201
|
Furusawa Y, Yamanouchi Y, Iizumi T, Zhao QL, Mitsuhashi Y, Morita A, Enomoto A, Tabuchi Y, Kondo T. Checkpoint kinase 2 is dispensable for regulation of the p53 response but is required for G 2/M arrest and cell survival in cells with p53 defects under heat stress. Apoptosis 2018; 22:1225-1234. [PMID: 28733865 DOI: 10.1007/s10495-017-1402-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Hyperthermia induced by heat stress (HS) is known to inhibit proliferation and induce cell death in cancer. We previously demonstrated that checkpoint kinase 1 (Chk1) contributes to G2/M arrest and cell survival under HS; however, the role of Chk2, a functional analog of Chk1, in regulation of the cell cycle and cell death under HS is still unknown. Here, we addressed the role of Chk2 using Molt-4 cells with p53-targeted shRNA (Molt-4/shp53) and parental control cells (Molt-4/V). Chk2 inhibition suppressed C-terminal acetylation of p53 and delayed the induction of p53-target genes in Molt-4/V cells under HS; however, Chk2 inhibition failed to inhibit apoptosis induced by HS, indicating that Chk2 was dispensable for p53-dependent apoptosis under HS. In contrast, Chk2 inhibition abrogated G2/M arrest and promoted cell death induced by HS in HeLa cells and Molt-4/shp53 cells. Thus, we demonstrated for the first time that Chk2 was required for cell cycle arrest and cell survival, particularly in cells with p53 defects under HS. These findings indicated that Chk2 may be a selective target for p53-mutated or -deficient cancer treated with hyperthermia.
Collapse
Affiliation(s)
- Yukihiro Furusawa
- Department of Liberal Arts and Sciences, Toyama Prefectural University, Kurokawa 5180, Toyama, 939-0398, Japan. .,Department of Radiological Sciences, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan.
| | - Yuka Yamanouchi
- Department of Liberal Arts and Sciences, Toyama Prefectural University, Kurokawa 5180, Toyama, 939-0398, Japan
| | - Takashi Iizumi
- Department of Radiological Sciences, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan.,Proton Beam Therapy Center, Tsukuba University Hospital Radiation Oncology, Tsukuba, Japan
| | - Qing-Li Zhao
- Department of Radiological Sciences, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan
| | - Yohei Mitsuhashi
- Department of Radiological Sciences, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan
| | - Akinori Morita
- Department of Radiological Science, Institute of Health Biosciences, The University of Tokushima Graduate School, Tokushima, Japan
| | - Atushi Enomoto
- Laboratory of Molecular Radiology, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| | - Yoshiaki Tabuchi
- Division of Molecular Genetic Research, Life Science Research Center, University of Toyama, Toyama, Japan
| | - Takashi Kondo
- Department of Radiological Sciences, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan
| |
Collapse
|
202
|
Sharma VK, Raimondi V, Ruggero K, Pise-Masison CA, Cavallari I, Silic-Benussi M, Ciminale V, D'Agostino DM. Expression of miR-34a in T-Cells Infected by Human T-Lymphotropic Virus 1. Front Microbiol 2018; 9:832. [PMID: 29780367 PMCID: PMC5945834 DOI: 10.3389/fmicb.2018.00832] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Accepted: 04/12/2018] [Indexed: 12/19/2022] Open
Abstract
Human T-lymphotropic virus 1 (HTLV-1) immortalizes T-cells and is the causative agent of adult T-cell leukemia/lymphoma (ATLL). HTLV-1 replication and transformation are governed by multiple interactions between viral regulatory proteins and host cell factors that remain to be fully elucidated. The present study investigated the impact of HTLV-1 infection on the expression of miR-34a, a microRNA whose expression is downregulated in many types of cancer. Results of RT-PCR assays showed that five out of six HTLV-1-positive cell lines expressed higher levels of miR-34a compared to normal PBMC or purified CD4+ T-cells. ATLL cell line ED, which did not express miR-34a, showed methylation of the miR-34a promoter. Newly infected PBMC and samples from 10 ATLL patients also showed a prominent increase in miR-34a expression compared to PBMC controls. The primary miR-34a transcript expressed in infected cell line C91PL contained binding motifs for NF-κB and p53. Pharmacological inhibition of NF-κB with Bay 11-7082 indicated that this pathway contributes to sustain miR-34a levels in infected cells. Treatment of infected cell lines with the p53 activator nutlin-3a resulted in a further increase in miR-34a levels, thus confirming it as a transcriptional target of p53. Nutlin-3a-treated cells showed downregulation of known miR-34a targets including the deacetylase SIRT1, which was accompanied by increased acetylation of p53, a substrate of SIRT1. Transfection of C91PL cells with a miR-34a mimic also led to downregulation of mRNA targets including SIRT1 as well as the pro-apoptotic factor BAX. Unlike nutlin-3a, the miR-34a mimic did not cause cell cycle arrest or reduce cell viability. On the other hand, sequestration of miR-34a with a sponge construct resulted in an increase in death of C91PL cells. These findings provide evidence for a functional role for miR-34a in fine-tuning the expression of target genes that influence the turnover of HTLV-1-infected cells.
Collapse
Affiliation(s)
- Varun K Sharma
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy
| | | | - Katia Ruggero
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy
| | - Cynthia A Pise-Masison
- Animal Models and Retroviral Vaccines Section, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | | | | | - Vincenzo Ciminale
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy.,Veneto Institute of Oncology IOV - IRCCS, Padova, Italy
| | | |
Collapse
|
203
|
Berberine protects renal tubular cells against hypoxia/reoxygenation injury via the Sirt1/p53 pathway. J Nat Med 2018; 72:715-723. [DOI: 10.1007/s11418-018-1210-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2017] [Accepted: 03/25/2018] [Indexed: 10/17/2022]
|
204
|
Bondy-Chorney E, Denoncourt A, Sai Y, Downey M. Nonhistone targets of KAT2A and KAT2B implicated in cancer biology 1. Biochem Cell Biol 2018; 97:30-45. [PMID: 29671337 DOI: 10.1139/bcb-2017-0297] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Lysine acetylation is a critical post-translation modification that can impact a protein's localization, stability, and function. Originally thought to only occur on histones, we now know thousands of nonhistone proteins are also acetylated. In conjunction with many other proteins, lysine acetyltransferases (KATs) are incorporated into large protein complexes that carry out these modifications. In this review we focus on the contribution of two KATs, KAT2A and KAT2B, and their potential roles in the development and progression of cancer. Systems biology demands that we take a broad look at protein function rather than focusing on individual pathways or targets. As such, in this review we examine KAT2A/2B-directed nonhistone protein acetylations in cancer in the context of the 10 "Hallmarks of Cancer", as defined by Hanahan and Weinberg. By focusing on specific examples of KAT2A/2B-directed acetylations with well-defined mechanisms or strong links to a cancer phenotype, we aim to reinforce the complex role that these enzymes play in cancer biology.
Collapse
Affiliation(s)
- Emma Bondy-Chorney
- Department of Cellular and Molecular Medicine and Ottawa Institute of Systems Biology, 451 Smyth Rd., Ottawa, ON KIH 8M5, Canada.,Department of Cellular and Molecular Medicine and Ottawa Institute of Systems Biology, 451 Smyth Rd., Ottawa, ON KIH 8M5, Canada
| | - Alix Denoncourt
- Department of Cellular and Molecular Medicine and Ottawa Institute of Systems Biology, 451 Smyth Rd., Ottawa, ON KIH 8M5, Canada.,Department of Cellular and Molecular Medicine and Ottawa Institute of Systems Biology, 451 Smyth Rd., Ottawa, ON KIH 8M5, Canada
| | - Yuka Sai
- Department of Cellular and Molecular Medicine and Ottawa Institute of Systems Biology, 451 Smyth Rd., Ottawa, ON KIH 8M5, Canada.,Department of Cellular and Molecular Medicine and Ottawa Institute of Systems Biology, 451 Smyth Rd., Ottawa, ON KIH 8M5, Canada
| | - Michael Downey
- Department of Cellular and Molecular Medicine and Ottawa Institute of Systems Biology, 451 Smyth Rd., Ottawa, ON KIH 8M5, Canada.,Department of Cellular and Molecular Medicine and Ottawa Institute of Systems Biology, 451 Smyth Rd., Ottawa, ON KIH 8M5, Canada
| |
Collapse
|
205
|
Dutto I, Scalera C, Prosperi E. CREBBP and p300 lysine acetyl transferases in the DNA damage response. Cell Mol Life Sci 2018; 75:1325-1338. [PMID: 29170789 PMCID: PMC11105205 DOI: 10.1007/s00018-017-2717-4] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2017] [Revised: 11/16/2017] [Accepted: 11/20/2017] [Indexed: 12/21/2022]
Abstract
The CREB-binding protein (CREBBP, or in short CBP) and p300 are lysine (K) acetyl transferases (KAT) belonging to the KAT3 family of proteins known to modify histones, as well as non-histone proteins, thereby regulating chromatin accessibility and transcription. Previous studies have indicated a tumor suppressor function for these enzymes. Recently, they have been found to acetylate key factors involved in DNA replication, and in different DNA repair processes, such as base excision repair, nucleotide excision repair, and non-homologous end joining. The growing list of CBP/p300 substrates now includes factors involved in DNA damage signaling, and in other pathways of the DNA damage response (DDR). This review will focus on the role of CBP and p300 in the acetylation of DDR proteins, and will discuss how this post-translational modification influences their functions at different levels, including catalytic activity, DNA binding, nuclear localization, and protein turnover. In addition, we will exemplify how these functions may be necessary to efficiently coordinate the spatio-temporal response to DNA damage. CBP and p300 may contribute to genome stability by fine-tuning the functions of DNA damage signaling and DNA repair factors, thereby expanding their role as tumor suppressors.
Collapse
Affiliation(s)
- Ilaria Dutto
- Istituto di Genetica Molecolare del CNR, Via Abbiategrasso 207, 27100, Pavia, Italy
- IRB, Carrer Baldiri Reixac 10, 08028, Barcelona, Spain
| | - Claudia Scalera
- Istituto di Genetica Molecolare del CNR, Via Abbiategrasso 207, 27100, Pavia, Italy
| | - Ennio Prosperi
- Istituto di Genetica Molecolare del CNR, Via Abbiategrasso 207, 27100, Pavia, Italy.
| |
Collapse
|
206
|
Wang L, Minchin RF, Butcher NJ. Arylamine N-acetyltransferase 1 protects against reactive oxygen species during glucose starvation: Role in the regulation of p53 stability. PLoS One 2018. [PMID: 29518119 PMCID: PMC5843258 DOI: 10.1371/journal.pone.0193560] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Human arylamine N-acetyltransferase 1 (NAT1) has been associated with cancer cell growth and invasion, but the underlying molecular mechanisms remain unknown. NAT1 is located on the short arm of chromosome 8 (8p21), a region that is commonly deleted in colon cancer. Previously, it was reported that HT-29 colon cancer cells, which have a large deletion at 8p21-22, show marked morphological changes, increased E-cadherin expression and altered cell-cell contact inhibition following down-regulation of NAT1 with shRNA. By contrast, no effects on growth were observed in HeLa cells. In the present study, cellular changes following knockout of NAT1 with CRISPR/Cas9 in HT-29 and HeLa cells were compared in the presence and absence of glucose. Cell growth decreased in both cell-lines during glucose starvation, but it was enhanced in HT-29 cells following NAT1 deletion. This was due to an increase in ROS production that induced cell apoptosis. Both ROS production and cell death were prevented by the glutathione precursor N-acetylcysteine. NAT1 knockout also resulted in a loss of the gain-of-function p53 protein in HT-29 cells. When p53 expression was inhibited with siRNA in parental HT-29 cells, ROS production and apoptosis increased to levels seen in the NAT1 knockout cells. The loss of p53 may explain the decreased colony formation and increased contact inhibition previously reported following NAT1 down-regulation in these cells. In conclusion, NAT1 is important in maintaining intracellular ROS, especially during glucose starvation, by stabilizing gain-of-function p53 in HT-29 cells. These results suggest that NAT1 may be a novel target to decrease intracellular gain-of -function p53.
Collapse
Affiliation(s)
- LiLi Wang
- Laboratory for Molecular and Cellular Pharmacology, School of Biomedical Sciences, University of Queensland, Brisbane, Australia
| | - Rodney F. Minchin
- Laboratory for Molecular and Cellular Pharmacology, School of Biomedical Sciences, University of Queensland, Brisbane, Australia
- * E-mail:
| | - Neville J. Butcher
- Laboratory for Molecular and Cellular Pharmacology, School of Biomedical Sciences, University of Queensland, Brisbane, Australia
| |
Collapse
|
207
|
SLAB51 Probiotic Formulation Activates SIRT1 Pathway Promoting Antioxidant and Neuroprotective Effects in an AD Mouse Model. Mol Neurobiol 2018; 55:7987-8000. [PMID: 29492848 PMCID: PMC6132798 DOI: 10.1007/s12035-018-0973-4] [Citation(s) in RCA: 180] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Accepted: 02/19/2018] [Indexed: 02/06/2023]
Abstract
The gut-brain axis is a bidirectional communication network functionally linking the gut and the central nervous system (CNS). Based on this, the rational manipulation of intestinal microbiota represents a novel attractive therapeutic strategy for the treatment of CNS-associated disorders. In this study, we explored the properties of a probiotic formulation (namely SLAB51) in counteracting brain oxidative damages associated with Alzheimer's disease (AD). Specifically, transgenic AD mice (3xTg-AD) were treated with SLAB51 and the effects on protein oxidation, neuronal antioxidant defence and repair systems were monitored, with the particular focus on the role of SIRT1-related pathways. We demonstrated that SLAB51 markedly reduced oxidative stress in AD mice brain by activating SIRT1-dependent mechanisms, thus representing a promising therapeutic adjuvant in AD treatment.
