1
|
Nishiyama M, Miki Y, Tanaka H, Yoshii M, Kuroda K, Kasashima H, Fukuoka T, Tamura T, Shibutani M, Toyokawa T, Lee S, Maeda K. Immunological Analysis of Prognostic Factors in Conversion Surgery Cases for Gastric Cancer. J Surg Res 2025; 306:533-542. [PMID: 39889314 DOI: 10.1016/j.jss.2024.12.053] [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: 06/03/2024] [Revised: 12/04/2024] [Accepted: 12/30/2024] [Indexed: 02/02/2025]
Abstract
INTRODUCTION In order to clarify the optimal strategy regarding conversion surgery (CS) for gastric cancer (GC) patients, we focused on clinicopathological findings, including immunological factors, related to the favorable prognosis in patients with stage IV GC who underwent CS. MATERIALS AND METHODS A total of 25 patients with Stage IV GC who underwent induction chemotherapy (IC) and CS at our hospital between 2010 and 2021 were enrolled in this study. Biopsy specimens before IC and surgical specimens were collected. Immunohistochemical staining was performed using programmed death-ligand 1 (PD-L1) antibody, translationally controlled tumor protein (TCTP) antibody, and CD20 antibody. Prognostic factors were investigated using clinicopathological factors as well as immunological factors such as PD-L1, TCTP, and CD20 expression. RESULTS cN0, ycStage1-2, R0-1 surgery, D2 lymph node dissection, ypN0, and ypStage1-2 were significantly associated with favorable overall survival. Among patients who underwent R0/1 surgery, only histological type was a significant prognostic factor for recurrence-free survival. Low PD-L1 expression before IC and high TCTP expression after IC were significantly associated with favorable recurrence-free survival. CONCLUSIONS In addition to clinical factors, high TCTP expression after IC was identified as a significant favorable prognostic factor, which could help in identifying candidates for CS in the future.
Collapse
Affiliation(s)
- Masaki Nishiyama
- Department of Gastroenterological Surgery, Osaka Metropolitan University Graduate School of Medicine, Osaka, Japan
| | - Yuichiro Miki
- Department of Gastroenterological Surgery, Osaka Metropolitan University Graduate School of Medicine, Osaka, Japan.
| | | | - Mami Yoshii
- Department of Gastroenterological Surgery, Osaka Metropolitan University Graduate School of Medicine, Osaka, Japan
| | - Kenji Kuroda
- Department of Gastroenterological Surgery, Osaka Metropolitan University Graduate School of Medicine, Osaka, Japan
| | - Hiroaki Kasashima
- Department of Gastroenterological Surgery, Osaka Metropolitan University Graduate School of Medicine, Osaka, Japan
| | - Tatsunari Fukuoka
- Department of Gastroenterological Surgery, Osaka Metropolitan University Graduate School of Medicine, Osaka, Japan
| | - Tatsuro Tamura
- Department of Gastroenterological Surgery, Osaka Metropolitan University Graduate School of Medicine, Osaka, Japan
| | - Masatsune Shibutani
- Department of Gastroenterological Surgery, Osaka Metropolitan University Graduate School of Medicine, Osaka, Japan
| | - Takahiro Toyokawa
- Department of Gastroenterological Surgery, Osaka Metropolitan University Graduate School of Medicine, Osaka, Japan
| | - Shigeru Lee
- Department of Gastroenterological Surgery, Osaka Metropolitan University Graduate School of Medicine, Osaka, Japan
| | - Kiyoshi Maeda
- Department of Gastroenterological Surgery, Osaka Metropolitan University Graduate School of Medicine, Osaka, Japan
| |
Collapse
|
2
|
Rho SB, Kim BR, Lee SH, Lee CH. Translationally Controlled Tumor Protein Enhances Angiogenesis in Ovarian Tumors by Activating Vascular Endothelial Growth Factor Receptor 2 Signaling. Biomol Ther (Seoul) 2025; 33:193-202. [PMID: 39664017 PMCID: PMC11704413 DOI: 10.4062/biomolther.2024.206] [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: 10/30/2024] [Revised: 11/25/2024] [Accepted: 11/25/2024] [Indexed: 12/13/2024] Open
Abstract
Translationally controlled tumor protein (TCTP) is a regulatory protein that plays pivotal roles in cellular processes including the cell cycle, apoptosis, microtubule stabilization, embryo development, stress responses, and cancer. However, the molecular mechanism by which it promotes tumor angiogenesis is still unclear. In this study, we explored the mechanisms underlying stimulation of angiogenesis by a novel TCTP. Recombinant TCTP enhanced vascular endothelial growth factor (VEGF)-induced endothelial cell migration, capillary-like tubular structure formation, and cell proliferation by interacting with VEGF receptor 2 (VEGFR-2) in vitro. In contrast, we showed that TCTP knockdown (using short interfering [si]TCTP) led to a decrease in ovarian tumor cells. We also examined the expression of VEGF and hypoxia inducible factor 1 (HIF-1α), an important angiogenic factor. The expression of VEGF as well as HIF-1α was dramatically decreased by siTCTP. Mechanistically, siTCTP inhibited VEGFR-2 tyrosine phosphorylation and phosphorylation of its downstream targets PI3K, Akt, and mTOR. Collectively, these findings indicate that TCTP can promote proliferation and angiogenesis via the VEGFR-2/PI3K and mTOR signaling pathways in ovarian tumor cells, providing new insight into the mechanism behind the involvement of TCTP in tumor angiogenesis.
Collapse
Affiliation(s)
- Seung Bae Rho
- Division of Cancer Biology, Research Institute, National Cancer Center, Goyang 10408, Republic of Korea
| | - Boh-Ram Kim
- College of Pharmacy, Dongguk University, Goyang 10326, Republic of Korea
| | - Seung-Hoon Lee
- Department of Life Science, Yong In University, Yongin 17092, Republic of Korea
| | - Chang Hoon Lee
- College of Pharmacy, Dongguk University, Goyang 10326, Republic of Korea
| |
Collapse
|
3
|
Targeting TCTP sensitizes tumor to T cell-mediated therapy by reversing immune-refractory phenotypes. Nat Commun 2022; 13:2127. [PMID: 35440620 PMCID: PMC9019109 DOI: 10.1038/s41467-022-29611-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Accepted: 03/25/2022] [Indexed: 01/07/2023] Open
Abstract
Immunotherapy has emerged as a powerful approach to cancer treatment. However, immunotherapeutic resistance limits its clinical application. Therefore, identifying immune-resistant factors, which can be targeted by clinically available drugs and it also can be a companion diagnostic marker, is needed to develop combination strategies. Here, using the transcriptome data of patients, and immune-refractory tumor models, we identify TCTP as an immune-resistance factor that correlates with clinical outcome of anti-PD-L1 therapy and confers immune-refractory phenotypes, decreased T cell trafficking to the tumor and resistance to cytotoxic T lymphocyte-mediated tumor cell killing. Mechanistically, TCTP activates the EGFR-AKT-MCL-1/CXCL10 pathway by phosphorylation-dependent interaction with Na, K ATPase. Furthermore, treatment with dihydroartenimsinin, the most effective agent impending the TCTP-mediated-refractoriness, synergizes with T cell-mediated therapy to control immune-refractory tumors. Thus, our findings suggest a role of TCTP in promoting immune-refractoriness, thereby encouraging a rationale for combination therapies to enhance the efficacy of T cell-mediated therapy. Translationally controlled tumor protein (TCTP) regulates several cellular processes, including apoptosis, and is overexpressed in several cancer types. Here, the authors report that high levels of TCTP are associated with poor response to anti-PD-L1 and that TCTP targeting increases the efficacy of T cell-mediated anti-tumor therapy.
Collapse
|
4
|
Dissection of the MKK3 Functions in Human Cancer: A Double-Edged Sword? Cancers (Basel) 2022; 14:cancers14030483. [PMID: 35158751 PMCID: PMC8833818 DOI: 10.3390/cancers14030483] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 01/02/2022] [Accepted: 01/17/2022] [Indexed: 01/27/2023] Open
Abstract
The role played by MKK3 in human cancer is controversial. MKK3 is an evolutionarily conserved protein kinase that activates in response to a variety of stimuli. Phosphorylates, specifically the p38MAPK family proteins, contribute to the regulation of a plethora of cellular processes such as proliferation, differentiation, apoptosis, invasion, and cell migration. Genes in carcinogenesis are classified as oncogenes and tumor suppressors; however, a clear distinction is not always easily made as it depends on the cell context and tissue specificity. The aim of this study is the examination of the potential contribution of MKK3 in cancer through a systematic analysis of the recent literature. The overall results reveal a complex scenario of MKK3’s involvement in cancer. The oncogenic functions of MKK3 were univocally documented in several solid tumors, such as colorectal, prostate cancer, and melanoma, while its tumor-suppressing functions were described in glioblastoma and gastric cancer. Furthermore, a dual role of MKK3 as an oncogene as well as tumor a suppressor has been described in breast, cervical, ovarian, liver, esophageal, and lung cancer. However, overall, more evidence points to its role as an oncogene in these diseases. This review indicates that the oncogenic and tumor-suppressing roles of MKK3 are strictly dependent on the tumor type and further suggests that MKK3 could represent an efficient putative molecular target that requires contextualization within a specific tumor type in order to adequately evaluate its potential effectiveness in designing novel anticancer therapies.
Collapse
|
5
|
Park S, Saravanakumar K, Sathiyaseelan A, Park S, Hu X, Wang MH. Cellular antioxidant properties of nontoxic exopolysaccharide extracted from Lactobacillales (Weissella cibaria) isolated from Korean kimchi. Lebensm Wiss Technol 2022. [DOI: 10.1016/j.lwt.2021.112727] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
6
|
Zhang N, Zhang X, Xu W, Zhang X, Mu Z. CircRNA_103948 inhibits autophagy in colorectal cancer in a ceRNA manner. Ann N Y Acad Sci 2021; 1503:88-101. [PMID: 34480353 DOI: 10.1111/nyas.14679] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 07/30/2021] [Accepted: 08/06/2021] [Indexed: 12/31/2022]
Abstract
Circular RNA (circRNA) is implicated in many types of cancer; however, the expression and role of circRNAs in colorectal cancer (CRC) remains poorly understood. In this study, a circRNA microarray assay was performed to detect abnormally expressed circRNAs in CRC, and tissue arrays were used to determine the prognosis for CRC patients. Cell counting kit-8, clone formation, wound healing, and transwell assays were used to evaluate cell functions in vitro, and a mouse subcutaneous tumor model was designed for in vivo analysis. Autophagy was observed using confocal laser scanning and transmission electron microscopy. The expression of circRNA, miRNA, and mRNA was detected using qPCR; western blot, RNA pull-down assay, RNA immunoprecipitation, and dual luciferase assessment were applied for mechanistic studies. We found that circRNA_103948 expression is upregulated in CRC tissues, compared with adjacent normal tissues, and associated with poor prognosis. Knockdown of circRNA_103948 suppressed CRC both in vitro and in vivo. Mechanistically, circRNA_103948 could directly bind to miR-1236-3p and relieve suppression of the target TPT1. Furthermore, circRNA_103948 inhibited autophagy of CRC cells. Taken together, circRNA_103948 knockdown inhibited CRC cell growth by targeting miR-1236-3p/TPT1 axis-mediated autophagy. Thus, the circRNA_103948/miR-1236-3p/TPT1 axis affects CRC progression via modulation of autophagy.
