1
|
Yoo W, Kim S, Noh K. SAMD13 serves as a useful prognostic biomarker for hepatocellular carcinoma. Eur J Med Res 2023; 28:514. [PMID: 37968735 PMCID: PMC10648382 DOI: 10.1186/s40001-023-01347-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 09/06/2023] [Indexed: 11/17/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is the most common form of liver cancer and the 5-year relative overall survival (OS) rate is less than 20%. Since there are no specific symptoms, most patients with HCC are diagnosed in an advanced stage with poor prognosis. Therefore, identifying novel prognostic biomarkers to improve the survival of patients with HCC is urgently needed. In the present study, we attempted to identify SAMD13 (Sterile Alpha Motif Domain-Containing Protein 13) as a novel biomarker associated with the prognosis of HCC using various bioinformatics tools. SAMD13 was found to be highly expressed pan-cancer; however, the SAMD13 expression was significantly correlated with the worst prognosis in HCC. Clinicopathological analysis revealed that SAMD13 upregulation was significantly associated with advanced HCC stage and high-grade tumor type. Simultaneously, high SAMD13 expression resulted in association with various immune markers in the immune cell subsets by TIMER databases and efficacy of immunotherapy. Methylation analysis showed SAMD13 was remarkably associated with prognosis. Furthermore, a six-hub gene signature associated with poor prognosis was correlated with the cell cycle, transcription, and epigenetic regulation and this analysis may support the connection between SAMD13 expression and drug-resistance. Our study illustrated the characteristics of SAMD13 role in patients with HCC using various bioinformatics tools and highlights its potential role as a therapeutic target and promising biomarker for prognosis in HCC.
Collapse
Affiliation(s)
- Wonbeak Yoo
- Personalized Genomic Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea
| | - Seokho Kim
- Department of Health Sciences, The Graduate School of Dong-A University, Busan, 49315, Republic of Korea.
- Department of Medicinal Biotechnology, College of Health Sciences, Dong-A University, 37, Nakdong-daero 550 beon-gil, Saha-gu, Busan, 49315, Republic of Korea.
| | - KyungHee Noh
- Bionanotechnology Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, 34141, Republic of Korea.
- Department of Nanobiotechnology, University of Science and Technology (UST), Daejeon, 34141, Republic of Korea.
| |
Collapse
|
2
|
Atay E, Bozkurt E, Ertekin A. Effect of tramadol hydrochloride on neural tube development in 48‐hr chick embryos: Argyrophilic nucleolar organizing region and genetic analysis study. Birth Defects Res 2022. [DOI: 10.1002/bdr2.2137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 11/15/2022] [Accepted: 11/21/2022] [Indexed: 12/23/2022]
Affiliation(s)
- Emre Atay
- Department of Anatomy, Faculty of Medicine Afyonkarahisar Health Sciences University Afyonkarahisar Turkey
| | - Erhan Bozkurt
- Department of Internal Medicine, Faculty of Medicine Afyonkarahisar Health Sciences University Afyonkarahisar Turkey
| | - Ayşe Ertekin
- Department of Emergency Medicine, Faculty of Medicine Afyonkarahisar Health Sciences University Afyonkarahisar Turkey
| |
Collapse
|
3
|
Jin F, Zhu Y, Liu M, Wang R, Cui Y, Wu Y, Liu G, Wang Y, Wang X, Ren Z. Babam2 negatively regulates osteoclastogenesis by interacting with Hey1 to inhibit Nfatc1 transcription. Int J Biol Sci 2022; 18:4482-4496. [PMID: 35864959 PMCID: PMC9295054 DOI: 10.7150/ijbs.72487] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 06/26/2022] [Indexed: 11/21/2022] Open
Abstract
Osteoclast-mediated excessive bone resorption was highly related to diverse bone diseases including osteoporosis. BRISC and BRCA1-A complex member 2 (Babam2) was an evolutionarily conserved protein that is highly expressed in bone tissues. However, whether Babam2 is involved in osteoclast formation is still unclear. In this study, we identify Babam2 as an essential negative regulator of osteoclast formation. We demonstrate that Babam2 knockdown significantly accelerated osteoclast formation and activity, while Babam2 overexpression blocked osteoclast formation and activity. Moreover, we demonstrate that the bone resorption activity was significantly downregulated in Babam2-transgenic mice as compared with wild-type littermates. Consistently, the bone mass of the Babam2-transgenic mice was increased. Furthermore, we found that Babam2-transgenic mice were protected from LPS-induced bone resorption activation and thus reduced the calvarial bone lesions. Mechanistically, we demonstrate that the inhibitory effects of Babam2 on osteoclast differentiation were dependent on Hey1. As silencing Hey1 largely diminished the effects of Babam2 on osteoclastogenesis. Finally, we show that Babam2 interacts with Hey1 to inhibit Nfatc1 transcription. In sum, our results suggested that Babam2 negatively regulates osteoclastogenesis and bone resorption by interacting with Hey1 to inhibit Nfatc1 transcription. Therefore, targeting Babam2 may be a novel therapeutic approach for osteoclast-related bone diseases.
Collapse
Affiliation(s)
- Fujun Jin
- Key Laboratory of Big Data-Based Precision Medicine, School of Engineering Medicine, Beihang University, Beijing 100191, China.,Guangzhou Jinan Biomedicine Research and Development Center, Institute of Biomedicine, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Yexuan Zhu
- Guangzhou Jinan Biomedicine Research and Development Center, Institute of Biomedicine, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Meijing Liu
- Key Laboratory of Big Data-Based Precision Medicine, School of Engineering Medicine, Beihang University, Beijing 100191, China
| | - Rongze Wang
- Guangzhou Jinan Biomedicine Research and Development Center, Institute of Biomedicine, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Yi Cui
- Key Laboratory of Big Data-Based Precision Medicine, School of Engineering Medicine, Beihang University, Beijing 100191, China
| | - Yanting Wu
- Guangzhou Jinan Biomedicine Research and Development Center, Institute of Biomedicine, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Gang Liu
- Department of Rehabilitation Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Yifei Wang
- Guangzhou Jinan Biomedicine Research and Development Center, Institute of Biomedicine, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Xiaogang Wang
- Key Laboratory of Big Data-Based Precision Medicine, School of Engineering Medicine, Beihang University, Beijing 100191, China
| | - Zhe Ren
- Guangzhou Jinan Biomedicine Research and Development Center, Institute of Biomedicine, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| |
Collapse
|
4
|
Yun M, Yingzi L, Jie G, Guanxin L, Zimei Z, Zhen C, Zhi L, Yingjie N, Lunquan S, Tao C, Yuezhen D, Chengzhi Z. PPDPF Promotes the Progression and acts as an Antiapoptotic Protein in Non-Small Cell Lung Cancer. Int J Biol Sci 2022; 18:214-228. [PMID: 34975328 PMCID: PMC8692159 DOI: 10.7150/ijbs.65654] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Accepted: 10/20/2021] [Indexed: 02/07/2023] Open
Abstract
Resistance to radiotherapy is frequently observed in the clinic and leads to poor prognosis of non-small cell lung cancer (NSCLC). How to overcome resistance to radiotherapy is a challenge in the treatment of NSCLC. In this study, PPDPF was found to be upregulated in NSCLC tissues and cell lines, and its expression negatively correlated with the overall survival of patients with NSCLC. PPDPF promoted the growth, colony formation and invasion of lung cancer cells. Moreover, knockout of PPDPF inhibited tumorigenesis in the KL (KrasG12D; LKB1f/f) mouse model of lung cancer. Additionally, overexpression of PPDPF led to radioresistance in lung cancer cells, and knockdown of PPDPF sensitized lung cancer cells to radiotherapy. Mechanistically, PPDPF interacted with BABAM2 (an antiapoptotic protein) and blocked its ubiquitination by MDM2, thus stabilizing BABAM2 and promoting the radioresistance of lung cancer cells. Our present study suggested PPDPF as a therapeutic target in NSCLC.
Collapse
Affiliation(s)
- Mu Yun
- Department of Oncology, Xiangya Cancer Center, Xiangya Hospital, Central South University, Changsha 410008, China.,Key Laboratory of Molecular Radiation Oncology Hunan Province, Changsha 410008, China
| | - Li Yingzi
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China
| | - Gao Jie
- Department of Oncology, Xiangya Cancer Center, Xiangya Hospital, Central South University, Changsha 410008, China.,Key Laboratory of Molecular Radiation Oncology Hunan Province, Changsha 410008, China.,Hunan International Science and Technology Collaboration Base of Precision Medicine for Cancer, Changsha 410008, China.,Center for Molecular Imaging of Central South University, Xiangya Hospital, Changsha 410008, China
| | - Liu Guanxin
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China
| | - Zeng Zimei
- Department of Oncology, Xiangya Cancer Center, Xiangya Hospital, Central South University, Changsha 410008, China.,Key Laboratory of Molecular Radiation Oncology Hunan Province, Changsha 410008, China.,Hunan International Science and Technology Collaboration Base of Precision Medicine for Cancer, Changsha 410008, China.,Center for Molecular Imaging of Central South University, Xiangya Hospital, Changsha 410008, China
| | - Cao Zhen
- Department of Oncology, Xiangya Cancer Center, Xiangya Hospital, Central South University, Changsha 410008, China.,Key Laboratory of Molecular Radiation Oncology Hunan Province, Changsha 410008, China.,Hunan International Science and Technology Collaboration Base of Precision Medicine for Cancer, Changsha 410008, China.,Center for Molecular Imaging of Central South University, Xiangya Hospital, Changsha 410008, China
| | - Li Zhi
- Department of Oncology, Xiangya Cancer Center, Xiangya Hospital, Central South University, Changsha 410008, China.,Key Laboratory of Molecular Radiation Oncology Hunan Province, Changsha 410008, China.,Hunan International Science and Technology Collaboration Base of Precision Medicine for Cancer, Changsha 410008, China.,Center for Molecular Imaging of Central South University, Xiangya Hospital, Changsha 410008, China
| | - Nie Yingjie
- NHC Key Laboratory of Pulmonary Immune-related Diseases, Guizhou Provincial People's Hospital; Guiyang 550000, China
| | - Sun Lunquan
- Department of Oncology, Xiangya Cancer Center, Xiangya Hospital, Central South University, Changsha 410008, China.,Key Laboratory of Molecular Radiation Oncology Hunan Province, Changsha 410008, China.,Hunan International Science and Technology Collaboration Base of Precision Medicine for Cancer, Changsha 410008, China.,Center for Molecular Imaging of Central South University, Xiangya Hospital, Changsha 410008, China
| | - Chen Tao
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China
| | - Deng Yuezhen
- Department of Oncology, Xiangya Cancer Center, Xiangya Hospital, Central South University, Changsha 410008, China.,Key Laboratory of Molecular Radiation Oncology Hunan Province, Changsha 410008, China.,Hunan International Science and Technology Collaboration Base of Precision Medicine for Cancer, Changsha 410008, China.,Center for Molecular Imaging of Central South University, Xiangya Hospital, Changsha 410008, China
| | - Zhou Chengzhi
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China
| |
Collapse
|
5
|
Yao W, Du X, Zhang J, Wang Y, Wang M, Pan Z, Li Q. SMAD4-induced knockdown of the antisense long noncoding RNA BRE-AS contributes to granulosa cell apoptosis. MOLECULAR THERAPY. NUCLEIC ACIDS 2021; 25:251-263. [PMID: 34458009 PMCID: PMC8368758 DOI: 10.1016/j.omtn.2021.05.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Accepted: 05/07/2021] [Indexed: 12/25/2022]
Abstract
Antisense long noncoding RNAs (AS-lncRNAs), a sub-class of lncRNAs, are transcribed in the opposite direction from their overlapping protein-coding genes and are implicated in various physiological and pathological processes. However, their role in female reproduction remains largely unknown. Here, we report that BRE-AS, an AS-lncRNA transcript from intron 10 of the protein-coding gene BRE, is involved in granulosa cell (GC) apoptosis. Based on our previous RNA sequencing data, we identified 28 AS-lncRNAs as important in the initiation of porcine follicular atresia, with BRE-AS showing the most significant upregulation in early atretic follicles. In this study, gain- and loss-of-function assays demonstrated that BRE-AS induces early apoptosis in GCs. Mechanistically, BRE-AS acts in cis to suppress the expression of BRE, an anti-apoptotic factor, via direct interaction with the pre-mRNA transcript of the latter, inducing increased GC apoptosis. Notably, we also found that BRE-AS was upregulated in SMAD4-silenced GCs. SMAD4 was identified as a transcriptional repressor of BRE-AS because it inhibits BRE-AS expression and BRE-AS-mediated GC apoptosis. In conclusion, we not only identified a novel AS-lncRNA related to the early apoptosis of GCs and initiation of follicular atresia but also described a novel regulatory pathway, SMAD4/BRE-AS/BRE, coordinating GC function and female fertility.