Collapse
|
208
|
Ghosh S, Wong SK, Jiang Z, Liu B, Wang Y, Hao Q, Gorbunova V, Liu X, Zhou Z. Haploinsufficiency of Trp53 dramatically extends the lifespan of Sirt6-deficient mice. eLife 2018; 7:32127. [PMID: 29474172 PMCID: PMC5825207 DOI: 10.7554/elife.32127] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Accepted: 02/01/2018] [Indexed: 12/13/2022] Open
Abstract
Mammalian sirtuin 6 (Sirt6) is a conserved NAD+-dependent deacylase and mono-ADP ribosylase that is known to be involved in DNA damage repair, metabolic homeostasis, inflammation, tumorigenesis, and aging. Loss of Sirt6 in mice results in accelerated aging and premature death within a month. Here, we show that haploinsufficiency (i.e., heterozygous deletion) of Trp53 dramatically extends the lifespan of both female and male Sirt6-deficient mice. Haploinsufficiency of Trp53 in Sirt6-deficient mice rescues several age-related phenotypes of Sirt6-deficient mice, including reduced body size and weight, lordokyphosis, colitis, premature senescence, apoptosis, and bone marrow stem cell decline. Mechanistically, SIRT6 deacetylates p53 at lysine 381 to negatively regulate the stability and activity of p53. These findings establish that elevated p53 activity contributes significantly to accelerated aging in Sirt6-deficient mice. Our study demonstrates that p53 is a substrate of SIRT6, and highlights the importance of SIRT6-p53 axis in the regulation of aging. Almost without exception, mammals age as they grow older. Older mammals are at greater risk of diseases like cancer, and have fewer stem cells that would otherwise help to keep their organs healthy. Some of the proteins that regulate and impact upon aging have been identified. One of these is an enzyme called SIRT6, which is thought to promote longevity. Mice without any SIRT6 suffer from severe premature aging, and rather than living up to two years like normal mice, SIRT6-deficient mice die within one month of their birth. The mutant mice also lose stem cells and exhibit signs of organ degeneration and body wasting. Another protein called p53 is well known for having the opposite effect to SIRT6: it accelerates aging and helps to prevent tumor growth. However, it was unclear if p53 is also involved in the processes that lead to the premature death of mice without SIRT6. Now, Ghosh et al. report that mouse cells and tissues without SIRT6 have more p53 compared to control samples. Biochemical experiments showed that the SIRT6 and p53 proteins physically interact, and that SIRT6 could use its enzymatic activity to remove a chemical modification, called acetylation, from p53. Without this specific acetylation, p53 became less stable and its levels dropped. Consequently, p53 was stabilized in the SIRT6-deficient cells. When Ghosh et al. deleted one copy of the gene that codes for p53 in SIRT6-deficient mice, mutant mice that had before only lived for a month now lived for up to sixteen months. Additionally, the mice were healthier, showing fewer signs of aging: for example, they had more immune cells and stem cells, straighter spines, and showed less gut inflammation and less body wasting. These findings suggest that SIRT6 does indeed inhibit p53 to counteract the normal aging process. Future experiments may explore if this regulation also holds true in human cells. Detailed knowledge of these molecular interactions could also open up more research into therapies against cancer and aging.
Collapse
Affiliation(s)
- Shrestha Ghosh
- School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong.,Shenzhen Institute of Research and Innovation, The University of Hong Kong, Pok Fu Lam, Hong Kong
| | - Sheung Kin Wong
- School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong.,Shenzhen Institute of Research and Innovation, The University of Hong Kong, Pok Fu Lam, Hong Kong
| | - Zhixin Jiang
- School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong.,Shenzhen Institute of Research and Innovation, The University of Hong Kong, Pok Fu Lam, Hong Kong
| | - Baohua Liu
- School of Medicine, Shenzhen University, Shenzhen, China
| | - Yi Wang
- School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong
| | - Quan Hao
- School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong
| | - Vera Gorbunova
- Rochester Aging Research Center, University of Rochester, Rochester, United States
| | - Xinguang Liu
- Institute for Aging Research, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, Dongguan, China
| | - Zhongjun Zhou
- School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong.,Shenzhen Institute of Research and Innovation, The University of Hong Kong, Pok Fu Lam, Hong Kong
| |
Collapse
|
209
|
ID4 regulates transcriptional activity of wild type and mutant p53 via K373 acetylation. Oncotarget 2018; 8:2536-2549. [PMID: 27911860 PMCID: PMC5356822 DOI: 10.18632/oncotarget.13701] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Accepted: 11/21/2016] [Indexed: 12/12/2022] Open
Abstract
Given that mutated p53 (50% of all human cancers) is over-expressed in many cancers, restoration of mutant p53 to its wild type biological function has been sought after as cancer therapy. The conformational flexibility has allowed to restore the normal biological function of mutant p53 by short peptides and small molecule compounds. Recently, studies have focused on physiological mechanisms such as acetylation of lysine residues to rescue the wild type activity of mutant p53. Using p53 null prostate cancer cell line we show that ID4 dependent acetylation promotes mutant p53 DNA-binding capabilities to its wild type consensus sequence, thus regulating p53-dependent target genes leading to subsequent cell cycle arrest and apoptosis. Specifically, by using wild type, mutant (P223L, V274F, R175H, R273H), acetylation mimics (K320Q and K373Q) and non-acetylation mimics (K320R and K373R) of p53, we identify that ID4 promotes acetylation of K373 and to a lesser extent K320, in turn restoring p53-dependent biological activities. Together, our data provides a molecular understanding of ID4 dependent acetylation that suggests a strategy of enhancing p53 acetylation at sites K373 and K320 that may serve as a viable mechanism of physiological restoration of mutant p53 to its wild type biological function.
Collapse
|
210
|
Wu CE, Esfandiari A, Ho YH, Wang N, Mahdi AK, Aptullahoglu E, Lovat P, Lunec J. Targeting negative regulation of p53 by MDM2 and WIP1 as a therapeutic strategy in cutaneous melanoma. Br J Cancer 2018; 118:495-508. [PMID: 29235570 PMCID: PMC5830592 DOI: 10.1038/bjc.2017.433] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Revised: 11/07/2017] [Accepted: 11/08/2017] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND Cutaneous melanoma is the most serious skin malignancy and new therapeutic strategies are needed for advanced melanoma. TP53 mutations are rare in cutaneous melanoma and hence activation of wild-type p53 is a potential therapeutic strategy in cutaneous melanoma. Here, we investigated the WIP1 inhibitor, GSK2830371, and MDM2-p53 binding antagonists (nutlin-3, RG7388 and HDM201) alone and in combination treatment in cutaneous melanoma cell lines and explored the mechanistic basis of these responses in relation to the genotype and induced gene expression profile of the cells. METHODS A panel of three p53WT (A375, WM35 and C8161) and three p53MUT (WM164, WM35-R and CHL-1) melanoma cell lines were used. The effects of MDM2 and WIP1 inhibition were evaluated by growth inhibition and clonogenic assays, immunoblotting, qRT-PCR gene expression profiling and flow cytometry. RESULTS GSK2830371, at doses (⩽10 μM) that alone had no growth-inhibitory or cytotoxic effects on the cells, nevertheless significantly potentiated the growth-inhibitory and clonogenic cell killing effects of MDM2 inhibitors in p53WT but not p53MUT melanoma cells, indicating the potentiation worked in a p53-dependent manner. The siRNA-mediated knockdown of p53 provided further evidence to support the p53 dependence. GSK2830371 increased p53 stabilisation through Ser15 phosphorylation and consequent Lys382 acetylation, and decreased ubiquitination and proteasome-dependent degradation when it was combined with MDM2 inhibitors. These changes were at least partly ATM mediated, shown by reversal with the ATM inhibitor (KU55933). GSK2830371 enhanced the induction of p53 transcriptional target genes, cell cycle arrest and apoptosis. CONCLUSIONS GSK2830371, a WIP1 inhibitor, at doses with no growth-inhibitory activity alone, potentiated the growth-inhibitory and cytotoxic activity of MDM2 inhibitors by increasing phosphorylation, acetylation and stabilisation of p53 in cutaneous melanoma cells in a functional p53-dependent manner.
Collapse
Affiliation(s)
- Chiao-En Wu
- Northern Institute for Cancer Research, School of Medicine, Newcastle University, Paul O'Gorman Building, Framlington Place, Newcastle-upon-Tyne NE2 4HH, UK
- Division of Hematology-Oncology, Department of Internal Medicine, Chang Gung Memorial Hospital at Linkou, Chang Gung University College of Medicine, Taoyuan 333, Taiwan
| | - Arman Esfandiari
- Northern Institute for Cancer Research, School of Medicine, Newcastle University, Paul O'Gorman Building, Framlington Place, Newcastle-upon-Tyne NE2 4HH, UK
| | - Yi-Hsuan Ho
- Northern Institute for Cancer Research, School of Medicine, Newcastle University, Paul O'Gorman Building, Framlington Place, Newcastle-upon-Tyne NE2 4HH, UK
| | - Nan Wang
- Arraygen UK Ltd, Devonshire Building, Newcastle University, Newcastle-upon-Tyne NE1 7RU, UK
| | - Ahmed Khairallah Mahdi
- Northern Institute for Cancer Research, School of Medicine, Newcastle University, Paul O'Gorman Building, Framlington Place, Newcastle-upon-Tyne NE2 4HH, UK
- Department of Pathology and Forensic Medicine, College of Medicine, Al-Nahrain University, Baghdad 10006, Iraq
| | - Erhan Aptullahoglu
- Northern Institute for Cancer Research, School of Medicine, Newcastle University, Paul O'Gorman Building, Framlington Place, Newcastle-upon-Tyne NE2 4HH, UK
| | - Penny Lovat
- Dermatological Sciences, Institute of Cellular Medicine, Newcastle University, Newcastle-upon-Tyne NE2 4HH, UK
| | - John Lunec
- Northern Institute for Cancer Research, School of Medicine, Newcastle University, Paul O'Gorman Building, Framlington Place, Newcastle-upon-Tyne NE2 4HH, UK
| |
Collapse
|
211
|
Di Martile M, Del Bufalo D, Trisciuoglio D. The multifaceted role of lysine acetylation in cancer: prognostic biomarker and therapeutic target. Oncotarget 2018; 7:55789-55810. [PMID: 27322556 PMCID: PMC5342454 DOI: 10.18632/oncotarget.10048] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Accepted: 06/01/2016] [Indexed: 12/28/2022] Open
Abstract
Lysine acetylation is a post-translational modification that regulates gene transcription by targeting histones as well as a variety of transcription factors in the nucleus. Recently, several reports have demonstrated that numerous cytosolic proteins are also acetylated and that this modification, affecting protein activity, localization and stability has profound consequences on their cellular functions. Interestingly, most non-histone proteins targeted by acetylation are relevant for tumorigenesis. In this review, we will analyze the functional implications of lysine acetylation in different cellular compartments, and will examine our current understanding of lysine acetyltransferases family, highlighting the biological role and prognostic value of these enzymes and their substrates in cancer. The latter part of the article will address challenges and current status of molecules targeting lysine acetyltransferase enzymes in cancer therapy.
Collapse
Affiliation(s)
- Marta Di Martile
- Preclinical Models and New Therapeutic Agents Unit, Research, Advanced Diagnostics and Technological Innovation Department, Regina Elena National Cancer Institute, Rome, Italy
| | - Donatella Del Bufalo
- Preclinical Models and New Therapeutic Agents Unit, Research, Advanced Diagnostics and Technological Innovation Department, Regina Elena National Cancer Institute, Rome, Italy
| | - Daniela Trisciuoglio
- Preclinical Models and New Therapeutic Agents Unit, Research, Advanced Diagnostics and Technological Innovation Department, Regina Elena National Cancer Institute, Rome, Italy
| |
Collapse
|
212
|
Goldstein I, Hager GL. The Three Ds of Transcription Activation by Glucagon: Direct, Delayed, and Dynamic. Endocrinology 2018; 159:206-216. [PMID: 29077799 PMCID: PMC6283435 DOI: 10.1210/en.2017-00521] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Accepted: 10/20/2017] [Indexed: 12/13/2022]
Abstract
Upon lowered blood glucose occurring during fasting, glucagon is secreted from pancreatic islets, exerting various metabolic effects to normalize glucose levels. A considerable portion of these effects is mediated by glucagon-activated transcription factors (TFs) in liver. Glucagon directly activates several TFs via immediate cyclic adenosine monophosphate (cAMP)- and calcium-dependent signaling events. Among these TFs, cAMP response element-binding protein (CREB) is a major factor. CREB recruits histone-modifying enzymes and cooperates with other TFs on the chromatin template to increase the rate of gene transcription. In addition to direct signal transduction, the transcriptional effects of glucagon are also influenced by dynamic TF cross talk. Specifically, assisted loading of one TF by a companion TF leads to increased binding and activity. Lastly, transcriptional regulation by glucagon is also exerted by TF cascades by which a primary TF induces the gene expression of secondary TFs that bring about their activity a few hours after the initial glucagon signal. This mechanism of a delayed response may be instrumental in establishing the temporal organization of the fasting response by which distinct metabolic events separate early from prolonged fasting. In this mini-review, we summarize recent advances and critical discoveries in glucagon-dependent gene regulation with a focus on direct TF activation, dynamic TF cross talk, and TF cascades.
Collapse
Affiliation(s)
- Ido Goldstein
- Laboratory of Receptor Biology and Gene Expression, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
- Correspondence: Gordon L. Hager, PhD, Laboratory of Receptor Biology and Gene Expression, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Building 41, Room B602, Bethesda, Maryland 20892. E-mail: ; or Ido Goldstein, PhD, Laboratory of Receptor Biology and Gene Expression, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Building 41, Room B307, Bethesda, Maryland 20892. E-mail:
| | - Gordon L Hager
- Laboratory of Receptor Biology and Gene Expression, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
- Correspondence: Gordon L. Hager, PhD, Laboratory of Receptor Biology and Gene Expression, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Building 41, Room B602, Bethesda, Maryland 20892. E-mail: ; or Ido Goldstein, PhD, Laboratory of Receptor Biology and Gene Expression, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Building 41, Room B307, Bethesda, Maryland 20892. E-mail:
| |
Collapse
|
213
|
Reynolds RH, Petersen MH, Willert CW, Heinrich M, Nymann N, Dall M, Treebak JT, Björkqvist M, Silahtaroglu A, Hasholt L, Nørremølle A. Perturbations in the p53/miR-34a/SIRT1 pathway in the R6/2 Huntington's disease model. Mol Cell Neurosci 2017; 88:118-129. [PMID: 29289683 DOI: 10.1016/j.mcn.2017.12.009] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Revised: 12/21/2017] [Accepted: 12/27/2017] [Indexed: 12/26/2022] Open
Abstract
The three factors, p53, the microRNA-34 family and Sirtuin 1 (SIRT1), interact in a positive feedback loop involved in cell cycle progression, cellular senescence and apoptosis. Each factor in this triad has roles in metabolic regulation, maintenance of mitochondrial function, and regulation of brain-derived neurotrophic factor (BDNF). Thus, this regulatory network holds potential importance for the pathophysiology of Huntington's disease (HD), an inherited neurodegenerative disorder in which both mitochondrial dysfunction and impaired neurotrophic signalling are observed. We investigated expression of the three members of this regulatory triad in the R6/2 HD mouse model. Compared to wild-type littermates, we found decreased levels of miR-34a-5p, increased SIRT1 mRNA and protein levels, and increased levels of p53 protein in brain tissue from R6/2 mice. The upregulation of SIRT1 did not appear to lead to an increased activity of the enzyme, as based on measures of p53 acetylation. In other words, the observed changes did not reflect the known interactions between these factors, indicating a general perturbation of the p53, miR-34a and SIRT1 pathway in HD. This is the first study investigating the entire triad during disease progression in an HD model. Given the importance of these three factors alone and within the triad, our results indicate that outside factors are regulating - or dysregulating - this pathway in HD.