Collapse
Affiliation(s)
- Nanyang Zhang
- Medical Research Center, the Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Xianxiang Zhang
- Department of Gastrointestinal Surgery, the Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Wenbing Xu
- Medical Research Center, the Affiliated Hospital of Qingdao University, Qingdao, Shandong, China.,Department of Reproductive Medicine, the Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Xiaoxiao Zhang
- Medical Research Center, the Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Zepeng Mu
- Medical Research Center, the Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| |
Collapse
|
7
|
Jeong M, Jeong MH, Kim JE, Cho S, Lee KJ, Park S, Sohn J, Park YG. TCTP protein degradation by targeting mTORC1 and signaling through S6K, Akt, and Plk1 sensitizes lung cancer cells to DNA-damaging drugs. Sci Rep 2021; 11:20812. [PMID: 34675258 PMCID: PMC8531033 DOI: 10.1038/s41598-021-00247-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Accepted: 10/08/2021] [Indexed: 11/14/2022] Open
Abstract
Translationally controlled tumor protein (TCTP) is expressed in many tissues, particularly in human tumors. It plays a role in malignant transformation, apoptosis prevention, and DNA damage repair. The signaling mechanisms underlying TCTP regulation in cancer are only partially understood. Here, we investigated the role of mTORC1 in regulating TCTP protein levels, thereby modulating chemosensitivity, in human lung cancer cells and an A549 lung cancer xenograft model. The inhibition of mTORC1, but not mTORC2, induced ubiquitin/proteasome-dependent TCTP degradation without a decrease in the mRNA level. PLK1 activity was required for TCTP ubiquitination and degradation and for its phosphorylation at Ser46 upon mTORC1 inhibition. Akt phosphorylation and activation was indispensable for rapamycin-induced TCTP degradation and PLK1 activation, and depended on S6K inhibition, but not mTORC2 activation. Furthermore, the minimal dose of rapamycin required to induce TCTP proteolysis enhanced the efficacy of DNA-damaging drugs, such as cisplatin and doxorubicin, through the induction of apoptotic cell death in vitro and in vivo. This synergistic cytotoxicity of these drugs was induced irrespective of the functional status of p53. These results demonstrate a new mechanism of TCTP regulation in which the mTORC1/S6K pathway inhibits a novel Akt/PLK1 signaling axis and thereby induces TCTP protein stabilization and confers resistance to DNA-damaging agents. The results of this study suggest a new therapeutic strategy for enhancing chemosensitivity in lung cancers regardless of the functional status of p53.
Collapse
Affiliation(s)
- Mini Jeong
- Department of Biochemistry and Molecular Biology, Korea University College of Medicine, 73 Koryodae-ro, Sungbuk-gu, Seoul, 02841, Republic of Korea
- Korean Institute of Molecular Medicine and Nutrition, Korea University College of Medicine, Seoul, Republic of Korea
- Division of Allergy and Clinical Immunology, Department of Internal Medicine, ASAN Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Mi Hyeon Jeong
- Department of Biochemistry and Molecular Biology, Korea University College of Medicine, 73 Koryodae-ro, Sungbuk-gu, Seoul, 02841, Republic of Korea
- Korean Institute of Molecular Medicine and Nutrition, Korea University College of Medicine, Seoul, Republic of Korea
| | - Jung Eun Kim
- Department of Biochemistry and Molecular Biology, Korea University College of Medicine, 73 Koryodae-ro, Sungbuk-gu, Seoul, 02841, Republic of Korea
- Korean Institute of Molecular Medicine and Nutrition, Korea University College of Medicine, Seoul, Republic of Korea
| | - Serin Cho
- Department of Biochemistry and Molecular Biology, Korea University College of Medicine, 73 Koryodae-ro, Sungbuk-gu, Seoul, 02841, Republic of Korea
- Korean Institute of Molecular Medicine and Nutrition, Korea University College of Medicine, Seoul, Republic of Korea
| | - Kyoung Jin Lee
- Department of Biochemistry and Molecular Biology, Korea University College of Medicine, 73 Koryodae-ro, Sungbuk-gu, Seoul, 02841, Republic of Korea
- Korean Institute of Molecular Medicine and Nutrition, Korea University College of Medicine, Seoul, Republic of Korea
| | - Serkin Park
- Department of Biochemistry and Molecular Biology, Korea University College of Medicine, 73 Koryodae-ro, Sungbuk-gu, Seoul, 02841, Republic of Korea
- Korean Institute of Molecular Medicine and Nutrition, Korea University College of Medicine, Seoul, Republic of Korea
| | - Jeongwon Sohn
- Department of Biochemistry and Molecular Biology, Korea University College of Medicine, 73 Koryodae-ro, Sungbuk-gu, Seoul, 02841, Republic of Korea
- Korean Institute of Molecular Medicine and Nutrition, Korea University College of Medicine, Seoul, Republic of Korea
| | - Yun Gyu Park
- Department of Biochemistry and Molecular Biology, Korea University College of Medicine, 73 Koryodae-ro, Sungbuk-gu, Seoul, 02841, Republic of Korea.
- Korean Institute of Molecular Medicine and Nutrition, Korea University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
8
|
Zhang L, Ye F, Zuo Z, Cao D, Peng Y, Li Z, Huang J, Duan L. Long noncoding RNA TPT1-AS1 promotes the progression and metastasis of colorectal cancer by upregulating the TPT1-mediated FAK and JAK-STAT3 signalling pathways. Aging (Albany NY) 2021; 13:3779-3797. [PMID: 33428595 PMCID: PMC7906141 DOI: 10.18632/aging.202339] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Accepted: 10/02/2020] [Indexed: 12/11/2022]
Abstract
Tumour protein translationally controlled 1 (TPT1) antisense RNA 1 (TPT1-AS1) is known to be involved in the development and metastasis of cervical and ovarian cancers; however, its biological role in colorectal cancer (CRC) remains unknown. This study aimed to determine the function and mechanism of action of TPT1-AS1 in the progression and metastasis of CRC. Elevated TPT1-AS1 levels were observed in CRC tissues. Furthermore, the high expression levels were found to be correlated with unfavourable clinicopathological characteristics in CRC. Cell function experiments demonstrated that TPT1-AS1 depletion impeded cell proliferation, migration and invasion and enhanced cell adhesion; it also attenuated tumorigenesis and metastasis in vivo. Additionally, TPT1-AS1 was predominately located in the nuclei of the cells and could upregulate the expression of TPT1 by recruiting mixed lineage leukaemia protein-1 (MLL1), which increased the trimethylation of H3K4 me3 in the TPT1 promoter region and subsequently activated FAK and JAK-STAT3 signalling cascades. The inhibition of FAK activation by PF573228 significantly attenuated the oncogenic effect of TPT1-AS1. These findings indicated that TPT1-AS1 promoted tumour progression and metastasis in CRC by upregulating TPT1 levels and activating the FAK and JAK-STAT3 signalling pathways. Thus, TPT1-AS1 may be considered as a potential therapeutic target for CRC.
Collapse
Affiliation(s)
- Leiyi Zhang
- Department of Minimally Invasive Surgery, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China
| | - Fei Ye
- Department of Minimally Invasive Surgery, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China
| | - Zhongkun Zuo
- Department of Minimally Invasive Surgery, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China
| | - Ding Cao
- Department of Minimally Invasive Surgery, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China
| | - Yu Peng
- Department of Minimally Invasive Surgery, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China
| | - Zedong Li
- Department of Minimally Invasive Surgery, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China
| | - Jiangsheng Huang
- Department of Minimally Invasive Surgery, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China
| | - Lunxi Duan
- Department of Minimally Invasive Surgery, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China
| |
Collapse
|
9
|
Kumar R, Saran S. Comparative modelling unravels the structural features of eukaryotic TCTP implicated in its multifunctional properties: an in silico approach. J Mol Model 2021; 27:20. [PMID: 33410974 DOI: 10.1007/s00894-020-04630-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Accepted: 12/01/2020] [Indexed: 10/22/2022]
Abstract
Comparative modelling helps compare the structure and functions of a given protein, to track the path of its origin and evolution and also guide in structure-based drug discovery. Presently, this has been applied for modelling the tertiary structure of highly conserved eukaryotic TCTP (translationally controlled tumour protein) which is involved in a plethora of functions during growth and development and also acts as a biomarker for many cancers like lung, breast, and prostate cancer. The modelled TCTP structures of different organisms belonging to the eukaryotic group showed similar spatial arrangement of structural units except loops and similar patterns of root mean square deviation (RMSD), root mean square fluctuation, and radius of gyration (Rg) inspected through molecular dynamics simulations. Essential dynamics (ED) analyses revealed different domains that exhibited different motions for the assistance in its multifunctional properties. Construction of a free-energy landscape (FEL) based on Rg versus RMSD was employed to characterize the folding behaviours of structures and observe that all proteins had nearly similar conformation and topologies, indicating common thermodynamic/kinetic pathways. A physico-chemical interaction study demonstrated the helices and sheets were well stabilized with ample amounts of bonding compared to turns or loops and charged residues were more accessible to solvent molecules. Hence, the current study reveals the important structural features of TCTP that aid in diverse functions in a wide range of organisms, thus extending our knowledge of TCTP and also providing a venue for designing the potent inhibitors against it.
Collapse
Affiliation(s)
- Rakesh Kumar
- School of Life Sciences, Jawaharlal Nehru University, New Delhi, 110067, India
| | - Shweta Saran
- School of Life Sciences, Jawaharlal Nehru University, New Delhi, 110067, India.
| |
Collapse
|
10
|
Zhang J, Gao S, Zhang Y, Yi H, Xu M, Xu J, Liu H, Ding Z, He H, Wang H, Hao Z, Sun L, Liu Y, Wei F. MiR-216a-5p inhibits tumorigenesis in Pancreatic Cancer by targeting TPT1/mTORC1 and is mediated by LINC01133. Int J Biol Sci 2020; 16:2612-2627. [PMID: 32792860 PMCID: PMC7415429 DOI: 10.7150/ijbs.46822] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Accepted: 07/01/2020] [Indexed: 12/25/2022] Open
Abstract
MiR-216a-5p has opposite effects on tumorigenesis and progression in the context of different tumors, acting as either a tumor suppressor or an oncogene. However, the expression and function of miR-216a-5p in pancreatic cancer (PC) is not well characterized. In this study, we found miR-216a-5p was significantly downregulated in PC tissues and cell lines, which showed a negative correlation with peripancreatic lymph, perineural invasion and TNM stage of PCs patients. We made use of functional assays to reveal that miR-216a-5p inhibited growth and migration of PC cells in vitro and in vivo. Then, by employing the bioinformatics analysis and luciferase reporter assay, we demonstrated TPT1 was a potential target of miR-216a-5p, which contributes to tumor malignance by mediating mTORC1 pathway-associated autophagy. Furthermore, bioinformatics analysis and RNA pulldown confirmed that miR-216a-5p was mediated by LINC01133, which sponge miR-216a-5p, as a competing endogenous RNA (ceRNA). Collectively, our study revealed an important role of LINC01133/miR-216a-5p/TPT1 axis in the genesis and progression of PCs, which provides potential biomarkers for clinical diagnosis and therapy of PCs.
