of Pathology, University of Cambridge, Cambridge, United Kingdom.
Washington University School of Medicine, Department of Pathology and Immunology, St. Louis, Missouri, USA.
Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom.
Department of Clinical Cell Biology, University of Southern Denmark, Vejle/Lillebælt Hospital, Institute of Regional Health Services Research, Vejle, Denmark.
Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Department of Microbiology, BK21 BioBC, and Graduate of Analytical Science and Technology, Chungnam National University, Yuseong-gu, Daejon, Republic of Korea.
Department of Cell Signaling, Tokyo Medical and Dental University, Tokyo, Japan.
Division of Endocrinology, Department of Medicine, University of Connecticut Health Center, Farmington, Connecticut, USA
| | - Taesoo Kim
- Department of Pathology, University of Cambridge, Cambridge, United Kingdom.
Washington University School of Medicine, Department of Pathology and Immunology, St. Louis, Missouri, USA.
Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom.
Department of Clinical Cell Biology, University of Southern Denmark, Vejle/Lillebælt Hospital, Institute of Regional Health Services Research, Vejle, Denmark.
Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Department of Microbiology, BK21 BioBC, and Graduate of Analytical Science and Technology, Chungnam National University, Yuseong-gu, Daejon, Republic of Korea.
Department of Cell Signaling, Tokyo Medical and Dental University, Tokyo, Japan.
Division of Endocrinology, Department of Medicine, University of Connecticut Health Center, Farmington, Connecticut, USA
| | - Jaerang Rho
- Department of Pathology, University of Cambridge, Cambridge, United Kingdom.
Washington University School of Medicine, Department of Pathology and Immunology, St. Louis, Missouri, USA.
Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom.
Department of Clinical Cell Biology, University of Southern Denmark, Vejle/Lillebælt Hospital, Institute of Regional Health Services Research, Vejle, Denmark.
Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Department of Microbiology, BK21 BioBC, and Graduate of Analytical Science and Technology, Chungnam National University, Yuseong-gu, Daejon, Republic of Korea.
Department of Cell Signaling, Tokyo Medical and Dental University, Tokyo, Japan.
Division of Endocrinology, Department of Medicine, University of Connecticut Health Center, Farmington, Connecticut, USA
| | - Takako Negishi-Koga
- Department of Pathology, University of Cambridge, Cambridge, United Kingdom.
Washington University School of Medicine, Department of Pathology and Immunology, St. Louis, Missouri, USA.
Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom.
Department of Clinical Cell Biology, University of Southern Denmark, Vejle/Lillebælt Hospital, Institute of Regional Health Services Research, Vejle, Denmark.
Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Department of Microbiology, BK21 BioBC, and Graduate of Analytical Science and Technology, Chungnam National University, Yuseong-gu, Daejon, Republic of Korea.
Department of Cell Signaling, Tokyo Medical and Dental University, Tokyo, Japan.
Division of Endocrinology, Department of Medicine, University of Connecticut Health Center, Farmington, Connecticut, USA
| | - Jean-Marie Delaisse
- Department of Pathology, University of Cambridge, Cambridge, United Kingdom.
Washington University School of Medicine, Department of Pathology and Immunology, St. Louis, Missouri, USA.
Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom.
Department of Clinical Cell Biology, University of Southern Denmark, Vejle/Lillebælt Hospital, Institute of Regional Health Services Research, Vejle, Denmark.
Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Department of Microbiology, BK21 BioBC, and Graduate of Analytical Science and Technology, Chungnam National University, Yuseong-gu, Daejon, Republic of Korea.
Department of Cell Signaling, Tokyo Medical and Dental University, Tokyo, Japan.
Division of Endocrinology, Department of Medicine, University of Connecticut Health Center, Farmington, Connecticut, USA
| | - Hiroshi Takayanagi
- Department of Pathology, University of Cambridge, Cambridge, United Kingdom.
Washington University School of Medicine, Department of Pathology and Immunology, St. Louis, Missouri, USA.
Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom.
Department of Clinical Cell Biology, University of Southern Denmark, Vejle/Lillebælt Hospital, Institute of Regional Health Services Research, Vejle, Denmark.
Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Department of Microbiology, BK21 BioBC, and Graduate of Analytical Science and Technology, Chungnam National University, Yuseong-gu, Daejon, Republic of Korea.
Department of Cell Signaling, Tokyo Medical and Dental University, Tokyo, Japan.