Collapse
Affiliation(s)
| | - Maria Hvidberg Petersen
- Department of Cellular and Molecular Medicine, University of Copenhagen, DK-2200 Copenhagen N, Denmark
| | - Cecilie Wennemoes Willert
- Department of Cellular and Molecular Medicine, University of Copenhagen, DK-2200 Copenhagen N, Denmark
| | - Marie Heinrich
- Department of Cellular and Molecular Medicine, University of Copenhagen, DK-2200 Copenhagen N, Denmark
| | - Nynne Nymann
- Department of Cellular and Molecular Medicine, University of Copenhagen, DK-2200 Copenhagen N, Denmark
| | - Morten Dall
- Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Integrative Physiology, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2200 Copenhagen N, Denmark
| | - Jonas T Treebak
- Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Integrative Physiology, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2200 Copenhagen N, Denmark
| | - Maria Björkqvist
- Brain Disease Biomarker Unit, Department of Experimental Medical Science, Wallenberg Neuroscience Centre, Lund University, 221 84 Lund, Sweden
| | - Asli Silahtaroglu
- Department of Cellular and Molecular Medicine, University of Copenhagen, DK-2200 Copenhagen N, Denmark
| | - Lis Hasholt
- Department of Cellular and Molecular Medicine, University of Copenhagen, DK-2200 Copenhagen N, Denmark
| | - Anne Nørremølle
- Department of Cellular and Molecular Medicine, University of Copenhagen, DK-2200 Copenhagen N, Denmark.
| |
Collapse
|
214
|
SIRT1 inhibition impairs non-homologous end joining DNA damage repair by increasing Ku70 acetylation in chronic myeloid leukemia cells. Oncotarget 2017; 7:13538-50. [PMID: 26646449 PMCID: PMC4924659 DOI: 10.18632/oncotarget.6455] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Accepted: 10/14/2015] [Indexed: 12/20/2022] Open
Abstract
Most chemotherapeutic agents for leukemia are DNA damaging agents. However, DNA lesions can be repaired by activities of DNA repair systems. Increasing evidence have shown that enhanced DNA damage repair capacity contributes to chemotherapy resistance in leukemia cells. Thus, targeting DNA repair mechanisms is a promising strategy for novel leukemia treatment. SIRT1 expressions were downregulated by lentivirus-delivered SIRT1 shRNA in myeloid leukemia cells. SIRT1 mRNA and protein levels were analyzed by real-time PCR and Western blot, respectively. Flow cytometry was carried out to analyze cell cycle progression, apoptosis and DNA damage repair efficiency. DNA damage levels were assessed by alkaline comet assay, and H2AX phosphorylation was analyzed by immunoblotting and immunofluorescence. A mouse leukemia model was established by transplanting lentivirus-infected K562 cells containing SIRT1 shRNA into sublethally irradiated NOD/SCID mice, and tumorigenesis was evaluated by detecting tumor weights and mice survival. SIRT1 expressions were upregulated in myeloid leukemic patients. Downregulation of SIRT1 by RNAi promoted etoposide-induced DNA damage in myeloid leukemia cells accompanied by reduced NHEJ activity, and increased Ku70 acetylation. Furthermore, SIRT1 knockdown resulted in cell cycle arrest, induction of apoptosis and reduction of K562 cell proliferation accompanied by enhanced p53 and FOXO1 acetylation in K562 cells after etoposide treatment. Importantly, SIRT1 downregulation reduced the tumorigenesis ability of K562 cells in mouse xenografts following chemotherapy treatment. These results revealed that SIRT1 promotes the NHEJ repair pathway by deacetylating Ku70 in K562 cells, suggesting that SIRT1 is a novel therapeutic target for treating myeloid leukemia.
Collapse
|
215
|
DasGupta T, Nweze EI, Yue H, Wang L, Jin J, Ghosh SK, Kawsar HI, Zender C, Androphy EJ, Weinberg A, McCormick TS, Jin G. Human papillomavirus oncogenic E6 protein regulates human β-defensin 3 (hBD3) expression via the tumor suppressor protein p53. Oncotarget 2017; 7:27430-44. [PMID: 27034006 PMCID: PMC5053661 DOI: 10.18632/oncotarget.8443] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2015] [Accepted: 03/17/2016] [Indexed: 01/24/2023] Open
Abstract
Human β-defensin-3 (hBD3) is an epithelial cell-derived innate immune regulatory molecule overexpressed in oral dysplastic lesions and fosters a tumor-promoting microenvironment. Expression of hBD3 is induced by the epidermal growth factor receptor signaling pathway. Here we describe a novel pathway through which the high-risk human papillomavirus type-16 (HPV-16) oncoprotein E6 induces hBD3 expression in mucosal keratinocytes. Ablation of E6 by siRNA induces the tumor suppressor p53 and diminishes hBD3 in HPV-16 positive CaSki cervical cancer cells and UM-SCC-104 head and neck cancer cells. Malignant cells in HPV-16-associated oropharyngeal cancer overexpress hBD3. HPV-16 E6 induces hBD3 mRNA expression, peptide production and gene promoter activity in mucosal keratinocytes. Reduction of cellular levels of p53 stimulates hBD3 expression, while activation of p53 by doxorubicin inhibits its expression in primary oral keratinocytes and CaSki cells, suggesting that p53 represses hBD3 expression. A p53 binding site in the hBD3 gene promoter has been identified by using electrophoretic mobility shift assays and chromatin immunoprecipitation (ChIP). In addition, the p63 protein isoform ΔNp63α, but not TAp63, stimulated transactivation of the hBD3 gene and was co-expressed with hBD3 in head and neck cancer specimens. Therefore, high-risk HPV E6 oncoproteins may stimulate hBD3 expression in tumor cells to facilitate tumorigenesis of HPV-associated head and neck cancer.
Collapse
Affiliation(s)
- Twishasri DasGupta
- Department of Biological Sciences, Case Western Reserve University School of Dental Medicine, Cleveland, OH, USA
| | - Emeka I Nweze
- Department of Biological Sciences, Case Western Reserve University School of Dental Medicine, Cleveland, OH, USA.,Present Address: University of Nigeria, Nsukka, Nigera
| | - Hong Yue
- Department of Biological Sciences, Case Western Reserve University School of Dental Medicine, Cleveland, OH, USA
| | - Liming Wang
- Center for Molecular Cancer Diagnosis Inc., Twinsburg, OH, USA
| | - Jessica Jin
- Human Developmental and Regenerative Biology, Harvard University, Cambridge, MA, USA
| | - Santosh K Ghosh
- Department of Biological Sciences, Case Western Reserve University School of Dental Medicine, Cleveland, OH, USA
| | - Hameem I Kawsar
- Department of Biological Sciences, Case Western Reserve University School of Dental Medicine, Cleveland, OH, USA.,Present Address: St. Luke's Hospital, Chesterfield, MO, USA
| | - Chad Zender
- Department of Otolaryngology-Head & Neck Surgery, University Hospitals Case Medical Center, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Elliot J Androphy
- Department of Dermatology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Aaron Weinberg
- Department of Biological Sciences, Case Western Reserve University School of Dental Medicine, Cleveland, OH, USA
| | - Thomas S McCormick
- Department of Dermatology, University Hospitals Case Medical Center, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Ge Jin
- Department of Biological Sciences, Case Western Reserve University School of Dental Medicine, Cleveland, OH, USA
| |
Collapse
|
216
|
Wood M, Rymarchyk S, Zheng S, Cen Y. Trichostatin A inhibits deacetylation of histone H3 and p53 by SIRT6. Arch Biochem Biophys 2017; 638:8-17. [PMID: 29233643 DOI: 10.1016/j.abb.2017.12.009] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Revised: 11/15/2017] [Accepted: 12/08/2017] [Indexed: 02/07/2023]
Abstract
SIRT6 is an epigenetic modification enzyme that regulates gene transcription through its deacetylase activity. In addition to histone protein, SIRT6 also modify other proteins and enzymes, some of which are central players in metabolic reprogramming and aging process. Therefore, SIRT6 has emerged as a therapeutic target for the treatment of metabolic disorder and age-related diseases. Here, we report that SIRT6 deacetylates lysine 382 of p53 in short synthetic peptide sequence and in full length p53. Further studies showed that the deacetylation of H3K9Ac and p53K382Ac are insensitive to nicotinamide inhibition, but are sensitive to trichostatin A (TSA) inhibition. Detailed kinetic analysis revealed that TSA competes with the peptide substrate for inhibition, and this inhibition is unique to SIRT6 in the sirtuin family. Taken together, this study not only suggests potential roles of SIRT6 in regulating apoptosis and stress resistance via direct deacetylation of p53, but also provides lead compound for the development of potent and selective SIRT6 inhibitors.
Collapse
Affiliation(s)
- Marci Wood
- Department of Pharmaceutical Sciences, Albany College of Pharmacy and Health Sciences, 261 Mountain View Drive, Colchester, VT 05446, USA
| | - Stacia Rymarchyk
- Department of Pharmaceutical Sciences, Albany College of Pharmacy and Health Sciences, 261 Mountain View Drive, Colchester, VT 05446, USA
| | - Song Zheng
- Department of Pharmaceutical Sciences, Albany College of Pharmacy and Health Sciences, 261 Mountain View Drive, Colchester, VT 05446, USA
| | - Yana Cen
- Department of Pharmaceutical Sciences, Albany College of Pharmacy and Health Sciences, 261 Mountain View Drive, Colchester, VT 05446, USA.
| |
Collapse
|
217
|
HDAC6 deficiency induces apoptosis in mesenchymal stem cells through p53 K120 acetylation. Biochem Biophys Res Commun 2017; 494:51-56. [DOI: 10.1016/j.bbrc.2017.10.087] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Accepted: 10/16/2017] [Indexed: 01/05/2023]
|
218
|
Suzuki M, Ikeda A, Bartlett JD. Sirt1 overexpression suppresses fluoride-induced p53 acetylation to alleviate fluoride toxicity in ameloblasts responsible for enamel formation. Arch Toxicol 2017; 92:1283-1293. [PMID: 29185024 DOI: 10.1007/s00204-017-2135-2] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Accepted: 11/23/2017] [Indexed: 12/21/2022]
Abstract
Low-dose fluoride is an effective caries prophylactic, but high-dose fluoride is an environmental health hazard that causes skeletal and dental fluorosis. Treatments to prevent fluorosis and the molecular pathways responsive to fluoride exposure remain to be elucidated. Previously we showed that fluoride activates SIRT1 as an adaptive response to protect cells. Here, we demonstrate that fluoride induced p53 acetylation (Ac-p53) [Lys379], which is a SIRT1 deacetylation target, in ameloblast-derived LS8 cells in vitro and in enamel organ in vivo. Here we assessed SIRT1 function on fluoride-induced Ac-p53 formation using CRISPR/Cas9-mediated Sirt1 knockout (LS8Sirt/KO) cells or CRISPR/dCas9/SAM-mediated Sirt1 overexpressing (LS8Sirt1/over) cells. NaF (5 mM) induced Ac-p53 formation and increased cell cycle arrest via Cdkn1a/p21 expression in Wild-type (WT) cells. However, fluoride-induced Ac-p53 was suppressed by the SIRT1 activator resveratrol (50 µM). Without fluoride, Ac-p53 persisted in LS8Sirt/KO cells, whereas it decreased in LS8Sirt1/over. Fluoride-induced Ac-p53 formation was also suppressed in LS8Sirt1/over cells. Compared to WT cells, fluoride-induced Cdkn1a/p21 expression was elevated in LS8Sirt/KO and these cells were more susceptible to fluoride-induced growth inhibition. In contrast, LS8Sirt1/over cells were significantly more resistant. In addition, fluoride-induced cytochrome-c release and caspase-3 activation were suppressed in LS8Sirt1/over cells. Fluoride induced expression of the DNA double strand break marker γH2AX in WT cells and this was augmented in LS8Sirt1/KO cells, but was attenuated in LS8Sirt1/over cells. Our results suggest that SIRT1 deacetylates Ac-p53 to mitigate fluoride-induced cell growth inhibition, mitochondrial damage, DNA damage and apoptosis. This is the first report implicating Ac-p53 in fluoride toxicity.
Collapse
Affiliation(s)
- Maiko Suzuki
- Division of Biosciences, College of Dentistry, The Ohio State University, Columbus, OH, 43210, USA
| | - Atsushi Ikeda
- Division of Biosciences, College of Dentistry, The Ohio State University, Columbus, OH, 43210, USA
| | - John D Bartlett
- Division of Biosciences, College of Dentistry, The Ohio State University, Columbus, OH, 43210, USA.
| |
Collapse
|
219
|
DNA damage responses and p53 in the aging process. Blood 2017; 131:488-495. [PMID: 29141944 DOI: 10.1182/blood-2017-07-746396] [Citation(s) in RCA: 226] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Accepted: 09/01/2017] [Indexed: 12/18/2022] Open
Abstract
The genome is constantly attacked by genotoxic insults. DNA damage has long been established as a cause of cancer development through its mutagenic consequences. Conversely, radiation therapy and chemotherapy induce DNA damage to drive cells into apoptosis or senescence as outcomes of the DNA damage response (DDR). More recently, DNA damage has been recognized as a causal factor for the aging process. The role of DNA damage in aging and age-related diseases is illustrated by numerous congenital progeroid syndromes that are caused by mutations in genome maintenance pathways. During the past 2 decades, understanding how DDR drives cancer development and contributes to the aging process has progressed rapidly. It turns out that the DDR factor p53 takes center stage during tumor development and also plays an important role in the aging process. Studies in metazoan models ranging from Caenorhabditis elegans to mammals have revealed cell-autonomous and systemic DDR mechanisms that orchestrate adaptive responses that augment maintenance of the aging organism amid gradually accumulating DNA damage.
Collapse
|
220
|
Shu Y, Ren L, Xie B, Liang Z, Chen J. MiR-204 enhances mitochondrial apoptosis in doxorubicin-treated prostate cancer cells by targeting SIRT1/p53 pathway. Oncotarget 2017; 8:97313-97322. [PMID: 29228612 PMCID: PMC5722564 DOI: 10.18632/oncotarget.21960] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Accepted: 09/24/2017] [Indexed: 12/24/2022] Open
Abstract
Chemotherapy is important for adjuvant treatment of prostate cancer. However, some cancer cells exhibited low sensitivity to chemotherapeutic agents. We are supposed to sensitize these prostate cancer cells to chemotherapeutic agents such as doxorubicin. Previous reports have suggested that microRNAs (miRNAs) regulate chemosensitivity in various cancers. In the present study, we observed that expression level of miR-204 was decreased in prostate cancer cell lines and patients’ tumors. Furthermore, we found that restore of miR-204 dramatically enhanced the cytotoxicity of doxorubicin (DOX) against prostate cancer cell lines C4-2 and LNCaP carrying wild type (WT) p53. Mechanically, miR-204 in prostate cancer cells targets SIRT1 which is a histone deacetylase, and thus decreasing deacetylation of p53. As the results, acetylated p53 induced by DOX upregulates the expression of Noxa and Puma followed by induction of mitochondrial apoptosis. These data demonstrate that restore of miR-204 in prostate cancer cells enhances the mitochondrial apoptosis induced by doxorubicin by targeting the SIRT1/p53 pathway.