Collapse
Affiliation(s)
- Jian Zhang
- Department of Hepatobiliary and Pancreas Surgery, Jilin University First Hospital, Changchun, China
| | - Shuohui Gao
- Department of Gastrointestinal Colorectal Surgery, China-Japan Union hospital of Jilin University, Changchun, China
| | - Yandong Zhang
- Department of Hepatobiliary and Pancreas Surgery, Jilin University First Hospital, Changchun, China
| | - Huixin Yi
- Genetic Engineering Laboratory of PLA, Institute of Military Veterinary Medicine, Academy of Military Medical Sciences, Changchun, China
| | - Mengxian Xu
- Genetic Engineering Laboratory of PLA, Institute of Military Veterinary Medicine, Academy of Military Medical Sciences, Changchun, China
| | - Jialun Xu
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University
| | - Huan Liu
- Department of Hepatobiliary and Pancreas Surgery, Jilin University First Hospital, Changchun, China
| | - Zhichen Ding
- Department of Hepatobiliary and Pancreas Surgery, Jilin University First Hospital, Changchun, China
| | - Hongbin He
- Ruminant Diseases Research Center, College of Life Sciences, Shandong Normal University, Jinan, China
| | - Hongmei Wang
- Ruminant Diseases Research Center, College of Life Sciences, Shandong Normal University, Jinan, China
| | - Zhuo Hao
- Genetic Engineering Laboratory of PLA, Institute of Military Veterinary Medicine, Academy of Military Medical Sciences, Changchun, China
| | - Liankun Sun
- Department of Pathophysiology, College of Basic Medicine Sciences, Jilin University, Changchun, China
| | - Yan Liu
- Genetic Engineering Laboratory of PLA, Institute of Military Veterinary Medicine, Academy of Military Medical Sciences, Changchun, China
| | - Feng Wei
- Department of Hepatobiliary and Pancreas Surgery, Jilin University First Hospital, Changchun, China
| |
Collapse
|
11
|
The role of translationally controlled tumor protein in proliferation of Drosophila intestinal stem cells. Proc Natl Acad Sci U S A 2019; 116:26591-26598. [PMID: 31843907 DOI: 10.1073/pnas.1910850116] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Translationally controlled tumor protein (TCTP) is a highly conserved protein functioning in multiple cellular processes, ranging from growth to immune responses. To explore the role of TCTP in tissue maintenance and regeneration, we employed the adult Drosophila midgut, where multiple signaling pathways interact to precisely regulate stem cell division for tissue homeostasis. Tctp levels were significantly increased in stem cells and enteroblasts upon tissue damage or activation of the Hippo pathway that promotes regeneration of intestinal epithelium. Stem cells with reduced Tctp levels failed to proliferate during normal tissue homeostasis and regeneration. Mechanistically, Tctp forms a complex with multiple proteins involved in translation and genetically interacts with ribosomal subunits. In addition, Tctp increases both Akt1 protein abundance and phosphorylation in vivo. Altogether, Tctp regulates stem cell proliferation by interacting with key growth regulatory signaling pathways and the translation process in vivo.
Collapse
|
12
|
Wu W, Gao H, Li X, Zhu Y, Peng S, Yu J, Zhan G, Wang J, Liu N, Guo X. LncRNA TPT1-AS1 promotes tumorigenesis and metastasis in epithelial ovarian cancer by inducing TPT1 expression. Cancer Sci 2019; 110:1587-1598. [PMID: 30941821 PMCID: PMC6500995 DOI: 10.1111/cas.14009] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Revised: 02/01/2019] [Accepted: 03/03/2019] [Indexed: 01/01/2023] Open
Abstract
Increasing numbers of studies have confirmed that long noncoding RNA (lncRNA) play a critical role in epithelial ovarian cancer (EOC) progression. However, the potential function of the lncRNA tumor protein translationally controlled 1 (TPT1) antisense RNA 1 (TPT1-AS1) in EOC is unclear. In this study, we aimed to uncover the biological roles and regulatory mechanisms of TPT1-AS1 in EOC progression and metastasis. First, TPT1-AS1 expression was significantly higher in EOC metastatic tissue and cell lines than in their respective control counterparts. In addition, ectopic TPT1-AS1 expression was strongly associated with unfavorable EOC clinicopathological features, including FIGO stage, tumor size and tumor differentiation. TPT1-AS1 overexpression remarkably induced cell proliferation, migration and invasion, and significantly attenuated cell adhesion ability in vitro and facilitated nude mouse subcutaneous xenograft growth and intraperitoneal metastasis in vivo, while the downregulation of TPT1-AS1 expression produced the opposite effect in vitro. Mechanistically, TPT1-AS1 was proven to be primarily distributed in EOC cell nuclei and positively modulated TPT1 promoter activity and transcription. Moreover, the oncogenic effects of TPT1-AS1 could be reversed by TPT1 depletion, and the PI3K/AKT signaling pathway downstream of TPT1 was also altered. These results suggested that TPT1-AS1 induced EOC tumor growth and metastasis through TPT1 and downstream PI3K/AKT signaling and that TPT1-AS1 may be a promising therapeutic target for EOC.
Collapse
MESH Headings
- Animals
- Female
- Humans
- Mice
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Carcinoma, Ovarian Epithelial/genetics
- Carcinoma, Ovarian Epithelial/metabolism
- Carcinoma, Ovarian Epithelial/pathology
- Cell Adhesion
- Cell Line, Tumor
- Cell Movement
- Cell Proliferation
- Cell Survival
- Gene Expression Regulation, Neoplastic
- Neoplasm Metastasis
- Neoplasm Transplantation
- Ovarian Neoplasms/genetics
- Ovarian Neoplasms/metabolism
- Ovarian Neoplasms/pathology
- Phosphatidylinositol 3-Kinases/metabolism
- Promoter Regions, Genetic
- Proto-Oncogene Proteins c-akt/metabolism
- RNA, Long Noncoding/genetics
- Signal Transduction
- Tumor Protein, Translationally-Controlled 1
- Up-Regulation
- RNA, Antisense
Collapse
Affiliation(s)
- Weimin Wu
- Department of Obstetrics and GynecologyShanghai First Maternity and Infant HospitalTongji University School of MedicineShanghaiChina
| | - Hao Gao
- Department of Obstetrics and GynecologyShanghai First Maternity and Infant HospitalTongji University School of MedicineShanghaiChina
| | - Xiaofeng Li
- Department of Obstetrics and GynecologyShanghai First Maternity and Infant HospitalTongji University School of MedicineShanghaiChina
| | - Yong Zhu
- Department of Obstetrics and GynecologyThe First Affiliated HospitalShihezi University School of MedicineXinjiangChina
| | - Shumin Peng
- Chongqing Health Center for Women and ChildrenChongqingChina
| | - Jing Yu
- Department of PathologyShanghai First Maternity and Infant HospitalTongji University School of MedicineShanghaiChina
| | - Guangxi Zhan
- Department of Obstetrics and GynecologyShanghai First Maternity and Infant HospitalTongji University School of MedicineShanghaiChina
| | - Jiapo Wang
- Department of Obstetrics and GynecologyShanghai First Maternity and Infant HospitalTongji University School of MedicineShanghaiChina
| | - Na Liu
- Department of Obstetrics and GynecologyShanghai First Maternity and Infant HospitalTongji University School of MedicineShanghaiChina
| | - Xiaoqing Guo
- Department of Obstetrics and GynecologyShanghai First Maternity and Infant HospitalTongji University School of MedicineShanghaiChina
| |
Collapse
|
13
|
Radiosensitivity of Cancer Cells Is Regulated by Translationally Controlled Tumor Protein. Cancers (Basel) 2019; 11:cancers11030386. [PMID: 30893896 PMCID: PMC6468585 DOI: 10.3390/cancers11030386] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Revised: 03/07/2019] [Accepted: 03/17/2019] [Indexed: 01/08/2023] Open
Abstract
Translationally controlled tumor protein (TCTP) is a ubiquitous multifunctional protein that is essential for cell survival. This study reveals that the regulation of radiosensitivity of cancer cells is yet another function of TCTP. The relationship between endogenous TCTP levels and sensitivity to radiation was examined in breast cancer cell lines (T47D, MDA-MB-231, and MCF7) and lung cancer cells lines (A549, H1299, and H460). Cancer cells with high expression levels of TCTP were more resistant to radiation. TCTP overexpression inhibited radiation-induced cell death, while silencing TCTP led to an increase in radiosensitivity. DNA damage in the irradiated TCTP-silenced A549 cells was greater than in irradiated control shRNA-transfected A549 cells. p53, a well-known reciprocal regulator of TCTP, was increased in irradiated TCTP down-regulated A549 cells. Moreover, introduction of p53 siRNA in TCTP knocked-down A549 cells abrogated the increased radiosensitivity induced by TCTP knockdown. An in vivo xenograft study also confirmed enhanced radiosensitivity in TCTP down-regulated A549 cells. These findings suggest that TCTP has the potential to serve as a therapeutic target to overcome radiation resistance in cancer, a major problem for the effective treatment of cancers.
Collapse
|
14
|
Ungewiß H, Rötzer V, Meir M, Fey C, Diefenbacher M, Schlegel N, Waschke J. Dsg2 via Src-mediated transactivation shapes EGFR signaling towards cell adhesion. Cell Mol Life Sci 2018; 75:4251-4268. [PMID: 29980799 PMCID: PMC11105603 DOI: 10.1007/s00018-018-2869-x] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Revised: 06/19/2018] [Accepted: 07/03/2018] [Indexed: 12/31/2022]
Abstract
Rapidly renewing epithelial tissues such as the intestinal epithelium require precise tuning of intercellular adhesion and proliferation to preserve barrier integrity. Here, we provide evidence that desmoglein 2 (Dsg2), an adhesion molecule of desmosomes, controls cell adhesion and proliferation via epidermal growth factor receptor (EGFR) signaling. Dsg2 is required for EGFR localization at intercellular junctions as well as for Src-mediated EGFR activation. Src binds to EGFR and is required for localization of EGFR and Dsg2 to cell-cell contacts. EGFR is critical for cell adhesion and barrier recovery. In line with this, Dsg2-deficient enterocytes display impaired barrier properties and increased cell proliferation. Mechanistically, Dsg2 directly interacts with EGFR and undergoes heterotypic-binding events on the surface of living enterocytes via its extracellular domain as revealed by atomic force microscopy. Thus, our study reveals a new mechanism by which Dsg2 via Src shapes EGFR function towards cell adhesion.
Collapse
Affiliation(s)
- Hanna Ungewiß
- Department I, Institute of Anatomy and Cell Biology, Ludwig Maximilians University Munich, Pettenkoferstr. 11, 80336, Munich, Germany
| | - Vera Rötzer
- Department I, Institute of Anatomy and Cell Biology, Ludwig Maximilians University Munich, Pettenkoferstr. 11, 80336, Munich, Germany
| | - Michael Meir
- Department of General, Visceral, Vascular and Paediatric Surgery, Julius-Maximilians-Universität, Oberdürrbacher Str. 6, 97080, Würzburg, Germany
| | - Christina Fey
- Department for Tissue Engineering and Regenerative Medicine, University Hospital Würzburg, Röntgenring 11, 97070, Würzburg, Germany
| | - Markus Diefenbacher
- Department of Biochemistry and Molecular Biochemistry, University of Würzburg, Am Hubland, 97074, Würzburg, Germany
| | - Nicolas Schlegel
- Department of General, Visceral, Vascular and Paediatric Surgery, Julius-Maximilians-Universität, Oberdürrbacher Str. 6, 97080, Würzburg, Germany
| | - Jens Waschke
- Department I, Institute of Anatomy and Cell Biology, Ludwig Maximilians University Munich, Pettenkoferstr. 11, 80336, Munich, Germany.
| |
Collapse
|
15
|
Some Biological Consequences of the Inhibition of Na,K-ATPase by Translationally Controlled Tumor Protein (TCTP). Int J Mol Sci 2018; 19:ijms19061657. [PMID: 29867020 PMCID: PMC6032315 DOI: 10.3390/ijms19061657] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Revised: 05/29/2018] [Accepted: 05/30/2018] [Indexed: 12/17/2022] Open
Abstract
Na,K-ATPase is an ionic pump that regulates the osmotic equilibrium and membrane potential of cells and also functions as a signal transducer. The interaction of Na,K-ATPase with translationally controlled tumor protein (TCTP) results, among others, in the inhibition of the former's pump activity and in the initiation of manifold biological and pathological phenomena. These phenomena include hypertension and cataract development in TCTP-overexpressing transgenic mice, as well as the induction of tumorigenesis signaling pathways and the activation of Src that ultimately leads to cell proliferation and migration. This review attempts to collate the biological effects of Na,K-ATPase and TCTP interaction and suggests that this interaction has the potential to serve as a possible therapeutic target for selected diseases.