Division of Endocrinology, Department of Medicine, University of Connecticut Health Center, Farmington, Connecticut, USA
| | - Joseph Lorenzo
- Department of Pathology, University of Cambridge, Cambridge, United Kingdom.
Washington University School of Medicine, Department of Pathology and Immunology, St. Louis, Missouri, USA.
Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom.
Department of Clinical Cell Biology, University of Southern Denmark, Vejle/Lillebælt Hospital, Institute of Regional Health Services Research, Vejle, Denmark.
Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Department of Microbiology, BK21 BioBC, and Graduate of Analytical Science and Technology, Chungnam National University, Yuseong-gu, Daejon, Republic of Korea.
Department of Cell Signaling, Tokyo Medical and Dental University, Tokyo, Japan.
Division of Endocrinology, Department of Medicine, University of Connecticut Health Center, Farmington, Connecticut, USA
| | - Marco Colonna
- Department of Pathology, University of Cambridge, Cambridge, United Kingdom.
Washington University School of Medicine, Department of Pathology and Immunology, St. Louis, Missouri, USA.
Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom.
Department of Clinical Cell Biology, University of Southern Denmark, Vejle/Lillebælt Hospital, Institute of Regional Health Services Research, Vejle, Denmark.
Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Department of Microbiology, BK21 BioBC, and Graduate of Analytical Science and Technology, Chungnam National University, Yuseong-gu, Daejon, Republic of Korea.
Department of Cell Signaling, Tokyo Medical and Dental University, Tokyo, Japan.
Division of Endocrinology, Department of Medicine, University of Connecticut Health Center, Farmington, Connecticut, USA
| | - Richard W. Farndale
- Department of Pathology, University of Cambridge, Cambridge, United Kingdom.
Washington University School of Medicine, Department of Pathology and Immunology, St. Louis, Missouri, USA.
Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom.
Department of Clinical Cell Biology, University of Southern Denmark, Vejle/Lillebælt Hospital, Institute of Regional Health Services Research, Vejle, Denmark.
Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Department of Microbiology, BK21 BioBC, and Graduate of Analytical Science and Technology, Chungnam National University, Yuseong-gu, Daejon, Republic of Korea.
Department of Cell Signaling, Tokyo Medical and Dental University, Tokyo, Japan.
Division of Endocrinology, Department of Medicine, University of Connecticut Health Center, Farmington, Connecticut, USA
| | - Yongwon Choi
- Department of Pathology, University of Cambridge, Cambridge, United Kingdom.
Washington University School of Medicine, Department of Pathology and Immunology, St. Louis, Missouri, USA.
Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom.
Department of Clinical Cell Biology, University of Southern Denmark, Vejle/Lillebælt Hospital, Institute of Regional Health Services Research, Vejle, Denmark.
Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Department of Microbiology, BK21 BioBC, and Graduate of Analytical Science and Technology, Chungnam National University, Yuseong-gu, Daejon, Republic of Korea.
Department of Cell Signaling, Tokyo Medical and Dental University, Tokyo, Japan.
Division of Endocrinology, Department of Medicine, University of Connecticut Health Center, Farmington, Connecticut, USA
| | - John Trowsdale
- Department of Pathology, University of Cambridge, Cambridge, United Kingdom.
Washington University School of Medicine, Department of Pathology and Immunology, St. Louis, Missouri, USA.
Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom.
Department of Clinical Cell Biology, University of Southern Denmark, Vejle/Lillebælt Hospital, Institute of Regional Health Services Research, Vejle, Denmark.
Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Department of Microbiology, BK21 BioBC, and Graduate of Analytical Science and Technology, Chungnam National University, Yuseong-gu, Daejon, Republic of Korea.
Department of Cell Signaling, Tokyo Medical and Dental University, Tokyo, Japan.