Collapse
Affiliation(s)
- Wang Yao
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Xing Du
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Jinbi Zhang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Yang Wang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Miaomiao Wang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Zengxiang Pan
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Qifa Li
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| |
Collapse
|
6
|
Chung CYT, Lo PHY, Lee KKH. Babam2 Regulates Cell Cycle Progression and Pluripotency in Mouse Embryonic Stem Cells as Revealed by Induced DNA Damage. Biomedicines 2020; 8:biomedicines8100397. [PMID: 33050379 PMCID: PMC7600899 DOI: 10.3390/biomedicines8100397] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 09/28/2020] [Accepted: 10/03/2020] [Indexed: 12/15/2022] Open
Abstract
BRISC and BRCA1-A complex member 2 (Babam2) plays an essential role in promoting cell cycle progression and preventing cellular senescence. Babam2-deficient fibroblasts show proliferation defect and premature senescence compared with their wild-type (WT) counterpart. Pluripotent mouse embryonic stem cells (mESCs) are known to have unlimited cell proliferation and self-renewal capability without entering cellular senescence. Therefore, studying the role of Babam2 in ESCs would enable us to understand the mechanism of Babam2 in cellular aging, cell cycle regulation, and pluripotency in ESCs. For this study, we generated Babam2 knockout (Babam2−/−) mESCs to investigate the function of Babam2 in mESCs. We demonstrated that the loss of Babam2 in mESCs leads to abnormal G1 phase retention in response to DNA damage induced by gamma irradiation or doxorubicin treatments. Key cell cycle regulators, CDC25A and CDK2, were found to be degraded in Babam2−/− mESCs following gamma irradiation. In addition, Babam2−/− mESCs expressed p53 strongly and significantly longer than in control mESCs, where p53 inhibited Nanog expression and G1/S cell cycle progression. The combined effects significantly reduced developmental pluripotency in Babam2−/− mESCs. In summary, Babam2 maintains cell cycle regulation and pluripotency in mESCs in response to induced DNA damage.
Collapse
Affiliation(s)
- Cheuk Yiu Tenny Chung
- MOE Key Laboratory for Regenerative Medicine, School of Biomedical Sciences, Chinese University of Hong Kong, Shatin, Hong Kong; (C.Y.T.C.); (P.H.Y.L.)
- Chinese University of Hong Kong-University of Southampton Joint Laboratory for Stem Cell and Regenerative Medicine, School of Biomedical Sciences, Chinese University of Hong Kong, Shatin, Hong Kong
| | - Paulisally Hau Yi Lo
- MOE Key Laboratory for Regenerative Medicine, School of Biomedical Sciences, Chinese University of Hong Kong, Shatin, Hong Kong; (C.Y.T.C.); (P.H.Y.L.)
- Chinese University of Hong Kong-University of Southampton Joint Laboratory for Stem Cell and Regenerative Medicine, School of Biomedical Sciences, Chinese University of Hong Kong, Shatin, Hong Kong
| | - Kenneth Ka Ho Lee
- MOE Key Laboratory for Regenerative Medicine, School of Biomedical Sciences, Chinese University of Hong Kong, Shatin, Hong Kong; (C.Y.T.C.); (P.H.Y.L.)
- Chinese University of Hong Kong-University of Southampton Joint Laboratory for Stem Cell and Regenerative Medicine, School of Biomedical Sciences, Chinese University of Hong Kong, Shatin, Hong Kong
- Correspondence:
| |
Collapse
|
7
|
Jin F, Zhu Y, Chen J, Wang R, Wang Y, Wu Y, Zhou P, Song X, Ren Z, Dong J. BRE Promotes Esophageal Squamous Cell Carcinoma Growth by Activating AKT Signaling. Front Oncol 2020; 10:1407. [PMID: 32850455 PMCID: PMC7431625 DOI: 10.3389/fonc.2020.01407] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Accepted: 07/03/2020] [Indexed: 01/09/2023] Open
Abstract
Brain and reproductive organ-expressed protein (BRE) is aberrantly expressed in multiple cancers; however, its expression pattern in human esophageal squamous cell carcinoma (ESCC) and its role in ESCC progression remain unclear. In this study, we aimed to investigate the expression pattern of BRE in human ESCC and its role in ESCC progression. BRE was overexpressed in ESCC tissues compared with that in the adjacent non-tumor tissues. Forced expression of BRE was sufficient to enhance ESCC cell growth by promoting cell cycle progression and anti-apoptosis. Silencing of BRE suppressed these malignant phenotypes of ESCC cells. Mechanistic evaluation revealed that BRE overexpression activated the phosphorylation of AKT, and inhibition of the AKT pathway by MK2206 decreased the BRE-induced cell growth and apoptotic resistance in ESCC cells, highlighting the critical role of AKT signaling in mediating the effects of BRE. Moreover, the effects of BRE on ESCC cell growth and AKT activation were verified in a xenograft model in vivo. The present results show that BRE is overexpressed in ESCC tissues and contributes to the growth of ESCC cells by activating AKT signaling both in vitro and in vivo and provide insight into the role of BRE in AKT signaling and ESCC pathogenesis.
Collapse
Affiliation(s)
- Fujun Jin
- Guangzhou Jinan Biomedicine Research and Development Center, College of Life Science and Technology, Institute of Biomedicine, Jinan University, Guangzhou, China.,Integrated Chinese and Western Medicine Postdoctoral Research Station, Jinan University, Guangzhou, China
| | - Yexuan Zhu
- Guangzhou Jinan Biomedicine Research and Development Center, College of Life Science and Technology, Institute of Biomedicine, Jinan University, Guangzhou, China
| | - Jingyi Chen
- Guangzhou Jinan Biomedicine Research and Development Center, College of Life Science and Technology, Institute of Biomedicine, Jinan University, Guangzhou, China
| | - Rongze Wang
- Guangzhou Jinan Biomedicine Research and Development Center, College of Life Science and Technology, Institute of Biomedicine, Jinan University, Guangzhou, China
| | - Yiliang Wang
- Guangzhou Jinan Biomedicine Research and Development Center, College of Life Science and Technology, Institute of Biomedicine, Jinan University, Guangzhou, China
| | - Yanting Wu
- Guangzhou Jinan Biomedicine Research and Development Center, College of Life Science and Technology, Institute of Biomedicine, Jinan University, Guangzhou, China
| | - Pengjun Zhou
- Guangzhou Jinan Biomedicine Research and Development Center, College of Life Science and Technology, Institute of Biomedicine, Jinan University, Guangzhou, China
| | - Xiaowei Song
- Guangzhou Jinan Biomedicine Research and Development Center, College of Life Science and Technology, Institute of Biomedicine, Jinan University, Guangzhou, China
| | - Zhe Ren
- Guangzhou Jinan Biomedicine Research and Development Center, College of Life Science and Technology, Institute of Biomedicine, Jinan University, Guangzhou, China
| | - Jun Dong
- Integrated Chinese and Western Medicine Postdoctoral Research Station, Jinan University, Guangzhou, China.,Department of Pathophysiology, School of Medicine, GHM Institute of CNS Regeneration, Jinan University, Guangzhou, China
| |
Collapse
|
8
|
Pun CCM, Lee KKH, Chui YL. C-terminal BRE inhibits cellular proliferation and increases sensitivity to chemotherapeutic drugs of MLL-AF9 acute myeloid leukemia cells. Leuk Lymphoma 2019; 60:3011-3019. [PMID: 31111759 DOI: 10.1080/10428194.2019.1616184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
BRE (Brain and Reproductive Organ-Expressed) is an anti-apoptotic protein and a core component of DNA-repair BRCA1-A complex. Microarray-detected high BRE gene expression has been found to be associated with better patient survival in AML (acute myeloid leukemia) with MLL-AF9 translocation, and radiotherapy-treated non-familial breast cancer. A recent finding suggests that the high BRE gene expression in MLL-AF9 AML could be attributed to the additional expression of a transcript variant encoding a novel C-terminal BRE isoform. Using THP-1 as the MLL-AF9 AML cell model, we found that ectopic expression of the C-terminal BRE, which could not form an intact BRCA1-A complex, indeed increased cellular sensitivity to chemotherapeutic drugs and inhibited cell proliferation, while the complete opposite was achieved by the ectopic expression of full-length BRE. Our findings suggest that the C-terminal BRE-encoding transcript could be responsible for better patient survival and may have therapeutic potential for cancer.
