1
|
Ghosh AK. Acetyltransferase p300 Is a Putative Epidrug Target for Amelioration of Cellular Aging-Related Cardiovascular Disease. Cells 2021; 10:cells10112839. [PMID: 34831061 PMCID: PMC8616404 DOI: 10.3390/cells10112839] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2021] [Revised: 10/15/2021] [Accepted: 10/18/2021] [Indexed: 12/31/2022] Open
Abstract
Cardiovascular disease is the leading cause of accelerated as well as chronological aging-related human morbidity and mortality worldwide. Genetic, immunologic, unhealthy lifestyles including daily consumption of high-carb/high-fat fast food, lack of exercise, drug addiction, cigarette smoke, alcoholism, and exposure to environmental pollutants like particulate matter (PM)-induced stresses contribute profoundly to accelerated and chronological cardiovascular aging and associated life threatening diseases. All these stressors alter gene expression epigenetically either through activation or repression of gene transcription via alteration of chromatin remodeling enzymes and chromatin landscape by DNA methylation or histone methylation or histone acetylation. Acetyltransferase p300, a major epigenetic writer of acetylation on histones and transcription factors, contributes significantly to modifications of chromatin landscape of genes involved in cellular aging and cardiovascular diseases. In this review, the key findings those implicate acetyltransferase p300 as a major contributor to cellular senescence or aging related cardiovascular pathologies including vascular dysfunction, cardiac hypertrophy, myocardial infarction, cardiac fibrosis, systolic/diastolic dysfunction, and aortic valve calcification are discussed. The efficacy of natural or synthetic small molecule inhibitor targeting acetyltransferase p300 in amelioration of stress-induced dysregulated gene expression, cellular aging, and cardiovascular disease in preclinical study is also discussed.
Collapse
Affiliation(s)
- Asish K Ghosh
- Feinberg Cardiovascular and Renal Research Institute, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| |
Collapse
|
2
|
Melekhova A, Baniahmad A. ING Tumour Suppressors and ING Splice Variants as Coregulators of the Androgen Receptor Signalling in Prostate Cancer. Cells 2021; 10:cells10102599. [PMID: 34685579 PMCID: PMC8533759 DOI: 10.3390/cells10102599] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 09/25/2021] [Accepted: 09/28/2021] [Indexed: 12/12/2022] Open
Abstract
Prevention and overcoming castration resistance of prostate cancer (PC) remains one of the main unsolved problems in modern oncology. Hence, many studies are focused on the investigation of novel androgen receptor (AR) regulators that could serve as potential drug targets in disease therapy. Among such factors, inhibitor of growth (ING) proteins were identified. Some ING proteins act as AR transcriptional coregulators, indicating their relevance for PC research. The ING family consists of five protein-coding genes from ING1 to ING5 and pseudogene INGX. The ING genes were revealed through their sequence homology to the first identified ING1 from an in vivo screen. ING factors are a part of histone modification complexes. With the help of the conserved plant homeodomain (PHD) motif, ING factors bind to Histone 3 Lysine 4 (H3K4) methylation mark with a stronger affinity to the highest methylation grade H3K4me3 and recruit histone acetyltransferases (HAT) and histone deacetylases (HDAC) to chromatin. ING1 and ING2 are core subunits of mSIN3a-HDAC corepressor complexes, whereas ING3–5 interact with different HAT complexes that serve as coactivators. ING members belong to type II tumour suppressors and are frequently downregulated in many types of malignancies, including PC. As the family name indicates, ING proteins are able to inhibit cell growth and tumour development via regulation of cell cycle and cancer-relevant pathways such as apoptosis, cellular senescence, DNA repair, cell migration, invasion, and angiogenesis. Many ING splice variants that enhance the diversity of ING activity were discovered. However, it seems that the existence of multiple ING splice variants is underestimated, since alternative splice variants, such as the AR coregulators ING1 and ING3, counteract full-length ING and thus play an opposite functional role. These results open a novel prospective investigation direction in understanding ING factors biology in PC and other malignancies.
Collapse
Affiliation(s)
| | - Aria Baniahmad
- Correspondence: ; Tel.: +49-3641-9396-820; Fax: +49-3641-9396-822
| |
Collapse
|
3
|
Shin EY, Soung NK, Schwartz MA, Kim EG. Altered endocytosis in cellular senescence. Ageing Res Rev 2021; 68:101332. [PMID: 33753287 PMCID: PMC8131247 DOI: 10.1016/j.arr.2021.101332] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 03/02/2021] [Accepted: 03/15/2021] [Indexed: 01/10/2023]
Abstract
Cellular senescence occurs in response to diverse stresses (e.g., telomere shortening, DNA damage, oxidative stress, oncogene activation). A growing body of evidence indicates that alterations in multiple components of endocytic pathways contribute to cellular senescence. Clathrin-mediated endocytosis (CME) and caveolae-mediated endocytosis (CavME) represent major types of endocytosis that are implicated in senescence. More recent research has also identified a chromatin modifier and tumor suppressor that contributes to the induction of senescence via altered endocytosis. Here, molecular regulators of aberrant endocytosis-induced senescence are reviewed and discussed in the context of their capacity to serve as senescence-inducing stressors or modifiers.
Collapse
Affiliation(s)
- Eun-Young Shin
- Department of Biochemistry, Chungbuk National University College of Medicine, Cheongju, 28644, South Korea
| | - Nak-Kyun Soung
- World Class Institute, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Ochang-eup, Cheongju, 28116, South Korea
| | - Martin Alexander Schwartz
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, And Department of Cell Biology, Yale University School of Medicine, New Haven, CT, 06511, USA; Wellcome Trust Centre for Cell-matrix Research, University of Manchester, Manchester, UK.
| | - Eung-Gook Kim
- Department of Biochemistry, Chungbuk National University College of Medicine, Cheongju, 28644, South Korea.