Division of Endocrinology, Department of Medicine, University of Connecticut Health Center, Farmington, Connecticut, USA
| |
Collapse
|
24
|
Liu F, Xu ZL, Qian XJ, Qiu WY, Huang H. Expression of Hsf1, Hsf2, and Phlda1 in cells undergoing cryptorchid-induced apoptosis in rat testes. Mol Reprod Dev 2011; 78:283-91. [DOI: 10.1002/mrd.21304] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2010] [Accepted: 02/22/2011] [Indexed: 11/11/2022]
|
25
|
Abstract
Activation-induced cell death (AICD) plays an essential role in the contraction of activated T cells after eradication of pathogen. Fas (APO-1/CD95) is one of the key cell surface proteins that mediate AICD in CD4(+) and CD8(+) T cells. Despite its prime importance in cell death, regulation of Fas expression in T cells is poorly understood. Here we show that Cyclon, a newly identified cytokine-inducible protein, is induced in T cells on T-cell receptor ligation and important for immune homeostasis. Transgenic expression of Cyclon ameliorated autoimmune phenotype in mice lacking subunits of IL-2R. Transgenic expression of Cyclon markedly enhanced AICD through increased expression of Fas whose expression is essential for Cyclon action. Finally, we demonstrated that activated but not resting CD4(+) T cells with targeted deletion of a Cyclon allele show reduced AICD and expression of Fas, indicating a critical role of Cyclon in Fas expression in activated T cells. We think that our data provide insight into expression regulation of Fas in T cells.
Collapse
|
26
|
Park ES, Choi S, Kim JM, Jeong Y, Choe J, Park CS, Choi Y, Rho J. Early embryonic lethality caused by targeted disruption of the TRAF-interacting protein (TRIP) gene. Biochem Biophys Res Commun 2007; 363:971-7. [PMID: 17927961 DOI: 10.1016/j.bbrc.2007.09.103] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2007] [Accepted: 09/20/2007] [Indexed: 10/22/2022]
Abstract
Tumor necrosis factor receptor (TNFR)-associated factors (TRAFs) are key adaptor molecules in the TNFR-signaling complexes that promote a wide variety of signaling cascades including cell proliferation, activation, differentiation, and apoptosis. TRAF-interacting protein (TRIP) is required for the inhibitory regulation of TNF-induced NF-kappaB signaling via the TNFR/TRAF-signaling complexes in vitro. TRIP also directly interacts with the familial cylindromatosis tumor suppressor gene (CYLD) and negatively regulates NF-kappaB activation in vitro. However, although there appears to be a relationship between TRIP, the TRAFs and also CYLD as modulators of NF-kappaB signaling in vitro, the functional role of TRIP in vivo is still unclear. To identify the role of TRIP in vivo, we have generated TRIP-deficient mice. Homozygous mouse embryos were found to die shortly after implantation due to proliferation defects and excessive cell death. These results indicate that TRIP is an essential factor during early mouse embryonic development in vivo.
Collapse
Affiliation(s)
- Eui-Soon Park
- Department of Microbiology and BK21 BioBC, Chungnam National University, Daejeon 305-764, Republic of Korea
| | | | | | | | | | | | | | | |
Collapse
|
27
|
Xi ZQ, Wang LY, Sun JJ, Liu XZ, Zhu X, Xiao F, Guan LF, Li JM, Wang L, Wang XF. TDAG51 in the anterior temporal neocortex of patients with intractable epilepsy. Neurosci Lett 2007; 425:53-8. [PMID: 17870236 DOI: 10.1016/j.neulet.2007.08.016] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2007] [Revised: 07/25/2007] [Accepted: 08/07/2007] [Indexed: 10/22/2022]
Abstract
TDAG51 (T cell death-associated gene 51) is an apoptosis-associated protein. Our aim was to investigate TDAG51 expression in the anterior temporal neocortex of patients with intractable epilepsy (IE), and then to discuss the possible role of TDAG51 in IE. Tissue samples from the anterior temporal neocortex of 33 patients who had surgery for IE were used to detect TDAG51 expression by immunohistochemistry, immunofluorescence, and Western blotting. We compared these tissues with nine histologically normal anterior temporal lobes from intracranial hypertension patients who had decompression procedures. TDAG51 was mainly expressed in the cytoplasm of neurons and glial cells. TDAG51 in IE was significantly higher than that in the controls. These findings were consistently observed using Western blotting, immunofluorescence, and immunohistochemistry techniques. TDAG51 in patients with IE was significantly higher when compared with levels in the controls. This finding suggests TDAG51 is consistent with a possible role of this gene in the evolution of the pathology in IE.