Collapse
Affiliation(s)
- Yan Shu
- Department of Urology, Tongde Hospital of Zhejiang Province, Hangzhou 310012, China
| | - Ligang Ren
- Department of Urology, Tongde Hospital of Zhejiang Province, Hangzhou 310012, China
| | - Bo Xie
- Department of Urology, Tongde Hospital of Zhejiang Province, Hangzhou 310012, China
| | - Zhen Liang
- Department of Urology, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Jing Chen
- Department of Urology, Tongde Hospital of Zhejiang Province, Hangzhou 310012, China
| |
Collapse
|
221
|
Advanced developmental toxicity test method based on embryoid body’s area. Reprod Toxicol 2017; 72:74-85. [DOI: 10.1016/j.reprotox.2017.06.185] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Revised: 06/25/2017] [Accepted: 06/29/2017] [Indexed: 11/18/2022]
|
222
|
ZNF509S1 downregulates PUMA by inhibiting p53K382 acetylation and p53-DNA binding. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2017; 1860:962-972. [DOI: 10.1016/j.bbagrm.2017.07.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Revised: 06/20/2017] [Accepted: 07/26/2017] [Indexed: 11/21/2022]
|
223
|
p53 stability is regulated by diverse deubiquitinating enzymes. Biochim Biophys Acta Rev Cancer 2017; 1868:404-411. [PMID: 28801249 DOI: 10.1016/j.bbcan.2017.08.001] [Citation(s) in RCA: 78] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Revised: 07/21/2017] [Accepted: 08/05/2017] [Indexed: 02/01/2023]
Abstract
The tumor suppressor protein p53 has a variety of roles in responses to various stress signals. In such responses, p53 activates specific transcriptional targets that control cell cycle arrest, DNA repair, angiogenesis, autophagy, metabolism, migration, aging, senescence, and apoptosis. Since p53 has been identified as the most frequently altered gene in human cancers, regulation and stabilization of its normal functions are important. Stability of p53 is regulated by the ubiquitin-proteasome pathway (UPP). Furthermore, it is readjusted by deubiquitination via deubiquitinating enzymes (DUBs) that can eliminate ubiquitin from p53. Diverse DUBs directly or indirectly affect the ubiquitination of p53 and, consequently, regulate various cellular processes associated with p53. As maintenance of p53 is regulated by a variety of DUBs, the interaction of DUBs and p53 can affect diseases such as cancer. Currently, DUBs have a central role in our understanding of various cancers, and some have potential in the development of effective therapeutic strategies. This review summarizes the current knowledge of p53 and of the interconnection between p53 and DUBs.
Collapse
|
224
|
Lu R, Bosland M, Xia Y, Zhang YG, Kato I, Sun J. Presence of Salmonella AvrA in colorectal tumor and its precursor lesions in mouse intestine and human specimens. Oncotarget 2017; 8:55104-55115. [PMID: 28903406 PMCID: PMC5589645 DOI: 10.18632/oncotarget.19052] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Accepted: 06/26/2017] [Indexed: 12/22/2022] Open
Abstract
Evidence directly supporting an association between Salmonella infection and colorectal cancer in human subjects is sparse. It has been well recognized that Salmonella infection increases the risk of gallbladder cancer. AvrA, a bacterial protein from Salmonella enterica, plays a crucial role in establishing chronic infection. To our knowledge, the presence of the bacterial AvrA has never been studied in human samples. Here, we demonstrated the presence and cellular localization of AvrA in inflamed, colorectal tumor and its precursor lesions, using both animal experimental infection models and human clinical specimens. We performed a newly developed AvrA serological assay and to determine the presence of anti-Salmonella AvrA antibody in chronic infected mouse serum samples. Further, we tested the presence of AvrA gene in healthy human fecal samples, in order to advance etiological studies of Salmonella AvrA in human population. Our study suggests a potential role of this bacterial protein in human colorectal cancer. Moreover, our new serological assay may serve a useful tool to identify individuals at increased risk for colorectal cancer.
Collapse
Affiliation(s)
- Rong Lu
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Maarten Bosland
- Department of Pathology, University of Illinois at Chicago, Chicago, IL, USA
| | - Yinglin Xia
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA.,Division of Academic Internal Medicine and Geriatrics, Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Yong-Guo Zhang
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Ikuko Kato
- Department of Oncology and Pathology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Jun Sun
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| |
Collapse
|
225
|
Yuan Y, Zhang A, Qi J, Wang H, Liu X, Zhao M, Duan S, Huang Z, Zhang C, Wu L, Zhang B, Zhang A, Xing C. p53/Drp1-dependent mitochondrial fission mediates aldosterone-induced podocyte injury and mitochondrial dysfunction. Am J Physiol Renal Physiol 2017; 314:F798-F808. [PMID: 28659272 DOI: 10.1152/ajprenal.00055.2017] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Mitochondrial dysfunction is increasingly recognized as an important factor in glomerular diseases. Previous study has shown that mitochondrial fission contributed to mitochondrial dysfunction. However, the mechanism of mitochondrial fission on mitochondrial dysfunction in aldosterone-induced podocyte injury remains ambiguous. This study aimed to investigate the pathogenic effect of mitochondrial fission both in vivo and in vitro. In an animal model of aldosterone-induced nephropathy, inhibition of the mitochondrial fission protein dynamin-related protein 1 (Drp1) suppressed aldosterone-induced podocyte injury. In cultured podocytes, aldosterone dose dependently induced Drp1 expression. Knockdown of Drp1 inhibited aldosterone-induced mitochondrial fission, mitochondrial dysfunction, and podocyte apoptosis. Furthermore, aldosterone dose dependently induced p53 expression. Knockdown of p53 inhibited aldosterone-induced Drp1 expression, mitochondrial dysfunction, and podocyte apoptosis. These findings implicated that aldosterone induced mitochondrial dysfunction and podocyte injury mediated by p53/Drp1-dependent mitochondrial fission, which may provide opportunities for therapeutic intervention for podocyte injury.
Collapse
Affiliation(s)
- Yanggang Yuan
- Department of Nephrology, the First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University , Nanjing , China
| | - Aiqing Zhang
- Department of Pediatric Nephrology, the Second Affiliated Hospital of Nanjing Medical University, Nanjing Medical University , Nanjing , China
| | - Jia Qi
- Department of Pharmacy, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine , Shanghai , China
| | - Hui Wang
- Department of Nephrology, the First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University , Nanjing , China
| | - Xi Liu
- Department of Nephrology, the First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University , Nanjing , China
| | - Min Zhao
- Department of Nephrology, Nanjing Children's Hospital, Nanjing Medical University , Nanjing , China
| | - Suyan Duan
- Department of Nephrology, the First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University , Nanjing , China
| | - Zhimin Huang
- Department of Nephrology, the First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University , Nanjing , China
| | - Chengning Zhang
- Department of Nephrology, the First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University , Nanjing , China
| | - Lin Wu
- Department of Nephrology, the First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University , Nanjing , China
| | - Bo Zhang
- Department of Nephrology, the First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University , Nanjing , China
| | - Aihua Zhang
- Department of Nephrology, Nanjing Children's Hospital, Nanjing Medical University , Nanjing , China
| | - Changying Xing
- Department of Nephrology, the First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University , Nanjing , China
| |
Collapse
|
226
|
Cubillos-Rojas M, Schneider T, Bartrons R, Ventura F, Rosa JL. NEURL4 regulates the transcriptional activity of tumor suppressor protein p53 by modulating its oligomerization. Oncotarget 2017; 8:61824-61836. [PMID: 28977907 PMCID: PMC5617467 DOI: 10.18632/oncotarget.18699] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2016] [Accepted: 05/22/2017] [Indexed: 01/23/2023] Open
Abstract
p53 is a transcription factor that regulates important cellular processes related to tumor suppression, including induction of senescence, apoptosis, and DNA repair as well as the inhibition of angiogenesis and cell migration. Therefore, it is critical to understand the molecular mechanism that regulates it. p53 tetramerization is a key step in its activation process and the regulation of this oligomerization, an important control point. The E3 ubiquitin ligase HERC2 controls the p53 transcriptional activity by regulation of its oligomerization state. HERC2-interacting proteins such as the adaptor-like protein with six neuralized domains NEURL4 are also candidates to regulate p53 activity. Here, we demonstrate the existence of an interaction network between NEURL4, HERC2 and p53 proteins. We report a functional interaction between NEURL4 and p53, involving the C-terminal region of p53 and the neuralized domains 3 and 4 of NEURL4. Through this interaction, NEURL4 regulates the transcriptional activity of p53. Thus, NEURL4 depletion reduced the transcriptional activity whereas NEURL4 overexpression increased it. In both cases, p53 stability was not affected. Although NEURL4 may interact with p53 independently of the E3 ubiquitin ligase HERC2, we observed that both proteins are needed to regulate the transcriptional activity of p53. Clonogenic assays confirmed the functional relevance of this interaction observing a decrease in cell growth by NEURL4 overexpression correlated to the increase of cellular cycle inhibitor p21 by p53 activation. Under these conditions, NEURL4 activated p53 oligomerization. All these findings identify NEURL4 as a novel regulator of the p53’s signaling.
Collapse
Affiliation(s)
- Monica Cubillos-Rojas
- Departament de Ciències Fisiològiques, Campus de Bellvitge, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), Universitat de Barcelona, L'Hospitalet de Llobregat, Barcelona E-08907, Spain
| | - Taiane Schneider
- Departament de Ciències Fisiològiques, Campus de Bellvitge, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), Universitat de Barcelona, L'Hospitalet de Llobregat, Barcelona E-08907, Spain
| | - Ramon Bartrons
- Departament de Ciències Fisiològiques, Campus de Bellvitge, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), Universitat de Barcelona, L'Hospitalet de Llobregat, Barcelona E-08907, Spain
| | - Francesc Ventura
- Departament de Ciències Fisiològiques, Campus de Bellvitge, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), Universitat de Barcelona, L'Hospitalet de Llobregat, Barcelona E-08907, Spain
| | - Jose Luis Rosa
- Departament de Ciències Fisiològiques, Campus de Bellvitge, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), Universitat de Barcelona, L'Hospitalet de Llobregat, Barcelona E-08907, Spain
| |
Collapse
|
227
|
Chen H, Zhang W, Cheng X, Guo L, Xie S, Ma Y, Guo N, Shi M. β2-AR activation induces chemoresistance by modulating p53 acetylation through upregulating Sirt1 in cervical cancer cells. Cancer Sci 2017; 108:1310-1317. [PMID: 28498637 PMCID: PMC5497720 DOI: 10.1111/cas.13275] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Revised: 04/27/2017] [Accepted: 04/30/2017] [Indexed: 01/12/2023] Open
Abstract
It has been suggested that β2‐adrenergic receptor (β2‐AR)‐mediated signaling induced by catecholamines regulates the degradation of p53. However, the underlying molecular mechanisms were not known. In the present study, we demonstrated that catecholamines upregulated the expression of silent information regulator 1 (Sirt1) through activating β2‐AR‐mediated signaling pathway, since selective β2‐AR antagonist ICI 118, 551 and non‐selective β‐blocker proprenolol effectively repressed isoproterenol (ISO)‐induced Sirt1 expression. Catecholamines inhibited doxorubicin (DOX)‐induced p53 acetylation and transcription‐activation activities by inducing the expression of Sirt1. Knockdown of the Sirt1 expression by the specific siRNA remarkably blocked the inhibitory effects of ISO on DOX‐induced p53 acetylation. In addition, we demonstrated that catecholamines induced resistance of cervical cancer cells to chemotherapeutics both in vitro and in vivo and that β2‐AR was overexpressed in cervical cancer tissues. Our data suggest that the p53‐dependent, chemotherapeutics‐induced cytotoxicity in cervical cancer cells may be compromised by catecholamines‐induced upregulation of the Sirt1 expression through activating the β2‐AR signaling.
Collapse
Affiliation(s)
- Hongyu Chen
- Institute of Basic Medical Sciences, Beijing, China
| | - Wei Zhang
- Laboratory of Cellular and Molecular Immunology, Medical School of Henan University, Kaifeng, China
| | - Xiang Cheng
- Institute of Basic Medical Sciences, Beijing, China
| | - Liang Guo
- Institute of Basic Medical Sciences, Beijing, China
| | - Shuai Xie
- Laboratory of Cellular and Molecular Immunology, Medical School of Henan University, Kaifeng, China
| | - Yuanfang Ma
- Laboratory of Cellular and Molecular Immunology, Medical School of Henan University, Kaifeng, China
| | - Ning Guo
- Institute of Basic Medical Sciences, Beijing, China
| | - Ming Shi
- Institute of Basic Medical Sciences, Beijing, China
| |
Collapse
|
228
|
SLC transporters as a novel class of tumour suppressors: identity, function and molecular mechanisms. Biochem J 2017; 473:1113-24. [PMID: 27118869 DOI: 10.1042/bj20150751] [Citation(s) in RCA: 82] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2015] [Accepted: 02/15/2016] [Indexed: 12/20/2022]
Abstract
The role of plasma membrane transporters in cancer is receiving increasing attention in recent years. Several transporters for essential nutrients are up-regulated in cancer and serve as tumour promoters. Transporters could also function as tumour suppressors. To date, four transporters belonging to the SLC gene family have been identified as tumour suppressors. SLC5A8 is a Na(+)-coupled transporter for monocarboxylates. Among its substrates are the bacterial fermentation products butyrate and propionate and the ubiquitous metabolite pyruvate. The tumour-suppressive function of this transporter relates to the ability of butyrate, propionate and pyruvate to inhibit histone deacetylases (HDAC). SLC5A8 functions as a tumour suppressor in most tissues studied thus far, and provides a molecular link to Warburg effect, a characteristic feature in most cancers. It also links colonic bacteria and dietary fibre to the host. SLC26A3 as a tumour suppressor is restricted to colon; it is a Cl(-)/HCO(-) 3 exchanger, facilitating the efflux of HCO(-) 3 The likely mechanism for the tumour-suppressive function of SLC26A3 is related to intracellular pH regulation. SLC39A1 is a Zn(2+) transporter and its role in tumour suppression has been shown in prostate. Zn(2+) is present at high concentrations in normal prostate where it elicits its tumour-suppressive function. SLC22A18 is possibly an organic cation transporter, but the identity of its physiological substrates is unknown. As such, there is no information on molecular pathways responsible for the tumour-suppressive function of this transporter. It is likely that additional SLC transporters will be discovered as tumour suppressors in the future.
Collapse
|
229
|
Angiolilli C, Baeten DL, Radstake TR, Reedquist KA. The acetyl code in rheumatoid arthritis and other rheumatic diseases. Epigenomics 2017; 9:447-461. [DOI: 10.2217/epi-2016-0136] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Growing evidence supports the idea that aberrancies in epigenetic processes contribute to the onset and progression of human immune-mediated inflammatory diseases, such as rheumatoid arthritis (RA). Epigenetic regulators of histone tail modifications play a role in chromatin accessibility and transcriptional responses to inflammatory stimuli. Among these, histone deacetylases (HDACs) regulate the acetylation status of histones and nonhistone proteins, essential for immune responses. Broad-spectrum HDAC inhibitors are well-known anti-inflammatory agents and reduce disease severity in animal models of arthritis; however, selective HDAC inhibitors remain poorly studied. In this review, we describe emerging findings regarding the aberrant acetyl code in RA and other rheumatic disorders which may help identify not only novel diagnostic and prognostic clinical biomarkers for RA, but also new targets for epigenetic pharmacological applications.