| |
Collapse
|
4
|
Resnik SR, Egger A, Abdo Abujamra B, Jozic I. Clinical Implications of Cellular Senescence on Wound Healing. CURRENT DERMATOLOGY REPORTS 2020. [DOI: 10.1007/s13671-020-00320-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
5
|
Dantas A, Al Shueili B, Yang Y, Nabbi A, Fink D, Riabowol K. Biological Functions of the ING Proteins. Cancers (Basel) 2019; 11:E1817. [PMID: 31752342 PMCID: PMC6896041 DOI: 10.3390/cancers11111817] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Accepted: 11/07/2019] [Indexed: 02/06/2023] Open
Abstract
The proteins belonging to the inhibitor of growth (ING) family of proteins serve as epigenetic readers of the H3K4Me3 histone mark of active gene transcription and target histone acetyltransferase (HAT) or histone deacetylase (HDAC) protein complexes, in order to alter local chromatin structure. These multidomain adaptor proteins interact with numerous other proteins to facilitate their localization and the regulation of numerous biochemical pathways that impinge upon biological functions. Knockout of some of the ING genes in murine models by various groups has verified their status as tumor suppressors, with ING1 knockout resulting in the formation of large clear-cell B-lymphomas and ING2 knockout increasing the frequency of ameloblastomas, among other phenotypic effects. ING4 knockout strongly affects innate immunity and angiogenesis, and INGs1, ING2, and ING4 have been reported to affect apoptosis in different cellular models. Although ING3 and ING5 knockouts have yet to be published, preliminary reports indicate that ING3 knockout results in embryonic lethality and that ING5 knockout may have postpartum effects on stem cell maintenance. In this review, we compile the known information on the domains of the INGs and the effects of altering ING protein expression, to better understand the functions of this adaptor protein family and its possible uses for targeted cancer therapy.
Collapse
Affiliation(s)
- Arthur Dantas
- Arnie Charbonneau Cancer Institute, Departments of Biochemistry and Molecular Biology and Oncology, University of Calgary, 374 HMRB, 3330 Hospital Dr. NW, Calgary, AB T2N 4N1, Canada; (A.D.); (B.A.S.); (Y.Y.)
| | - Buthaina Al Shueili
- Arnie Charbonneau Cancer Institute, Departments of Biochemistry and Molecular Biology and Oncology, University of Calgary, 374 HMRB, 3330 Hospital Dr. NW, Calgary, AB T2N 4N1, Canada; (A.D.); (B.A.S.); (Y.Y.)
| | - Yang Yang
- Arnie Charbonneau Cancer Institute, Departments of Biochemistry and Molecular Biology and Oncology, University of Calgary, 374 HMRB, 3330 Hospital Dr. NW, Calgary, AB T2N 4N1, Canada; (A.D.); (B.A.S.); (Y.Y.)
| | - Arash Nabbi
- Princess Margaret Cancer Centre, Toronto, ON M5G 2M9, Canada
| | - Dieter Fink
- Institute of Laboratory Animal Science, Department for Biomedical Sciences, University of Veterinary Medicine Vienna, 1210 Vienna, Austria;
| | - Karl Riabowol
- Arnie Charbonneau Cancer Institute, Departments of Biochemistry and Molecular Biology and Oncology, University of Calgary, 374 HMRB, 3330 Hospital Dr. NW, Calgary, AB T2N 4N1, Canada; (A.D.); (B.A.S.); (Y.Y.)
| |
Collapse
|
6
|
Zheng Q, Liu P, Gao G, Yuan J, Wang P, Huang J, Xie L, Lu X, Di F, Tong T, Chen J, Lu Z, Guan J, Wang G. Mitochondrion-processed TERC regulates senescence without affecting telomerase activities. Protein Cell 2019; 10:631-648. [PMID: 30788732 PMCID: PMC6711880 DOI: 10.1007/s13238-019-0612-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2018] [Accepted: 01/15/2019] [Indexed: 12/22/2022] Open
Abstract
Mitochondrial dysfunctions play major roles in ageing. How mitochondrial stresses invoke downstream responses and how specificity of the signaling is achieved, however, remains unclear. We have previously discovered that the RNA component of Telomerase TERC is imported into mitochondria, processed to a shorter form TERC-53, and then exported back to the cytosol. Cytosolic TERC-53 levels respond to mitochondrial functions, but have no direct effect on these functions, suggesting that cytosolic TERC-53 functions downstream of mitochondria as a signal of mitochondrial functions. Here, we show that cytosolic TERC-53 plays a regulatory role on cellular senescence and is involved in cognition decline in 10 months old mice, independent of its telomerase function. Manipulation of cytosolic TERC-53 levels affects cellular senescence and cognition decline in 10 months old mouse hippocampi without affecting telomerase activity, and most importantly, affects cellular senescence in terc−/− cells. These findings uncover a senescence-related regulatory pathway with a non-coding RNA as the signal in mammals.
Collapse
Affiliation(s)
- Qian Zheng
- MOE Key laboratory of Bioinformatics, Cell Biology and Development Center, School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Peipei Liu
- MOE Key laboratory of Bioinformatics, Cell Biology and Development Center, School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Ge Gao
- MOE Key laboratory of Bioinformatics, Cell Biology and Development Center, School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Jiapei Yuan
- MOE Key laboratory of Bioinformatics, Cell Biology and Development Center, School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Pengfeng Wang
- Peking University Research Center on Aging, Beijing, 100191, China
| | - Jinliang Huang
- MOE Key laboratory of Bioinformatics, Cell Biology and Development Center, School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Leiming Xie
- MOE Key laboratory of Bioinformatics, Cell Biology and Development Center, School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Xinping Lu
- MOE Key laboratory of Bioinformatics, Cell Biology and Development Center, School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Fan Di
- MOE Key laboratory of Bioinformatics, Cell Biology and Development Center, School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Tanjun Tong
- Peking University Research Center on Aging, Beijing, 100191, China.,Department of Biochemistry and Molecular Biology, Peking University Health Science Center, Beijing, 100191, China
| | - Jun Chen
- Peking University Research Center on Aging, Beijing, 100191, China.,Department of Biochemistry and Molecular Biology, Peking University Health Science Center, Beijing, 100191, China
| | - Zhi Lu
- MOE Key laboratory of Bioinformatics, Cell Biology and Development Center, School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Jisong Guan
- MOE Key laboratory of Bioinformatics, Cell Biology and Development Center, School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Geng Wang
- MOE Key laboratory of Bioinformatics, Cell Biology and Development Center, School of Life Sciences, Tsinghua University, Beijing, 100084, China.