Collapse
Affiliation(s)
- Zhi-qin Xi
- Department of Neurology, the First Affiliated Hospital, Chongqing Medical University, 1 You Yi Road, Chongqing 400016, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Nagai MA, Fregnani JHTG, Netto MM, Brentani MM, Soares FA. Down-regulation of PHLDA1 gene expression is associated with breast cancer progression. Breast Cancer Res Treat 2007; 106:49-56. [PMID: 17211533 DOI: 10.1007/s10549-006-9475-6] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2006] [Accepted: 11/27/2006] [Indexed: 01/24/2023]
Abstract
In a previous study, using differential display reverse transcriptase-PCR (DDRT-PCR) we showed that down-regulation of the PHLDA1 (pleckstrin homology-like domain, family A, member 1; also named TDAG51) mRNA was down-regulated in breast tumors compared with normal breast tissue. The present study was conducted to determine the expression pattern and predictive prognostic value of PHLDA1 in breast cancer. A series of 720 primary invasive breast tumors were examined for PHLDA1 expression. PHLDA1 mRNA expression was determined in 74 breast tumors using quantitative Real Time PCR analysis (qPCR). PHLDA1 protein expression was evaluated by immunohistochemistry (IHC) using Tissue Microarrays (TMA) containing 699 primary invasive breast tumors. Reduced PHLDA1 mRNA expression was identified in 72% (53/74) of the primary breast tumors analyzed. Seventy-three percent (512/699) of cases analyzed showed negative PHLDA1 protein expression. Down-regulation of PHLDA1 protein was a strong predictor of poor prognosis for breast cancer patients. Breast cancer patients with tumors that were negative for PHLDA1 protein expression had shorter disease free survival (P < 0.001) and overall survival (P < 0.001) than patients with tumors that were positive for PHLDA1 protein expression. In addition patients with tumors exhibiting reduced PHLDA1 expression and paucity for ER had the worse outcome (P < 0.001). Multivariate analysis indicated that PHLDA1 protein expression is an independent prognostic factor of patient survival. To our knowledge, the expression pattern of PHLDA1 in breast cancer has not previously been investigated. Our results provide strong evidence that reduced PHLDA1 expression is important in breast cancer progression and could serve as useful prognostic marker of disease outcome.
Collapse
Affiliation(s)
- Maria Aparecida Nagai
- Disciplina de Oncologia, Departamento de Radiologia, Faculdade de Medicina, Universidade de São Paulo, CEP 01246-903, São Paulo, Brazil.
| | | | | | | | | |
Collapse
|
29
|
Lee SH, Rho J, Jeong D, Sul JY, Kim T, Kim N, Kang JS, Miyamoto T, Suda T, Lee SK, Pignolo RJ, Koczon-Jaremko B, Lorenzo J, Choi Y. v-ATPase V0 subunit d2-deficient mice exhibit impaired osteoclast fusion and increased bone formation. Nat Med 2006; 12:1403-9. [PMID: 17128270 DOI: 10.1038/nm1514] [Citation(s) in RCA: 430] [Impact Index Per Article: 23.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2006] [Accepted: 10/31/2006] [Indexed: 12/22/2022]
Abstract
Matrix-producing osteoblasts and bone-resorbing osteoclasts maintain bone homeostasis. Osteoclasts are multinucleated, giant cells of hematopoietic origin formed by the fusion of mononuclear pre-osteoclasts derived from myeloid cells. Fusion-mediated giant cell formation is critical for osteoclast maturation; without it, bone resorption is inefficient. To understand how osteoclasts differ from other myeloid lineage cells, we previously compared global mRNA expression patterns in these cells and identified genes of unknown function predominantly expressed in osteoclasts, one of which is the d2 isoform of vacuolar (H(+)) ATPase (v-ATPase) V(0) domain (Atp6v0d2). Here we show that inactivation of Atp6v0d2 in mice results in markedly increased bone mass due to defective osteoclasts and enhanced bone formation. Atp6v0d2 deficiency did not affect differentiation or the v-ATPase activity of osteoclasts. Rather, Atp6v0d2 was required for efficient pre-osteoclast fusion. Increased bone formation was probably due to osteoblast-extrinsic factors, as Atp6v02 was not expressed in osteoblasts and their differentiation ex vivo was not altered in the absence of Atp6v02. Our results identify Atp6v0d2 as a regulator of osteoclast fusion and bone formation, and provide genetic data showing that it is possible to simultaneously inhibit osteoclast maturation and stimulate bone formation by therapeutically targeting the function of a single gene.