Collapse
Affiliation(s)
| | - Kenneth Ka-Ho Lee
- School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, China
| | - Yiu-Loon Chui
- Department of Chemical Pathology, The Chinese University of Hong Kong, Shatin, China
| |
Collapse
|
9
|
Lohitesh K, Chowdhury R, Mukherjee S. Resistance a major hindrance to chemotherapy in hepatocellular carcinoma: an insight. Cancer Cell Int 2018; 18:44. [PMID: 29568237 PMCID: PMC5859782 DOI: 10.1186/s12935-018-0538-7] [Citation(s) in RCA: 179] [Impact Index Per Article: 29.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Accepted: 03/12/2018] [Indexed: 12/18/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the leading causes of cancer mortality, accounting for almost 90% of total liver cancer burden. Surgical resection followed by adjuvant and systemic chemotherapy are the most meticulously followed treatment procedures but the complex etiology and high metastatic potential of the disease renders surgical treatment futile in majority of the cases. Another hindrance to the scenario is the acquired resistance to drugs resulting in relapse of the disease. Hence, to provide insights into development of novel therapeutic targets and diagnostic biomarkers, this review focuses on the various molecular mechanisms underlying chemoresistance in HCC. We have provided a comprehensive summary of the various strategies adopted by HCC cells, extending from apoptosis evasion, autophagy activation, drug expulsion to epigenetic transformation as modes of therapy resistance. The role of stem cells in imparting chemoresistance is also discussed. Furthermore, the review also focuses on how this knowledge might be exploited for the development of an effective, prospective therapy against HCC.
Collapse
Affiliation(s)
- K Lohitesh
- Department of Biological-Sciences, Birla Institute of Technology and Sciences (BITS), Campus, VidyaVihar, Pilani, Rajasthan 333031 India
| | - Rajdeep Chowdhury
- Department of Biological-Sciences, Birla Institute of Technology and Sciences (BITS), Campus, VidyaVihar, Pilani, Rajasthan 333031 India
| | - Sudeshna Mukherjee
- Department of Biological-Sciences, Birla Institute of Technology and Sciences (BITS), Campus, VidyaVihar, Pilani, Rajasthan 333031 India
| |
Collapse
|
10
|
BRE/BRCC45 regulates CDC25A stability by recruiting USP7 in response to DNA damage. Nat Commun 2018; 9:537. [PMID: 29416040 PMCID: PMC5803202 DOI: 10.1038/s41467-018-03020-6] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Accepted: 01/12/2018] [Indexed: 01/07/2023] Open
Abstract
BRCA2 is essential for maintaining genomic integrity. BRCA2-deficient primary cells are either not viable or exhibit severe proliferation defects. Yet, BRCA2 deficiency contributes to tumorigenesis. It is believed that mutations in genes such as TRP53 allow BRCA2 heterozygous cells to overcome growth arrest when they undergo loss of heterozygosity. Here, we report the use of an insertional mutagenesis screen to identify a role for BRE (Brain and Reproductive organ Expressed, also known as BRCC45), known to be a part of the BRCA1-DNA damage sensing complex, in the survival of BRCA2-deficient mouse ES cells. Cell viability by BRE overexpression is mediated by deregulation of CDC25A phosphatase, a key cell cycle regulator and an oncogene. We show that BRE facilitates deubiquitylation of CDC25A by recruiting ubiquitin-specific-processing protease 7 (USP7) in the presence of DNA damage. Additionally, we uncovered the role of CDC25A in BRCA-mediated tumorigenesis, which can have implications in cancer treatment. Loss of BRCA2 leads to cancer formation. Here, the authors use an insertional mutagenesis approach and identify a multiprotein complex consisting of BRE, USP7 and CDC25A that can support the survival of BRCA2-deficient cells.
Collapse
|
11
|
Marneth AE, Prange KHM, Al Hinai ASA, Bergevoet SM, Tesi N, Janssen-Megens EM, Kim B, Sharifi N, Yaspo ML, Kuster J, Sanders MA, Stoetman ECG, Knijnenburg J, Arentsen-Peters TCJM, Zwaan CM, Stunnenberg HG, van den Heuvel-Eibrink MM, Haferlach T, Fornerod M, Jansen JH, Valk PJM, van der Reijden BA, Martens JHA. C-terminal BRE overexpression in 11q23-rearranged and t(8;16) acute myeloid leukemia is caused by intragenic transcription initiation. Leukemia 2017; 32:828-836. [PMID: 28871137 DOI: 10.1038/leu.2017.280] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Revised: 07/16/2017] [Accepted: 08/10/2017] [Indexed: 01/05/2023]
Abstract
Overexpression of the BRE (brain and reproductive organ-expressed) gene defines a distinct pediatric and adult acute myeloid leukemia (AML) subgroup. Here we identify a promoter enriched for active chromatin marks in BRE intron 4 causing strong biallelic expression of a previously unknown C-terminal BRE transcript. This transcript starts with BRE intron 4 sequences spliced to exon 5 and downstream sequences, and if translated might code for an N terminally truncated BRE protein. Remarkably, the new BRE transcript was highly expressed in over 50% of 11q23/KMT2A (lysine methyl transferase 2A)-rearranged and t(8;16)/KAT6A-CREBBP cases, while it was virtually absent from other AML subsets and normal tissues. In gene reporter assays, the leukemia-specific fusion protein KMT2A-MLLT3 transactivated the intragenic BRE promoter. Further epigenome analyses revealed 97 additional intragenic promoter marks frequently bound by KMT2A in AML with C-terminal BRE expression. The corresponding genes may be part of a context-dependent KMT2A-MLLT3-driven oncogenic program, because they were higher expressed in this AML subtype compared with other groups. C-terminal BRE might be an important contributor to this program because in a case with relapsed AML, we observed an ins(11;2) fusing CHORDC1 to BRE at the region where intragenic transcription starts in KMT2A-rearranged and KAT6A-CREBBP AML.
Collapse
Affiliation(s)
- A E Marneth
- Department of Laboratory Medicine, Laboratory of Hematology, Radboud Institute for Molecular Life Sciences (RIMLS), Radboud University Medical Center, Nijmegen, The Netherlands
| | - K H M Prange
- Department of Molecular Biology, Faculty of Science, RIMLS, Radboud University, Nijmegen, The Netherlands
| | - A S A Al Hinai
- Department of Hematology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - S M Bergevoet
- Department of Laboratory Medicine, Laboratory of Hematology, Radboud Institute for Molecular Life Sciences (RIMLS), Radboud University Medical Center, Nijmegen, The Netherlands
| | - N Tesi
- Department of Molecular Biology, Faculty of Science, RIMLS, Radboud University, Nijmegen, The Netherlands
| | - E M Janssen-Megens
- Department of Molecular Biology, Faculty of Science, RIMLS, Radboud University, Nijmegen, The Netherlands
| | - B Kim
- Department of Molecular Biology, Faculty of Science, RIMLS, Radboud University, Nijmegen, The Netherlands
| | - N Sharifi
- Department of Molecular Biology, Faculty of Science, RIMLS, Radboud University, Nijmegen, The Netherlands
| | - M L Yaspo
- Max Planck Institute for Molecular Genetics, Berlin, Germany
| | - J Kuster
- Department of Molecular Biology, Faculty of Science, RIMLS, Radboud University, Nijmegen, The Netherlands
| | - M A Sanders
- Department of Hematology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - E C G Stoetman
- Department of Hematology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - J Knijnenburg
- Department of Clinical Genetics, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - T C J M Arentsen-Peters
- Pediatric Oncology/Hematology, Erasmus University Medical Center-Sophia Children's Hospital, Rotterdam, The Netherlands
| | - C M Zwaan
- Pediatric Oncology/Hematology, Erasmus University Medical Center-Sophia Children's Hospital, Rotterdam, The Netherlands
| | - H G Stunnenberg
- Department of Molecular Biology, Faculty of Science, RIMLS, Radboud University, Nijmegen, The Netherlands
| | - M M van den Heuvel-Eibrink
- Pediatric Oncology/Hematology, Erasmus University Medical Center-Sophia Children's Hospital, Rotterdam, The Netherlands.,Princess Maxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - T Haferlach
- MLL Munich Leukemia Laboratory, Munich, Germany
| | - M Fornerod
- Pediatric Oncology/Hematology, Erasmus University Medical Center-Sophia Children's Hospital, Rotterdam, The Netherlands
| | - J H Jansen
- Department of Laboratory Medicine, Laboratory of Hematology, Radboud Institute for Molecular Life Sciences (RIMLS), Radboud University Medical Center, Nijmegen, The Netherlands
| | - P J M Valk
- Department of Hematology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - B A van der Reijden
- Department of Laboratory Medicine, Laboratory of Hematology, Radboud Institute for Molecular Life Sciences (RIMLS), Radboud University Medical Center, Nijmegen, The Netherlands
| | - J H A Martens
- Department of Molecular Biology, Faculty of Science, RIMLS, Radboud University, Nijmegen, The Netherlands
| |
Collapse
|
12
|
Jin F, Wang Y, Wang X, Wu Y, Wang X, Liu Q, Zhu Y, Liu E, Fan J, Wang Y. Bre Enhances Osteoblastic Differentiation by Promoting the Mdm2-Mediated Degradation of p53. Stem Cells 2017; 35:1760-1772. [PMID: 28436570 DOI: 10.1002/stem.2620] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Revised: 03/12/2017] [Accepted: 03/21/2017] [Indexed: 01/04/2023]
Abstract
Bre is a conserved cellular protein expressed in various tissues. Its major function includes DNA damage repair and anti-apoptosis. Recent studies indicate that Bre is potentially involved in stem cell differentiation although pathophysiological significance along with the molecular mechanisms is still unclear. Here, we report that Bre protein was substantially expressed in the bone tissue and its expression was highly upregulated during the osteogenic differentiation. To test a hypothesis that Bre plays functional roles in the process of osteogenic differentiation, we examined the expression of Bre in an osteoporosis mouse model. Compared with the normal bone tissue, Bre expression in osteoporotic bone was also significantly reduced. Moreover, knockdown of Bre in the mouse bone marrow mesenchymal cells significantly reduced the expression of osteogenic marker genes, the alkaline phosphatase activity, and the mineralization capacity, while overexpression of Bre greatly promoted the osteogenesis both in vitro and in vivo. Interestingly, we founded that knockdown of Bre led to activation of the p53 signaling pathways exhibited by increased p53, p21, and Mdm2. However, when we inhibited the p53 by siRNA silencing or pifithrin-α, the impaired osteogenesis caused by Bre knockdown was greatly restored. Finally, we found that Bre promoted the Mdm2-mediated p53 ubiquitination and degradation by physically interacting with p53. Taken together, our results revealed a novel function of Bre in osteoblast differentiation through modulating the stability of p53. Stem Cells 2017;35:1760-1772.