| |
Collapse
|
7
|
Bertschmann J, Thalappilly S, Riabowol K. The ING1a model of rapid cell senescence. Mech Ageing Dev 2019; 177:109-117. [DOI: 10.1016/j.mad.2018.06.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2018] [Revised: 05/21/2018] [Accepted: 06/16/2018] [Indexed: 12/17/2022]
|
8
|
Yan H, Wu A. FOXO1 is crucial in glioblastoma cell tumorigenesis and regulates the expression of SIRT1 to suppress senescence in the brain. Mol Med Rep 2017; 17:2535-2542. [PMID: 29207098 DOI: 10.3892/mmr.2017.8146] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Accepted: 08/14/2017] [Indexed: 11/06/2022] Open
Abstract
In the present study, the role of Forkhead Box O1 (FOXO1) in glioblastoma (GBM) cell tumorigenesis was examined and the underlying mechanisms were investigated. Reverse transcription‑quantitative polymerase chain reaction and western blot analyses were used to analyze the expression of FOXO1 in GBM cell lines (LN18 and T98G) and tissues. Compared with the control groups, FOXO1 was significantly downregulated in the GBM tissues and GBM cell lines (P<0.05). The effects of the expression of FOXO1 on GBM cell proliferation and cell cycle were examined using flow cytometry. The overexpression of FOXO1 markedly inhibited LN18 and T98G cell proliferation and arrested cell cycle at the G0/G1 phase. In addition, FOXO1 facilitated cell senescence through regulation of the expression of sirtuin 1. Epithelial‑mesenchymal transition (EMT) is a complex process, which affects cell growth, invasion and metastasis. The results of the present study revealed that FOXO1 inhibited EMT and metastasis in GBM. These finding revealed a novel mechanism of FOXO1 in the suppression of tumorigenesis and metastasis of GBM cells and suggested that FOXO1 may be a potential therapeutic target for treating GBM.
Collapse
Affiliation(s)
- Han Yan
- Department of Neurosurgery, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Anhua Wu
- Department of Neurosurgery, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| |
Collapse
|
9
|
Zhao G, Wang H, Xu C, Wang P, Chen J, Wang P, Sun Z, Su Y, Wang Z, Han L, Tong T. SIRT6 delays cellular senescence by promoting p27Kip1 ubiquitin-proteasome degradation. Aging (Albany NY) 2017; 8:2308-2323. [PMID: 27794562 PMCID: PMC5115890 DOI: 10.18632/aging.101038] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2016] [Accepted: 09/04/2016] [Indexed: 12/15/2022]
Abstract
Sirtuin6(SIRT6) has been implicated as a key factor in aging and aging-related diseases. However, the role of SIRT6 in cellular senescence has not been fully understood. Here, we show that SIRT6 repressed the expression of p27Kip1 (p27) in cellular senescence. The expression of SIRT6 was reduced during cellular senescence, whereas enforced SIRT6 expression promoted cell proliferation and antagonized cellular senescence. In addition, we demonstrated that SIRT6 promoted p27 degradation by proteasome and SIRT6 decreased the acetylation level and protein half-life of p27. p27 acetylation increased its protein stability. Furthermore, SIRT6 directly interacted with p27. Importantly, p27 was strongly acetylated and had a prolonged protein half-life with the reduction of SIRT6 when cells were senescent, compared with those young cells. Finally, SIRT6 markedly rescued senescence induced by p27. Our findings indicate that SIRT6 decreases p27 acetylation, leading to its degradation via ubiquitin-proteasome pathway and then delays cellular senescence.
Collapse
Affiliation(s)
- Ganye Zhao
- Peking University Research Center on Aging, Department of Biochemistry and Molecular Biology, Peking University Health Science Center, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Beijing, 100191, China
| | - Hui Wang
- Peking University Research Center on Aging, Department of Biochemistry and Molecular Biology, Peking University Health Science Center, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Beijing, 100191, China
| | - Chenzhong Xu
- Peking University Research Center on Aging, Department of Biochemistry and Molecular Biology, Peking University Health Science Center, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Beijing, 100191, China
| | - Pan Wang
- Peking University Research Center on Aging, Department of Biochemistry and Molecular Biology, Peking University Health Science Center, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Beijing, 100191, China
| | - Jun Chen
- Peking University Research Center on Aging, Department of Biochemistry and Molecular Biology, Peking University Health Science Center, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Beijing, 100191, China
| | - Pengfeng Wang
- Peking University Research Center on Aging, Department of Biochemistry and Molecular Biology, Peking University Health Science Center, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Beijing, 100191, China
| | - Zhaomeng Sun
- Peking University Research Center on Aging, Department of Biochemistry and Molecular Biology, Peking University Health Science Center, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Beijing, 100191, China
| | - Yuanyuan Su
- Peking University Research Center on Aging, Department of Biochemistry and Molecular Biology, Peking University Health Science Center, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Beijing, 100191, China
| | - Zhao Wang
- MOE Key Laboratory of Protein Sciences, Department of Pharmacology, School of Medicine, Tsinghua University, Beijing, 100084, China
| | - Limin Han
- Peking University Research Center on Aging, Department of Biochemistry and Molecular Biology, Peking University Health Science Center, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Beijing, 100191, China
| | - Tanjun Tong
- Peking University Research Center on Aging, Department of Biochemistry and Molecular Biology, Peking University Health Science Center, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Beijing, 100191, China
| |
Collapse
|
10
|
Deschênes M, Chabot B. The emerging role of alternative splicing in senescence and aging. Aging Cell 2017; 16:918-933. [PMID: 28703423 PMCID: PMC5595669 DOI: 10.1111/acel.12646] [Citation(s) in RCA: 126] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/12/2017] [Indexed: 12/22/2022] Open
Abstract
Deregulation of precursor mRNA splicing is associated with many illnesses and has been linked to age-related chronic diseases. Here we review recent progress documenting how defects in the machinery that performs intron removal and controls splice site selection contribute to cellular senescence and organismal aging. We discuss the functional association linking p53, IGF-1, SIRT1, and ING-1 splice variants with senescence and aging, and review a selection of splicing defects occurring in accelerated aging (progeria), vascular aging, and Alzheimer's disease. Overall, it is becoming increasingly clear that changes in the activity of splicing factors and in the production of key splice variants can impact cellular senescence and the aging phenotype.