Collapse
Affiliation(s)
- Chiara Angiolilli
- Laboratory of Translational Immunology & Department of Rheumatology & Clinical Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
- Amsterdam Rheumatology & Immunology Center, Department of Clinical Immunology & Rheumatology, Department of Experimental Immunology Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Dominique L Baeten
- Amsterdam Rheumatology & Immunology Center, Department of Clinical Immunology & Rheumatology, Department of Experimental Immunology Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Timothy R Radstake
- Laboratory of Translational Immunology & Department of Rheumatology & Clinical Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Kris A Reedquist
- Laboratory of Translational Immunology & Department of Rheumatology & Clinical Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
| |
Collapse
|
230
|
Kwok J, O'Shea M, Hume DA, Lengeling A. Jmjd6, a JmjC Dioxygenase with Many Interaction Partners and Pleiotropic Functions. Front Genet 2017; 8:32. [PMID: 28360925 PMCID: PMC5352680 DOI: 10.3389/fgene.2017.00032] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Accepted: 02/27/2017] [Indexed: 12/20/2022] Open
Abstract
Lysyl hydroxylation and arginyl demethylation are post-translational events that are important for many cellular processes. The jumonji domain containing protein 6 (JMJD6) has been reported to catalyze both lysyl hydroxylation and arginyl demethylation on diverse protein substrates. It also interacts directly with RNA. This review summarizes knowledge of JMJD6 functions that have emerged in the last 15 years and considers how a single Jumonji C (JmjC) domain-containing enzyme can target so many different substrates. New links and synergies between the three main proposed functions of Jmjd6 in histone demethylation, promoter proximal pause release of polymerase II and RNA splicing are discussed. The physiological context of the described molecular functions is considered and recently described novel roles for JMJD6 in cancer and immune biology are reviewed. The increased knowledge of JMJD6 functions has wider implications for our general understanding of the JmjC protein family of which JMJD6 is a member.
Collapse
Affiliation(s)
- Janice Kwok
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh Edinburgh, UK
| | - Marie O'Shea
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh Edinburgh, UK
| | - David A Hume
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh Edinburgh, UK
| | - Andreas Lengeling
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh Edinburgh, UK
| |
Collapse
|
231
|
Molecular mechanisms involved in gliomagenesis. Brain Tumor Pathol 2017; 34:1-7. [DOI: 10.1007/s10014-017-0278-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2016] [Accepted: 01/12/2017] [Indexed: 10/20/2022]
|
232
|
Ye Q, Pang S, Zhang W, Guo X, Wang J, Zhang Y, Liu Y, Wu X, Jiang F. Therapeutic Targeting of RNA Polymerase I With the Small-Molecule CX-5461 for Prevention of Arterial Injury-Induced Neointimal Hyperplasia. Arterioscler Thromb Vasc Biol 2017; 37:476-484. [PMID: 28062495 DOI: 10.1161/atvbaha.116.308401] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Accepted: 12/21/2016] [Indexed: 01/09/2023]
Abstract
OBJECTIVE RNA polymerase I (Pol I)-dependent rRNA synthesis is a determinant factor in ribosome biogenesis and thus cell proliferation. The importance of dysregulated Pol I activity in cardiovascular disease, however, has not been recognized. Here, we tested the hypothesis that specific inhibition of Pol I might prevent arterial injury-induced neointimal hyperplasia. APPROACH AND RESULTS CX-5461 is a novel selective Pol I inhibitor. Using this tool, we demonstrated that local inhibition of Pol I blocked balloon injury-induced neointima formation in rat carotid arteries in vivo. Neointimal development was associated with augmented rDNA transcriptional activity as evidenced by the increased phosphorylation of upstream binding factor-1. The beneficial effect of CX-5461 was mainly mediated by inducing G2/M cell cycle arrest of proliferating smooth muscle cells without obvious apoptosis. CX-5461 did not induce p53 stabilization but increased p53 phosphorylation and acetylation and activated the ataxia telangiectasia mutated/ataxia telangiectasia and Rad3-related (ATR) pathway. Inhibition of ATR, but not of ataxia telangiectasia mutated, abolished the cytostatic effect of CX-5461 and p53 phosphorylation. In addition, inhibition of p53 or knockdown of the p53 target GADD45 mimicked the effect of ATR inhibition. In vivo experiments showed that the levels of phospho-p53 and acetyl-p53, and activity of the ataxia telangiectasia mutated/ATR pathway were all augmented in CX-5461-treated vessels. CONCLUSIONS Pol I can be therapeutically targeted to inhibit the growth of neointima, supporting that Pol I is a novel biological target for preventing arterial restenosis. Mechanistically, Pol I inhibition elicited G2/M cell cycle arrest in smooth muscle cells via activation of the ATR-p53 axis.
Collapse
Affiliation(s)
- Qing Ye
- From the School of Basic Medicine, Shandong University, Jinan, Shandong Province, China (Q.Y., S.P., W.Z., X.G., J.W., Y.L., F.J.); Key Laboratory of Cardiovascular Remodeling and Function Research & The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, Jinan, Shandong, China (X.W.); and Department of Cardiology, Qing Dao Central Hospital, Qing Dao, Shandong Province, China (Y.Z.)
| | - Shu Pang
- From the School of Basic Medicine, Shandong University, Jinan, Shandong Province, China (Q.Y., S.P., W.Z., X.G., J.W., Y.L., F.J.); Key Laboratory of Cardiovascular Remodeling and Function Research & The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, Jinan, Shandong, China (X.W.); and Department of Cardiology, Qing Dao Central Hospital, Qing Dao, Shandong Province, China (Y.Z.)
| | - Wenjing Zhang
- From the School of Basic Medicine, Shandong University, Jinan, Shandong Province, China (Q.Y., S.P., W.Z., X.G., J.W., Y.L., F.J.); Key Laboratory of Cardiovascular Remodeling and Function Research & The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, Jinan, Shandong, China (X.W.); and Department of Cardiology, Qing Dao Central Hospital, Qing Dao, Shandong Province, China (Y.Z.)
| | - Xiaotong Guo
- From the School of Basic Medicine, Shandong University, Jinan, Shandong Province, China (Q.Y., S.P., W.Z., X.G., J.W., Y.L., F.J.); Key Laboratory of Cardiovascular Remodeling and Function Research & The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, Jinan, Shandong, China (X.W.); and Department of Cardiology, Qing Dao Central Hospital, Qing Dao, Shandong Province, China (Y.Z.)
| | - Jianli Wang
- From the School of Basic Medicine, Shandong University, Jinan, Shandong Province, China (Q.Y., S.P., W.Z., X.G., J.W., Y.L., F.J.); Key Laboratory of Cardiovascular Remodeling and Function Research & The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, Jinan, Shandong, China (X.W.); and Department of Cardiology, Qing Dao Central Hospital, Qing Dao, Shandong Province, China (Y.Z.)
| | - Yongtao Zhang
- From the School of Basic Medicine, Shandong University, Jinan, Shandong Province, China (Q.Y., S.P., W.Z., X.G., J.W., Y.L., F.J.); Key Laboratory of Cardiovascular Remodeling and Function Research & The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, Jinan, Shandong, China (X.W.); and Department of Cardiology, Qing Dao Central Hospital, Qing Dao, Shandong Province, China (Y.Z.)
| | - Yang Liu
- From the School of Basic Medicine, Shandong University, Jinan, Shandong Province, China (Q.Y., S.P., W.Z., X.G., J.W., Y.L., F.J.); Key Laboratory of Cardiovascular Remodeling and Function Research & The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, Jinan, Shandong, China (X.W.); and Department of Cardiology, Qing Dao Central Hospital, Qing Dao, Shandong Province, China (Y.Z.)
| | - Xiao Wu
- From the School of Basic Medicine, Shandong University, Jinan, Shandong Province, China (Q.Y., S.P., W.Z., X.G., J.W., Y.L., F.J.); Key Laboratory of Cardiovascular Remodeling and Function Research & The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, Jinan, Shandong, China (X.W.); and Department of Cardiology, Qing Dao Central Hospital, Qing Dao, Shandong Province, China (Y.Z.)
| | - Fan Jiang
- From the School of Basic Medicine, Shandong University, Jinan, Shandong Province, China (Q.Y., S.P., W.Z., X.G., J.W., Y.L., F.J.); Key Laboratory of Cardiovascular Remodeling and Function Research & The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, Jinan, Shandong, China (X.W.); and Department of Cardiology, Qing Dao Central Hospital, Qing Dao, Shandong Province, China (Y.Z.).
| |
Collapse
|
233
|
Wright DG, Marchal C, Hoang K, Ankney JA, Nguyen ST, Rushing AW, Polakowski N, Miotto B, Lemasson I. Human T-cell leukemia virus type-1-encoded protein HBZ represses p53 function by inhibiting the acetyltransferase activity of p300/CBP and HBO1. Oncotarget 2016; 7:1687-706. [PMID: 26625199 PMCID: PMC4811490 DOI: 10.18632/oncotarget.6424] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2015] [Accepted: 11/15/2015] [Indexed: 01/31/2023] Open
Abstract
Adult T-cell leukemia (ATL) is an often fatal malignancy caused by infection with the complex retrovirus, human T-cell Leukemia Virus, type 1 (HTLV-1). In ATL patient samples, the tumor suppressor, p53, is infrequently mutated; however, it has been shown to be inactivated by the viral protein, Tax. Here, we show that another HTLV-1 protein, HBZ, represses p53 activity. In HCT116 p53+/+ cells treated with the DNA-damaging agent, etoposide, HBZ reduced p53-mediated activation of p21/CDKN1A and GADD45A expression, which was associated with a delay in G2 phase-arrest. These effects were attributed to direct inhibition of the histone acetyltransferase (HAT) activity of p300/CBP by HBZ, causing a reduction in p53 acetylation, which has be linked to decreased p53 activity. In addition, HBZ bound to, and inhibited the HAT activity of HBO1. Although HBO1 did not acetylate p53, it acted as a coactivator for p53 at the p21/CDKN1A promoter. Therefore, through interactions with two separate HAT proteins, HBZ impairs the ability of p53 to activate transcription. This mechanism may explain how p53 activity is restricted in ATL cells that do not express Tax due to modifications of the HTLV-1 provirus, which accounts for a majority of patient samples.
Collapse
Affiliation(s)
- Diana G Wright
- Brody School of Medicine, Department of Microbiology and Immunology, East Carolina University, Greenville, NC, USA
| | - Claire Marchal
- Université Paris Diderot, Sorbonne Paris Cité, Epigenetics and Cell Fate, UMR 7216, CNRS, Paris, France
| | - Kimson Hoang
- Brody School of Medicine, Department of Microbiology and Immunology, East Carolina University, Greenville, NC, USA
| | - John A Ankney
- Brody School of Medicine, Department of Microbiology and Immunology, East Carolina University, Greenville, NC, USA.,Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Stephanie T Nguyen
- Brody School of Medicine, Department of Microbiology and Immunology, East Carolina University, Greenville, NC, USA
| | - Amanda W Rushing
- Brody School of Medicine, Department of Microbiology and Immunology, East Carolina University, Greenville, NC, USA
| | - Nicholas Polakowski
- Brody School of Medicine, Department of Microbiology and Immunology, East Carolina University, Greenville, NC, USA
| | - Benoit Miotto
- Université Paris Diderot, Sorbonne Paris Cité, Epigenetics and Cell Fate, UMR 7216, CNRS, Paris, France.,INSERM, U1016, Institut Cochin, Paris, France.,CNRS, UMR8104, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Isabelle Lemasson
- Brody School of Medicine, Department of Microbiology and Immunology, East Carolina University, Greenville, NC, USA
| |
Collapse
|
234
|
Park JB. Javamide-I-O-methyl ester increases p53 acetylation and induces cell death via activating caspase 3/7 in monocytic THP-1 cells. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2016; 23:1647-1652. [PMID: 27823629 DOI: 10.1016/j.phymed.2016.10.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Revised: 09/22/2016] [Accepted: 10/07/2016] [Indexed: 06/06/2023]
Abstract
BACKGROUND Javamide-I and-II are phenolic amide compounds found in Coffea sp. Previous study suggested that javamide-II may be a potent compound with Sirt1/2 inhibition activity. PURPOSE However, the effects of javamide-I and the its O-methyl ester on Sirt inhibition, p53 acetylation and cell death have not been investigated. METHODS The isolation and synthesis of javamide-I and its O-methyl ester analogue were confirmed by NMR. Their potential effects on Sirt1/2/3, p53 acetylation and cell death were examined using sirt assay, silico analysis, Western blot, caspase 3/7 and apoptotic assay methods. RESULTS Javamide-I and its O-methyl ester demonstrated a similar inhibition pattern; Sirt1 (IC50 of 19µM) better than Sirt2 (IC50 of 104µM) and Sirt3 (IC50 of 160µM). However, javamide-I and its O-methyl ester were found to inhibit Sirt1 better than Sirt2, which is different from javamide-II able to inhibit Sirt2 stronger than Sirt1. In silico analysis, javamide-I and its O-methyl ester showed a competitive binding pattern against NAD+, which was also supported by the kinetic analysis with Ki=20.1µM for javamide-I and 19.5µM for the O-methyl ester. However, the O-methyl ester increased p53 acetylation better than javamide-I in monocytic THP-1 cells. Since caspase 3/7 activation is often followed by the p53 activation, we investigated their effects on caspase 3/7, and found that O-methyl ester again activated caspase 3/7 greater than javamide-I in the cells, eventually leading to apoptotic cell death. CONCLUSION These data suggest that the O-methyl modification in javamide-I may play a critical role in increasing p53 acetylation, activating caspase, and eventually inducing the THP-1 cell death.
Collapse
Affiliation(s)
- Jae B Park
- Diet, Genomics, and Immunology Laboratory, Bldg. 307C, Rm. 131, BHNRC, ARS, USDA, Beltsville, MD 20705, United States.
| |
Collapse
|
235
|
Liu MY, Wang WZ, Liao FF, Wu QQ, Lin XH, Chen YH, Cheng L, Jin XB, Zhu JY. Selective and effective targeting of chronic myeloid leukemia stem cells by topoisomerase II inhibitor etoposide in combination with imatinib mesylate in vitro. Cell Biol Int 2016; 41:16-23. [PMID: 27677634 DOI: 10.1002/cbin.10686] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Accepted: 09/24/2016] [Indexed: 11/06/2022]
Abstract
Imatinib mesylate (IM) and other BCR-ABL tyrosine kinase inhibitors (TKIs) have improved chronic myeloid leukemia (CML) patient survival markedly but fail to eradicate quiescent CML leukemia stem cells (LSCs). Thus, strategies targeting LSCs are required to induce long-term remission and achieve cure. Here, we investigated the ability of topoisomerase II (Top II) inhibitor etoposide (Eto) to target CML LSCs. Treatment with Eto combined with IM markedly induced apoptosis in primitive CML CD34+ CD38- stem cells resistant to eradication by IM alone, but not in normal hematopoietic stem cells, CML and normal mature CD34- cells, and other leukemia and lymphoma cell lines. The interaction of IM and Eto significantly inhibited phosphorylation of PDK1, AKT, GSK3, S6, and ERK proteins; increased the expression of pro-apoptotic gene Bax; and decreased the expression of anti-apoptotic gene c-Myc in CML CD34+ cells. Top II inhibitors treatment represents an attractive approach for targeting LSCs in CML patients undergoing TKIs monotherapy.