Collapse
|
16
|
Aberrant expression of translationally controlled tumor protein (TCTP) can lead to radioactive susceptibility and chemosensitivity in lung cancer cells. Oncotarget 2017; 8:101922-101935. [PMID: 29254214 PMCID: PMC5731924 DOI: 10.18632/oncotarget.21747] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Accepted: 07/29/2017] [Indexed: 01/15/2023] Open
Abstract
Translationally controlled tumor protein (TCTP) is an evolutionally highly conserved protein which has been implicated as a biomarker for cancer cell reversion although the mechanism is not very clear. This makes it a potential target for cancer therapy. P53 tumor suppressor protein is important in regulating cell growth, it can induce either growth arrest or programmed cell death (apoptosis). TCTP and P53 has been reported that can regulate the protein level of each other. Here we proved that TCTP is a malignancy state keeper in lung cancer and lower level of TCTP protein made cells more sensitive to stressful condition. No obvious difference has been observed from wildtype and the TCTP knockdown lung cancer cells (A549) when located in the normal circumstances. While under the stressful condition, the existence of higher protein level of TCTP can protect cells from apoptosis. TCTP and P53 formed a feedback signal pathway and through it to regulate the downstream Akt signal pathways to make the lung cancer cells keep a higher metabolism level and protect cancer cells from apoptosis induced by outside stress.
Collapse
|
17
|
You Y, Tan J, Gong Y, Dai H, Chen H, Xu X, Yang A, Zhang Y, Bie P. MicroRNA-216b-5p Functions as a Tumor-suppressive RNA by Targeting TPT1 in Pancreatic Cancer Cells. J Cancer 2017; 8:2854-2865. [PMID: 28928875 PMCID: PMC5604218 DOI: 10.7150/jca.18931] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2016] [Accepted: 05/01/2017] [Indexed: 01/06/2023] Open
Abstract
MicroRNAs (miRNAs) are increasingly recognized as being involved in pancreatic cancer progression by directly regulating the expression of their targets. In this study, we showed that miR-216b-5p expression was significantly decreased in pancreatic cancer tissues and cell lines. In addition, low miR-216b-5p expression was significantly associated with large tumor size and advanced TNM stage. Meanwhile, both Kaplan-Meier and multivariate survival analysis showed that decreased miR-216b-5p expression was associated with overall survival. miR-216b-5p over-expression repressed pancreatic cancer cell proliferation and induced cell cycle arrest and cell apoptosis in vitro and inhibited tumorigenesis in vivo. The translationally controlled tumor protein (TPT1) was identified as a novel direct target of miR-216b-5p. miR-216b-5p up-regulation suppressed TPT1 expression. Moreover, TPT1 mRNA expression levels were increased in pancreatic cancer tissues, and were inversely correlated with miR-216b-5p expression. TPT1 down-regulation had similar effects as miR-216b-5p up-regulation on pancreatic cancer cell progression. The restoration of TPT1 reversed the effect of miR-216b-5p on pancreatic cancer cell progression. Furthermore, we found that miR-216b-5p up-regulation suppressed Pim-3, Cyclin B1, p-Bad and Bcl-xL protein expression. However, the effect of miR-216b-5p up-regulation was partly reversed by TPT1 up-regulation in vitro. Taken together, our findings suggested that miR-216b-5p functions as a potential tumor suppressor by regulating TPT1 in pancreatic cancer cells, and it may represent a potential therapeutic target for patients with pancreatic cancer.
Collapse
Affiliation(s)
- Yu You
- Department of Hepatobiliary Surgery, Southwest Hospital, Third Military Medical University, Chongqing, 400038, China
| | - Jiaxin Tan
- Department of Hepatobiliary Surgery, Southwest Hospital, Third Military Medical University, Chongqing, 400038, China
| | - Yi Gong
- Department of Hepatobiliary Surgery, Southwest Hospital, Third Military Medical University, Chongqing, 400038, China
| | - Haisu Dai
- Department of Hepatobiliary Surgery, Southwest Hospital, Third Military Medical University, Chongqing, 400038, China
| | - Haowei Chen
- Department of Hepatobiliary Surgery, Southwest Hospital, Third Military Medical University, Chongqing, 400038, China
| | - Xuejun Xu
- Department of Hepatobiliary Surgery, Southwest Hospital, Third Military Medical University, Chongqing, 400038, China
| | - Aigang Yang
- Department of Hepatobiliary Surgery, Southwest Hospital, Third Military Medical University, Chongqing, 400038, China
| | - Yujun Zhang
- Department of Hepatobiliary Surgery, Southwest Hospital, Third Military Medical University, Chongqing, 400038, China
| | - Ping Bie
- Department of Hepatobiliary Surgery, Southwest Hospital, Third Military Medical University, Chongqing, 400038, China
| |
Collapse
|
18
|
Function of translationally controlled tumor protein (TCTP) in Eudrilus eugeniae regeneration. PLoS One 2017; 12:e0175319. [PMID: 28403226 PMCID: PMC5389791 DOI: 10.1371/journal.pone.0175319] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Accepted: 03/23/2017] [Indexed: 01/07/2023] Open
Abstract
TCTP (Translationally Controlled Tumour Protein) is a multifunctional protein that plays a role in the development, immune system, tumour reversion, and maintenance of stem cells. The mRNA of the Tpt1 gene is over-expressed during liver regeneration. But, the function of the protein in regeneration is not known. To study the role of the protein in regeneration, the earthworm Eudrilus eugeniae was chosen. First, the full length cDNA of the Tpt1 gene was sequenced. The size of the cDNA is 504 bp and the protein has 167 amino acids. The highest level of TCTP expression was documented in the worm after three days of regeneration. The protein was found to be expressed specifically in the epithelial layer of the skin. During regeneration, the protein expression was found to be the highest at the tip of blastema. The pharmacological suppression of TCTP using nutlin-3 and TCTP RNAi experiments resulted in the failure of the regeneration process. The suppression of TCTP caused the arrest of proliferation in posterior amputated worms. The severe cell death was documented in the amputated region of nutlin-3 injected worm. The silencing of TCTP has blocked the modification of clitellar segments. The experiments confirm that TCTP has major functions in the upstream signalling of cell proliferation in the early regeneration process in E. eugeniae.
Collapse
|
19
|
Bae SY, Byun S, Bae SH, Min DS, Woo HA, Lee K. TPT1 (tumor protein, translationally-controlled 1) negatively regulates autophagy through the BECN1 interactome and an MTORC1-mediated pathway. Autophagy 2017; 13:820-833. [PMID: 28409693 DOI: 10.1080/15548627.2017.1287650] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
TPT1/TCTP (tumor protein, translationally-controlled 1) is highly expressed in tumor cells, known to participate in various cellular activities including protein synthesis, growth and cell survival. In addition, TPT1 was identified as a direct target of the tumor suppressor TP53/p53 although little is known about the mechanism underlying the anti-survival function of TPT1. Here, we describe a role of TPT1 in the regulation of the MTORC1 pathway through modulating the molecular machinery of macroautophagy/autophagy. TPT1 inhibition induced cellular autophagy via the MTORC1 and AMPK pathways, which are inhibited and activated, respectively, during treatment with the MTOR inhibitor rapamycin. We also found that the depletion of TPT1 potentiated rapamycin-induced autophagy by synergizing with MTORC1 inhibition. We further demonstrated that TPT1 knockdown altered the BECN1 interactome, a representative MTOR-independent pathway, to stimulate autophagosome formation, via downregulating BCL2 expression through activating MAPK8/JNK1, and thereby enhancing BECN1-phosphatidylinositol 3-kinase (PtdIns3K)-UVRAG complex formation. Furthermore, reduced TPT1 promoted autophagic flux by modulating not only early steps of autophagy but also autophagosome maturation. Consistent with in vitro findings, in vivo organ analysis using Tpt1 heterozygote knockout mice showed that autophagy is enhanced because of haploinsufficient TPT1 expression. Overall, our study demonstrated the novel role of TPT1 as a negative regulator of autophagy that may have potential use in manipulating various diseases associated with autophagic dysfunction.
Collapse
Affiliation(s)
- Seong-Yeon Bae
- a Graduate School of Pharmaceutical Sciences , College of Pharmacy, Ewha Womans University , Seoul , Korea
| | - Sanguine Byun
- b Division of Bioengineering , College of Life and Sciences and Bioengineering, Incheon National University , Incheon , Korea
| | - Soo Han Bae
- c Severance Biomedical Science Institute, Yonsei Biomedical Research Institute, Yonsei University College of Medicine , Seoul , Korea
| | - Do Sik Min
- d Department of Molecular Biology , College of Natural Sciences, Pusan National University , Busan , Korea
| | - Hyun Ae Woo
- a Graduate School of Pharmaceutical Sciences , College of Pharmacy, Ewha Womans University , Seoul , Korea
| | - Kyunglim Lee
- a Graduate School of Pharmaceutical Sciences , College of Pharmacy, Ewha Womans University , Seoul , Korea
| |
Collapse
|
20
|
Zhang J, Shim G, de Toledo SM, Azzam EI. The Translationally Controlled Tumor Protein and the Cellular Response to Ionizing Radiation-Induced DNA Damage. Results Probl Cell Differ 2017; 64:227-253. [DOI: 10.1007/978-3-319-67591-6_12] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/11/2023]
|
21
|
Function of Translationally Controlled Tumor Protein in Organ Growth: Lessons from Drosophila Studies. Results Probl Cell Differ 2017; 64:173-191. [PMID: 29149408 DOI: 10.1007/978-3-319-67591-6_8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Regulation of cell growth and proliferation is crucial for development and function of organs in all animals. Genetic defects in growth control can lead to developmental disorders and cancers. Translationally controlled tumor protein (TCTP) is a family of evolutionarily conserved proteins implicated in cancer. Recent studies have revealed multiple roles of TCTP in diverse cellular events, but TCTP functions in vivo are poorly understood in vertebrate systems. We have used Drosophila melanogaster, the fruit fly, as a model organism for genetic dissection of Tctp function. Our studies have shown that Tctp is essential for organ development by regulating growth signaling. Furthermore, it is required for genome stability by promoting DNA repair and chromatin remodeling in the nucleus. Thus, Tctp acts as a multifaceted cytosolic and nuclear factor for regulating organ growth and genome stability. In this chapter, we describe an overview of our findings on Tctp functions in Drosophila and discuss their implications in cancer.
Collapse
|
22
|
Bommer UA. The Translational Controlled Tumour Protein TCTP: Biological Functions and Regulation. Results Probl Cell Differ 2017; 64:69-126. [PMID: 29149404 DOI: 10.1007/978-3-319-67591-6_4] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
The Translational Controlled Tumour Protein TCTP (gene symbol TPT1, also called P21, P23, Q23, fortilin or histamine-releasing factor, HRF) is a highly conserved protein present in essentially all eukaryotic organisms and involved in many fundamental cell biological and disease processes. It was first discovered about 35 years ago, and it took an extended period of time for its multiple functions to be revealed, and even today we do not yet fully understand all the details. Having witnessed most of this history, in this chapter, I give a brief overview and review the current knowledge on the structure, biological functions, disease involvements and cellular regulation of this protein.TCTP is able to interact with a large number of other proteins and is therefore involved in many core cell biological processes, predominantly in the response to cellular stresses, such as oxidative stress, heat shock, genotoxic stress, imbalance of ion metabolism as well as other conditions. Mechanistically, TCTP acts as an anti-apoptotic protein, and it is involved in DNA-damage repair and in cellular autophagy. Thus, broadly speaking, TCTP can be considered a cytoprotective protein. In addition, TCTP facilitates cell division through stabilising the mitotic spindle and cell growth through modulating growth signalling pathways and through its interaction with the proteosynthetic machinery of the cell. Due to its activities, both as an anti-apoptotic protein and in promoting cell growth and division, TCTP is also essential in the early development of both animals and plants.Apart from its involvement in various biological processes at the cellular level, TCTP can also act as an extracellular protein and as such has been involved in modulating whole-body defence processes, namely in the mammalian immune system. Extracellular TCTP, typically in its dimerised form, is able to induce the release of cytokines and other signalling molecules from various types of immune cells. There are also several examples, where TCTP was shown to be involved in antiviral/antibacterial defence in lower animals. In plants, the protein appears to have a protective effect against phytotoxic stresses, such as flooding, draught, too high or low temperature, salt stress or exposure to heavy metals. The finding for the latter stress condition is corroborated by earlier reports that TCTP levels are considerably up-regulated upon exposure of earthworms to high levels of heavy metals.Given the involvement of TCTP in many biological processes aimed at maintaining cellular or whole-body homeostasis, it is not surprising that dysregulation of TCTP levels may promote a range of disease processes, foremost cancer. Indeed a large body of evidence now supports a role of TCTP in at least the most predominant types of human cancers. Typically, this can be ascribed to both the anti-apoptotic activity of the protein and to its function in promoting cell growth and division. However, TCTP also appears to be involved in the later stages of cancer progression, such as invasion and metastasis. Hence, high TCTP levels in tumour tissues are often associated with a poor patient outcome. Due to its multiple roles in cancer progression, TCTP has been proposed as a potential target for the development of new anti-cancer strategies in recent pilot studies. Apart from its role in cancer, TCTP dysregulation has been reported to contribute to certain processes in the development of diabetes, as well as in diseases associated with the cardiovascular system.Since cellular TCTP levels are highly regulated, e.g. in response to cell stress or to growth signalling, and because deregulation of this protein contributes to many disease processes, a detailed understanding of regulatory processes that impinge on TCTP levels is required. The last section of this chapter summarises our current knowledge on the mechanisms that may be involved in the regulation of TCTP levels. Essentially, expression of the TPT1 gene is regulated at both the transcriptional and the translational level, the latter being particularly advantageous when a rapid adjustment of cellular TCTP levels is required, for example in cell stress responses. Other regulatory mechanisms, such as protein stability regulation, may also contribute to the regulation of overall TCTP levels.