Collapse
Affiliation(s)
- Mathieu Deschênes
- Department of Microbiology and Infectious DiseasesFaculty of Medicine and Health SciencesUniversité de SherbrookeSherbrookeQuebecJ1E 4K8Canada
| | - Benoit Chabot
- Department of Microbiology and Infectious DiseasesFaculty of Medicine and Health SciencesUniversité de SherbrookeSherbrookeQuebecJ1E 4K8Canada
| |
Collapse
|
11
|
Sun R, Zhu B, Xiong K, Sun Y, Shi D, Chen L, Zhang Y, Li Z, Xue L. Senescence as a novel mechanism involved in β-adrenergic receptor mediated cardiac hypertrophy. PLoS One 2017; 12:e0182668. [PMID: 28783759 PMCID: PMC5544424 DOI: 10.1371/journal.pone.0182668] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Accepted: 07/14/2017] [Indexed: 01/15/2023] Open
Abstract
Pathological cardiac hypertrophy used to be elucidated by biomechanical, stretch-sensitive or neurohumoral mechanisms. However, a series of hints have indicated that hypertrophy process simulates senescence program. However, further evidence need to be pursued. To verify this hypothesis and examine whether cardiac senescence is a novel mechanism of hypertrophy induced by isoproterenol, 2-month-old male Sprague Dawley rats were subjected to isoproterenol infusion (0.25mg/kg/day) for 7 days by subcutaneous injection). Key characteristics of senescence (senescence-associated β-galactosidase activity, lipofuscin, expression of cyclin-dependent kinase inhibitors) were examined in cardiac hypertrophy model. Senescence-like phenotype, such as increased senescence-associated β-galactosidase activity, accumulation of lipofuscin and high levels of cyclin-dependent kinase inhibitors (e.g. p16, p19, p21 and p53) was found along the process of cardiac hypertrophy. Cardiac-specific transcription factor GATA4 increased in isoproterenol-treated cardiomyocytes as well. We further found that myocardial hypertrophy could be inhibited by resveratrol, an anti-aging compound, in a dose-dependent manner. Our results showed for the first time that cardiac senescence is involved in the process of pathological cardiac hypertrophy induced by isoproterenol.
Collapse
Affiliation(s)
- Rongrong Sun
- Department of Biochemistry and Molecular Biology, Peking University Health Science Center, Beijing, China
- Institute of Vascular Medicine, Peking University Third Hospital, Beijing, China
| | - Baoling Zhu
- Institute of Vascular Medicine, Peking University Third Hospital, Beijing, China
| | - Kai Xiong
- Medical Research Center, Peking University Third Hospital, Beijing, China
| | - Yan Sun
- Medical Research Center, Peking University Third Hospital, Beijing, China
| | - Dandan Shi
- Medical Research Center, Peking University Third Hospital, Beijing, China
| | - Li Chen
- Medical Research Center, Peking University Third Hospital, Beijing, China
| | - Youyi Zhang
- Institute of Vascular Medicine, Peking University Third Hospital, Beijing, China
| | - Zijian Li
- Institute of Vascular Medicine, Peking University Third Hospital, Beijing, China
- * E-mail: (LX); (ZL)
| | - Lixiang Xue
- Department of Biochemistry and Molecular Biology, Peking University Health Science Center, Beijing, China
- Medical Research Center, Peking University Third Hospital, Beijing, China
- Department of Radiation Oncology, Peking University Third Hospital, Beijing, China
- * E-mail: (LX); (ZL)
| |
Collapse
|
12
|
Zhang R, Jin J, Shi J, Hou Y. INGs are potential drug targets for cancer. J Cancer Res Clin Oncol 2017; 143:189-197. [PMID: 27544390 DOI: 10.1007/s00432-016-2219-z] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Accepted: 08/09/2016] [Indexed: 12/11/2022]
Abstract
PURPOSE The inhibitor of growth (ING) family consists of ING1, ING2, ING3, ING4 and ING5, which function as the type II tumor suppressors. INGs regulate cell proliferation, senescence, apoptosis, differentiation, angiogenesis, DNA repair, metastasis, and invasion by multiple pathways. In addition, INGs increase cancer cell sensitivity for chemotherapy and radiotherapy, while clinical observations show that INGs are frequently lost in some types of cancers. The aim of the study was to summarize the recent progress regarding INGs regulating tumor progression. METHODS The literatures of INGs regulating tumor progression were searched and assayed. RESULTS The regulating signaling pathways of ING1, ING2, ING3 or ING4 on tumor progression were shown. The mechanisms of INGs on tumor suppression were also assayed. CONCLUSIONS This review better summarized the signaling mechanism of INGs on tumor suppression, which provides a candidate therapy strategy for cancers.
Collapse
Affiliation(s)
- Runyun Zhang
- Department of Oncology, Affiliated Wujin People's Hospital, Jiangsu University, Changzhou, 212017, People's Republic of China
- Institute of Life Sciences, Jiangsu University, Zhenjiang, 212013, People's Republic of China
| | - Jianhua Jin
- Department of Oncology, Affiliated Wujin People's Hospital, Jiangsu University, Changzhou, 212017, People's Republic of China
| | - Juanjuan Shi
- Institute of Life Sciences, Jiangsu University, Zhenjiang, 212013, People's Republic of China.
| | - Yongzhong Hou
- Department of Oncology, Affiliated Wujin People's Hospital, Jiangsu University, Changzhou, 212017, People's Republic of China.
- Institute of Life Sciences, Jiangsu University, Zhenjiang, 212013, People's Republic of China.
| |
Collapse
|
13
|
KUNTER İ, KANDEMİŞ E, ALOTAİBİ H, CANDA T, ERDAL BAĞRIYANIK E. Alteration in the subcellular location of the inhibitor of growth proteinp33(ING1b) in estrogen receptor alpha positive breast carcinoma cells. Turk J Biol 2017. [DOI: 10.3906/biy-1602-95] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
|
14
|
AL Shabanah OA, Alotaibi MR, Al Rejaie SS, Alhoshani AR, Almutairi MM, Alshammari MA, Hafez MM. Inhibitory Effect of Ginseng on Breast Cancer Cell Line Growth Via Up-Regulation of Cyclin Dependent Kinase Inhibitor, p21 and p53. Asian Pac J Cancer Prev 2016; 17:4965-4971. [PMID: 28032724 PMCID: PMC5454704 DOI: 10.22034/apjcp.2016.17.11.4965] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Objective: Breast cancer is global female health problem worldwide. Most of the currently used agents for breast cancer treatment have toxic side-effects. Ginseng root, an oriental medicine, has many health benefits and may exhibit direct anti-cancer properties. This study was performed to assess the effects of ginseng on breast cancer cell lines. Materials and Methods: Cytotoxicity of ginseng extract was measured by MTT assay after exposure of MDA-MB-231, MCF-10A and MCF-7 breast cancer cells to concentrations of 0.25, 0.5, 1, 1.5, 2 and 2.5 mg/well. Expression levels of p21WAF, p16INK4A, Bcl-2, Bax and P53 genes were analyzed by quantitative real time PCR. Results: The treatment resulted in inhibition of cell proliferation in a dose-and time-dependent manner. p53, p21WAF1and p16INK4A expression levels were up-regulated in ginseng treated MDA-MB-231 and MCF-7 cancer cells compared to untreated controls and in MCF-10A cells. The expression levels of Bcl2 in the MDA-MB-231 and MCF-7 cells were down-regulated. In contrast, that of Bax was significantly up-regulated. Conclusion: The results of this study revealed that ginseng may inhibit breast cancer cell growth by activation of the apoptotic pathway.