Collapse
Affiliation(s)
- Seoung-Hoon Lee
- Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania 19104, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Hayashida N, Inouye S, Fujimoto M, Tanaka Y, Izu H, Takaki E, Ichikawa H, Rho J, Nakai A. A novel HSF1-mediated death pathway that is suppressed by heat shock proteins. EMBO J 2006; 25:4773-83. [PMID: 17024176 PMCID: PMC1618102 DOI: 10.1038/sj.emboj.7601370] [Citation(s) in RCA: 78] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2006] [Accepted: 09/04/2006] [Indexed: 12/24/2022] Open
Abstract
Heat shock response is an adoptive response to proteotoxic stress, and a major heat shock transcription factor 1 (HSF1) has been believed to protect cells from cell death by inducing heat shock proteins (Hsps) that assist protein folding and prevent protein denaturation. However, it is revealed recently that HSF1 also promotes cell death of male germ cells. Here, we found a proapoptotic Tdag51 (T-cell death associated gene 51) gene as a direct target gene of HSF1. Heat shock and other stresses induced different levels of Hsps and Tdag51, which depend on cell types. Hsps bound directly to the N-terminal pleckstrin-homology like (PHL) domain of Tdag51, and suppressed death activity of the C-terminal proline/glutamine/histidine-rich domain. Tdag51, but not major Hsps, were induced in male germ cells exposed to high temperatures. Analysis of Tdag51-null testes showed that Tdag51 played substantial roles in promoting heat shock-induced cell death in vivo. These data suggest that cell fate on proteotoxic condition is determined at least by balance between Hsp and Tdag51 levels, which are differently regulated by HSF1.
Collapse
Affiliation(s)
- Naoki Hayashida
- Department of Biochemistry and Molecular Biology, Yamaguchi University School of Medicine, Ube, Japan
| | - Sachiye Inouye
- Department of Biochemistry and Molecular Biology, Yamaguchi University School of Medicine, Ube, Japan
| | - Mitsuaki Fujimoto
- Department of Biochemistry and Molecular Biology, Yamaguchi University School of Medicine, Ube, Japan
| | - Yasunori Tanaka
- Department of Biochemistry and Molecular Biology, Yamaguchi University School of Medicine, Ube, Japan
| | - Hanae Izu
- Department of Biochemistry and Molecular Biology, Yamaguchi University School of Medicine, Ube, Japan
| | - Eiichi Takaki
- Department of Biochemistry and Molecular Biology, Yamaguchi University School of Medicine, Ube, Japan
| | - Hitoshi Ichikawa
- ancer Transcriptome Project, National Cancer Center Research Institute, Chuo-ku, Tokyo, Japan
| | - Jaerang Rho
- Department of Microbiology, Natural Science College, Chungnam National University, Yuseong-gu, Daejeon, Korea
| | - Akira Nakai
- Department of Biochemistry and Molecular Biology, Yamaguchi University School of Medicine, Ube, Japan
- Department of Biochemistry and Molecular Biology, Yamaguchi University School of Medicine, Minami-Kogushi 1-1-1, Ube 755-8505, Japan. Tel.: 81 836 22 2214; Fax: 81 836 22 2315; E-mail:
| |
Collapse
|
31
|
Wang Z, Rong YP, Malone MH, Davis MC, Zhong F, Distelhorst CW. Thioredoxin-interacting protein (txnip) is a glucocorticoid-regulated primary response gene involved in mediating glucocorticoid-induced apoptosis. Oncogene 2006; 25:1903-13. [PMID: 16301999 DOI: 10.1038/sj.onc.1209218] [Citation(s) in RCA: 89] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Glucocorticoid hormones induce apoptosis in lymphoid cells. This process is transcriptionally regulated and requires de novo RNA/protein synthesis. However, the full spectrum of glucocorticoid-regulated genes mediating this cell death process is unknown. Through gene expression profiling we discovered that the expression of thioredoxin-intereacting protein (txnip) mRNA is significantly induced by the glucocorticoid hormone dexamethasone not only in the murine T-cell lymphoma line WEHI7.2, but also in normal mouse thymocytes. This result was confirmed by Northern blot analysis in multiple models of dexamethasone-induced apoptosis. The induction of txnip mRNA by dexamethasone appears to be mediated through the glucocorticoid receptor as it is blocked in the presence of RU486, a glucocorticoid receptor antagonist. Deletion and mutation analysis of the txnip promoter identified a functional glucocorticoid response element in the txnip promoter. Reporter assays demonstrated that this glucocorticoid response element was necessary and sufficient for induction of txnip by dexamethasone. Expression of a GFP-TXNIP fusion protein was sufficient to induce apoptosis in WEHI7.2 cells, and repression of endogenous txnip by RNA interference inhibited dexamethasone-induced apoptosis in WEHI7.2 cells. Together, these findings indicate that txnip is a novel glucocorticoid-induced primary target gene involved in mediating glucocorticoid-induced apoptosis.