Collapse
Affiliation(s)
- Ulrich-Axel Bommer
- School of Medicine, Graduate Medicine, University of Wollongong, Wollongong, NSW, 2522, Australia.
| |
Collapse
|
23
|
Amphiregulin enhances intercellular adhesion molecule-1 expression and promotes tumor metastasis in human osteosarcoma. Oncotarget 2016; 6:40880-95. [PMID: 26503469 PMCID: PMC4747375 DOI: 10.18632/oncotarget.5679] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2015] [Accepted: 09/13/2015] [Indexed: 02/07/2023] Open
Abstract
Osteosarcoma is a common, high malignant, and metastatic bone cancer. Amphiregulin (AREG) has been associated with cancer cellular activities. However, the effect of AREG on metastasis activity in human osteosarcoma cells has yet to be determined. We determined that AREG increases the expression of intercellular adhesion molecule-1 (ICAM-1) through PI3K/Akt signaling pathway via its interaction with the epidermal growth factor receptor, thus resulting in the enhanced cell migration of osteosarcoma. Furthermore, AREG stimulation increased the association of NF-κB to ICAM-1 promoter which then up-regulated ICAM-1 expression. Finally, we observed that shRNA silencing of AREG decreased osteosarcoma metastasis in vivo. Our findings revealed a relationship between osteosarcoma metastatic potential and AREG expression and the modulating effect of AREG on ICAM-1 expression.
Collapse
|
24
|
Li S, Chen M, Xiong Q, Zhang J, Cui Z, Ge F. Characterization of the Translationally Controlled Tumor Protein (TCTP) Interactome Reveals Novel Binding Partners in Human Cancer Cells. J Proteome Res 2016; 15:3741-3751. [PMID: 27607350 DOI: 10.1021/acs.jproteome.6b00556] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Translationally controlled tumor protein (TCTP) is a highly conserved housekeeping protein present in eukaryotic organisms. It is involved in regulating many fundamental processes and plays a critical role in tumor reversion and tumorigenesis. Increasing evidence suggests that TCTP plays a role in the regulation of cell fate determination and is a promising therapeutic target for cancer. To decipher the exact mechanisms by which TCTP functions and how all these functions are integrated, we analyzed the interactome of TCTP in HeLa cells by coimmunoprecipitation (IP) and mass spectrometry (MS). A total of 98 proteins were identified. We confirmed the in vitro and in vivo association of TCTP with six of the identified binding proteins using reciprocal IP and bimolecular fluorescence complementation (BiFC) analysis, respectively. Moreover, TCTP interacted with Y-box-binding protein 1 (YBX1), and their interaction was localized to the N-terminal region of TCTP and the 1-129 amino acid (aa) residues of YBX1. The YBX1 protein plays an important role in cell proliferation, RNA splicing, DNA repair, drug resistance, and stress response to extracellular signals. These data suggest that the interaction of TCTP with YBX1 might cooperate or coordinate their functions in the control of diverse regulatory pathways in cancer cells. Taken together, our results not only reveal a large number of TCTP-associated proteins that possess pleiotropic functions, but also provide novel insights into the molecular mechanisms of TCTP in tumorigenesis.
Collapse
Affiliation(s)
- Siting Li
- Graduate University, Chinese Academy of Sciences , Beijing 100049, China
| | - Minghai Chen
- Graduate University, Chinese Academy of Sciences , Beijing 100049, China
| | | | | | | | | |
Collapse
|
25
|
Prasad S, Gupta SC, Tyagi AK. Reactive oxygen species (ROS) and cancer: Role of antioxidative nutraceuticals. Cancer Lett 2016; 387:95-105. [PMID: 27037062 DOI: 10.1016/j.canlet.2016.03.042] [Citation(s) in RCA: 638] [Impact Index Per Article: 70.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Revised: 03/22/2016] [Accepted: 03/23/2016] [Indexed: 12/21/2022]
Abstract
Extensive research over the past half a century indicates that reactive oxygen species (ROS) play an important role in cancer. Although low levels of ROS can be beneficial, excessive accumulation can promote cancer. One characteristic of cancer cells that distinguishes them from normal cells is their ability to produce increased numbers of ROS and their increased dependence on an antioxidant defense system. ROS are produced as a byproduct intracellularly by mitochondria and other cellular elements and exogenously by pollutants, tobacco, smoke, drugs, xenobiotics, and radiation. ROS modulate various cell signaling pathways, which are primarily mediated through the transcription factors NF-κB and STAT3, hypoxia-inducible factor-1α, kinases, growth factors, cytokines and other proteins, and enzymes; these pathways have been linked to cellular transformation, inflammation, tumor survival, proliferation, invasion, angiogenesis, and metastasis of cancer. ROS are also associated with epigenetic changes in genes, which is helpful in diagnosing diseases. This review considers the role of ROS in the various stages of cancer development. Finally, we provide evidence that nutraceuticals derived from Mother Nature are highly effective in eliminating cancer cells.
Collapse
Affiliation(s)
- Sahdeo Prasad
- Cytokine Research Laboratory, Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA.
| | - Subash C Gupta
- Cytokine Research Laboratory, Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
| | - Amit K Tyagi
- Cytokine Research Laboratory, Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
| |
Collapse
|
26
|
Kloc M, Liu Y, Zhang L, Tejpal N, Kubiak J, Ghobrial R, Li X. TCTP Silencing in Ovarian Cancer Cells Results in Actin Cytoskeleton Remodeling and Motility Increase. ACTA ACUST UNITED AC 2015. [DOI: 10.6000/1927-7229.2015.04.04.1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022]
|
27
|
Translationally controlled tumor protein induces epithelial to mesenchymal transition and promotes cell migration, invasion and metastasis. Sci Rep 2015; 5:8061. [PMID: 25622969 PMCID: PMC4306963 DOI: 10.1038/srep08061] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2014] [Accepted: 12/29/2014] [Indexed: 12/21/2022] Open
Abstract
Translationally controlled tumor protein (TCTP), is a highly conserved protein involved in fundamental processes, such as cell proliferation and growth, tumorigenesis, apoptosis, pluripotency, and cell cycle regulation. TCTP also inhibits Na,K-ATPase whose subunits have been suggested as a marker of epithelial-to-mesenchymal transition (EMT), a crucial step during tumor invasiveness, metastasis and fibrosis. We hypothesized that, TCTP might also serve as an EMT inducer. This study attempts to verify this hypothesis. We found that overexpression of TCTP in a porcine renal proximal tubule cell line, LLC-PK1, induced EMT-like phenotypes with the expected morphological changes and appearance of EMT related markers. Conversely, depletion of TCTP reversed the induction of these EMT phenotypes. TCTP overexpression also enhanced cell migration via activation of mTORC2/Akt/GSK3β/β-catenin, and invasiveness by activating MMP-9. Moreover, TCTP depletion in melanoma cells significantly reduced pulmonary metastasis by inhibiting the development of mesenchymal-like phenotypes. Overall, these findings support our hypothesis that TCTP is a positive regulator of EMT and suggest that modulation of TCTP expression is a potential approach to inhibit the invasiveness and migration of cancer cells and the attendant pathologic processes including metastasis.
Collapse
|
28
|
Osteoclastogenic activity of translationally-controlled tumor protein (TCTP) with reciprocal repression of p21. FEBS Lett 2014; 588:4026-31. [PMID: 25263704 DOI: 10.1016/j.febslet.2014.09.022] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2014] [Revised: 08/29/2014] [Accepted: 09/16/2014] [Indexed: 11/24/2022]
Abstract
Translationally-controlled tumor protein (TCTP) plays a role in a number of cellular processes, but there is limited information about its function in cell differentiation. Previous observations of a twofold induction of TCTP mRNA during osteoclast differentiation prompted us to investigate its involvement in osteoclast differentiation. The osteoclastogenicity of TCTP gradually expressed during osteoclast differentiation was confirmed in mouse and human cells using loss-of-function studies and TCTP heterogeneous mice and transgenic mice. Higher expression ratios of TCTP to p21 could represent TCTP-mediated phenotypic induction of osteoclast differentiation accompanied by p21 down-regulation, attenuating the proliferation of osteoclast precursor cells.
Collapse
|
29
|
Yan X, Liang F, Li D, Zheng J. Ouabain elicits human glioblastoma cells apoptosis by generating reactive oxygen species in ERK-p66SHC-dependent pathway. Mol Cell Biochem 2014; 398:95-104. [PMID: 25217205 DOI: 10.1007/s11010-014-2208-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2014] [Accepted: 08/30/2014] [Indexed: 01/26/2023]
Abstract
Excessive reactive oxygen species (ROS) generation has been implicated as one of main agents in ouabain-induced anticancer effect. Unfortunately, the signaling pathways under it are not very clarified. In the present study, we investigated the molecular mechanism involved in ouabain-induced ROS generation and cell apoptosis on human U373MG and U87MG glioma cells. Ouabain-induced glioblastoma cells apoptosis and increased ROS generation. Clearance ROS by three different ROS scavenger partly, but not totally, reversed ouabain's effect on cell apoptosis. Ouabain-induced ROS generation was not regulated by calcium overload, reduced nicotinamide adenine dinucleotide phosphate oxidation, but by p66Shc phosphorylation. Ouabain treatment increased p66Shc Ser36 phosphorylation. Knockdown of p66Shc by siRNA significantly inhibited ROS generations in response to ouabain. Ouabain-induced p66Shc phosphorylation through Src/Ras/extracellular signal-regulated kinase signal pathway. Our results uncovered a novel signaling pathway with p66Shc, ouabain-induced ROS generation, and glioblastoma cell apoptosis.