Collapse
Affiliation(s)
- Othman A AL Shabanah
- College of Pharmacy, Pharmacology and Toxicology Department, Kind Saud University, Riyadh, Kingdom of Saudi Arabia.
| | | | | | | | | | | | | |
Collapse
|
15
|
Xu Q, Liu M, Zhang J, Xue L, Zhang G, Hu C, Wang Z, He S, Chen L, Ma K, Liu X, Zhao Y, Lv N, Liang S, Zhu H, Xu N. Overexpression of KLF4 promotes cell senescence through microRNA-203-survivin-p21 pathway. Oncotarget 2016; 7:60290-60302. [PMID: 27531889 PMCID: PMC5312384 DOI: 10.18632/oncotarget.11200] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2015] [Accepted: 07/26/2016] [Indexed: 02/05/2023] Open
Abstract
Krüppel-like factor 4 (KLF4) is a transcription factor and functions as a tumor suppressor or tumor promoter in different cancer types. KLF4 regulates many gene expression, thus affects the process of cell proliferation, differentiation, and apoptosis. Recently, KLF4 was reported to induce senescence during the generation of induced pluripotent stem (iPS) cells, but the exact mechanism is still unclear. In this study, we constructed two doxycycline-inducing KLF4 cell models, and demonstrated overexpression of KLF4 could promote cell senescence, detected by senescence-associated β-galactosidase activity assay. Then we confirmed that p21, a key effector of senescence, was directly induced by KLF4. KLF4 could also inhibit survivin, which could indirectly induce p21. By miRNA microarray, we found a series of miRNAs regulated by KLF4 and involved in senescence. We demonstrated that KLF4 could upregulate miR-203, and miR-203 contributed to senescence through miR-203-survivin-p21 pathway. Our results suggest that KLF4 could promote cell senescence through a complex network: miR-203, survivin, and p21, which were all regulated by overexpression of KLF4 and contributed to cell senescence.
Collapse
Affiliation(s)
- Qing Xu
- Laboratory of Cell and Molecular Biology and State Key Laboratory of Molecular Oncology, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Mei Liu
- Laboratory of Cell and Molecular Biology and State Key Laboratory of Molecular Oncology, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ju Zhang
- Division of Proteomics, Beijing Institute of Genomics, Chinese Academy of Science, Beijing, China
| | - Liyan Xue
- Department of Pathology, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Guo Zhang
- Laboratory of Cell and Molecular Biology and State Key Laboratory of Molecular Oncology, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Chenfei Hu
- Laboratory of Cell and Molecular Biology and State Key Laboratory of Molecular Oncology, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zaozao Wang
- Laboratory of Cell and Molecular Biology and State Key Laboratory of Molecular Oncology, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Shun He
- Laboratory of Cell and Molecular Biology and State Key Laboratory of Molecular Oncology, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Lechuang Chen
- Laboratory of Cell and Molecular Biology and State Key Laboratory of Molecular Oncology, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Kai Ma
- Laboratory of Cell and Molecular Biology and State Key Laboratory of Molecular Oncology, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xianghe Liu
- Laboratory of Cell and Molecular Biology and State Key Laboratory of Molecular Oncology, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yahui Zhao
- Laboratory of Cell and Molecular Biology and State Key Laboratory of Molecular Oncology, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ning Lv
- Department of Pathology, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Shufang Liang
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, P. R. China
| | - Hongxia Zhu
- Laboratory of Cell and Molecular Biology and State Key Laboratory of Molecular Oncology, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ningzhi Xu
- Laboratory of Cell and Molecular Biology and State Key Laboratory of Molecular Oncology, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, P. R. China
| |
Collapse
|
16
|
Esmaeili M, Jennek S, Ludwig S, Klitzsch A, Kraft F, Melle C, Baniahmad A. The tumor suppressor ING1b is a novel corepressor for the androgen receptor and induces cellular senescence in prostate cancer cells. J Mol Cell Biol 2016; 8:207-20. [PMID: 26993046 DOI: 10.1093/jmcb/mjw007] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Accepted: 12/10/2015] [Indexed: 12/28/2022] Open
Abstract
The androgen receptor (AR) signaling is critical for prostate cancer (PCa) progression to the castration-resistant stage with poor clinical outcome. Altered function of AR-interacting factors may contribute to castration-resistant PCa (CRPCa). Inhibitor of growth 1 (ING1) is a tumor suppressor that regulates various cellular processes including cell proliferation. Interestingly, ING1 expression is upregulated in senescent primary human prostate cells; however, its role in AR signaling in PCa was unknown. Using a proteomic approach by surface-enhanced laser desorption ionization-mass spectrometry (SELDI-MS) combined with immunological techniques, we provide here evidence that ING1b interacts in vivo with the AR. The interaction was confirmed by co-immunoprecipitation, in vitro GST-pull-down, and quantitative intracellular colocalization analyses. Functionally, ING1b inhibits AR-responsive promoters and endogenous key AR target genes in the human PCa LNCaP cells. Conversely, ING1b knockout (KO) mouse embryonic fibroblasts (MEFs) exhibit enhanced AR activity, suggesting that the interaction with ING1b represses the AR-mediated transcription. Also, data suggest that ING1b expression is downregulated in CRPCa cells compared with androgen-dependent LNCaP cells. Interestingly, its ectopic expression induces cellular senescence and reduces cell migration in both androgen-dependent and CRPCa cells. Intriguingly, ING1b can also inhibit androgen-induced growth in LNCaP cells in a similar manner as AR antagonists. Moreover, ING1b upregulates different cell cycle inhibitors including p27(KIP1), which is a novel target for ING1b. Taken together, our findings reveal a novel corepressor function of ING1b on various AR functions, thereby inhibiting PCa cell growth.