Collapse
Affiliation(s)
- Fujun Jin
- Guangzhou Jinan Biomedicine Research and Development Center, Institute of Biomedicine, College of Life Science and Technology, Jinan University, Guangzhou, People's Republic of China
| | - Yiliang Wang
- Guangzhou Jinan Biomedicine Research and Development Center, Institute of Biomedicine, College of Life Science and Technology, Jinan University, Guangzhou, People's Republic of China
| | - Xiaojing Wang
- Research Institute of Atherosclerotic Disease, Laboratory Animal Center, School of Medicine, Xi'an Jiaotong University, Xi'an, People's Republic of China
| | - Yanting Wu
- Guangzhou Jinan Biomedicine Research and Development Center, Institute of Biomedicine, College of Life Science and Technology, Jinan University, Guangzhou, People's Republic of China
| | - Xiaoyan Wang
- Guangzhou Jinan Biomedicine Research and Development Center, Institute of Biomedicine, College of Life Science and Technology, Jinan University, Guangzhou, People's Republic of China
| | - Qiuying Liu
- Guangzhou Jinan Biomedicine Research and Development Center, Institute of Biomedicine, College of Life Science and Technology, Jinan University, Guangzhou, People's Republic of China
| | - Yexuan Zhu
- Guangzhou Jinan Biomedicine Research and Development Center, Institute of Biomedicine, College of Life Science and Technology, Jinan University, Guangzhou, People's Republic of China
| | - Enqi Liu
- Research Institute of Atherosclerotic Disease, Laboratory Animal Center, School of Medicine, Xi'an Jiaotong University, Xi'an, People's Republic of China
| | - Jianglin Fan
- Department of Molecular Pathology, Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi, Yamanashi, Japan
| | - Yifei Wang
- Guangzhou Jinan Biomedicine Research and Development Center, Institute of Biomedicine, College of Life Science and Technology, Jinan University, Guangzhou, People's Republic of China
| |
Collapse
|
13
|
Macrolide antibiotics differentially influence human HepG2 cytotoxicity and modulate intrinsic/extrinsic apoptotic pathways in rat hepatocellular carcinoma model. Naunyn Schmiedebergs Arch Pharmacol 2017; 390:379-395. [DOI: 10.1007/s00210-016-1337-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Accepted: 12/29/2016] [Indexed: 01/20/2023]
|
14
|
Xu LM, Chen L, Li F, Zhang R, Li ZY, Chen FF, Jiang XD. Over-expression of the long non-coding RNA HOTTIP inhibits glioma cell growth by BRE. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2016; 35:162. [PMID: 27733185 PMCID: PMC5062847 DOI: 10.1186/s13046-016-0431-y] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Accepted: 09/20/2016] [Indexed: 12/14/2022]
Abstract
Background Gliomas are the most common type of primary brain tumour in the central nervous system of adults. The long non-coding RNA (lncRNA) HOXA transcript at the distal tip (HOTTIP) is transcribed from the 5′ tip of the HOXA locus. HOTTIP has recently been shown to be dysregulated and play an important role in the progression of several cancers. However, little is known about whether and how HOTTIP regulates glioma development. Methods In this study, we assayed the expression of HOTTIP in glioma tissue samples and glioma cell lines using real-time polymerase chain reaction and defined the biological functions of HOTTIP using the CCK-8 assay, flow cytometry, terminal deoxynucleotidyl transferase dUTP nick end labelling (TUNEL assay) and tumour formation assay in a nude mouse model. Finally, we discovered the underlying mechanism using the Apoptosis PCR 384HT Array, Western blot, RNA immunoprecipitation (RIP) and luciferase reporter assay. Results HOTTIP was aberrantly down-regulated in glioma tissues and glioma cell lines (U87-MG, U118-MG, U251 and A172), and over-expression of HOTTIP inhibited the growth of glioma cell lines in vitro and in vivo. Furthermore, HOTTIP could directly bind to the brain and reproductive expression (BRE) gene and down-regulate BRE gene expression. In addition, we further verified that over-expression of the BRE gene promoted the growth of glioma cell lines in vitro. Finally, over-expression of HOTTIP significantly suppressed the expression of the cyclin A and CDK2 proteins and increased the expression of the P53 protein. However, we found that the over-expression of BRE significantly increased the expression of the cyclin A and CDK2 proteins and suppressed the expression of the P53 protein. Taken together, these findings suggested that high levels of HOTTIP reduced glioma cell growth. Additionally, the mechanism of HOTTIP-mediated reduction of glioma cell growth may involve the suppression of cyclin A and CDK2 protein expression, which increases P53 protein expression via the down-regulation of BRE. Conclusions Our studies demonstrated that over-expression of HOTTIP promotes cell apoptosis and inhibits cell growth in U118-MG and U87-MG human glioma cell lines by down-regulating BRE expression to regulate the expression of P53, CDK2 and Cyclin A proteins. The data described in this study indicate that HOTTIP is an interesting candidate for further functional studies in glioma and demonstrate the potential application of HOTTIP in glioma therapy. Electronic supplementary material The online version of this article (doi:10.1186/s13046-016-0431-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Li-Min Xu
- The National Key Clinic Specialty, The Neurosurgery Institute of Guangdong Province, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, China
| | - Lei Chen
- Department of Neurosurgery, Shenzhen Second People' s Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, 518029, China
| | - Feng Li
- The National Key Clinic Specialty, The Neurosurgery Institute of Guangdong Province, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, China
| | - Run Zhang
- The National Key Clinic Specialty, The Neurosurgery Institute of Guangdong Province, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, China
| | - Zong-Yang Li
- Department of Neurosurgery, Shenzhen Second People' s Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, 518029, China
| | - Fan-Fan Chen
- Department of Neurosurgery, Guangzhou First People's Hospital, Guangzhou, 510180, China
| | - Xiao-Dan Jiang
- The National Key Clinic Specialty, The Neurosurgery Institute of Guangdong Province, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, China.
| |
Collapse
|
15
|
Shi W, Tang MK, Yao Y, Tang C, Chui YL, Lee KKH. BRE plays an essential role in preventing replicative and DNA damage-induced premature senescence. Sci Rep 2016; 6:23506. [PMID: 27001068 PMCID: PMC4802329 DOI: 10.1038/srep23506] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2015] [Accepted: 03/08/2016] [Indexed: 11/20/2022] Open
Abstract
The BRE gene, alias BRCC45, produces a 44 kDa protein that is normally distributed in both cytoplasm and nucleus. In this study, we used adult fibroblasts isolated from wild-type (WT) and BRE knockout (BRE−/−) mice to investigate the functional role of BRE in DNA repair and cellular senescence. We compared WT with BRE−/− fibroblasts at different cell passages and observed that the mutant fibroblasts entered replicative senescence earlier than the WT fibroblasts. With the use of gamma irradiation to induce DNA damage in fibroblasts, the percentage of SA-β-Gal+ cells was significantly higher in BRE−/− fibroblasts compared with WT cells, suggesting that BRE is also associated with DNA damage-induced premature senescence. We also demonstrated that the gamma irradiation induced γ-H2AX foci, a DNA damage marker, persisted significantly longer in BRE−/− fibroblasts than in WT fibroblasts, confirming that the DNA repair process is impaired in the absence of BRE. In addition, the BRCA1-A complex recruitment and homologous recombination (HR)-dependent DNA repair process upon DNA damage were impaired in BRE−/− fibroblasts. Taken together, our results demonstrate a role for BRE in both replicative senescence and DNA damage-induced premature senescence. This can be attributed to BRE being required for BRCA1-A complex-driven HR DNA repair.
Collapse
Affiliation(s)
- Wenting Shi
- Stem Cell and Regeneration Thematic Research Programme, School of Biomedical Sciences, Chinese University of Hong Kong, Hong Kong, People's Republic of China
| | - Mei Kuen Tang
- Stem Cell and Regeneration Thematic Research Programme, School of Biomedical Sciences, Chinese University of Hong Kong, Hong Kong, People's Republic of China
| | - Yao Yao
- Stem Cell and Regeneration Thematic Research Programme, School of Biomedical Sciences, Chinese University of Hong Kong, Hong Kong, People's Republic of China
| | - Chengcheng Tang
- Stem Cell and Regeneration Thematic Research Programme, School of Biomedical Sciences, Chinese University of Hong Kong, Hong Kong, People's Republic of China
| | - Yiu Loon Chui
- Department of Chemical Pathology, Chinese University of Hong Kong, Hong Kong, People's Republic of China
| | - Kenneth Ka Ho Lee
- Stem Cell and Regeneration Thematic Research Programme, School of Biomedical Sciences, Chinese University of Hong Kong, Hong Kong, People's Republic of China
| |
Collapse
|
16
|
Xiao L, Lee KKH. BRE facilitates skeletal muscle regeneration by promoting satellite cell motility and differentiation. Biol Open 2016; 5:100-11. [PMID: 26740569 PMCID: PMC4823978 DOI: 10.1242/bio.012450] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The function of the Bre gene in satellite cells was investigated during skeletal muscle regeneration. The tibialis anterior leg muscle was experimentally injured in Bre knockout mutant (BRE-KO) mice. It was established that the accompanying muscle regeneration was impaired as compared with their normal wild-type counterparts (BRE-WT). There were significantly fewer pax7+ satellite cells and smaller newly formed myofibers present in the injury sites of BRE-KO mice. Bre was required for satellite cell fusion and myofiber formation. The cell fusion index and average length of newly-formed BRE-KO myofibers were found to be significantly reduced as compared with BRE-WT myofibers. It is well established that satellite cells are highly invasive which confers on them the homing ability to reach the muscle injury sites. Hence, we tracked the migratory behavior of these cells using time-lapse microscopy. Image analysis revealed no difference in directionality of movement between BRE-KO and BRE-WT satellite cells but there was a significant decrease in the velocity of BRE-KO cell movement. Moreover, chemotactic migration assays indicated that BRE-KO satellite cells were significantly less responsive to chemoattractant SDF-1α than BRE-WT satellite cells. We also established that BRE normally protects CXCR4 from SDF-1α-induced degradation. In sum, BRE facilitates skeletal muscle regeneration by enhancing satellite cell motility, homing and fusion. Summary: BRE facilitates skeletal muscle regeneration by promoting satellite cell motility and differentiation, probably by protecting CXCR4 from degradation.