Collapse
Affiliation(s)
- Xiaofei Yan
- Department of Biochemistry and Molecular Biology, Medical School, Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, People's Republic of China,
| | | | | | | |
Collapse
|
30
|
Li L, Li X, Han X, Yang T, Fu J, Zhang Y, Gou W. An ovarian cancer model with positive ER: Reversion of ER antagonist resistance by Src blockade. Oncol Rep 2014; 32:943-50. [PMID: 24968896 DOI: 10.3892/or.2014.3284] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2014] [Accepted: 04/16/2014] [Indexed: 11/06/2022] Open
Abstract
Approximately 60% of ovarian cancers are positive for the estrogen receptor (ER); however, ER-targeted treatment is disappointing due to drug resistance as compared with breast cancer. In estrogen-sensitive cancers, estrogen activates Src to phosphorylate p27 promoting its degradation and increasing cell cycle progression. Since Src is frequently activated in ovarian cancers, we investigated whether combined Src and ER blockade by saracatinib and fulvestrant would circumvent anti-estrogen resistance. In 20 out of 40 enrolled patients with immunohistochemically ER-positive ovarian cancer, phosphorylated Src (p-Src) at the site of 416 tyrosine was expressed with a propensity for metastasis and a poorer disease-free survival (DFS) at 3 years following ER antagonist treatment. The effects of ER and Src blockade on cell cycle were assayed in estrogen receptor α (ERα)-positive ovarian cancer. We observed that Src activity was fairly greater in anti-estrogen-resistant ovarian cancer cells than that in the anti-estrogen-sensitive cell line. Estrogen activated Src via ER-Src binding and ER translocation from cytoplasm to nucleus. Mitogenesis was mediated via ERα, not ERβ. Combined saracatinib and fulvestrant increased p27 and inhibited cell cycle progression. Furthermore, dual therapy induced autophagy and inhibited ovarian cancer xenograft growth more effectively than monotherapy. Saracatinib facilitated the therapeutic effects of fulvestrant by antagonizing the estrogen-mediated Src activation. These are supportive of further preclinical assessment of combined fulvestrant and saracatinib in patients with ovarian cancer.
Collapse
Affiliation(s)
- Long Li
- Department of Gynaecology and Obstetrics, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Xiaojun Li
- Department of Second Thoracosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Xiaobing Han
- Department of Gynaecology and Obstetrics, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Ting Yang
- Department of Gynaecology and Obstetrics, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Jing Fu
- Department of Gynaecology and Obstetrics, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Yunfeng Zhang
- Department of Second Thoracosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Wenli Gou
- Department of Gynaecology and Obstetrics, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| |
Collapse
|
31
|
Wang X, Ji X, Chen J, Yan D, Zhang Z, Wang Q, Xi X, Feng Y. SOX2 enhances the migration and invasion of ovarian cancer cells via Src kinase. PLoS One 2014; 9:e99594. [PMID: 24937695 PMCID: PMC4061006 DOI: 10.1371/journal.pone.0099594] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2014] [Accepted: 05/15/2014] [Indexed: 02/07/2023] Open
Abstract
Ovarian cancer is the leading cause of death among gynecologic cancers and is the fifth leading cause of all cancer-related deaths among women. The development of novel molecular targets is therefore important to many patients. Recently, the SRY-related transcription factor SOX2 has been widely reported to be involved in multiple pathophysiological diseases, including maintenance of stem cell characteristics and carcinogenesis. Up to now, SOX2 has been mainly shown to promote the development of cancer, although its inhibitory roles in cancer have also been reported. However, the role of SOX2 in ovarian cancer is largely unknown. In the present study, we detected the expression of SOX2 in 64 human serous ovarian carcinoma (SOC) tissues and paired corresponding metastatic specimens using immunohistochemistry. The results showed that the expression of SOX2 in primary tumors is much lower than that in the corresponding metastatic lesions. We further found that SOX2 overexpression promotes proliferation, migration and invasion, while inhibiting adhesion abilities of SOC cells. Finally, we found that SOX2 targets Src kinase, a non-receptor tyrosine kinase that regulates cell migration, invasion and adhesion in SOC cells. Together, these results suggested that Src kinase is a key molecule in SOX2-mediated migration and invasion of SOC cells.
Collapse
Affiliation(s)
- Xiaojie Wang
- Department of Obstetrics and Gynecology, Shanghai Jiao Tong University Affiliated First People's Hospital, Shanghai, China
| | - Xiaoning Ji
- Department of Obstetrics and Gynecology, Shanghai Jiao Tong University Affiliated First People's Hospital, Shanghai, China
| | - Jiazhou Chen
- Department of Obstetrics and Gynecology, Shanghai Jiao Tong University Affiliated First People's Hospital, Shanghai, China
| | - Dong Yan
- Department of Obstetrics and Gynecology, Shanghai Jiao Tong University Affiliated First People's Hospital, Shanghai, China
| | - Zhenbo Zhang
- Department of Obstetrics and Gynecology, Shanghai Jiao Tong University Affiliated First People's Hospital, Shanghai, China
| | - Qifeng Wang
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xiaowei Xi
- Department of Obstetrics and Gynecology, Shanghai Jiao Tong University Affiliated First People's Hospital, Shanghai, China
| | - Youji Feng
- Department of Obstetrics and Gynecology, Shanghai Jiao Tong University Affiliated First People's Hospital, Shanghai, China
| |
Collapse
|
32
|
Chen K, Huang C, Yuan J, Cheng H, Zhou R. Long-term artificial selection reveals a role of TCTP in autophagy in mammalian cells. Mol Biol Evol 2014; 31:2194-211. [PMID: 24890374 DOI: 10.1093/molbev/msu181] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Understanding genomic variation and detecting selection signatures in a genome under selection have been great challenges for a century. Activation, development/exhaustion of primordial follicles in mammalian ovary determines reproductive success, menopause/end of female reproductive life. However, molecular mechanisms underlying oogenesis, particularly under artificial selection, are largely unknown. We report that a proteome-wide scan for selection signatures in the genome over 9,000 years of artificial pressure on the ovary revealed a general picture of selection signatures in the genome, especially genomic variations through artificial selection were detected in promoter and intron regions. Crossbreeding between domestic and wild species results in more than half of the protein spots exhibiting heterosis. Translationally controlled tumor protein (TCTP) is upregulated by artificial selection and positively regulates autophagy through the AMP-activated protein kinase pathway. Notably, TCTP interacts with ATG16 complex. In addition to cytoplasmic autophagy, nucleophagy occurs in the nuclei of granulosa and cumulus cells in ovaries, indicating an importance of the nuclear material for degradation by nucleophagy. Our findings provide insight into cellular and molecular mechanisms relevant for improvement of ovary functions, and identify selection signatures in the genome for ovary function over long-term artificial selection pressure.
Collapse
Affiliation(s)
- Ke Chen
- Department of Genetics, College of Life Sciences, Wuhan University, Wuhan, People's Republic of China
| | - Chunhua Huang
- Department of Genetics, College of Life Sciences, Wuhan University, Wuhan, People's Republic of China
| | - Jia Yuan
- Department of Genetics, College of Life Sciences, Wuhan University, Wuhan, People's Republic of China
| | - Hanhua Cheng
- Department of Genetics, College of Life Sciences, Wuhan University, Wuhan, People's Republic of China
| | - Rongjia Zhou
- Department of Genetics, College of Life Sciences, Wuhan University, Wuhan, People's Republic of China
| |
Collapse
|
33
|
Hiepen C, Benn A, Denkis A, Lukonin I, Weise C, Boergermann JH, Knaus P. BMP2-induced chemotaxis requires PI3K p55γ/p110α-dependent phosphatidylinositol (3,4,5)-triphosphate production and LL5β recruitment at the cytocortex. BMC Biol 2014; 12:43. [PMID: 24885555 PMCID: PMC4071339 DOI: 10.1186/1741-7007-12-43] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2014] [Accepted: 05/13/2014] [Indexed: 01/13/2023] Open
Abstract
Background BMP-induced chemotaxis of mesenchymal progenitors is fundamental for vertebrate development, disease and tissue repair. BMP2 induces Smad and non-Smad signalling. Whereas signal transduction via Smads lead to transcriptional responses, non-Smad signalling induces both, transcriptional and immediate/early non-transcriptional responses. However, the molecular mechanisms by which BMP2 facilitates planar cell polarity, cortical actin rearrangements, lamellipodia formation and chemotaxis of mesenchymal progenitors are poorly understood. Our aim was to uncover the molecular mechanism by which BMP2 facilitates chemotaxis via the BMP2-dependent activation of PI3K and spatiotemporal control of PIP3 production important for actin rearrangements at the mesenchymal cell cytocortex. Results We unveiled the molecular mechanism by which BMP2 induces non-Smad signalling by PI3K and the role of the second messenger PIP3 in BMP2-induced planar cell polarity, cortical actin reorganisation and lamellipodia formation. By using protein interaction studies, we identified the class Ia PI3K regulatory subunit p55γ to act as a specific and non-redundant binding partner for BMP receptor type II (BMPRII) in concert with the catalytic subunit p110α. We mapped the PI3K interaction to a region within the BMPRII kinase. Either BMP2 stimulation or increasing amounts of BMPRI facilitated p55γ association with BMPRII, but BMPRII kinase activity was not required for the interaction. We visualised BMP2-dependent PIP3 production via PI3K p55γ/p110α and were able to localise PIP3 to the leading edge of intact cells during the process of BMP2-induced planar cell polarity and actin dependent lamellipodia formation. Using mass spectrometry, we found the highly PIP3-sensitive PH-domain protein LL5β to act as a novel BMP2 effector in orchestrating cortical actin rearrangements. By use of live cell imaging we found that knock-down of p55γ or LL5β or pharmacological inhibition of PI3K impaired BMP2-induced migratory responses. Conclusions Our results provide evidence for an important contribution of the BMP2-PI3K (p55γ/p110α)- PIP3-LL5β signalling axis in mesenchymal progenitor cell chemotaxis. We demonstrate molecular insights into BMP2-induced PI3K signalling on the level of actin reorganisation at the leading edge cytocortex. These findings are important to better understand BMP2–induced cytoskeletal reorganisation and chemotaxis of mesenchymal progenitors in different physiological or pathophysiological contexts.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Petra Knaus
- Institute for Chemistry and Biochemistry, Freie Universität Βerlin, 14195 Berlin, Germany.
| |
Collapse
|
34
|
Interaction of translationally controlled tumor protein with Apaf-1 is involved in the development of chemoresistance in HeLa cells. BMC Cancer 2014; 14:165. [PMID: 24606760 PMCID: PMC4015309 DOI: 10.1186/1471-2407-14-165] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2013] [Accepted: 02/27/2014] [Indexed: 11/11/2022] Open
Abstract
Background Translationally controlled tumor protein (TCTP), alternatively called fortilin, is believed to be involved in the development of the chemoresistance of tumor cells against anticancer drugs such as etoposide, taxol, and oxaliplatin, the underlying mechanisms of which still remain elusive. Methods Cell death analysis of TCTP-overexpressing HeLa cells was performed following etoposide treatment to assess the mitochondria-dependent apoptosis. Apoptotic pathway was analyzed through measuring the cleavage of epidermal growth factor receptor (EGFR) and phospholipase C-γ (PLC-γ), caspase activation, mitochondrial membrane perturbation, and cytochrome c release by flow cytometry and western blotting. To clarify the role of TCTP in the inhibition of apoptosome, in vitro apoptosome reconstitution and immunoprecipitation was used. Pull-down assay and silver staining using the variants of Apaf-1 protein was applied to identify the domain that is responsible for its interaction with TCTP. Results In the present study, we confirmed that adenoviral overexpression of TCTP protects HeLa cells from cell death induced by cytotoxic drugs such as taxol and etoposide. TCTP antagonized the mitochondria-dependent apoptotic pathway following etoposide treatment, including mitochondrial membrane damage and resultant cytochrome c release, activation of caspase-9, and -3, and eventually, the cleavage of EGFR and PLC-γ. More importantly, TCTP interacts with the caspase recruitment domain (CARD) of Apaf-1 and is incorporated into the heptameric Apaf-1 complex, and that C-terminal cleaved TCTP specifically associates with Apaf-1 of apoptosome in apoptosome-forming condition thereby inhibiting the amplification of caspase cascade. Conclusions TCTP protects the cancer cells from etoposide-induced cell death by inhibiting the mitochondria-mediated apoptotic pathway. Interaction of TCTP with Apaf-1 in apoptosome is involved in the molecular mechanism of TCTP-induced chemoresistance. These findings suggest that TCTP may serve as a therapeutic target for chemoresistance in cancer treatment.