Collapse
Affiliation(s)
- Z Wang
- Department of Medicine and Pharmacology, Comprehensive Cancer Center, Case Western Reserve University School of Medicine and University Hospitals of Cleveland, Cleveland, OH 44106-7285, USA
| | | | | | | | | | | |
Collapse
|
32
|
Austin RC, Lentz SR, Werstuck GH. Role of hyperhomocysteinemia in endothelial dysfunction and atherothrombotic disease. Cell Death Differ 2005; 11 Suppl 1:S56-64. [PMID: 15243582 DOI: 10.1038/sj.cdd.4401451] [Citation(s) in RCA: 276] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Hyperhomocysteinemia (HHcy) is an independent risk factor for cardiovascular disease, including ischemic heart disease, stroke, and peripheral vascular disease. Mutations in the enzymes responsible for homocysteine metabolism, particularly cystathionine beta-synthase (CBS) or 5,10-methylenetetrahydrofolate reductase (MTHFR), result in severe forms of HHcy. Additionally, nutritional deficiencies in B vitamin cofactors required for homocysteine metabolism, including folic acid, vitamin B6 (pyridoxal phosphate), and/or B12 (methylcobalamin), can induce HHcy. Studies using animal models of genetic- and diet-induced HHcy have recently demonstrated a causal relationship between HHcy, endothelial dysfunction, and accelerated atherosclerosis. Dietary enrichment in B vitamins attenuates these adverse effects of HHcy. Although oxidative stress and activation of proinflammatory factors have been proposed to explain the atherogenic effects of HHcy, recent in vitro and in vivo studies demonstrate that HHcy induces endoplasmic reticulum (ER) stress, leading to activation of the unfolded protein response (UPR). This review summarizes the current role of HHcy in endothelial dysfunction and explores the cellular mechanisms, including ER stress, that contribute to atherothrombosis.
Collapse
Affiliation(s)
- R C Austin
- Department of Pathology and Molecular Medicine, McMaster University and the Henderson Research Centre, Hamilton, Ontario, Canada.
| | | | | |
Collapse
|
33
|
Oberg HH, Sipos B, Kalthoff H, Janssen O, Kabelitz D. Regulation of T-cell death-associated gene 51 (TDAG51) expression in human T-cells. Cell Death Differ 2005; 11:674-84. [PMID: 15002043 DOI: 10.1038/sj.cdd.4401407] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
T-cell death-associated gene 51 (TDAG51) has been described to regulate T-cell receptor/CD3-dependent induction of CD95/Fas and subsequent activation-induced cell death (AICD) in a murine T-cell hybridoma. Using well-defined pharmacological inhibitors, we investigated the regulation of TDAG51 expression in human T-cells and the correlation with cell death. TDAG51 was induced in resting T-cells, lymphoid cell lines and AICD-susceptible as well as AICD-resistant T-cell clones, and induction was inhibited by MAP-kinase inhibitors and PKC inhibitor Gö6983. No correlation between the effects of inhibitors on TDAG51 expression and cell death was observed. The constitutive TDAG51 expression in five pancreatic carcinoma cell lines was reduced by MAP-kinase inhibitors but not by Gö6983. Furthermore, the inducible overexpression of TDAG51 in TetOn Jurkat cells did not modulate cellular proliferation, phorbolester/ionomycin-induced growth arrest, or the expression of various cell surface molecules. Our results indicate that the expression of TDAG51 in human T-cells does not correlate with AICD.
Collapse
Affiliation(s)
- H-H Oberg
- Institute of Immunology, University Hospital Schleswig-Holstein Campus Kiel, Kiel, Germany
| | | | | | | | | |
Collapse
|
34
|
Meier-Noorden M, Flindt S, Kalinke U, Hinz T. A CpG-rich bidirectional promoter induces the T-cell death-associated gene 51 and downregulates an inversely oriented transcript during early T-cell activation. Gene 2004; 338:197-207. [PMID: 15315823 DOI: 10.1016/j.gene.2004.05.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2004] [Revised: 04/16/2004] [Accepted: 05/06/2004] [Indexed: 11/18/2022]
Abstract
The human T-cell death-associated gene 51 (TDAG51) is upregulated upon lymphocyte stimulation and in the context of ER stress. Moreover, TDAG51 plays a role in programmed cell death and tumorigenesis. We performed an extensive TDAG51 promoter analysis and found a strong CpG-rich bidirectional promoter within the first 582 nucleotides of the TDAG51 reference DNA complementary to RNA (cDNA). Upon stimulation of primary human T cells, this promoter modulated the downregulation of a newly detected head-to-head oriented transcript. Mapping of the transcription start points revealed that the 5' regions of the TDAG51 mRNA and of the newly identified transcript did not overlap in T cells. Thus, the TDAG51 locus shows an operon-like organization of two head-to-head oriented transcripts that are inversely regulated in T lymphocytes by a CpG-rich bidirectional promoter.