Collapse
Affiliation(s)
- Mohsen Esmaeili
- Institute of Human Genetics, Jena University Hospital, Jena, Germany
| | - Susanne Jennek
- Institute of Human Genetics, Jena University Hospital, Jena, Germany
| | - Susann Ludwig
- Institute of Human Genetics, Jena University Hospital, Jena, Germany
| | | | - Florian Kraft
- Institute of Human Genetics, Jena University Hospital, Jena, Germany
| | - Christian Melle
- Biomolecular Photonics Group, Jena University Hospital, Jena, Germany
| | - Aria Baniahmad
- Institute of Human Genetics, Jena University Hospital, Jena, Germany
| |
Collapse
|
17
|
Thakur S, Nabbi A, Klimowicz A, Riabowol K. Stromal ING1 expression induces a secretory phenotype and correlates with breast cancer patient survival. Mol Cancer 2015; 14:164. [PMID: 26306560 PMCID: PMC4549945 DOI: 10.1186/s12943-015-0434-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2015] [Accepted: 08/13/2015] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND Previous studies have established that levels of the Inhibitor of Growth 1(ING1) tumor suppressor are reduced in a significant proportion of different cancer types. Here we analyzed levels of ING1 in breast cancer patients to determine its prognostic significance as a biomarker for breast cancer prognosis. METHODS We used automated quantitative analysis (AQUA) to determine the levels of ING1 in the tumor associated stromal cells of 462 breast cancer samples. To better understand how high ING1 levels affect nearby epithelium, we measured the levels of cytokines and secreted matrix metalloproteases (MMPs), using an ELISA based assay in mammary fibroblasts overexpressing ING1. These cells were also used in a 3-dimensional co-culture with MCF7 cells to determine the effect of released MMPs and other cytokines on growing colonies. RESULTS We find that high levels of ING1 in stroma are associated with tumor grade (p = 0.001) and size (p = 0.02), and inversely associated with patient survival (p = 0.0001) in luminal, but not in non-luminal cancers, suggesting that high stromal ING1 promotes cancer development. In this group of patients ING1 could also predict patient survival and act as a biomarker (HR = 2.125). While ING1 increased or decreased the expression of different cytokines, ING1 also increased the levels of MMP1, MMP3 and MMP10 by 5-8 fold, and concomitantly decreased levels of the tissue inhibitors of metalloproteases TIMP2, TIMP3 and TIMP4 by 1.5-3.3 fold, resulting in significant increases in MMP activity as determined by zymography. Co-culturing of MCF7 cells with stromal cells expressing ING1 in 3-dimensional organoid cultures suggested that MCF7 colonies were less well defined, suggesting that secreted MMPs might promote migration. CONCLUSION These data indicate that stromal ING1 expression can predict the survival of patients with luminal breast cancer. High levels of ING1 in stromal cells can promote the development of breast cancer through increased expression and release of MMPs and down regulation of TIMPs, which may be an underlying mechanism of reduced patient survival.
Collapse
Affiliation(s)
- Satbir Thakur
- Department of Biochemistry and Molecular Biology, University of Calgary, 3330 Hospital Drive, Calgary, T2N 4N1, AB, Canada
| | - Arash Nabbi
- Department of Medical Science, University of Calgary, 3330 Hospital Drive, Calgary, T2N 4N1, AB, Canada
| | - Alexander Klimowicz
- Functional Tissue Imaging Unit, Translational Research Laboratory, Tom Baker Cancer Center, 1331- 29 street NW, Calgary, T2N 4N2, AB, Canada.,Present Address: Boehringer Ingelheim Pharmaceuticals Inc, 900 Ridgebury Road, Ridgefield, CT, 06877, USA
| | - Karl Riabowol
- Department of Biochemistry and Molecular Biology, University of Calgary, 3330 Hospital Drive, Calgary, T2N 4N1, AB, Canada. .,Department of Oncology, University of Calgary, 311 HMRB, 3330 Hospital Drive, NW, Calgary, T2N 4N1, AB, Canada.
| |
Collapse
|
18
|
Jia AI, Lv Y, Guo X, Ren LI, Qin J. Ectopic expression of p33 ING1b suppresses proliferation and induces apoptosis in colonic adenocarcinoma cells. Oncol Lett 2015; 10:1517-1522. [PMID: 26622701 DOI: 10.3892/ol.2015.3385] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2014] [Accepted: 05/12/2015] [Indexed: 12/12/2022] Open
Abstract
Inhibitor of growth 1b (ING1b) is considered to be a class II tumor suppressor gene. Although decreased expression of p33ING1b has previously been reported in colorectal cancer (CRC), its role in CRC has remained to be elucidated. The present study was designed to assess the function of p33ING1b in CRC and to further evaluate its underlying mechanisms of action. Western blot analysis confirmed that ING1b gene expression was significantly decreased in CRC tissues compared with that of adjacent non-tumorous colorectal tissues. Furthermore, recombinant adenovirus-mediated ectopic expression of p33ING1b resulted in growth inhibition, G1-phase cell cycle arrest and apoptosis in the SW480, HT29 and LoVo colorectal adenocarcinoma cell lines. The results suggested that the downregulation of ING1b contributes to colorectal carcinogenesis and that ectopic expression of ING1b may be a potentially useful therapeutic approach for CRC.
Collapse
Affiliation(s)
- A I Jia
- Department of Gastroenterology, The First Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Yifei Lv
- Department of Gastroenterology, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi 710068, P.R. China
| | - Xueyan Guo
- Department of Gastroenterology, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi 710068, P.R. China
| | - L I Ren
- Department of Gastroenterology, The First Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Jie Qin
- Department of Gastroenterology, The First Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| |
Collapse
|
19
|
Llanos AA, Dumitrescu RG, Brasky TM, Liu Z, Mason JB, Marian C, Makambi KH, Spear SL, Kallakury BVS, Freudenheim JL, Shields PG. Relationships among folate, alcohol consumption, gene variants in one-carbon metabolism and p16INK4a methylation and expression in healthy breast tissues. Carcinogenesis 2014; 36:60-7. [PMID: 25344837 DOI: 10.1093/carcin/bgu219] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
p16(INK4a) is a tumor suppressor gene, frequently hypermethylated in breast cancer; this epigenetic silencing of p16(INK4a) occurs early in carcinogenesis. The risk factors and functional consequences of p16(INK4a) methylation are unknown. Alcohol consumption, a breast cancer risk factor, impedes folate metabolism and may thereby alter gene methylation since folate plays a pivotal role in DNA methylation. In a cross-sectional study of 138 women with no history of breast cancer who underwent reduction mammoplasty, we studied breast cancer risk factors, plasma and breast folate concentrations, variation in one-carbon metabolism genes, p16(INK4a) promoter methylation and P16 protein expression. Logistic regression was used to estimate multivariable-adjusted odds ratios (OR) and 95% confidence intervals (CI). p16(INK4a) methylation was negatively correlated with P16 expression (r = -0.28; P = 0.002). Alcohol consumption was associated with lower breast folate (P = 0.03), higher p16(INK4a) promoter methylation (P = 0.007) and less P16 expression (P = 0.002). Higher breast folate concentrations were associated with lower p16(INK4a) promoter methylation (P = 0.06). Genetic variation in MTRR (rs1801394) and MTHFD1 (rs1950902) was associated with higher p16 (INK4a) promoter methylation (OR = 2.66, 95% CI: 1.11-6.42 and OR = 2.72, 95% CI: 1.12-6.66, respectively), whereas variation in TYMS (rs502396) was associated with less P16 protein expression (OR = 0.22, 95% CI: 0.05-0.99). Given that this is the first study to indicate that alcohol consumption, breast folate and variation in one-carbon metabolism genes are associated with p16(INK4a) promoter methylation and P16 protein expression in healthy tissues; these findings require replication.