Collapse
Affiliation(s)
- Man-Yu Liu
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, P. R. China.,Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances, Guangdong Pharmaceutical University, Guangzhou Higher Education Mega Center, Guangzhou, 510006, P. R. China
| | - Wei-Zhang Wang
- Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances, Guangdong Pharmaceutical University, Guangzhou Higher Education Mega Center, Guangzhou, 510006, P. R. China.,Department of Biochemistry and Molecular Biology, School of Basic Courses, Guangdong Pharmaceutical University, Guangzhou, P. R. China
| | - Fen-Fang Liao
- Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances, Guangdong Pharmaceutical University, Guangzhou Higher Education Mega Center, Guangzhou, 510006, P. R. China.,Department of Biochemistry and Molecular Biology, School of Basic Courses, Guangdong Pharmaceutical University, Guangzhou, P. R. China
| | - Qing-Qing Wu
- Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances, Guangdong Pharmaceutical University, Guangzhou Higher Education Mega Center, Guangzhou, 510006, P. R. China.,Department of Biochemistry and Molecular Biology, School of Basic Courses, Guangdong Pharmaceutical University, Guangzhou, P. R. China
| | - Xiang-Hua Lin
- Department of Clinical Laboratory, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, P. R. China
| | - Yong-Hen Chen
- Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances, Guangdong Pharmaceutical University, Guangzhou Higher Education Mega Center, Guangzhou, 510006, P. R. China.,Department of Biochemistry and Molecular Biology, School of Basic Courses, Guangdong Pharmaceutical University, Guangzhou, P. R. China
| | - Lin Cheng
- Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances, Guangdong Pharmaceutical University, Guangzhou Higher Education Mega Center, Guangzhou, 510006, P. R. China.,Department of Biochemistry and Molecular Biology, School of Basic Courses, Guangdong Pharmaceutical University, Guangzhou, P. R. China
| | - Xiao-Bao Jin
- Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances, Guangdong Pharmaceutical University, Guangzhou Higher Education Mega Center, Guangzhou, 510006, P. R. China
| | - Jia-Yong Zhu
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, P. R. China.,Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances, Guangdong Pharmaceutical University, Guangzhou Higher Education Mega Center, Guangzhou, 510006, P. R. China
| |
Collapse
|
236
|
Hwang SJ, Park HG, Park Y, Lee HJ. An α-quaternary chiral latam derivative, YH-304 as a novel broad-spectrum anticancer agent. Int J Oncol 2016; 49:2480-2486. [PMID: 27748805 DOI: 10.3892/ijo.2016.3726] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Accepted: 09/30/2016] [Indexed: 11/06/2022] Open
Abstract
Previously, we reported that α-quaternary chiral lactam derivatives have broad spectrum anticancer activity. However, the underlying molecular mechanisms and its relevance are largely unknown. In the present study, we report progress on α-quaternary chiral lactam analogues that address this, focusing on the novel analogue YH-304 as a candidate to broadly target human cancer cells. The effect of YH-304 on cell transformation was assessed by clonogenic assay in non-small cell lung cancer cells (NSCLCs) A549 and 226B. Proapoptotic activity of YH-304 was determined by TUNEL assay and cleaved PARP, cleaved caspase-9, and Bax as markers for apoptosis. The p53-dependency and therapeutic spectrum of YH-304 was assessed by western blot analysis, real-time PCR, and cell viability assays in cells expressing endogenous wild or mutant p53. The effect of YH-304 on angiogenesis in vivo was examined by bFGF-mediated angiogenesis assay in zebrafish. Finally, the effect of YH-304 on AKT and ERK activation (phosphorylation) as a putative mechanism underlying the effect of YH-304 on bFGF-mediated angiogenesis was assessed using western blotting. We found that YH-304 significantly decreases the colony-forming activities of both A549 and 226B cells, inducing cellular apoptosis. Unlike nutlin-3 (p53 pathway activator), YH-304 did not affect the expression levels of p53 and its target gene such as p21 and thus showed p53-independent anticancer activity with broad spectrum. In addition, YH-304 inhibited bFGF-induced angiogenesis in vivo through mediating AKT and ERK signaling pathway, which plays an important role in bFGF activation and angiogenesis. Taken together, our data indicate that YH-304 may represent a novel therapeutic option for the treatment of cancer in a p53-independent manner.
Collapse
Affiliation(s)
- Su Jung Hwang
- College of Pharmacy and Inje Institute of Pharmaceutical Sciences and Research, Inje University, Gimhae, Gyeongnam 621-749, Republic of Korea
| | - Hyeung-Geun Park
- Research Institute of Pharmaceutical Science and College of Pharmacy, Seoul National University, Seoul 151-742, Republic of Korea
| | - Yohan Park
- College of Pharmacy and Inje Institute of Pharmaceutical Sciences and Research, Inje University, Gimhae, Gyeongnam 621-749, Republic of Korea
| | - Hyo-Jong Lee
- College of Pharmacy and Inje Institute of Pharmaceutical Sciences and Research, Inje University, Gimhae, Gyeongnam 621-749, Republic of Korea
| |
Collapse
|
237
|
HDAC1 and HDAC2 integrate the expression of p53 mutants in pancreatic cancer. Oncogene 2016; 36:1804-1815. [PMID: 27721407 DOI: 10.1038/onc.2016.344] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2015] [Revised: 08/05/2016] [Accepted: 08/15/2016] [Indexed: 12/25/2022]
Abstract
Mutation of p53 is a frequent genetic lesion in pancreatic cancer being an unmet clinical challenge. Mutants of p53 have lost the tumour-suppressive functions of wild type p53. In addition, p53 mutants exert tumour-promoting functions, qualifying them as important therapeutic targets. Here, we show that the class I histone deacetylases HDAC1 and HDAC2 contribute to maintain the expression of p53 mutants in human and genetically defined murine pancreatic cancer cells. Our data reveal that the inhibition of these HDACs with small molecule HDAC inhibitors (HDACi), as well as the specific genetic elimination of HDAC1 and HDAC2, reduce the expression of mutant p53 mRNA and protein levels. We further show that HDAC1, HDAC2 and MYC directly bind to the TP53 gene and that MYC recruitment drops upon HDAC inhibitor treatment. Therefore, our results illustrate a previously unrecognized class I HDAC-dependent control of the TP53 gene and provide evidence for a contribution of MYC. A combined approach targeting HDAC1/HDAC2 and MYC may present a novel and molecularly defined strategy to target mutant p53 in pancreatic cancer.
Collapse
|
238
|
Hrgovic I, Doll M, Kleemann J, Wang XF, Zoeller N, Pinter A, Kippenberger S, Kaufmann R, Meissner M. The histone deacetylase inhibitor trichostatin a decreases lymphangiogenesis by inducing apoptosis and cell cycle arrest via p21-dependent pathways. BMC Cancer 2016; 16:763. [PMID: 27716272 PMCID: PMC5045659 DOI: 10.1186/s12885-016-2807-y] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Accepted: 09/23/2016] [Indexed: 01/05/2023] Open
Abstract
Background The formation of new lymphatic vessels provides an additional route for tumour cells to metastasize. Therefore, inhibiting lymphangiogenesis represents an interesting target in cancer therapy. First evidence suggests that histone deacetylase inhibitors (HDACi) may mediate part of their antitumor effects by interfering with lymphangiogenesis. However, the underlying mechanisms of HDACi induced anti-lymphangiogenic properties are not fully investigated so far and in part remain unknown. Methods Human lymphatic endothelial cells (LEC) were cultured in vitro and treated with or without HDACi. Effects of HDACi on proliferation and cell cycle progress were analysed by BrdU assay and flow cytometry. Apoptosis was measured by quantifying mono- and oligonucleosomes in the cytoplasmic fraction of cell lysates. In vitro lymphangiogenesis was investigated using the Matrigel short term lymphangiogenesis assay. The effects of TSA on cell cycle regulatory proteins and apoptosis-related proteins were examined by western blotting, immunofluorescence staining and semi-quantitative RT-PCR. Protein- and mRNA half-life of p21 were analysed by western blotting and quantitative RT-PCR. The activity of the p21 promoter was determined using a dual luciferase assay and DNA-binding activity of Sp1/3 was investigated using EMSA. Furthermore, siRNA assays were performed to analyse the role of p21 and p53 on TSA-mediated anti-lymphangiogenic effects. Results We found that HDACi inhibited cell proliferation and that the pan-HDACi TSA induced G0/G1 arrest in LEC. Cell cycle arrest was accompanied by up-regulation of p21, p27 and p53. Additionally, we observed that p21 protein accumulated in cellular nuclei after treatment with TSA. Moreover, we found that p21 mRNA was significantly up-regulated by TSA, while the protein and mRNA half-life remained largely unaffected. The promoter activity of p21 was enhanced by TSA indicating a transcriptional mechanism. Subsequent EMSA analyses showed increased constitutive Sp1/3-dependent DNA binding in response to HDACi. We demonstrated that p53 was not required for TSA induced p21 expression and growth inhibition of LECs. Interestingly, siRNA-mediated p21 depletion almost completely reversed the anti-proliferative effects of TSA in LEC. In addition, TSA induced apoptosis by cytochrome c release contributed to activating caspases-9, −7 and −3 and downregulating the anti-apoptotic proteins cIAP-1 and −2. Conclusions In conclusion, we demonstrate that TSA - a pan-HDACi - has distinct anti-lymphangiogenic effects in primary human lymphatic endothelial cells by activating intrinsic apoptotic pathway and cell cycle arrest via p21-dependent pathways. Electronic supplementary material The online version of this article (doi:10.1186/s12885-016-2807-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Igor Hrgovic
- Department of Dermatology, Venereology and Allergology, Goethe University, Theodor-Stern Kai 7, Frankfurt/Main, 60590, Germany. .,Klinik für Dermatologie, Venerologie und Allergologie, Klinikum der J. W. Goethe-Universität, Theodor-Stern-Kai 7, Frankfurt am Main, D-60590, Germany.
| | - Monika Doll
- Department of Dermatology, Venereology and Allergology, Goethe University, Theodor-Stern Kai 7, Frankfurt/Main, 60590, Germany
| | - Johannes Kleemann
- Department of Dermatology, Venereology and Allergology, Goethe University, Theodor-Stern Kai 7, Frankfurt/Main, 60590, Germany
| | - Xiao-Fan Wang
- Department of Pharmacology & Cancer Biology, Duke University School of Medicine, C218 LSRC, Box 3813, Durham, NC, 27710, USA
| | - Nadja Zoeller
- Department of Dermatology, Venereology and Allergology, Goethe University, Theodor-Stern Kai 7, Frankfurt/Main, 60590, Germany
| | - Andreas Pinter
- Department of Dermatology, Venereology and Allergology, Goethe University, Theodor-Stern Kai 7, Frankfurt/Main, 60590, Germany
| | - Stefan Kippenberger
- Department of Dermatology, Venereology and Allergology, Goethe University, Theodor-Stern Kai 7, Frankfurt/Main, 60590, Germany
| | - Roland Kaufmann
- Department of Dermatology, Venereology and Allergology, Goethe University, Theodor-Stern Kai 7, Frankfurt/Main, 60590, Germany
| | - Markus Meissner
- Department of Dermatology, Venereology and Allergology, Goethe University, Theodor-Stern Kai 7, Frankfurt/Main, 60590, Germany
| |
Collapse
|
239
|
Corpas R, Revilla S, Ursulet S, Castro-Freire M, Kaliman P, Petegnief V, Giménez-Llort L, Sarkis C, Pallàs M, Sanfeliu C. SIRT1 Overexpression in Mouse Hippocampus Induces Cognitive Enhancement Through Proteostatic and Neurotrophic Mechanisms. Mol Neurobiol 2016; 54:5604-5619. [PMID: 27614878 DOI: 10.1007/s12035-016-0087-9] [Citation(s) in RCA: 79] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Accepted: 08/26/2016] [Indexed: 01/08/2023]
Abstract
SIRT1 induces cell survival and has shown neuroprotection against amyloid and tau pathologies in Alzheimer's disease (AD). However, protective effects against memory loss or the enhancement of cognitive functions have not yet been proven. We aimed to investigate the benefits induced by SIRT1 overexpression in the hippocampus of the AD mouse model 3xTg-AD and in control non-transgenic mice. A lentiviral vector encoding mouse SIRT1 or GFP, selectively transducing neurons, was injected into the dorsal CA1 hippocampal area of 4-month-old mice. Six-month overexpression of SIRT1 fully preserved learning and memory in 10-month-old 3xTg-AD mice. Remarkably, SIRT1 also induced cognitive enhancement in healthy non-transgenic mice. Neuron cultures of 3xTg-AD mice, which show traits of AD-like pathology, and neuron cultures from non-transgenic mice were also transduced with lentiviral vectors to analyze beneficial SIRT1 mechanisms. We uncovered novel pathways of SIRT1 neuroprotection through enhancement of cell proteostatic mechanisms and activation of neurotrophic factors not previously reported such as GDNF, present in both AD-like and healthy neurons. Therefore, SIRT1 may increase neuron function and resilience against AD.