Collapse
Affiliation(s)
- Marc Meier-Noorden
- Division of Immunology, Paul-Ehrlich-Institut, Paul-Ehrlich-Str. 51-59, D-63225 Langen, Germany
| | | | | | | |
Collapse
|
35
|
Toyoshima Y, Karas M, Yakar S, Dupont J, LeRoith D. TDAG51 mediates the effects of insulin-like growth factor I (IGF-I) on cell survival. J Biol Chem 2004; 279:25898-904. [PMID: 15037619 DOI: 10.1074/jbc.m400661200] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Insulin-like growth factor-I (IGF-I) receptors and insulin receptors belong to the same subfamily of receptor tyrosine kinases and share a similar set of intracellular signaling pathways, despite their distinct biological actions. In the present study, we evaluated T cell death-associated gene 51 (TDAG51), which we previously identified by cDNA microarray analysis as a gene specifically induced by IGF-I. We characterized the signaling pathways by which IGF-I induces TDAG51 gene expression and the functional role of TDAG51 in IGF-I signaling in NIH-3T3 (NWTb3) cells, which overexpress the human IGF-I receptor. Treatment with IGF-I increased TDAG51 mRNA and protein levels in NWTb3 cells. This effect of IGF-I was specifically mediated by the IGF-IR, because IGF-I did not induce TDAG51 expression in NIH-3T3 cells overexpressing a dominant-negative IGF-I receptor. Through the use of specific inhibitors of various protein kinases, we found that IGF-I induced TDAG51 expression via the p38 MAPK pathway. The ERK, JNK, and phosphatidylinositol 3-kinase pathways were not involved in IGF-I-induced regulation of TDAG51. To assess the role of TDAG51 in IGF-I signaling, we used small interfering RNA (siRNA) expression vectors directed at two different target sites to reduce the level of TDAG51 protein. In cells expressing these siRNA vectors, TDAG51 protein levels were decreased by 75-80%. Furthermore, TDAG51 siRNA expression abolished the ability of IGF-I to rescue cells from serum starvation-induced apoptosis. These findings suggest that TDAG51 plays an important role in the anti-apoptotic effects of IGF-I.
Collapse
Affiliation(s)
- Yuka Toyoshima
- Section on Molecular and Cellular Physiology, Diabetes Branch, NIDDK, National Institutes of Health, Bethesda, Maryland 20892-1758, USA
| | | | | | | | | |
Collapse
|
36
|
Pandini G, Medico E, Conte E, Sciacca L, Vigneri R, Belfiore A. Differential gene expression induced by insulin and insulin-like growth factor-II through the insulin receptor isoform A. J Biol Chem 2003; 278:42178-89. [PMID: 12881524 DOI: 10.1074/jbc.m304980200] [Citation(s) in RCA: 83] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The human insulin receptor (IR) exists in two isoforms (IR-A and IR-B). IR-A is a short isoform, generated by the skipping of exon 11, a small exon encoding for 12 amino acid residues at the carboxyl terminus of the IR alpha-subunit. Recently, we found that IR-A is the predominant isoform in fetal tissues and malignant cells and binds with a high affinity not only insulin but also insulin-like growth factor-II (IGF-II). To investigate whether the activation of IR-A by the two ligands differentially activate post-receptor molecular mechanisms, we studied gene expression in response to IR-A activation by either insulin or IGF-II, using microarray technology. To avoid the interfering effect of the IGF-IR, IGF-II binding to the IR-A was studied in IGF-IR-deficient murine fibroblasts (R- cells) transfected with the human IR-A cDNA (R-/IR-A cells). Gene expression was studied at 0.5, 3, and 8 h. We found that 214 transcripts were similarly regulated by insulin and IGF-II, whereas 45 genes were differentially transcribed. Eighteen of these differentially regulated genes were responsive to only one of the two ligands (12 to insulin and 6 to IGF-II). Twenty-seven transcripts were regulated by both insulin and IGF-II, but a significant difference between the two ligands was present at least in one time point. Interestingly, IGF-II was a more potent and/or persistent regulator than insulin for these genes. Results were validated by measuring the expression of 12 genes by quantitative real-time reverse transcriptase-PCR. In conclusion, we show that insulin and IGF-II, acting via the same receptor, may differentially affect gene expression in cells. These studies provide a molecular basis for understanding some of the biological differences between the two ligands and may help to clarify the biological role of IR-A in embryonic/fetal growth and the selective biological advantage that malignant cells producing IGF-II may acquire via IR-A overexpression.