Collapse
|
35
|
Acunzo J, Baylot V, So A, Rocchi P. TCTP as therapeutic target in cancers. Cancer Treat Rev 2014; 40:760-9. [PMID: 24650927 DOI: 10.1016/j.ctrv.2014.02.007] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2013] [Revised: 02/19/2014] [Accepted: 02/21/2014] [Indexed: 01/07/2023]
Abstract
The translationally controlled tumor protein (TCTP) is a highly conserved protein present in eukaryotic organisms. This protein, located both in the cytoplasmic and the nucleus, is expressed in various tissues and is regulated in response to a wide range of extracellular stimuli. TCTP interacts with itself and other protein including MCL1 and p53. TCTP has been shown to play an important role in physiological events, such as cell proliferation, cell death and immune responses but also in stress response and tumor reversion. Moreover, TCTP expression is associated with malignancy and chemoresistance. In this review, we will evaluate pathways regulated by TCTP and current inhibitory strategy to target TCTP in cancerous diseases.
Collapse
Affiliation(s)
- Julie Acunzo
- (a)Inserm, U1068, CRCM, Marseille F-13009, France; (b)Institut Paoli-Calmettes, Marseille F-13009, France; (c)Aix-Marseille Univ., Marseille F-13284, France; (d)CNRS, UMR7258, Marseille F-13009, France
| | - Virginie Baylot
- (a)Inserm, U1068, CRCM, Marseille F-13009, France; (b)Institut Paoli-Calmettes, Marseille F-13009, France; (c)Aix-Marseille Univ., Marseille F-13284, France; (d)CNRS, UMR7258, Marseille F-13009, France
| | - Alan So
- (e)University of British Columbia, The Vancouver Prostate Centre 2660- Oak St Vancouver, BC V6H3Z6, Canada
| | - Palma Rocchi
- (a)Inserm, U1068, CRCM, Marseille F-13009, France; (b)Institut Paoli-Calmettes, Marseille F-13009, France; (c)Aix-Marseille Univ., Marseille F-13284, France; (d)CNRS, UMR7258, Marseille F-13009, France.
| |
Collapse
|
36
|
Tsai MJ, Yang-Yen HF, Chiang MK, Wang MJ, Wu SS, Chen SH. TCTP is essential for β-cell proliferation and mass expansion during development and β-cell adaptation in response to insulin resistance. Endocrinology 2014; 155:392-404. [PMID: 24248465 DOI: 10.1210/en.2013-1663] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The perinatal period is critical for β-cell mass establishment, which is characterized by a transient burst in proliferation to increase β-cell mass in response to the need for glucose homeostasis throughout life. In adulthood, the ability of β-cells to grow, proliferate, and expand their mass is also characteristic of pathological states of insulin resistance. Translationally controlled tumor-associated protein (TCTP), an evolutionarily highly conserved protein that is implicated in cell growth and proliferation, has been identified as a novel glucose-regulated survival-supporting protein in pancreatic β-cells. In this study, the enhanced β-cell proliferation detected both during the perinatal developmental period and in insulin-resistant states in high-fat diet-fed mice was found to parallel the expression of TCTP in pancreatic β-cells. Specific knockout of TCTP in β-cells led to increased expression of total and nuclear Forkhead box protein O1 and tumor suppressor protein 53, and decreased expression of p70S6 kinase phosphorylation and cyclin D2 and cyclin-dependent kinase 2. This resulted in decreased β-cell proliferation and growth, reduced β-cell mass, and insulin secretion. Together, these effects led to hyperglycemia. These observations suggest that TCTP is essential for β-cell mass expansion during development and β-cell adaptation in response to insulin resistance.
Collapse
Affiliation(s)
- Ming-Jen Tsai
- PhD Program in Pharmacology and Toxicology (M.J.-T., S.-H.C.) and Department of Pharmacology (S.-S.W., S.-H.C.), School of Medicine, Tzu Chi University, Hualien 970, Taiwan; Departments of Emergency Medicine (M.J.-T.) and Medical Research (M.-J.W.), Neuro-Medical Scientific Center, Buddhist Tzu Chi General Hospital, Hualien 970, Taiwan; Institutes of Molecular Biology (H.-F.Y.-Y.), Academia Sinica, Taipei 115, Taiwan; and Department of Life Science and Institute of Molecular Biology (M.-K.C.), National Chung-Cheng University, Chia-Yi 621, Taiwan
| | | | | | | | | | | |
Collapse
|
37
|
Gu X, Yao L, Ma G, Cui L, Li Y, Liang W, Zhao B, Li K. TCTP promotes glioma cell proliferation in vitro and in vivo via enhanced β-catenin/TCF-4 transcription. Neuro Oncol 2013; 16:217-27. [PMID: 24311645 DOI: 10.1093/neuonc/not194] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Background The translationally controlled tumor protein (TCTP) is a multifunctional protein that plays important roles in immune responses, cell proliferation, tumorigenicity and cell apoptosis. Here, we examined the clinical value of TCTP in glioma patient survival and investigated the functional roles and mechanism of TCTP in glioma development. Methods TCTP expression was determined through immunohistochemical staining, immunoblotting, and quantitative real-time PCR (qRT-PCR). TCTP or TCF-4 expression was silenced using short hairpin (sh) RNA. In vitro cell proliferation was detected using MTT, BrdU and colony formation assays, and in vivo tumor growth was performed using the xenograft model. TCTP/TCF-4/β-catenin association was detected using a co-immunoprecipitation (co-IP) assay. TCF-4 transcription activity was detected using a TOPflash/FOPflash report gene assay. Wnt/β-catenin-targeted gene expression was detected through Western blotting. Results TCTP protein levels were significantly elevated in high-grade gliomas compared with low-grade gliomas and normal brain tissues. Importantly, the expression of TCTP was significantly associated with poorer overall survival and disease-free survival, and TCTP also reduced the survival rate after treatment with radiotherapy and temozolomide (RT-TMZ) for glioma patients. The ectopic expression of TCTP enhanced glioma cell proliferation both in vitro and in vivo, whereas the knockdown of TCTP inhibited this effect. Similarly, the overexpression of TCTP increased β-catenin binding to TCF-4, TOPflash report gene transcription activity, and the expression of Wnt/β-catenin signaling target genes including c-Myc and cyclin D1; notably, the knockdown of TCTP reduced these effects. The knockdown of TCF-4 using shRNA rescued the enhanced cell proliferation induced by the overexpression of TCTP. Conclusion TCTP is associated with reduced survival of glioma patients and induces glioma tumor growth through enhanced Wnt/β-catenin signaling.
Collapse
Affiliation(s)
- Xuefeng Gu
- Corresponding authors: Keshen Li, MD, PhD, Institute of Neurology, Guangdong Medical College, Zhanjiang 524001, China. ); Bin Zhao, MD, PhD, Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Affiliated Hospital of Guangdong Medical College, Zhanjiang 524001, China (
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Cellular functions regulated by phosphorylation of EGFR on Tyr845. Int J Mol Sci 2013; 14:10761-90. [PMID: 23702846 PMCID: PMC3709701 DOI: 10.3390/ijms140610761] [Citation(s) in RCA: 79] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2013] [Revised: 05/06/2013] [Accepted: 05/13/2013] [Indexed: 11/17/2022] Open
Abstract
The Src gene product (Src) and the epidermal growth factor receptor (EGFR) are prototypes of oncogene products and function primarily as a cytoplasmic non-receptor tyrosine kinase and a transmembrane receptor tyrosine kinase, respectively. The identification of Src and EGFR, and the subsequent extensive investigations of these proteins have long provided cutting edge research in cancer and other molecular and cellular biological studies. In 1995, we reported that the human epidermoid carcinoma cells, A431, contain a small fraction of Src and EGFR in which these two kinase were in physical association with each other, and that Src phosphorylates EGFR on tyrosine 845 (Y845) in the Src-EGFR complex. Y845 of EGFR is located in the activation segment of the kinase domain, where many protein kinases contain kinase-activating autophosphorylation sites (e.g., cAMP-dependent protein kinase, Src family kinases, transmembrane receptor type tyrosine kinases) or trans-phosphorylation sites (e.g., cyclin-dependent protein kinase, mitogen-activated protein kinase, Akt protein kinase). A number of studies have demonstrated that Y845 phosphorylation serves an important role in cancer as well as normal cells. Here we compile the experimental facts involving Src phosphorylation of EGFR on Y845, by which cell proliferation, cell cycle control, mitochondrial regulation of cell metabolism, gamete activation and other cellular functions are regulated. We also discuss the physiological relevance, as well as structural insights of the Y845 phosphorylation.
Collapse
|
39
|
4-Bromo-2-(piperidin-1-yl)thiazol-5-yl-phenyl methanone (12b) inhibits Na+/K(+)-ATPase and Ras oncogene activity in cancer cells. Eur J Med Chem 2013; 63:213-23. [PMID: 23474907 DOI: 10.1016/j.ejmech.2013.01.046] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2012] [Revised: 01/16/2013] [Accepted: 01/22/2013] [Indexed: 11/23/2022]
Abstract
The in vitro growth inhibitory activity of 26 thiazoles (including 4-halogeno-2,5-disubtituted-1,3-thiazoles) and 5 thienothiazoles was assessed on a panel of 6 human cancer cell lines, including glioma cell lines. (4-Chloro-2-(piperidin-1-yl)thiazol-5-yl)(phenyl)methanone (12a) and (4-bromo-2-(piperidin-1-yl)thiazol-5-yl)(phenyl)methanone (12b) displayed ~10 times greater in vitro growth inhibitory activity than perillyl alcohol (POH), which therapeutically benefits glioma patients through the inhibition of both alpha-1 Na(+)/K(+)-ATPase (NAK) and Ras oncogene activity. The in vitro cytostatic activities (as revealed by quantitative videomicroscopy) displayed by 12a and 12b were independent of the intrinsic resistance to pro-apoptotic stimuli associated with cancer cells. Compounds 12a and 12b displayed relatively similar inhibitory activities on purified guinea pig brain preparations that mainly express NAK alpha-2 and alpha-3 subunits, whereas only compound 12b was efficacious against purified guinea pig kidney preparations that mainly express the NAK alpha-1 subunit, which is also expressed in gliomas, melanomas and non-small-cell lung cancers NSCLCs.
Collapse
|
40
|
Reinhard L, Tidow H, Clausen MJ, Nissen P. Na(+),K (+)-ATPase as a docking station: protein-protein complexes of the Na(+),K (+)-ATPase. Cell Mol Life Sci 2013; 70:205-22. [PMID: 22695678 PMCID: PMC11113973 DOI: 10.1007/s00018-012-1039-9] [Citation(s) in RCA: 118] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2012] [Revised: 05/13/2012] [Accepted: 05/23/2012] [Indexed: 12/13/2022]
Abstract
The Na(+),K(+)-ATPase, or sodium pump, is well known for its role in ion transport across the plasma membrane of animal cells. It carries out the transport of Na(+) ions out of the cell and of K(+) ions into the cell and thus maintains electrolyte and fluid balance. In addition to the fundamental ion-pumping function of the Na(+),K(+)-ATPase, recent work has suggested additional roles for Na(+),K(+)-ATPase in signal transduction and biomembrane structure. Several signaling pathways have been found to involve Na(+),K(+)-ATPase, which serves as a docking station for a fast-growing number of protein interaction partners. In this review, we focus on Na(+),K(+)-ATPase as a signal transducer, but also briefly discuss other Na(+),K(+)-ATPase protein-protein interactions, providing a comprehensive overview of the diverse signaling functions ascribed to this well-known enzyme.