Collapse
Affiliation(s)
- Rubén Corpas
- Institut d'Investigacions Biomèdiques de Barcelona (IIBB) - CSIC, C/Rosselló 161, 6th floor, 08036, Barcelona, Spain
| | - Susana Revilla
- Institut d'Investigacions Biomèdiques de Barcelona (IIBB) - CSIC, C/Rosselló 161, 6th floor, 08036, Barcelona, Spain
| | | | - Marco Castro-Freire
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036, Barcelona, Spain
| | - Perla Kaliman
- Institut d'Investigacions Biomèdiques de Barcelona (IIBB) - CSIC, C/Rosselló 161, 6th floor, 08036, Barcelona, Spain
| | - Valérie Petegnief
- Institut d'Investigacions Biomèdiques de Barcelona (IIBB) - CSIC, C/Rosselló 161, 6th floor, 08036, Barcelona, Spain
| | - Lydia Giménez-Llort
- Department of Psychiatry and Forensic Medicine, Universitat Autònoma de Barcelona, 08193, Bellaterra, Spain
| | | | - Mercè Pallàs
- Facultat de Farmàcia, Institut de Neurociències, Universitat de Barcelona and CIBERNED, 08028, Barcelona, Spain
| | - Coral Sanfeliu
- Institut d'Investigacions Biomèdiques de Barcelona (IIBB) - CSIC, C/Rosselló 161, 6th floor, 08036, Barcelona, Spain. .,Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036, Barcelona, Spain.
| |
Collapse
|
240
|
Cui Y, Liu N, Ma F, Sun W, Wu H, Xu Z, Yuan H. Role of histone modification in 12‑lipoxygenase‑associated p21 gene regulation. Mol Med Rep 2016; 14:3978-84. [PMID: 27600103 DOI: 10.3892/mmr.2016.5724] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Accepted: 06/21/2016] [Indexed: 11/05/2022] Open
Abstract
The aim of the present study was to examine the impacts and mechanisms of 12‑lipoxygenase (12‑LO) and its metabolites on the acetylation and methylation of histone‑3‑lysine (H3K) in the p21 gene. Rat mesangial cells (MCs) were selected for use in the present study. A chromatin immunoprecipitation assay, reverse transcription‑quantitative polymerase chain reaction analysis and a luciferase assay were used to detect transcriptional activities, the acetylation (Ac) of H3K (H3KAc), p21 promoter methylation (Me) and the transcription regions induced by 12 (S)‑hydroxyeicosatetraenoic acid (HETE). The cells were transfected to induce the overexpression of p300 to examine changes in 12 (S)‑HETE‑associated p21 regulation and epigenetic modifications. 12 (S)‑HETE enhanced p21 transcriptional activity and mRNA expression. In the promoter regions, P1 and P2, and the T1 transcription region, 12 (S)‑HETE induced significant H3K9 Ac and H3K4 Me1 epigenetic modifications, however, no changes were observed in the T2 region. By contrast, 12 (S)‑HETE treatment markedly prevented H3K9Me3 at the p21 promoter, suggesting that complex Me was involved in 12 (S)‑HETE‑associated p21 regulation. Furthermore, the overexpression of p300 markedly enhanced basal and 12 (S)‑HETE‑associated p21 transcriptional regulation in the MCs. 12 (S)‑HETE treatment also induced histone acetyltransferase p300 occupancy in the p21 promoter, and reduced the nuclear expression and occupancy of lysine‑specific demethylase (LSD1) in the p21 promoter. 12 (S)‑HETE induced p300 occupancy, and reduced the nuclear expression and occupancy of LSD1 in the p21 promoter. Therefore, enhanced H3K9Ac and H3K4Me1 in the p21 promoter and transcription regions, and decreased H3K9Me3 in the p21 promoter increased the expression of p21.
Collapse
Affiliation(s)
- Yingchun Cui
- Department of Nephrology, Second Affiliated Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Nian Liu
- Center of Urology, First Affiliated Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Fuzhe Ma
- Department of Nephrology, First Affiliated Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Weixia Sun
- Department of Nephrology, First Affiliated Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Hao Wu
- Department of Nephrology, First Affiliated Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Zhonggao Xu
- Department of Nephrology, First Affiliated Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Hang Yuan
- Department of Nephrology, First Affiliated Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| |
Collapse
|
241
|
Gong EY, Shin YJ, Hwang IY, Kim JH, Kim SM, Moon JH, Shin JS, Lee DH, Hur DY, Jin DH, Hong SW, Lee WK, Lee WJ. Combined treatment with vitamin C and sulindac synergistically induces p53- and ROS-dependent apoptosis in human colon cancer cells. Toxicol Lett 2016; 258:126-133. [PMID: 27339904 DOI: 10.1016/j.toxlet.2016.06.019] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Revised: 06/09/2016] [Accepted: 06/18/2016] [Indexed: 12/22/2022]
Abstract
Sulindac has anti-neoplastic properties against colorectal cancers; however, its use as a chemopreventive agent has been limited due to toxicity and efficacy concerns. Combinatorial treatment of colorectal cancers has been attempted to maximize anti-cancer efficacy with minimal side effects by administrating NSAIDs in combination with other inhibitory compounds or drugs such as l-ascorbic acid (vitamin C), which is known to exhibit cytotoxicity towards various cancer cells at high concentrations. In this study, we evaluated a combinatorial strategy utilizing sulindac and vitamin C. The death of HCT116 cells upon combination therapy occurred via a p53-mediated mechanism. The combination therapeutic resistance developed in isogenic p53 null HCT116 cells and siRNA-mediated p53 knockdown HCT116 cells, but the exogenous expression of p53 in p53 null isogenic cells resulted in the induction of cell death. In addition, we investigated an increased level of intracellular ROS (reactive oxygen species), which was preceded by p53 activation. The expression level of PUMA (p53-upregulated modulator of apoptosis), but not Bim, was significantly increased in HCT116 cells in response to the combination treatment. Taken together, our results demonstrate that combination therapy with sulindac and vitamin C could be a novel anti-cancer therapeutic strategy for p53 wild type colon cancers.
Collapse
Affiliation(s)
- Eun-Yeung Gong
- Asan Institute of Life Sciences, Asan Medical Center, 88 Olympic-ro 43-gil, Songpa-gu, Seoul 05505, Republic of Korea; Department of Convergence Medicine, University of Ulsan College of Medicine, Asan Medical Center, 88 Olympic-ro 43-gil, Songpa-gu, Seoul 05505, Republic of Korea
| | - Yu Jin Shin
- Asan Institute of Life Sciences, Asan Medical Center, 88 Olympic-ro 43-gil, Songpa-gu, Seoul 05505, Republic of Korea; Department of Convergence Medicine, University of Ulsan College of Medicine, Asan Medical Center, 88 Olympic-ro 43-gil, Songpa-gu, Seoul 05505, Republic of Korea
| | - Ih-Yeon Hwang
- Asan Institute of Life Sciences, Asan Medical Center, 88 Olympic-ro 43-gil, Songpa-gu, Seoul 05505, Republic of Korea; Department of Convergence Medicine, University of Ulsan College of Medicine, Asan Medical Center, 88 Olympic-ro 43-gil, Songpa-gu, Seoul 05505, Republic of Korea
| | - Jeong Hee Kim
- Asan Institute of Life Sciences, Asan Medical Center, 88 Olympic-ro 43-gil, Songpa-gu, Seoul 05505, Republic of Korea; Department of Convergence Medicine, University of Ulsan College of Medicine, Asan Medical Center, 88 Olympic-ro 43-gil, Songpa-gu, Seoul 05505, Republic of Korea
| | - Seung-Mi Kim
- Asan Institute of Life Sciences, Asan Medical Center, 88 Olympic-ro 43-gil, Songpa-gu, Seoul 05505, Republic of Korea; Department of Convergence Medicine, University of Ulsan College of Medicine, Asan Medical Center, 88 Olympic-ro 43-gil, Songpa-gu, Seoul 05505, Republic of Korea; Division of Biosciences and Bioinformatics, Myongji University, 116 Myongji-ro, Cheoin-gu, Yongin, Gyeonggi-do 17058, Republic of Korea
| | - Jai-Hee Moon
- Asan Institute of Life Sciences, Asan Medical Center, 88 Olympic-ro 43-gil, Songpa-gu, Seoul 05505, Republic of Korea; Department of Convergence Medicine, University of Ulsan College of Medicine, Asan Medical Center, 88 Olympic-ro 43-gil, Songpa-gu, Seoul 05505, Republic of Korea
| | - Jae-Sik Shin
- Asan Institute of Life Sciences, Asan Medical Center, 88 Olympic-ro 43-gil, Songpa-gu, Seoul 05505, Republic of Korea; Department of Convergence Medicine, University of Ulsan College of Medicine, Asan Medical Center, 88 Olympic-ro 43-gil, Songpa-gu, Seoul 05505, Republic of Korea
| | - Dae-Hee Lee
- Asan Institute of Life Sciences, Asan Medical Center, 88 Olympic-ro 43-gil, Songpa-gu, Seoul 05505, Republic of Korea; Department of Convergence Medicine, University of Ulsan College of Medicine, Asan Medical Center, 88 Olympic-ro 43-gil, Songpa-gu, Seoul 05505, Republic of Korea
| | - Dae Young Hur
- Department of Anatomy, Inje University College of Medicine, 75 Bokji-ro, Busanjin-gu, Busan 47392, Republic of Korea
| | - Dong-Hoon Jin
- Asan Institute of Life Sciences, Asan Medical Center, 88 Olympic-ro 43-gil, Songpa-gu, Seoul 05505, Republic of Korea; Department of Convergence Medicine, University of Ulsan College of Medicine, Asan Medical Center, 88 Olympic-ro 43-gil, Songpa-gu, Seoul 05505, Republic of Korea
| | - Seung-Woo Hong
- Asan Institute of Life Sciences, Asan Medical Center, 88 Olympic-ro 43-gil, Songpa-gu, Seoul 05505, Republic of Korea; Department of Anatomy, Inje University College of Medicine, 75 Bokji-ro, Busanjin-gu, Busan 47392, Republic of Korea.
| | - Won Keun Lee
- Division of Biosciences and Bioinformatics, Myongji University, 116 Myongji-ro, Cheoin-gu, Yongin, Gyeonggi-do 17058, Republic of Korea.
| | - Wang-Jae Lee
- Department of Anatomy and Tumor Immunity Medical Research Center, Seoul National University College of Medicine, 103, Daehak-ro, Jongno-gu, Seoul 03080, Republic of Korea
| |
Collapse
|
242
|
Abstract
It is now clear that functional p53 is critical to protect the genome from alterations that lead to tumorigenesis. However, with the myriad of cellular stresses and pathways linked to p53 activation, much remains unknown about how p53 maintains genome stability and the proteins involved. The current understanding of the multiple ways p53 contributes to genome stability and how two of its negative regulators, Mdm2 and Mdmx, induce genome instability will be described.
Collapse
Affiliation(s)
- Christine M Eischen
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee 37212
| |
Collapse
|
243
|
Knyphausen P, de Boor S, Kuhlmann N, Scislowski L, Extra A, Baldus L, Schacherl M, Baumann U, Neundorf I, Lammers M. Insights into Lysine Deacetylation of Natively Folded Substrate Proteins by Sirtuins. J Biol Chem 2016; 291:14677-94. [PMID: 27226597 DOI: 10.1074/jbc.m116.726307] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Indexed: 11/06/2022] Open
Abstract
Sirtuins are NAD(+)-dependent lysine deacylases, regulating a variety of cellular processes. The nuclear Sirt1, the cytosolic Sirt2, and the mitochondrial Sirt3 are robust deacetylases, whereas the other sirtuins have preferences for longer acyl chains. Most previous studies investigated sirtuin-catalyzed deacylation on peptide substrates only. We used the genetic code expansion concept to produce natively folded, site-specific, and lysine-acetylated Sirt1-3 substrate proteins, namely Ras-related nuclear, p53, PEPCK1, superoxide dismutase, cyclophilin D, and Hsp10, and analyzed the deacetylation reaction. Some acetylated proteins such as Ras-related nuclear, p53, and Hsp10 were robustly deacetylated by Sirt1-3. However, other reported sirtuin substrate proteins such as cyclophilin D, superoxide dismutase, and PEPCK1 were not deacetylated. Using a structural and functional approach, we describe the ability of Sirt1-3 to deacetylate two adjacent acetylated lysine residues. The dynamics of this process have implications for the lifetime of acetyl modifications on di-lysine acetylation sites and thus constitute a new mechanism for the regulation of proteins by acetylation. Our studies support that, besides the primary sequence context, the protein structure is a major determinant of sirtuin substrate specificity.
Collapse
Affiliation(s)
- Philipp Knyphausen
- From the Institute for Genetics and Cologne Excellence Cluster on Cellular Stress Responses in Aging-associated Diseases (CECAD), Joseph-Stelzmann-Strasse 26, University of Cologne, 50931 Cologne and
| | - Susanne de Boor
- From the Institute for Genetics and Cologne Excellence Cluster on Cellular Stress Responses in Aging-associated Diseases (CECAD), Joseph-Stelzmann-Strasse 26, University of Cologne, 50931 Cologne and
| | - Nora Kuhlmann
- From the Institute for Genetics and Cologne Excellence Cluster on Cellular Stress Responses in Aging-associated Diseases (CECAD), Joseph-Stelzmann-Strasse 26, University of Cologne, 50931 Cologne and
| | - Lukas Scislowski
- From the Institute for Genetics and Cologne Excellence Cluster on Cellular Stress Responses in Aging-associated Diseases (CECAD), Joseph-Stelzmann-Strasse 26, University of Cologne, 50931 Cologne and
| | - Antje Extra
- From the Institute for Genetics and Cologne Excellence Cluster on Cellular Stress Responses in Aging-associated Diseases (CECAD), Joseph-Stelzmann-Strasse 26, University of Cologne, 50931 Cologne and
| | - Linda Baldus
- From the Institute for Genetics and Cologne Excellence Cluster on Cellular Stress Responses in Aging-associated Diseases (CECAD), Joseph-Stelzmann-Strasse 26, University of Cologne, 50931 Cologne and
| | - Magdalena Schacherl
- the Institute for Biochemistry, Zülpicher Strasse 47b, University of Cologne, 50674 Cologne, Germany
| | - Ulrich Baumann
- the Institute for Biochemistry, Zülpicher Strasse 47b, University of Cologne, 50674 Cologne, Germany
| | - Ines Neundorf
- the Institute for Biochemistry, Zülpicher Strasse 47b, University of Cologne, 50674 Cologne, Germany
| | - Michael Lammers
- From the Institute for Genetics and Cologne Excellence Cluster on Cellular Stress Responses in Aging-associated Diseases (CECAD), Joseph-Stelzmann-Strasse 26, University of Cologne, 50931 Cologne and
| |
Collapse
|
244
|
Newbold A, Falkenberg KJ, Prince HM, Johnstone RW. How do tumor cells respond to HDAC inhibition? FEBS J 2016; 283:4032-4046. [PMID: 27112360 DOI: 10.1111/febs.13746] [Citation(s) in RCA: 90] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Revised: 03/30/2016] [Accepted: 04/22/2016] [Indexed: 02/06/2023]
Abstract
It is now well recognized that mutations, deregulated expression, and aberrant recruitment of epigenetic readers, writers, and erasers are fundamentally important processes in the onset and maintenance of many human tumors. The molecular, biological, and biochemical characteristics of a particular class of epigenetic erasers, the histone deacetylases (HDACs), have been extensively studied and small-molecule HDAC inhibitors (HDACis) have now been clinically approved for the treatment of human hemopoietic malignancies. This review explores our current understanding of the biological and molecular effects on tumor cells following HDACi treatment. The predominant responses include induction of tumor cell death and inhibition of proliferation that in experimental models have been linked to therapeutic efficacy. However, tumor cell-intrinsic responses to HDACi, including modulating tumor immunogenicity have also been described and may have substantial roles in mediating the antitumor effects of HDACi. We posit that the field has failed to fully reconcile the biological consequences of exposure to HDACis with the molecular events that underpin these responses, however progress is being made. Understanding the pleiotrophic activities of HDACis on tumor cells will hopefully fast track the development of more potent and selective HDACi that may be used alone or in combination to improve patient outcomes.