Collapse
Affiliation(s)
- Lihai Xiao
- Stem Cell and Regeneration Thematic Research Programme, School of Biomedical Sciences, Chinese University of Hong Kong, Hong Kong
| | - Kenneth Ka Ho Lee
- Stem Cell and Regeneration Thematic Research Programme, School of Biomedical Sciences, Chinese University of Hong Kong, Hong Kong
| |
Collapse
|
17
|
Abstract
Hepatocellular carcinoma (HCC) is a major health problem. In human hepatocarcinogenesis, the balance between cell death and proliferation is deregulated, tipping the scales for a situation where antiapoptotic signals are overpowering the death-triggering stimuli. HCC cells harbor a wide variety of mutations that alter the regulation of apoptosis and hence the response to chemotherapeutical drugs, making them resistant to the proapoptotic signals. Considering all these modifications found in HCC cells, therapeutic approaches need to be carefully studied in order to specifically target the antiapoptotic signals. This review deals with the recent relevant contributions reporting molecular alterations for HCC that lead to a deregulation of apoptosis, as well as the challenge of death-inducing chemotherapeutics in current HCC treatment.
Collapse
Affiliation(s)
- Joaquim Moreno-Càceres
- Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain.,Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain
| | - Isabel Fabregat
- Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain.,Department of Physiological Sciences II, University of Barcelona, Spain.,Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain.,Department of Physiological Sciences II, University of Barcelona, Spain
| |
Collapse
|
18
|
Li Y, Qi K, Zu L, Wang M, Wang Y, Zhou Q. Anti-apoptotic brain and reproductive organ-expressed proteins enhance cisplatin resistance in lung cancer cells via the protein kinase B signaling pathway. Thorac Cancer 2015; 7:190-8. [PMID: 27042221 PMCID: PMC4773300 DOI: 10.1111/1759-7714.12313] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2015] [Accepted: 08/12/2015] [Indexed: 01/24/2023] Open
Abstract
Background Cisplatin‐based chemotherapy is the standard first‐line treatment for non‐small‐cell lung cancers (NSCLCs); however, the long‐term therapeutic effect is reduced by chemoresistance. Brain and reproductive organ‐expressed (BRE) proteins are overexpressed in several cancers and have an anti‐apoptotic function. However, their biological role in the development of the chemoresistant phenotype of human NSCLC remains unknown. We investigate the differential expression of the BRE gene in human lung adenocarcinoma cell lines A549 and the cisplatin‐resistant variant A549/cisplatin (DDP), and the mechanisms of cisplatin‐resistance induced by the BRE gene. Methods Cell counting kit‐8 assay was employed to determine the sensitivity of A549 and A549/DDP cell lines to cisplatin. BRE expression was measured using quantitative real time‐polymerase chain reaction and western blot analysis. The apoptosis rate of lung adenocarcinoma cells was determined by flow cytometry. Results BRE expression in A549 cells, derived from human lung cells, was markedly decreased compared with parental cisplatin‐resistant A549/DDP cells at messenger ribonucleic acid and protein levels. BRE overexpression in A549 significantly decreased sensitivity to DDP by inhibiting cell apoptosis. Conversely, BRE knockdown in A549/DDP cells increased their chemosensitivity. Importantly, we demonstrate that BRE overexpression induces the expression of phosphoprotein kinase B (p‐Akt) in lung cancer cells, while BRE silencing inhibits p‐Akt expression. Furthermore, downregulation of p‐Akt by LY294002 reversed the DDP resistance induced by BRE by increasing apoptosis. BRE enhances the DDP resistance of lung cancer cells through the Akt signaling pathway. Conclusion Our findings provide new insight into the mechanism of DDP resistance in NSCLC cells and suggest BRE as an attractive new target for NSCLC treatment.
Collapse
Affiliation(s)
- Yang Li
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Tianjin Lung Cancer Institute Tianjin Medical University General Hospital Tianjin China
| | - Kang Qi
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Tianjin Lung Cancer Institute Tianjin Medical University General Hospital Tianjin China
| | - Lingling Zu
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Tianjin Lung Cancer Institute Tianjin Medical University General Hospital Tianjin China
| | - Min Wang
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Tianjin Lung Cancer Institute Tianjin Medical University General Hospital Tianjin China
| | - Yuli Wang
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Tianjin Lung Cancer Institute Tianjin Medical University General Hospital Tianjin China
| | - Qinghua Zhou
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Tianjin Lung Cancer Institute Tianjin Medical University General Hospital Tianjin China
| |
Collapse
|
19
|
Oliveira RJ, Navarro SD, de Lima DP, Meza A, Pesarini JR, da Silva Gomes R, Karaziack CB, de Oliveira Mauro M, Cunha-Laura AL, Monreal ACD, Romão W, Júnior VL, Beatriz A. A novel cytosporone 3-Heptyl-4,6-dihydroxy-3H-isobenzofuran-1-one: synthesis; toxicological, apoptotic and immunomodulatory properties; and potentiation of mutagenic damage. BMC Cancer 2015; 15:561. [PMID: 26228542 PMCID: PMC4520062 DOI: 10.1186/s12885-015-1532-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2014] [Accepted: 07/02/2015] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND A large number of studies are attempting to identify alternative products from natural sources or synthesized compounds that effectively interact with cancer cells without causing adverse effects on healthy cells. Resorcinolic lipids are a class of bioactive compounds that possess anticancer activity and are able to interact with the lipid bilayer. Therefore, the objective of this study was to synthesize a novel resorcinolic lipid and test its biological proprieties. METHODS We aimed to synthesize a novel resorcinolic lipid belonging to the class of cytosporones, AMS049 (3-Heptyl-4,6-dihydroxy-3H-isobenzofuran-1-one) and to evaluate the toxicity of two concentrations of this lipid (7.5 and 10 mg/kg) by determining its genotoxic, mutagenic, immunomodulatory, and apoptotic effects, as well as any biochemical and histopathological alterations in mice treated with cyclophosphamide. The results were analyzed by ANOVA followed by the Tukey test A . level of significance of p < 0.05 was adopted. RESULTS The new cytosporone AMS049 was synthesized in only three steps and in satisfactory yields. The results indicate that the compound is neither genotoxic nor mutagenic and does not alter biochemical parameters. The histological alterations observed in the liver and kidneys did not compromise the function of these organs. Histology of the spleen suggested immunomodulation, although no changes were observed in splenic phagocytosis or differential blood cell count. The results also show that AMS049 potentiates the mutagenic effect of the chemotherapy drug cyclophosphamide and that the combination induces apoptosis. CONCLUSION These facts indicate a potential therapeutic application of this novel cytosporone as an important chemotherapeutic adjuvant.
Collapse
Affiliation(s)
- Rodrigo Juliano Oliveira
- Centro de Estudos em Células Tronco, Terapia Celular e Genética Toxicológica - CeTroGen, Hospital Universitário "Maria Aparecida Pedrossian" - HUMAP, Empresa Brasileira de Serviços Hospitalares - EBSERH, Campo Grande, MS, Brazil. .,Programa de Mestrado em Farmácia, Centro de Ciências Biológicas e da Saúde - CCBS, Universidade Federal de Mato Grosso do Sul - UFMS, Campo Grande, MS, Brazil. .,Programa de Pós-graduação em Saúde e Desenvolvimento na Região Centro-Oeste, Faculdade de Medicina "Dr. Hélio Mandetta" - FAMED, Universidade Federal de Mato Grosso do Sul - UFMS, Campo Grande, MS, Brazil.
| | - Stephanie Dynczuki Navarro
- Centro de Estudos em Células Tronco, Terapia Celular e Genética Toxicológica - CeTroGen, Hospital Universitário "Maria Aparecida Pedrossian" - HUMAP, Empresa Brasileira de Serviços Hospitalares - EBSERH, Campo Grande, MS, Brazil. .,Programa de Mestrado em Farmácia, Centro de Ciências Biológicas e da Saúde - CCBS, Universidade Federal de Mato Grosso do Sul - UFMS, Campo Grande, MS, Brazil.
| | - Dênis Pires de Lima
- Programa de Mestrado em Farmácia, Centro de Ciências Biológicas e da Saúde - CCBS, Universidade Federal de Mato Grosso do Sul - UFMS, Campo Grande, MS, Brazil. .,Programa de Pós-graduação em Química, Instituto de Química, Universidade Federal de Mato Grosso do Sul - UFMS, Campo Grande, MS, Brazil.
| | - Alisson Meza
- Programa de Pós-graduação em Química, Instituto de Química, Universidade Federal de Mato Grosso do Sul - UFMS, Campo Grande, MS, Brazil.
| | - João Renato Pesarini
- Centro de Estudos em Células Tronco, Terapia Celular e Genética Toxicológica - CeTroGen, Hospital Universitário "Maria Aparecida Pedrossian" - HUMAP, Empresa Brasileira de Serviços Hospitalares - EBSERH, Campo Grande, MS, Brazil. .,Programa de Pós-graduação em Saúde e Desenvolvimento na Região Centro-Oeste, Faculdade de Medicina "Dr. Hélio Mandetta" - FAMED, Universidade Federal de Mato Grosso do Sul - UFMS, Campo Grande, MS, Brazil.
| | - Roberto da Silva Gomes
- Programa de Pós-graduação em Química, Instituto de Química, Universidade Federal de Mato Grosso do Sul - UFMS, Campo Grande, MS, Brazil. .,Faculdade de Ciências Exatas e Tecnologia - FACET, Universidade Federal da Grande Dourados - UFGD, Dourados, MS, Brazil.
| | - Caroline Bilhar Karaziack
- Programa de Pós-graduação em Química, Instituto de Química, Universidade Federal de Mato Grosso do Sul - UFMS, Campo Grande, MS, Brazil.