Collapse
Affiliation(s)
- Linda Reinhard
- Danish National Research Foundation, Centre for Membrane Pumps in Cells and Disease-PUMPKIN, Department of Molecular Biology and Genetics, 8000 Aarhus C, Denmark
| | - Henning Tidow
- Danish National Research Foundation, Centre for Membrane Pumps in Cells and Disease-PUMPKIN, Department of Molecular Biology and Genetics, 8000 Aarhus C, Denmark
| | - Michael J. Clausen
- Danish National Research Foundation, Centre for Membrane Pumps in Cells and Disease-PUMPKIN, Department of Molecular Biology and Genetics, 8000 Aarhus C, Denmark
| | - Poul Nissen
- Danish National Research Foundation, Centre for Membrane Pumps in Cells and Disease-PUMPKIN, Department of Molecular Biology and Genetics, 8000 Aarhus C, Denmark
| |
Collapse
|
41
|
Lo WY, Wang HJ, Chiu CW, Chen SF. miR-27b-regulated TCTP as a novel plasma biomarker for oral cancer: from quantitative proteomics to post-transcriptional study. J Proteomics 2012; 77:154-66. [PMID: 22902387 DOI: 10.1016/j.jprot.2012.07.039] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2012] [Revised: 07/20/2012] [Accepted: 07/23/2012] [Indexed: 01/12/2023]
Abstract
We combined an iTRAQ-based quantitative proteomic analysis and the miRNA determination to profile potentially novel biomarker from oral cancer. There are 757 and 674 unique proteins identified from proteomic analysis, and 13 proteins displayed consistent underexpression (<0.67 fold) in normal tissues in comparison with the corresponding tumor tissues. After preliminary screening, EGFR, OAT, TPT1, ITGA6, G3BP1 and CB39L were the six genes validated in the 37 oral cancer patients (T1, n=10; T2, n=10; T3, n=10 and T4, n=7). The TPT1, ITGA6 and CAB39L genes were displayed the higher transcriptions level in the tumor tissues and the TPT1, ITGA6 and CAB39L proteins were also shown overexpression in the tumor tissues from the same patients. The miR-19a, 19b, 27a, 27b, 186, 203 and 377 transcripts were predicted and the miR-27b level was shown to significantly reduce in the tumor tissues and the plasma of OSCC patients. In the in vitro study, the overexpression of miR-27b only significantly decreased TCTP protein and gene levels in both HSC-3 and Cal-27 cell lines. Our results demonstrate that human miR-27b regulates the expression of the TCTP tumor protein, and circulating miR-27b may be useful as a biomarker for oral cancer research.
Collapse
Affiliation(s)
- Wan-Yu Lo
- Graduate Institute of Integrated Medicine, China Medical University, Taichung, Taiwan
| | | | | | | |
Collapse
|
42
|
Maeng J, Kim HY, Shin DH, Lee K. Transduction of translationally controlled tumor protein employing TCTP-derived protein transduction domain. Anal Biochem 2012; 435:47-53. [PMID: 23256924 DOI: 10.1016/j.ab.2012.11.029] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2012] [Revised: 11/26/2012] [Accepted: 11/26/2012] [Indexed: 11/26/2022]
Abstract
Protein transduction domains (PTDs), which are cell-penetrating peptides, have been employed for delivery of various cargos. We previously showed that the N-terminal fragment of translationally controlled tumor protein functions as a PTD (TCTP-PTD) by as yet poorly understood mechanisms. In this study, we generated several green fluorescent protein (GFP)-tagged TCTP fusion proteins by conjugating a single PTD or tandem PTDs at the N-terminus, the C-terminus, and both termini and compared their transduction efficiencies in human lung adenocarcinoma A549 cells to determine whether the protein transducing function of TCTP depends on the location or the number of PTD moieties in the TCTP molecule. Fluorimetric analysis and Western blotting assays revealed that TCTP-GFP fusion protein containing one or two TCTP-PTDs at its N-terminus showed more efficient cellular entry than either the C-terminal TCTP-PTD or TCTP-PTD with PTDs located at both the N- and C-terminals. This study demonstrates the feasibility of transduction of TCTP target cells employing its TCTP-PTD by simple co-incubation with purified proteins.
Collapse
Affiliation(s)
- Jeehye Maeng
- College of Pharmacy, Center for Cell Signaling and Drug Discovery Research, Ewha Womans University, Seoul 120-750, Republic of Korea
| | | | | | | |
Collapse
|
43
|
Zhao H, Wu G, Cao X. EGFR dependent subcellular communication was responsible for morphine mediated AC superactivation. Cell Signal 2012; 25:417-28. [PMID: 23142605 DOI: 10.1016/j.cellsig.2012.10.016] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2012] [Revised: 10/01/2012] [Accepted: 10/01/2012] [Indexed: 10/27/2022]
Abstract
Compensatory adenylyl cyclase (AC) superactivation has been postulated to be responsible for the development of morphine tolerance and dependence, the underlying mechanism was demonstrated to comprise c-Src-dependent upregulation of AC5 within the lipid rafts. In the present study, we demonstrated that chronic morphine treatment sensitized EGFR signaling by augmenting EGFR phosphorylation and translocation into ER, which was essential for CRT-MOR tethering within the lipid rafts and AC5 superactivation. Intriguingly, synaptic clustering of CRT-MOR was dependent on EGFR phosphorylation and presumed to implicate in alignment and organization of synaptic compartments. Taken together, our data raised the possibility that an adaptive change in MOR and EGFR signal systems might establish CRT related subcellular communication, the signaling network within brain synaptic zone was proposed to implicate in morphine tolerance and dependence.
Collapse
Affiliation(s)
- Hui Zhao
- Department of Integrative Medicine and Neurobiology, National Key lab of Medical Neurobiology, Institute of Brain Research Sciences, Shanghai Medical College, Fudan University, PR China.
| | | | | |
Collapse
|
44
|
Biological effects of Mammalian translationally controlled tumor protein (TCTP) on cell death, proliferation, and tumorigenesis. Biochem Res Int 2012; 2012:204960. [PMID: 22675633 PMCID: PMC3364544 DOI: 10.1155/2012/204960] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2012] [Accepted: 03/20/2012] [Indexed: 12/22/2022] Open
Abstract
Translationally controlled tumor protein (TCTP) is a highly conserved protein found in eukaryotes, across animal and plant kingdoms and even in yeast. Mammalian TCTP is ubiquitously expressed in various tissues and cell types. TCTP is a multifunctional protein which plays important roles in a number of cell physiological events, such as immune responses, cell proliferation, tumorigenicity, and cell death, including apoptosis. Recent identification of TCTP as an antiapoptotic protein has attracted interest of many researchers in the field. The mechanism of antiapoptotic activity, however, has not been solved completely, and TCTP might inhibit other types of cell death. Cell death (including apoptosis) is closely linked to proliferation and tumorigenesis. In this context, we review recent findings regarding the role of TCTP in cell death, proliferation, and tumorigenesis and discuss the mechanisms.
Collapse
|
45
|
Kim DK, Nam BY, Li JJ, Park JT, Lee SH, Kim DH, Kim JY, Kang HY, Han SH, Yoo TH, Han DS, Kang SW. Translationally controlled tumour protein is associated with podocyte hypertrophy in a mouse model of type 1 diabetes. Diabetologia 2012; 55:1205-17. [PMID: 22311416 DOI: 10.1007/s00125-012-2467-7] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2011] [Accepted: 12/31/2011] [Indexed: 02/06/2023]
Abstract
AIMS/HYPOTHESIS Translationally controlled tumour protein (TCTP) is thought to be involved in cell growth by regulating mTOR complex 1 (mTORC1) signalling. As diabetes characteristically induces podocyte hypertrophy and mTORC1 has been implicated in this process, TCTP may have a role in the pathogenesis of diabetes-induced podocyte hypertrophy. METHODS We investigated the effects and molecular mechanisms of TCTP in diabetic mice and in high glucose-stimulated cultured podocytes. To characterise the role of TCTP, we conducted lentivirus-mediated gene silencing of TCTP both in vivo and in vitro. RESULTS Glomerular production of TCTP was significantly higher in streptozotocin induced-diabetic DBA/2J mice than in control animals. Double-immunofluorescence staining for TCTP and synaptopodin revealed that podocyte was the principal cell responsible for this increase. TCTP knockdown attenuated the activation of mTORC1 downstream effectors and the overproduction of cyclin-dependent kinase inhibitors (CKIs) in diabetic glomeruli, along with a reduction in proteinuria and a decrease in the sizes of podocytes as well as glomeruli. In addition, knockdown of TCTP in db/db mice prevented the development of diabetic nephropathy, as indicated by the amelioration of proteinuria, mesangial expansion, podocytopenia and glomerulosclerosis. In accordance with the in vivo data, TCTP inhibition abrogated high glucose-induced hypertrophy in cultured podocytes, which was accompanied by the downregulation of mTORC1 effectors and CKIs. CONCLUSIONS/INTERPRETATION These findings suggest that TCTP might play an important role in the process of podocyte hypertrophy under diabetic conditions via the regulation of mTORC1 activity and the induction of cell-cycle arrest.
Collapse
Affiliation(s)
- D K Kim
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Abstract
Reactive oxygen species (ROS) are a group of molecules produced in the cell through metabolism of oxygen. Endogenous ROS such as hydrogen peroxide (H2O2) have long been recognised as destructive molecules. The well-established roles they have in the phagosome and genomic instability has led to the characterisation of these molecules as non-specific agents of destruction. Interestingly, there is a growing body of literature suggesting a less sinister role for this Jekyll and Hyde molecule. It is now evident that at lower physiological levels, H2O2 can act as a classical intracellular signalling molecule regulating kinase-driven pathways. The newly discovered biological functions attributed to ROS include proliferation, migration, anoikis, survival and autophagy. Furthermore, recent advances in detection and quantification of ROS-family members have revealed that the diverse functions of ROS can be determined by the subcellular source, location and duration of these molecules within the cell. In light of this confounding paradox, we will examine the factors and circumstances that determine whether H2O2 acts in a pro-survival or deleterious manner.
Collapse
|
47
|
Li S, Chen X, Ding Y, Liu X, Wang Y, He J. Expression of translationally controlled tumor protein (TCTP) in the uterus of mice of early pregnancy and its possible significance during embryo implantation. Hum Reprod 2011; 26:2972-80. [PMID: 21865235 DOI: 10.1093/humrep/der275] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND Translationally controlled tumor protein (TCTP) is a highly conserved, growth-related protein. Previous studies showed that TCTP is involved in many biological processes and it is essential for early embryo development and proliferation of embryonic stem cells. However, whether TCTP plays a role during embryo implantation remains unclear. This paper examines the expression and the role of TCTP in the uterus of mice during early pregnancy. METHODS The expression of TCTP in the uterus of mice during early pregnancy was examined by quantitative real-time PCR, immunohistochemistry and western blot. A functional study of TCTP in embryo implantation of mice was also performed by intrauterine injection with antisense oligodeoxynucleotides (A-ODNs) of TCTP on day 3 (D3) of pregnancy. RESULTS The TCTP mRNA levels were significantly upgraded from D3 to D5 of pregnancy and reached maximum levels on D5, then dramatically decreased on D6 and D7. The levels of the TCTP protein detected by western blot were consistent with those of the mRNA. Immunohistochemistry analysis showed that the TCTP protein was mainly located in the luminal and the glandular epithelium on D1 and D2 of pregnancy and reached maximum levels on D5 in the luminal and glandular epithelium and in the stromal cells. The levels of TCTP in the pseudo-pregnant uterus of mice were lower than those of pregnant mice on D4 and D6. Furthermore, inhibiting the TCTP expression by intrauterine injection with A-ODNs of TCTP on D3 of pregnancy significantly reduced the number of the implanted embryos compared with the control. CONCLUSIONS This study demonstrated that TCTP may play a significant role in embryo implantation in mice.
Collapse
Affiliation(s)
- Shuang Li
- Laboratory of Reproductive Biology, School of Public Health, Chongqing Medical University, Box 197, 1 Yixueyuan Road, Yuzhong District, 400016 Chongqing, PR China
| | | | | | | | | | | |
Collapse
|