Collapse
Affiliation(s)
- Adana A Llanos
- Division of Population Sciences, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43201, USA, Department of Epidemiology, RBHS-School of Public Health and Rutgers Cancer Institute of New Jersey, Rutgers University, New Brunswick, NJ 08903, USA
| | - Ramona G Dumitrescu
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC 20057, USA, Department of Medical Genetics and Epidemiology, Basic Sciences Program, Saba University School of Medicine, Saba, Dutch Caribbean, The Netherlands
| | - Theodore M Brasky
- Division of Cancer Prevention and Control, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Zhenhua Liu
- Human Nutrition Research Center, Tufts University, Boston, MA 02111, USA, Department of Nutrition, School of Public Health and Health Sciences, University of Massachusetts, Amherst, MA 01003, USA
| | - Joel B Mason
- Human Nutrition Research Center, Tufts University, Boston, MA 02111, USA
| | - Catalin Marian
- Division of Cancer Prevention and Control, College of Medicine, The Ohio State University, Columbus, OH 43210, USA, Department of Biochemistry and Pharmacology, University of Medicine and Pharmacy Timisoara, Timisoara, Romania
| | - Kepher H Makambi
- Department of Biostatistics, Bioinformatics and Biomathematics, Georgetown University, Washington, DC 20057, USA
| | - Scott L Spear
- Department of Plastic Surgery, Georgetown University, Washington, DC 20057, USA
| | | | - Jo L Freudenheim
- Department of Epidemiology and Environmental Health, School of Public Health and Health Professions, University at Buffalo, Buffalo, NY 14214, USA
| | - Peter G Shields
- Division of Cancer Prevention and Control, College of Medicine, The Ohio State University, Columbus, OH 43210, USA,
| |
Collapse
|
20
|
Rotte A, Li G, Bhandaru M. Tumor suppressor Ing1b facilitates DNA repair and prevents oxidative stress induced cell death. Apoptosis 2014; 19:518-26. [PMID: 24242916 DOI: 10.1007/s10495-013-0940-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Inhibitor of growth (ING) family of proteins are known to coordinate with histone acetyltransferases and regulate the key events of cell cycle and DNA repair. Previous work from our lab showed that Ing1b regulated the nucleotide excision repair by facilitating histone acetylation and subsequent chromatin relaxation. Further, it was also shown that Ing1b protected the cells from genomic instability induced cell death by promoting ubiquitination of proliferating cell nuclear antigen (PCNA). In the present study we explored the role of Ing1b in the repair of oxidized DNA and prevention of oxidative stress induced genotoxic cell death. Using HCT116 cells we show that Ing1b protein expression is induced by treatment with H2O2. Ing1b lacking cells showed decreased ability to repair the oxidized DNA. PCNA monoubiquitination, a critical event of DNA repair was blunted in Ing1b knock down cells and augmented in Ing1b over expressing cells. Moreover, oxidative stress induced cell death was higher in cells lacking Ing1b whereas it was lower in Ing1b over expressing cells. Finally we show that inhibition of histone deacetylases, rescued the Ing1b knock down cells from cytotoxic effects of H2O2 treatment.
Collapse
Affiliation(s)
- Anand Rotte
- Department of Dermatology and Skin Science, University of British Columbia, Research Pavilion, 828 West, 10th Avenue, Vancouver, BC, V5Z 1L8, Canada,
| | | | | |
Collapse
|
21
|
Keep-ING balance: tumor suppression by epigenetic regulation. FEBS Lett 2014; 588:2728-42. [PMID: 24632289 DOI: 10.1016/j.febslet.2014.03.011] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2014] [Accepted: 03/06/2014] [Indexed: 12/26/2022]
Abstract
Cancer cells accumulate genetic and epigenetic changes that alter gene expression to drive tumorigenesis. Epigenetic silencing of tumor suppressor, cell cycle, differentiation and DNA repair genes contributes to neoplastic transformation. The ING (inhibitor of growth) proteins (ING1-ING5) have emerged as a versatile family of growth regulators, phospholipid effectors, histone mark sensors and core components of HDAC1/2 - and several HAT chromatin-modifying complexes. This review will describe the characteristic pathways by which ING family proteins differentially affect the Hallmarks of Cancer and highlight the various epigenetic mechanisms by which they regulate gene expression. Finally, we will discuss their potentials as biomarkers and therapeutic targets in epigenetic treatment strategies.
Collapse
|
22
|
Guérillon C, Larrieu D, Pedeux R. ING1 and ING2: multifaceted tumor suppressor genes. Cell Mol Life Sci 2013; 70:3753-72. [PMID: 23412501 PMCID: PMC11113716 DOI: 10.1007/s00018-013-1270-z] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2012] [Revised: 01/14/2013] [Accepted: 01/17/2013] [Indexed: 01/27/2023]
Abstract
Inhibitor of Growth 1 (ING1) was identified and characterized as a "candidate" tumor suppressor gene in 1996. Subsequently, four more genes, also characterized as "candidate" tumor suppressor genes, were identified by homology search: ING2, ING3, ING4, and ING5. The ING proteins are characterized by a high homology in their C-terminal domain, which contains a Nuclear Localization Sequence and a Plant HomeoDomain (PHD), which has a high affinity to Histone 3 tri-methylated on lysine 4 (H3K4Me3). The ING proteins have been involved in the control of cell growth, senescence, apoptosis, chromatin remodeling, and DNA repair. Within the ING family, ING1 and ING2 form a subgroup since they are evolutionarily and functionally close. In yeast, only one gene, Pho23, is related to ING1 and ING2 and possesses also a PHD. Recently, the ING1 and ING2 tumor suppressor status has been fully established since several studies have described the loss of ING1 and ING2 protein expression in human tumors and both ING1 and ING2 knockout mice were reported to have spontaneously developed tumors, B cell lymphomas, and soft tissue sarcomas, respectively. In this review, we will describe for the first time what is known about the ING1 and ING2 genes, proteins, their regulations in both human and mice, and their status in human tumors. Furthermore, we explore the current knowledge about identified functions involving ING1 and ING2 in tumor suppression pathways especially in the control of cell cycle and in genome stability.