Collapse
Affiliation(s)
- Giuseppe Pandini
- Dipartimento di Medicina Interna e di Medicine Specialistiche, Cattedra di Endocrinologia, University of Catania, USL 34, Ospedale Garibaldi, 95123 Catania, Italy
| | | | | | | | | | | |
Collapse
|
37
|
Hossain GS, van Thienen JV, Werstuck GH, Zhou J, Sood SK, Dickhout JG, de Koning ABL, Tang D, Wu D, Falk E, Poddar R, Jacobsen DW, Zhang K, Kaufman RJ, Austin RC. TDAG51 is induced by homocysteine, promotes detachment-mediated programmed cell death, and contributes to the cevelopment of atherosclerosis in hyperhomocysteinemia. J Biol Chem 2003; 278:30317-27. [PMID: 12738777 DOI: 10.1074/jbc.m212897200] [Citation(s) in RCA: 161] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Hyperhomocysteinemia is an independent risk factor for cardiovascular disease and accelerates atherosclerosis in apoE-/- mice. Despite the observations that homocysteine causes endoplasmic reticulum (ER) stress and programmed cell death (PCD) in cultured human vascular endothelial cells, the cellular factors responsible for this effect and their relevance to atherogenesis have not been completely elucidated. We report here that homocysteine induces the expression of T-cell death-associated gene 51 (TDAG51), a member of the pleckstrin homology-related domain family, in cultured human vascular endothelial cells. This effect was observed for other ER stress-inducing agents, including dithiothreitol and tunicamycin. TDAG51 expression was attenuated in homozygous A/A mutant eukaryotic translation initiation factor 2 alpha mouse embryonic fibroblasts treated with homocysteine or tunicamycin, suggesting that ER stress-induced phosphorylation of eukaryotic translation initiation factor 2 alpha is required for TDAG51 transcriptional activation. Transient overexpression of TDAG51 elicited significant changes in cell morphology, decreased cell adhesion, and promoted detachment-mediated PCD. In support of these in vitro findings, TDAG51 expression was increased and correlated with PCD in the atherosclerotic lesions from apoE-/- mice fed hyperhomocysteinemic diets, compared with mice fed a control diet. Collectively, these findings provide evidence that TDAG51 is induced by homocysteine, promotes detachment-mediated PCD, and contributes to the development of atherosclerosis observed in hyperhomocysteinemia.
Collapse
MESH Headings
- Animals
- Apoptosis
- Arteriosclerosis/pathology
- Blotting, Northern
- Cell Adhesion
- Cell Nucleus/metabolism
- Cells, Cultured
- Cloning, Molecular
- Crosses, Genetic
- DNA, Complementary/metabolism
- Endoplasmic Reticulum/metabolism
- Endothelium, Vascular/cytology
- Fluorescent Antibody Technique, Indirect
- Gene Expression Profiling
- Gene Library
- Homocysteine/chemistry
- Homocysteine/metabolism
- Humans
- Hyperhomocysteinemia/complications
- Hyperhomocysteinemia/pathology
- In Situ Nick-End Labeling
- Mice
- Mice, Inbred C57BL
- Mice, Transgenic
- Phosphorylation
- Protein Binding
- Protein Structure, Tertiary
- RNA, Messenger/metabolism
- Subcellular Fractions
- Time Factors
- Tissue Distribution
- Transcription Factors/chemistry
- Transcription Factors/metabolism
- Transcriptional Activation
- Transfection
- Tunicamycin/pharmacology
- Umbilical Veins
Collapse
Affiliation(s)
- Gazi S Hossain
- Department of Pathology and Molecular Medicine, McMaster University and the Henderson Research Centre, Hamilton, Ontario L8V 1C3, Canada
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|