| | - Mariana de Oliveira Mauro
- Centro de Estudos em Células Tronco, Terapia Celular e Genética Toxicológica - CeTroGen, Hospital Universitário "Maria Aparecida Pedrossian" - HUMAP, Empresa Brasileira de Serviços Hospitalares - EBSERH, Campo Grande, MS, Brazil. .,Programa de Doutorado em Biotecnologia e Biodiversidade - Rede Pró Centro-Oeste, Universidade Federal de Mato Grosso do Sul - UFMS, Campo Grande, MS, Brazil.
| | - Andréa Luiza Cunha-Laura
- Centro de Ciências Biológicas e da Saúde - CCBS, Universidade Federal de Mato Grosso do Sul - UFMS, Campo Grande, MS, Brazil.
| | - Antônio Carlos Duenhas Monreal
- Centro de Ciências Biológicas e da Saúde - CCBS, Universidade Federal de Mato Grosso do Sul - UFMS, Campo Grande, MS, Brazil.
| | - Wanderson Romão
- Departamento de Química, Universidade Federal do Espírito Santo - UFES, Vitória, ES, Brazil.
| | - Valdemar Lacerda Júnior
- Departamento de Química, Universidade Federal do Espírito Santo - UFES, Vitória, ES, Brazil.
| | - Adilson Beatriz
- Programa de Mestrado em Farmácia, Centro de Ciências Biológicas e da Saúde - CCBS, Universidade Federal de Mato Grosso do Sul - UFMS, Campo Grande, MS, Brazil. .,Programa de Pós-graduação em Química, Instituto de Química, Universidade Federal de Mato Grosso do Sul - UFMS, Campo Grande, MS, Brazil.
| |
Collapse
|
20
|
Anti-apoptotic protein BRE/BRCC45 attenuates apoptosis through maintaining the expression of caspase inhibitor XIAP in mouse Lewis lung carcinoma D122 cells. Apoptosis 2014; 19:829-40. [PMID: 24395041 DOI: 10.1007/s10495-013-0963-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Brain and Reproductive Organ Expressed (BRE), or BRCC45, is a death receptor-associated antiapoptotic protein, which is also involved in DNA-damage repair, and K63-specific deubiquitination. BRE overexpression attenuates both death receptor- and stress-induced apoptosis, promotes experimental tumor growth, and is associated with human hepatocellular and esophageal carcinoma. How BRE mediates its antiapoptotic function is unknown. Here we report based on the use of a mouse Lewis lung carcinoma cell line D122 that BRE has an essential role in maintaining the cellular protein level of XIAP, which is the most potent endogenous inhibitor of the caspases functioning in both extrinsic and intrinsic apoptosis. shRNA-mediated exhaustive depletion of BRE sensitized D122 cells to apoptosis induced not only by etopoxide, but also by TNF-α even in the absence of cycloheximide, which blocks the synthesis of antiapoptotic proteins by TNF-α-activated NF-κB pathway. In BRE-depleted cells, protein level of XIAP was downregulated, but not the levels of other antiapoptotic proteins, cIAP-1, 2, and cFLIP, regulated by the same NF-κB pathway. Reconstitution of BRE restored XIAP levels and increased resistance to apoptosis. XIAP mRNA level was also reduced in the BRE-depleted cells, but the level of reduction was less profound than that of the protein level. However, BRE could not delay protein turnover of XIAP. Depletion of BRE also increased tumor cell apoptosis, and decreased both local and metastatic tumor growth. Taken together, these findings indicate that BRE and its XIAP-sustaining mechanism could represent novel targets for anti-cancer therapy.
Collapse
|
21
|
Hao L, Luan J, Zhang D, Li C, Guo H, Qi L, Liu X, Li T, Zhang Q. Research on the in vitro anticancer activity and in vivo tissue distribution of Amoitone B nanocrystals. Colloids Surf B Biointerfaces 2014; 117:258-66. [PMID: 24657612 DOI: 10.1016/j.colsurfb.2014.02.042] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2013] [Revised: 02/13/2014] [Accepted: 02/16/2014] [Indexed: 11/18/2022]
Abstract
Amoitone B, a natural agonist to Nur77, is a promising anticancer drug. However, its application is seriously restricted due to the water-insolubility and short biological half-life. Amoitone B nanocrystals (AmB-NC) were formulated by microfluidization method to overcome the above obstacles. This study aims to evaluate the cytotoxicity and tissue distribution of AmB-NC. MTT [3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide] assay revealed the improved in vitro antitumor activity of AmB-NC against stomach, colon, liver and lung cancer cells compared with Amoitone B solution. Meanwhile, observation of morphological changes, cell cycle and apoptosis examination using flow cytometry exhibited that AmB-NC could induce G1 cycle arrest and markedly enhance the apoptosis of human gastric cancer BGC-823 cell line. Tissue distribution study demonstrated that AmB-NC had a higher distribution in liver and lung, which was helpful for relevant cancer treatment. In conclusion, AmB-NC could be a potential delivery system for treatment of human cancer.
Collapse
Affiliation(s)
- Leilei Hao
- Department of Pharmaceutics, College of Pharmacy, Shandong University, 44 Wenhua Xilu, Jinan 250012, PR China
| | - Jingjing Luan
- Department of Pharmaceutics, College of Pharmacy, Shandong University, 44 Wenhua Xilu, Jinan 250012, PR China
| | - Dianrui Zhang
- Department of Pharmaceutics, College of Pharmacy, Shandong University, 44 Wenhua Xilu, Jinan 250012, PR China.
| | - Caiyun Li
- Department of Pharmaceutics, College of Pharmacy, Shandong University, 44 Wenhua Xilu, Jinan 250012, PR China
| | - Hejian Guo
- Department of Pharmaceutics, College of Pharmacy, Shandong University, 44 Wenhua Xilu, Jinan 250012, PR China
| | - Lisi Qi
- Department of Pharmaceutics, College of Pharmacy, Shandong University, 44 Wenhua Xilu, Jinan 250012, PR China
| | - Xinquan Liu
- Department of Pharmaceutics, College of Pharmacy, Shandong University, 44 Wenhua Xilu, Jinan 250012, PR China
| | - Tingting Li
- Department of Pharmaceutics, College of Pharmacy, Shandong University, 44 Wenhua Xilu, Jinan 250012, PR China
| | - Qiang Zhang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, 38 Xueyuan Road, Beijing 100083, PR China
| |
Collapse
|
22
|
Navarro SD, Beatriz A, Meza A, Pesarini JR, Gomes RDS, Karaziack CB, Cunha-Laura AL, Monreal ACD, Romão W, Lacerda Júnior V, Mauro MDO, Oliveira RJ. A new synthetic resorcinolic lipid 3-Heptyl-3,4,6-trimethoxy-3H-isobenzofuran-1-one: Evaluation of toxicology and ability to potentiate the mutagenic and apoptotic effects of cyclophosphamide. Eur J Med Chem 2014; 75:132-42. [DOI: 10.1016/j.ejmech.2014.01.057] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2013] [Revised: 01/27/2014] [Accepted: 01/29/2014] [Indexed: 01/21/2023]
|
23
|
Chen E, Tang MK, Yao Y, Yau WWY, Lo LM, Yang X, Chui YL, Chan J, Lee KKH. Silencing BRE expression in human umbilical cord perivascular (HUCPV) progenitor cells accelerates osteogenic and chondrogenic differentiation. PLoS One 2013; 8:e67896. [PMID: 23935848 PMCID: PMC3720665 DOI: 10.1371/journal.pone.0067896] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2012] [Accepted: 05/23/2013] [Indexed: 01/27/2023] Open
Abstract
BRE is a multifunctional adapter protein involved in DNA repair, cell survival and stress response. To date, most studies of this protein have been focused in the tumor model. The role of BRE in stem cell biology has never been investigated. Therefore, we have used HUCPV progenitor cells to elucidate the function of BRE. HUCPV cells are multipotent fetal progenitor cells which possess the ability to differentiate into a multitude of mesenchymal cell lineages when chemically induced and can be more easily amplified in culture. In this study, we have established that BRE expression was normally expressed in HUCPV cells but become down-regulated when the cells were induced to differentiate. In addition, silencing BRE expression, using BRE-siRNAs, in HUCPV cells could accelerate induced chondrogenic and osteogenic differentiation. Hence, we postulated that BRE played an important role in maintaining the stemness of HUCPV cells. We used microarray analysis to examine the transcriptome of BRE-silenced cells. BRE-silencing negatively regulated OCT4, FGF5 and FOXO1A. BRE-silencing also altered the expression of epigenetic genes and components of the TGF-β/BMP and FGF signaling pathways which are crucially involved in maintaining stem cell self-renewal. Comparative proteomic profiling also revealed that BRE-silencing resulted in decreased expressions of actin-binding proteins. In sum, we propose that BRE acts like an adaptor protein that promotes stemness and at the same time inhibits the differentiation of HUCPV cells.