Collapse
Affiliation(s)
- Claire Guérillon
- INSERM U917, Faculté de Médecine de Rennes, Microenvironnement et Cancer, Building 2, Room 117, 2 avenue du Professeur Léon Bernard, 35043 Rennes, France
- Université de Rennes 1, Rennes, France
| | - Delphine Larrieu
- The Wellcome Trust and Cancer Research UK Gurdon Institute, University of Cambridge, Tennis Court Road, Cambridge, CB2 1QN UK
| | - Rémy Pedeux
- INSERM U917, Faculté de Médecine de Rennes, Microenvironnement et Cancer, Building 2, Room 117, 2 avenue du Professeur Léon Bernard, 35043 Rennes, France
- Université de Rennes 1, Rennes, France
- Etablissement Français du Sang, Rennes, France
| |
Collapse
|
23
|
Jin J, Iakova P, Breaux M, Sullivan E, Jawanmardi N, Chen D, Jiang Y, Medrano EM, Timchenko NA. Increased expression of enzymes of triglyceride synthesis is essential for the development of hepatic steatosis. Cell Rep 2013; 3:831-43. [PMID: 23499441 DOI: 10.1016/j.celrep.2013.02.009] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2012] [Revised: 12/12/2012] [Accepted: 02/06/2013] [Indexed: 12/14/2022] Open
Abstract
Molecular mechanisms underpinning nonalcoholic fatty liver disease (NAFLD) are not well understood. The earliest step of NAFLD is hepatic steatosis, which is one of the main characteristics of aging liver. Here, we present a molecular scenario of age-related liver steatosis. We show that C/EBPα-S193D knockin mice have age-associated epigenetic changes and develop hepatic steatosis at 2 months of age. The underlying mechanism of the hepatic steatosis in old wild-type (WT) mice and in young S193D mice includes increased amounts of tripartite p300-C/EBPα/β complexes that activate promoters of five genes that drive triglyceride synthesis. Knockdown of p300 in old WT mice inhibits hepatic steatosis. Indeed, transgenic mice expressing dominant-negative p300 have fewer C/EBPα/β-p300 complexes and do not develop age-dependent hepatic steatosis. Notably, the p300-C/EBPα/β pathway is activated in the livers of patients with NAFLD. Thus, our results show that p300 and C/EBP proteins are essential participants in hepatic steatosis.
Collapse
Affiliation(s)
- Jingling Jin
- Huffington Center on Aging and Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX 77030, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
24
|
The ING1a tumor suppressor regulates endocytosis to induce cellular senescence via the Rb-E2F pathway. PLoS Biol 2013; 11:e1001502. [PMID: 23472054 PMCID: PMC3589274 DOI: 10.1371/journal.pbio.1001502] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2012] [Accepted: 01/24/2013] [Indexed: 02/07/2023] Open
Abstract
An age-associated isoform of ING1, ING1a, induces cell senescence by altering endocytosis, subsequently activating the retinoblastoma tumor suppressor. The INhibitor of Growth (ING) proteins act as type II tumor suppressors and epigenetic regulators, being stoichiometric members of histone acetyltransferase and histone deacetylase complexes. Expression of the alternatively spliced ING1a tumor suppressor increases >10-fold during replicative senescence. ING1a overexpression inhibits growth; induces a large flattened cell morphology and the expression of senescence-associated β-galactosidase; increases Rb, p16, and cyclin D1 levels; and results in the accumulation of senescence-associated heterochromatic foci. Here we identify ING1a-regulated genes and find that ING1a induces the expression of a disproportionate number of genes whose products encode proteins involved in endocytosis. Intersectin 2 (ITSN2) is most affected by ING1a, being rapidly induced >25-fold. Overexpression of ITSN2 independently induces expression of the p16 and p57KIP2 cyclin-dependent kinase inhibitors, which act to block Rb inactivation, acting as downstream effectors of ING1a. ITSN2 is also induced in normally senescing cells, consistent with elevated levels of ING1a inducing ITSN2 as part of a normal senescence program. Inhibition of endocytosis or altering the stoichiometry of endosome components such as Rab family members similarly induces senescence. Knockdown of ITSN2 also blocks the ability of ING1a to induce a senescent phenotype, confirming that ITSN2 is a major transducer of ING1a-induced senescence signaling. These data identify a pathway by which ING1a induces senescence and indicate that altered endocytosis activates the Rb pathway, subsequently effecting a senescent phenotype. Alternative splicing of several genes including the p16 and p53 tumor suppressors has been reported to increase during replicative senescence of normal diploid cells, but the biological functions of most alternative transcripts are unknown. We have found that a splicing product of the ING1 epigenetic regulator, ING1a, also increases during senescence; moreover, forced expression of ING1a at these levels in otherwise growth-competent cells can induce senescence. In this study we have determined that a major mechanism by which ING1a induces senescence is through inhibiting endocytosis; this subsequently activates the retinoblastoma (Rb) tumor suppressor pathway by increasing Rb levels and preventing its inactivation through multiple mechanisms. Our study also establishes a link between endocytosis and oxidative stress and suggests that multiple mechanisms that induce cellular senescence may do so by inhibiting normal endocytic processes, thereby affecting normal signal transduction pathways including those mitogenic pathways required for cell growth.
Collapse
|
25
|
Lu W, Zhang Y, Liu D, Songyang Z, Wan M. Telomeres-structure, function, and regulation. Exp Cell Res 2012; 319:133-41. [PMID: 23006819 DOI: 10.1016/j.yexcr.2012.09.005] [Citation(s) in RCA: 170] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2012] [Accepted: 09/13/2012] [Indexed: 12/15/2022]
Abstract
In mammals, maintenance of the linear chromosome ends (or telomeres) involves faithful replication of genetic materials and protection against DNA damage signals, to ensure genome stability and integrity. These tasks are carried out by the telomerase holoenzyme and a unique nucleoprotein structure in which an array of telomere-associated proteins bind to telomeric DNA to form special protein/DNA complexes. The telomerase complex, which is comprised of telomeric reverse transcriptase (TERT), telomeric RNA component (TERC), and other assistant factors, is responsible for adding telomeric repeats to the ends of chromosomes. Without proper telomere maintenance, telomere length will shorten with successive round of DNA replication due to the so-called end replication problem. Aberrant regulation of telomeric proteins and/or telomerase may lead to abnormalities that can result in diseases such as dyskeratosis congenita (DC) and cancers. Understanding the mechanisms that regulate telomere homeostasis and the factors that contribute to telomere dysfunction should aid us in developing diagnostic and therapeutic tools for these diseases.
Collapse
Affiliation(s)
- Weisi Lu
- State Key Laboratory for Biocontrol, SYSU, Guangzhou, PR China
| | | | | | | | | |
Collapse
|