Collapse
Affiliation(s)
- Andrea Newbold
- Cancer Therapeutics Program, The Peter MacCallum Cancer Centre, East Melbourne, Vic., Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Vic., Australia
| | | | - H Miles Prince
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Vic., Australia.,Division of Cancer Medicine, The Peter MacCallum Cancer Centre, East Melbourne, Vic., Australia
| | - Ricky W Johnstone
- Cancer Therapeutics Program, The Peter MacCallum Cancer Centre, East Melbourne, Vic., Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Vic., Australia
| |
Collapse
|
245
|
Kurosawa N, Wakata Y, Inobe T, Kitamura H, Yoshioka M, Matsuzawa S, Kishi Y, Isobe M. Novel method for the high-throughput production of phosphorylation site-specific monoclonal antibodies. Sci Rep 2016; 6:25174. [PMID: 27125496 PMCID: PMC4850396 DOI: 10.1038/srep25174] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Accepted: 04/11/2016] [Indexed: 12/03/2022] Open
Abstract
Threonine phosphorylation accounts for 10% of all phosphorylation sites compared with 0.05% for tyrosine and 90% for serine. Although monoclonal antibody generation for phospho-serine and -tyrosine proteins is progressing, there has been limited success regarding the production of monoclonal antibodies against phospho-threonine proteins. We developed a novel strategy for generating phosphorylation site-specific monoclonal antibodies by cloning immunoglobulin genes from single plasma cells that were fixed, intracellularly stained with fluorescently labeled peptides and sorted without causing RNA degradation. Our high-throughput fluorescence activated cell sorting-based strategy, which targets abundant intracellular immunoglobulin as a tag for fluorescently labeled antigens, greatly increases the sensitivity and specificity of antigen-specific plasma cell isolation, enabling the high-efficiency production of monoclonal antibodies with desired antigen specificity. This approach yielded yet-undescribed guinea pig monoclonal antibodies against threonine 18-phosphorylated p53 and threonine 68-phosphorylated CHK2 with high affinity and specificity. Our method has the potential to allow the generation of monoclonal antibodies against a variety of phosphorylated proteins.
Collapse
Affiliation(s)
- Nobuyuki Kurosawa
- Laboratory of Molecular and Cellular Biology, Graduate School of Science and Engineering for Research, University of Toyama, 3190 Gofuku, Toyama-shi, Toyama, 930-8555, Japan
| | - Yuka Wakata
- Laboratory of Molecular and Cellular Biology, Graduate School of Science and Engineering for Research, University of Toyama, 3190 Gofuku, Toyama-shi, Toyama, 930-8555, Japan
| | - Tomonao Inobe
- Frontier Research Core for Life Sciences, University of Toyama, 3190 Gofuku, Toyama-shi, Toyama, 930-8555, Japan
| | - Haruki Kitamura
- Graduate School of Science and Engineering for Education, University of Toyama, Toyama-shi, Toyama, 930-8555, Japan
| | - Megumi Yoshioka
- Laboratory of Molecular and Cellular Biology, Graduate School of Science and Engineering for Research, University of Toyama, 3190 Gofuku, Toyama-shi, Toyama, 930-8555, Japan
| | - Shun Matsuzawa
- Medical &Biological Laboratories Co., Ltd., 15-502 Akaho, Komagane, Nagano, 399-4117, Japan
| | - Yoshihiro Kishi
- Medical &Biological Laboratories Co., Ltd., 15-502 Akaho, Komagane, Nagano, 399-4117, Japan
| | - Masaharu Isobe
- Laboratory of Molecular and Cellular Biology, Graduate School of Science and Engineering for Research, University of Toyama, 3190 Gofuku, Toyama-shi, Toyama, 930-8555, Japan
| |
Collapse
|
246
|
Franci G, Manfroni G, Cannalire R, Felicetti T, Tabarrini O, Salvato A, Barreca ML, Altucci L, Cecchetti V. Tumour cell population growth inhibition and cell death induction of functionalized 6-aminoquinolone derivatives. Cell Prolif 2016; 48:705-17. [PMID: 26597381 DOI: 10.1111/cpr.12224] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2015] [Accepted: 08/17/2015] [Indexed: 12/17/2022] Open
Abstract
OBJECTIVES A number of previous studies has provided evidence that the well-known anti-bacterial quinolones may have potential as anti-cancer drugs. The aim of this study was to evaluate potential anti-tumour activity and selectivity of a set of 6-aminoquinolones showing some chemical similarity to naphthyridone derivative CX-5461, recently described as innovative anti-cancer agent. MATERIALS AND METHODS In-house quinolones 1-8 and ad hoc synthesized derivatives 9-13 were tested on Michigan Cancer Foundation-7 (MCF-7) breast cancer cells and mesenchymal progenitor (MePR2B) cell lines, analysing their effects on the cell cycle and cell death using FACS methodology. Activation of p53 was evaluated by western blotting. RESULTS Benzyl esters 4, 5 and their amide counterparts 12, 13 drastically modulated MCF-7 cell cycles inducing DNA fragmentation and cell death, thus proving to be potential anti-tumour compounds. When assayed in non-tumour MePR2B cells, compounds 4 and 5 were cytotoxic while 12 and 13 had a certain degree of selectivity, with compound 12 emerging as the most promising. Western blot analysis revealed that severe p53-K382ac activation was promoted by benzylester 5. In contrast, amide 12 exerted only a moderate effect which was, however, comparable to that of suberoylanilide hydoxamic acid (SAHA). CONCLUSIONS Taken together, these results further reinforce evidence that quinolones have potential as anti-cancer agents. Future work will be focused on understanding compound 12 mechanisms of action, and to obtain more potent and selective compounds.
Collapse
Affiliation(s)
- G Franci
- Dipartimento di Biochimica, Biofisica e Patologia Generale, Seconda Università degli Studi di Napoli, 80138, Napoli, Italy
| | - G Manfroni
- Dipartimento di Scienze Farmaceutiche, Università degli Studi di Perugia, 06123, Perugia, Italy
| | - R Cannalire
- Dipartimento di Scienze Farmaceutiche, Università degli Studi di Perugia, 06123, Perugia, Italy
| | - T Felicetti
- Dipartimento di Scienze Farmaceutiche, Università degli Studi di Perugia, 06123, Perugia, Italy
| | - O Tabarrini
- Dipartimento di Scienze Farmaceutiche, Università degli Studi di Perugia, 06123, Perugia, Italy
| | - A Salvato
- Dipartimento di Biochimica, Biofisica e Patologia Generale, Seconda Università degli Studi di Napoli, 80138, Napoli, Italy
| | - M L Barreca
- Dipartimento di Scienze Farmaceutiche, Università degli Studi di Perugia, 06123, Perugia, Italy
| | - L Altucci
- Dipartimento di Biochimica, Biofisica e Patologia Generale, Seconda Università degli Studi di Napoli, 80138, Napoli, Italy.,Istituto di Genetica e Biofisica, IGB, 'Adriano Buzzati Traverso' Via P. Castellino, 80131, Napoli, Italy
| | - V Cecchetti
- Dipartimento di Scienze Farmaceutiche, Università degli Studi di Perugia, 06123, Perugia, Italy
| |
Collapse
|
247
|
Woschek M, Kneip N, Jurida K, Marzi I, Relja B. Simvastatin Reduces Cancerogenic Potential of Renal Cancer Cells via Geranylgeranyl Pyrophosphate and Mevalonate Pathway. Nutr Cancer 2016; 68:420-7. [PMID: 27042994 DOI: 10.1080/01635581.2016.1152383] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Simvastatin is a cholesterol-lowering drug, inhibiting 3-hydroxy-3-methylglutaryl-coenzyme CoA (HMG-CoA) reductase. Previous studies have indicated the anticancerous effects of simvastatin. Here, we evaluated the anticancerous potential of simvastatin in renal cell carcinoma (RCC) cell lines. RCC occurs with an incidence of 2-3% of all cancer entities with high chemoresistance rate. Therefore, the understanding of underlying mechanisms for RCC activity and the development of alternative therapies are essential. Human RCC cell lines Caki-1 and KTC-26 were treated with simvastatin (16 or 33 µM) for 48 or 72 h. The effects of the downstream substrates mevalonate (MA), farnesyl pyrophosphate (FPP), and geranylgeranyl pyrophosphate (GGPP) were evaluated using add-back experiments. Cell growth was assessed using MTT assay. Apoptosis and cell cycle were analyzed by flow cytometry. Apoptosis-involved proteins were evaluated by Western blot. Simvastatin caused dose- and time-dependent inhibition of RCC cell growth by cell cycle arrest and apoptosis induction. Substitution of MA or GGPP abolished these effects to a large extent. These findings suggest that the antiproliferative effects of simvastatin are not only mediated through cholesterol deprivation but also by prenylation-associated mechanisms, thereby providing new insights into tumor-suppressive ability of simvastatin and into novel additive treatment options in the management of RCC.
Collapse
Affiliation(s)
- Mathias Woschek
- a Department of Trauma , Hand and Reconstructive Surgery, University Hospital Frankfurt, Goethe University , Frankfurt am Main , Germany
| | - Niels Kneip
- a Department of Trauma , Hand and Reconstructive Surgery, University Hospital Frankfurt, Goethe University , Frankfurt am Main , Germany
| | - Katrin Jurida
- a Department of Trauma , Hand and Reconstructive Surgery, University Hospital Frankfurt, Goethe University , Frankfurt am Main , Germany
| | - Ingo Marzi
- a Department of Trauma , Hand and Reconstructive Surgery, University Hospital Frankfurt, Goethe University , Frankfurt am Main , Germany
| | - Borna Relja
- a Department of Trauma , Hand and Reconstructive Surgery, University Hospital Frankfurt, Goethe University , Frankfurt am Main , Germany
| |
Collapse
|
248
|
Sørensen BH, Nielsen D, Thorsteinsdottir UA, Hoffmann EK, Lambert IH. Downregulation of LRRC8A protects human ovarian and alveolar carcinoma cells against Cisplatin-induced expression of p53, MDM2, p21Waf1/Cip1, and Caspase-9/-3 activation. Am J Physiol Cell Physiol 2016; 310:C857-73. [PMID: 26984736 PMCID: PMC4935196 DOI: 10.1152/ajpcell.00256.2015] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Accepted: 03/13/2016] [Indexed: 11/22/2022]
Abstract
The leucine-rich repeat containing 8A (LRRC8A) protein is an essential component of the volume-sensitive organic anion channel (VSOAC), and using pharmacological anion channel inhibitors (NS3728, DIDS) and LRRC8A siRNA we have investigated its role in development of Cisplatin resistance in human ovarian (A2780) and alveolar (A549) carcinoma cells. In Cisplatin-sensitive cells Cisplatin treatment increases p53-protein level as well as downstream signaling, e.g., expression of p21Waf1/Cip1, Bax, Noxa, MDM2, and activation of Caspase-9/-3. In contrast, Cisplatin-resistant cells do not enter apoptosis, i.e., their p53 and downstream signaling are reduced and caspase activity unaltered following Cisplatin exposure. Reduced LRRC8A expression and VSOAC activity are previously shown to correlate with Cisplatin resistance, and here we demonstrate that pharmacological inhibition and transient knockdown of LRRC8A reduce the protein level of p53, MDM2, and p21Waf1/Cip1 as well as Caspase-9/-3 activation in Cisplatin-sensitive cells. Cisplatin resistance is accompanied by reduction in total LRRC8A expression (A2780) or LRRC8A expression in the plasma membrane (A549). Activation of Caspase-3 dependent apoptosis by TNFα-exposure or hyperosmotic cell shrinkage is almost unaffected by pharmacological anion channel inhibition. Our data indicate 1) that expression/activity of LRRC8A is essential for Cisplatin-induced increase in p53 protein level and its downstream signaling, i.e., Caspase-9/-3 activation, expression of p21Waf1/Cip1 and MDM2; and 2) that downregulation of LRRC8A-dependent osmolyte transporters contributes to acquirement of Cisplatin resistance in ovarian and lung carcinoma cells. Activation of LRRC8A-containing channels is upstream to apoptotic volume decrease as hypertonic cell shrinkage induces apoptosis independent of the presence of LRRC8A.
Collapse
Affiliation(s)
- Belinda Halling Sørensen
- Department of Biology, Section of Cell Biology and Physiology, The August Krogh Building, University of Copenhagen, Copenhagen, Denmark
| | - Dorthe Nielsen
- Department of Biology, Section of Cell Biology and Physiology, The August Krogh Building, University of Copenhagen, Copenhagen, Denmark
| | - Unnur Arna Thorsteinsdottir
- Department of Biology, Section of Cell Biology and Physiology, The August Krogh Building, University of Copenhagen, Copenhagen, Denmark
| | - Else Kay Hoffmann
- Department of Biology, Section of Cell Biology and Physiology, The August Krogh Building, University of Copenhagen, Copenhagen, Denmark
| | - Ian Henry Lambert
- Department of Biology, Section of Cell Biology and Physiology, The August Krogh Building, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
249
|
Apoptosis, autophagy and unfolded protein response pathways in Arbovirus replication and pathogenesis. Expert Rev Mol Med 2016; 18:e1. [PMID: 26781343 PMCID: PMC4836210 DOI: 10.1017/erm.2015.19] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Arboviruses are pathogens that widely affect the health of people in different communities around the world. Recently, a few successful approaches toward production of effective vaccines against some of these pathogens have been developed, but treatment and prevention of the resulting diseases remain a major health and research concern. The arbovirus infection and replication processes are complex, and many factors are involved in their regulation. Apoptosis, autophagy and the unfolded protein response (UPR) are three mechanisms that are involved in pathogenesis of many viruses. In this review, we focus on the importance of these pathways in the arbovirus replication and infection processes. We provide a brief introduction on how apoptosis, autophagy and the UPR are initiated and regulated, and then discuss the involvement of these pathways in regulation of arbovirus pathogenesis.
Collapse
|
250
|
Differential Proteomic Analysis of Human Placenta-Derived Mesenchymal Stem Cells Cultured on Normal Tissue Culture Surface and Hyaluronan-Coated Surface. Stem Cells Int 2015; 2016:2809192. [PMID: 27057169 PMCID: PMC4709773 DOI: 10.1155/2016/2809192] [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: 05/16/2015] [Revised: 09/07/2015] [Accepted: 10/07/2015] [Indexed: 12/15/2022] Open
Abstract
Our previous results showed that hyaluronan (HA) preserved human placenta-derived mesenchymal stem cells (PDMSC) in a slow cell cycling mode similar to quiescence, the pristine state of stem cells in vivo, and HA was found to prevent murine adipose-derived mesenchymal stem cells from senescence. Here, stable isotope labeling by amino acid in cell culture (SILAC) proteomic profiling was used to evaluate the effects of HA on aging phenomenon in stem cells, comparing (1) old and young passage PDMSC cultured on normal tissue culture surface (TCS); (2) old passage on HA-coated surface (CHA) compared to TCS; (3) old and young passage on CHA. The results indicated that senescence-associated protein transgelin (TAGLN) was upregulated in old TCS. Protein CYR61, reportedly senescence-related, was downregulated in old CHA compared to old TCS. The SIRT1-interacting Nicotinamide phosphoribosyltransferase (NAMPT) increased by 2.23-fold in old CHA compared to old TCS, and is 0.48-fold lower in old TCS compared to young TCS. Results also indicated that components of endoplasmic reticulum associated degradation (ERAD) pathway were upregulated in old CHA compared to old TCS cells, potentially for overcoming stress to maintain cell function and suppress senescence. Our data points to pathways that may be targeted by HA to maintain stem cells youth.
Collapse
|