Collapse
Affiliation(s)
- Elve Chen
- Stem Cell and Regeneration Thematic Research Programme, School of Biomedical Sciences, Chinese University of Hong Kong, Hong Kong, People's Republic of China
| | - Mei Kuen Tang
- Stem Cell and Regeneration Thematic Research Programme, School of Biomedical Sciences, Chinese University of Hong Kong, Hong Kong, People's Republic of China
| | - Yao Yao
- Stem Cell and Regeneration Thematic Research Programme, School of Biomedical Sciences, Chinese University of Hong Kong, Hong Kong, People's Republic of China
| | - Winifred Wing Yiu Yau
- Stem Cell and Regeneration Thematic Research Programme, School of Biomedical Sciences, Chinese University of Hong Kong, Hong Kong, People's Republic of China
| | - Lok Man Lo
- Stem Cell and Regeneration Thematic Research Programme, School of Biomedical Sciences, Chinese University of Hong Kong, Hong Kong, People's Republic of China
| | - Xuesong Yang
- Key Laboratory for Regenerative Medicine Ministry of Education, Jinan University, Guangzhou, People's Republic of China
| | - Yiu Loon Chui
- Department of Chemical Pathology, Chinese University of Hong Kong, Chinese University of Hong Kong, Hong Kong, People's Republic of China
| | - John Chan
- Key Laboratory for Regenerative Medicine Ministry of Education, Jinan University, Guangzhou, People's Republic of China
| | - Kenneth Ka Ho Lee
- Stem Cell and Regeneration Thematic Research Programme, School of Biomedical Sciences, Chinese University of Hong Kong, Hong Kong, People's Republic of China
- Key Laboratory for Regenerative Medicine Ministry of Education, Jinan University, Guangzhou, People's Republic of China
- School of Pharmacy and Life Sciences, Robert Gordon University, Aberdeen, Scotland, United Kingdom
- * E-mail:
| |
Collapse
|
24
|
Hao L, Wang X, Zhang D, Xu Q, Song S, Wang F, Li C, Guo H, Liu Y, Zheng D, Zhang Q. Studies on the preparation, characterization and pharmacokinetics of Amoitone B nanocrystals. Int J Pharm 2012; 433:157-64. [PMID: 22579996 DOI: 10.1016/j.ijpharm.2012.05.002] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2012] [Revised: 04/27/2012] [Accepted: 05/03/2012] [Indexed: 11/18/2022]
Abstract
Amoitone B, as a new derivative of cytosporone B, has been proved to be a natural agonist for Nur77. It exhibits remarkable anticancer activity in vivo and has the potential to be a therapeutic agent for cancer treatment. However, the poor solubility and dissolution rate result in low therapeutic index for injection and low bioavailability for oral administration, therefore limiting its application. In order to magnify the clinical use of Amoitone B, nanocrystal was selected as an application technology to solve the above problems. In this study, the optimized Amoitone B nanocrystals with small and uniform particle size were successfully prepared by microfluidization method and investigated by morphology, size distribution, and zeta potential. The differential scanning calorimetry (DSC) and X-ray diffraction (XRD) confirmed there was no crystalline state changed in the size reduction process. For Amoitone B nanocrystals, an accelerated dissolution velocity and increased saturation solubility were achieved in vitro and a markedly different pharmacokinetic property in vivo was exhibited with retarded clearance and magnified AUC compared with Amoitone B solution. These results implied that developing Amoitone B as nanocrystals is a promising choice for intravenous delivery and further application for cancer therapy.
Collapse
Affiliation(s)
- Leilei Hao
- Department of Pharmaceutics, College of Pharmacy, Shandong University, 44 Wenhua Xilu, Jinan 250012, PR China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Mechanisms and biomarkers of apoptosis in liver disease and fibrosis. Int J Hepatol 2012; 2012:648915. [PMID: 22567408 PMCID: PMC3332069 DOI: 10.1155/2012/648915] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2011] [Accepted: 01/24/2012] [Indexed: 02/07/2023] Open
Abstract
Liver fibrosis and cirrhosis are a major cause of morbidity and mortality worldwide. Development of the fibrotic scar is an outcome of chronic liver diseases of varying aetiologies including alcoholic liver disease (ALD) nonalcoholic liver disease (NAFLD) including non-alcoholic steatohepatitis (NASH) viral hepatitis B and C (HBV, HCV). The critical step in the development of scar is activation of hepatic stellate cells (HSCs), which become the primary source of extracellular matrix. Aberrant apoptosis is a feature of chronic liver diseases and is associated with worsening stages of fibrosis. However, apoptosis is also the main mechanism promoting the resolution of fibrosis, and spontaneous or targeted apoptosis of HSC is associated with regression of fibrosis in animal models and patients with chronic liver disease. Given the importance of apoptosis in disease progression and resolution, there is much interest in precisely delineating the mechanisms involved and also developing biomarkers that accurately reflect the underlying pathogenesis. Here, we review the mechanisms driving apoptosis in development of liver disease and use of apoptosis -related biomarkers to aid in clinical diagnosis. Finally, we will also examine the recent literature regarding new insights into mechanisms involved in apoptosis of activated HSCs as possible method of fibrosis regression.
Collapse
|
26
|
Yao LM, He JP, Chen HZ, Wang Y, Wang WJ, Wu R, Yu CD, Wu Q. Orphan receptor TR3 participates in cisplatin-induced apoptosis via Chk2 phosphorylation to repress intestinal tumorigenesis. Carcinogenesis 2011; 33:301-11. [DOI: 10.1093/carcin/bgr287] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
|
27
|
Gene expression profiling of peripheral blood mononuclear cells from children with active hemophagocytic lymphohistiocytosis. Blood 2011; 117:e151-60. [PMID: 21325597 DOI: 10.1182/blood-2010-08-300046] [Citation(s) in RCA: 79] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Familial hemophagocytic lymphohistiocytosis (FHL) is a rare, genetically heterogeneous autosomal recessive immune disorder that results when the critical regulatory pathways that mediate immune defense mechanisms and the natural termination of immune/inflammatory responses are disrupted or overwhelmed. To advance the understanding of FHL, we performed gene expression profiling of peripheral blood mononuclear cells from 11 children with untreated FHL. Total RNA was isolated and gene expression levels were determined using microarray analysis. Comparisons between patients with FHL and normal pediatric controls (n = 30) identified 915 down-regulated and 550 up-regulated genes with more than or equal to 2.5-fold difference in expression (P ≤ .05). The expression of genes associated with natural killer cell functions, innate and adaptive immune responses, proapoptotic proteins, and B- and T-cell differentiation were down-regulated in patients with FHL. Genes associated with the canonical pathways of interleukin-6 (IL-6), IL-10 IL-1, IL-8, TREM1, LXR/RXR activation, and PPAR signaling and genes encoding of antiapoptotic proteins were overexpressed in patients with FHL. This first study of genome-wide expression profiling in children with FHL demonstrates the complexity of gene expression patterns, which underlie the immunobiology of FHL.
Collapse
|
28
|
Potential targets for molecular imaging of apoptosis resistance in hepatocellular carcinoma. Biomed Imaging Interv J 2011; 7:e5. [PMID: 21655114 PMCID: PMC3107687 DOI: 10.2349/biij.7.1.e5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2010] [Revised: 08/25/2010] [Accepted: 09/22/2010] [Indexed: 12/16/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the most common cancers, which is mainly a concern in Southeast Asia. Apoptosis resistance in HCC is one of the significant factors for hepatocarcinogenesis and tumour progression. Recent advances of apoptosis resistance mechanisms in HCC could serve as potential targets for molecular imaging, which would be of considerable value to explore the molecular processes involved in HCC progression and to evaluate responses of certain anti-HCC therapies. Disruptions in the balance of anti-apoptotic and pro-apoptotic processes have been found to be involved in apoptosis resistance in HCC. Loss of response to death receptors, transformation of growth factor-β induced apoptosis, upregulation of anti-apoptotic Bcl-2 subgroup, as well as downregulation of pro-apoptotic Bax subgroup and BH3-only subgroup, are associated with apoptosis resistance in HCC. Mutation of p53 gene, dysregulation of NF-κB and survivin are also of interest because of their contribution to HCC development. In this review, the aim is to identify potential targets for molecular imaging of apoptosis resistance in HCC.
Collapse
|
29
|
Balgobind BV, Zwaan CM, Reinhardt D, Arentsen-Peters TJCM, Hollink IHIM, de Haas V, Kaspers GJL, de Bont ESJM, Baruchel A, Stary J, Meyer C, Marschalek R, Creutzig U, den Boer ML, Pieters R, van den Heuvel-Eibrink MM. High BRE expression in pediatric MLL-rearranged AML is associated with favorable outcome. Leukemia 2010; 24:2048-55. [PMID: 20861917 DOI: 10.1038/leu.2010.211] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Translocations involving the mixed lineage leukemia (MLL) gene, localized at 11q23, frequently occur in pediatric acute myeloid leukemia (AML). We recently reported differences in prognosis between the different translocation partners, suggesting differences in biological background. To unravel the latter, we used microarrays to generate gene expression profiles of 245 pediatric AML cases, including 53 MLL-rearranged cases. Thereby, we identified a specific gene expression signature for t(9;11)(p22;q23), and identified BRE (brain and reproductive organ expressed) to be discriminative for t(9;11)(p22;q23) (P<0.001) when compared with other MLL subtypes. Patients with high BRE expression showed a significantly better 3-year relapse-free survival (pRFS) (80±13 vs 30±10%, P=0.02) within MLL-rearranged AML cases. Moreover, multivariate analysis identified high BRE expression as an independent favorable prognostic factor within pediatric AML for RFS (HR=0.2, P=0.04). No significant differences were identified for 3-year event-free survival or for 3-year overall survival. Forced expression of BRE did not result in altered cell proliferation, apoptosis or drug sensitivity, which could explain the favorable outcome. In conclusion, overexpression of the BRE gene is predominantly found in MLL-rearranged AML with t(9;11)(p22;q23). Although further investigation for the role of BRE in leukemogenesis and outcome is warranted, high BRE expression is an independent prognostic factor for pRFS in pediatric AML.
Collapse
Affiliation(s)
- B V Balgobind
- Pediatric Oncology/Hematology, Erasmus MC-Sophia Children's Hospital, Rotterdam, The Netherlands
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Liu JJ, Zeng HN, Zhang LR, Zhan YY, Chen Y, Wang Y, Wang J, Xiang SH, Liu WJ, Wang WJ, Chen HZ, Shen YM, Su WJ, Huang PQ, Zhang HK, Wu Q. A unique pharmacophore for activation of the nuclear orphan receptor Nur77 in vivo and in vitro. Cancer Res 2010; 70:3628-37. [PMID: 20388790 DOI: 10.1158/0008-5472.can-09-3160] [Citation(s) in RCA: 80] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Nur77 is a steroid orphan receptor that plays a critical role in regulating proliferation, differentiation, and apoptosis, including acting as a switch for Bcl-2 function. We previously reported that the octaketide cytosporone B (Csn-B) is a natural agonist for Nur77. In this study, we synthesized a series of Csn-B analogues and performed a structure-activity analysis that suggested criteria for the development of a unique pharmacophore to activate Nur77. The components of the pharmacophore necessary for binding Nur77 included the benzene ring, the phenolic hydroxyl group, and the acyl chain of the Csn-B scaffold, whereas the key feature for activating the biological function of Nur77 was the ester group. Csn-B analogues that bound Nur77 tightly not only stimulated its transactivation activity but also initiated mitochondrial apoptosis by means of novel cross-talk between Nur77 and BRE, an antiapoptotic protein regulated at the transcriptional level. Notably, the derivative n-amyl 2-[3,5-dihydroxy-2-(1-nonanoyl)phenyl]acetate exhibited greater antitumor activity in vivo than its parent compounds, highlighting particular interest in this compound. Our findings describe a pathway for rational design of Csn-B-derived Nur77 agonists as a new class of potent and effective antitumor agents.
Collapse
Affiliation(s)
- Jing-jing Liu
- Key Laboratory of the Ministry of Education for Cell Biology and Tumor Cell Engineering, School of Life Sciences, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, Fujian Province, PR China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Chan JYH, Li L, Miao J, Cai DQ, Lee KKH, Chui YL. Differential expression of a novel gene BRE (TNFRSF1A modulator/BRCC45) in response to stress and biological signals. Mol Biol Rep 2010; 37:363-8. [PMID: 19757177 DOI: 10.1007/s11033-009-9796-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2009] [Accepted: 09/02/2009] [Indexed: 01/09/2023]
Abstract
Stress-responsive genes play critical roles in many biological functions that includes apoptosis, survival, differentiation and regeneration. We have identified a novel stress-responsive gene called BRE which interacts with TNF-receptor-1 and blocks the apoptotic effect of TNF-alpha. BRE enhances tumor growth in vivo and is up-regulated in hepatocellular and esophageal carcinomas. BRE also regulates the ubiquitination of the DNA repair complex BRCC, and the synthesis of steroid hormones. Here, we examined BRE-mRNA in cells after treatments with UV and ionizing radiation (IR). UV and IR treatment alone suppressed BRE-mRNA levels by more than 90% at 24 h, while hydroxyurea, fluorodeoxyuridine, aphidicolin, known inhibitors of S-phase DNA synthesis, had no significant effect. BRE protein expression was unaltered in cells treated with TNF-alpha, Interleukin-1 and Dexamethasone, while a threefold increase was observed following chorionic gonadotropin exposure. Although BRE plays a regulatory role in many different pathways, yet its expression is apparently under very stringent control.
Collapse
Affiliation(s)
- John Yeuk-Hon Chan
- Key Joint CUHK-JiNan University Laboratories for Regenerative Medicine, Ministry of Education, JiNan University, Guang Zhou, Guang Dong, China.
| | | | | | | | | | | |
Collapse
|
32
|
Chui YL, Ching AKK, Chen S, Yip FP, Rowlands DK, James AE, Lee KKH, Chan JYH. BRE over-expression promotes growth of hepatocellular carcinoma. Biochem Biophys Res Commun 2009; 391:1522-5. [PMID: 20035718 DOI: 10.1016/j.bbrc.2009.12.111] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2009] [Accepted: 12/18/2009] [Indexed: 11/16/2022]
Abstract
BRE, also known as TNFRSF1A modulator and BRCC45, is an evolutionarily highly conserved protein. It is a death receptor-associated protein in cytoplasm and a component of BRCA1/2-containing DNA repair complex in nucleus. BRE was found to have anti-apoptotic activity. Over-expression of BRE by transfection promoted survival of cell lines against apoptotic induction; whereas depletion of the protein by siRNA resulted in the opposite. In vivo anti-apoptotic activity of BRE was demonstrated by significant attenuation of Fas-induced acute fulminant hepatitis in transgenic mice expressing the human protein specifically in the liver. BRE was also implicated in tumor promotion by the accelerated tumor growth of Lewis Lung carcinoma transfected with human BRE; and by high expression of BRE specifically in the tumoral regions of human hepatocellular carcinoma (HCC). The present study was to test directly if transgenic expression of BRE in livers could promote HCC development in neonatal diethylnitrosamine model. By 8months after tumor induction, the maximal sizes of tumor nodules of transgenic mice were significantly larger than those of the non-transgenic controls, although the numbers of tumor nodules between the two groups did not significantly differ. Importantly, as in human HCC, the mouse endogenous BRE level was up-regulated in mouse HCC nodules. These results show that BRE over-expression can indeed promote growth, though not initiation, of liver tumors. Furthermore, the common occurrence of BRE over-expression in human and mouse HCC suggests that up-regulation of BRE is functionally important in liver tumor development.
Collapse
Affiliation(s)
- Yiu-Loon Chui
- Department of Chemical Pathology and Sir Y.K. Pao Centre for Cancer, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong SAR, China.
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Tang MK, Liu G, Hou Z, Chui YL, Chan JYH, Lee KKH. Livers overexpressing BRE transgene are under heightened state of stress-response, as revealed by comparative proteomics. Proteomics Clin Appl 2009; 3:1362-70. [PMID: 21136956 DOI: 10.1002/prca.200900097] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2009] [Revised: 08/17/2009] [Accepted: 08/18/2009] [Indexed: 12/27/2022]
Affiliation(s)
- Mei-Kuen Tang
- Health and Social Science Team, Open University of Hong Kong LiPACE, Hong Kong
| | | | | | | | | | | |
Collapse
|
34
|
Abstract
Hepatocellular carcinoma (HCC) is a major health problem, being the sixth most common cancer world-wide. Dysregulation of the balance between proliferation and cell death represents a pro-tumorigenic principle in human hepatocarcinogenesis. This review updates the recent relevant contributions reporting molecular alterations for HCC that induce an imbalance in the regulation of apoptosis. Alterations in the expression and/or activation of p53 are frequent in HCC cells, which confer on them resistance to chemotherapeutic drugs. Many HCCs are also insensitive to apoptosis induced either by death receptor ligands, such as FasL or TRAIL, or by transforming growth factor-beta (TGF-β). Although the expression of some pro-apoptotic genes is decreased, the balance between death and survival is dysregulated in HCC mainly due to overactivation of anti-apoptotic pathways. Indeed, some molecules involved in counteracting apoptosis, such as Bcl-XL, Mcl-1, c-IAP1, XIAP or survivin are over-expressed in HCC cells. Furthermore, some growth factors that mediate cell survival are up-regulated in HCC, as well as the molecules involved in the machinery responsible for cleavage of their pro-forms to an active peptide. The expression and/or activation of the JAK/STAT, PI3K/AKT and RAS/ERKs pathways are enhanced in many HCC cells, conferring on them resistance to apoptotic stimuli. Finally, recent evidence indicates that inflammatory processes, as well as the epithelial-mesenchymal transitions that occur in HCC cells to facilitate their dissemination, are related to cell survival. Therefore, therapeutic strategies to selectively inhibit anti-apoptotic signals in liver tumor cells have the potential to provide powerful tools to treat HCC.
Collapse
|
35
|
Hevel JM, Olson-Buelow LC, Ganesan B, Stevens JR, Hardman JP, Aust AE. Novel functional view of the crocidolite asbestos-treated A549 human lung epithelial transcriptome reveals an intricate network of pathways with opposing functions. BMC Genomics 2008; 9:376. [PMID: 18687144 PMCID: PMC2533023 DOI: 10.1186/1471-2164-9-376] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2007] [Accepted: 08/07/2008] [Indexed: 01/09/2023] Open
Abstract
Background Although exposure to asbestos is now regulated, patients continue to be diagnosed with mesothelioma, asbestosis, fibrosis and lung carcinoma because of the long latent period between exposure and clinical disease. Asbestosis is observed in approximately 200,000 patients annually and asbestos-related deaths are estimated at 4,000 annually[1]. Although advances have been made using single gene/gene product or pathway studies, the complexity of the response to asbestos and the many unanswered questions suggested the need for a systems biology approach. The objective of this study was to generate a comprehensive view of the transcriptional changes induced by crocidolite asbestos in A549 human lung epithelial cells. Results A statistically robust, comprehensive data set documenting the crocidolite-induced changes in the A549 transcriptome was collected. A systems biology approach involving global observations from gene ontological analyses coupled with functional network analyses was used to explore the effects of crocidolite in the context of known molecular interactions. The analyses uniquely document a transcriptome with function-based networks in cell death, cancer, cell cycle, cellular growth, proliferation, and gene expression. These functional modules show signs of a complex interplay between signaling pathways consisting of both novel and previously described asbestos-related genes/gene products. These networks allowed for the identification of novel, putative crocidolite-related genes, leading to several new hypotheses regarding genes that are important for the asbestos response. The global analysis revealed a transcriptome that bears signatures of both apoptosis/cell death and cell survival/proliferation. Conclusion Our analyses demonstrate the power of combining a statistically robust, comprehensive dataset and a functional network genomics approach to 1) identify and explore relationships between genes of known importance 2) identify novel candidate genes, and 3) observe the complex interplay between genes/gene products that function in seemingly different processes. This study represents the first function-based global approach toward understanding the response of human lung epithelial cells to the carcinogen crocidolite. Importantly, our investigation paints a much broader landscape for the crocidolite response than was previously appreciated and reveals novel paths to study. Our graphical representations of the function-based global network will be a valuable resource to model new research findings.
Collapse
Affiliation(s)
- Joan M Hevel
- Department of Chemistry and Biochemistry, Utah State University, Logan, USA.
| | | | | | | | | | | |
Collapse
|
36
|
Chen HB, Pan K, Tang MK, Chui YL, Chen L, Su ZJ, Shen ZY, Li EM, Xie W, Lee KKH. Comparative proteomic analysis reveals differentially expressed proteins regulated by a potential tumor promoter, BRE, in human esophageal carcinoma cells. Biochem Cell Biol 2008; 86:302-11. [PMID: 18756325 DOI: 10.1139/o08-069] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Esophageal tumorigenesis is a complex and cascading process, involving the interaction of many genes and proteins. In this study, we have used the comparative proteomic approach to identify tumor-associated proteins and explore the carcinogenic mechanisms. Two-dimensional electrophoresis (2-DE) and MALDI-TOF MS analysis of esophageal carcinoma and control cells revealed 10 proteins that were upregulated. A further 10 proteins were downregulated. Among these 20 differentially expressed proteins, brain and reproductive organ-expressed (BRE) protein was identified as a potential tumor promoter. It was high expressed by the esophageal carcinoma cells, as confirmed by RT-PCR and immunoblotting. BRE has been reported to be a stress-responsive protein. To gain further insight into its function, BRE expression was silenced in esophageal carcinoma cells using BRE-specific small interference RNA. It was discovered that silencing BRE expression downregulated prohibitin expression, but upregulated tumor-suppressor p53 expression. Furthermore, cyclin A and CDK2 expressions were suppressed suggesting that BRE inhibited cell proliferation. These results implied that BRE plays a significant role in mediating antiapoptotic and proliferative responses in esophageal carcinoma cells.
Collapse
Affiliation(s)
- Hai Bin Chen
- Department of Histology and Embryology, Shantou University Medical College, Shantou, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|