1
|
Dantas A, Shueili BA, Park J, Abdullah S, Bertschmann J, Krowicki H, Djamshidi M, Yang Y, Blote K, Thalappilly S, Riabowol K. Tissue-specific localization of the ING4 targeting subunit of the HBO1 histone acetyltransferase in the cytoplasm and nucleus of secretory cells. Histochem Cell Biol 2025; 163:56. [PMID: 40402282 DOI: 10.1007/s00418-025-02385-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/01/2025] [Indexed: 05/23/2025]
Abstract
Members of the INhibitor of Growth protein family (ING1-5) function as epigenetic regulators by targeting different histone acetyltransferase (HAT) and histone deacetylase (HDAC) complexes to the H3K4Me3 mark of active transcription. The INGs recognize H3K4Me3 by specific interaction with their well-conserved plant homeodomains, and affinity can be increased by interactions between DNA and disordered regions within the ING proteins. They are classified as type II tumor suppressors since they are downregulated in numerous cancer types and knockout of ING family members results in tumorigenesis. ING4 targets the HBO1 HAT complex, which is known to affect acetylation of the H4 core nucleosomal histone, to affect local chromatin structure and knockout results in deficient innate immunity. Reports indicating roles in cell cycle regulation, tumor suppression, and apoptosis suggest that ING4 may be a promising target for cancer treatment by targeting pathways of innate immunity. Given the relatedness between ING4 and the closely related ING5 proteins, we have developed and characterized two mouse monoclonal antibodies to specifically recognize human and mouse ING4, but not ING5, to more accurately characterize ING4 levels by western, immunofluorescence and immunohistochemical assays. Using them, we show that ING4 differentially partitions between the nucleus and cytoplasm in different tissues and localizes largely to the cytoplasm of cells having a secretory role in different tissue types.
Collapse
Affiliation(s)
- Arthur Dantas
- Robson DNA Sciences Centre, Calgary, Canada
- Arnie Charbonneau Cancer Institute, Calgary, Canada
- Faculty of Health Sciences, School of Human Kinetics, University of Ottawa, Ottawa, ON, Canada
| | - Buthaina Al Shueili
- Robson DNA Sciences Centre, Calgary, Canada
- Arnie Charbonneau Cancer Institute, Calgary, Canada
- Department of Biology, College of Science, Sultan Qaboos University, Muscat, Oman
| | - Jeongah Park
- Robson DNA Sciences Centre, Calgary, Canada
- Arnie Charbonneau Cancer Institute, Calgary, Canada
- University of Alberta, Edmonton, AB, Canada
| | - Suleyman Abdullah
- Robson DNA Sciences Centre, Calgary, Canada
- Arnie Charbonneau Cancer Institute, Calgary, Canada
- University of Western Ontario, London, ON, Canada
| | - Jessica Bertschmann
- Robson DNA Sciences Centre, Calgary, Canada
- Arnie Charbonneau Cancer Institute, Calgary, Canada
- Departments of Biochemistry and Molecular Biology, Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, T2N 4N1, Canada
| | - Hokan Krowicki
- Robson DNA Sciences Centre, Calgary, Canada
- Arnie Charbonneau Cancer Institute, Calgary, Canada
| | - Mahbod Djamshidi
- Robson DNA Sciences Centre, Calgary, Canada
- Arnie Charbonneau Cancer Institute, Calgary, Canada
- Departments of Biochemistry and Molecular Biology, Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, T2N 4N1, Canada
| | - Yang Yang
- Robson DNA Sciences Centre, Calgary, Canada
- Arnie Charbonneau Cancer Institute, Calgary, Canada
| | - Karen Blote
- Robson DNA Sciences Centre, Calgary, Canada
- Arnie Charbonneau Cancer Institute, Calgary, Canada
| | - Subhash Thalappilly
- Robson DNA Sciences Centre, Calgary, Canada
- National University of Singapore, Queenstown, Singapore
| | - Karl Riabowol
- Robson DNA Sciences Centre, Calgary, Canada.
- Arnie Charbonneau Cancer Institute, Calgary, Canada.
- Departments of Biochemistry and Molecular Biology, Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, T2N 4N1, Canada.
| |
Collapse
|
2
|
Mayo-Smith M, Poulet A, Zhang L, Peng Y, Goldstone D, Putterill J. Medicago Mting1 Mting2 double knockout mutants are extremely dwarfed and never flower implicating essential MtING functions in growth and flowering. BMC PLANT BIOLOGY 2025; 25:410. [PMID: 40169950 PMCID: PMC11960017 DOI: 10.1186/s12870-025-06432-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Accepted: 03/19/2025] [Indexed: 04/03/2025]
Abstract
BACKGROUND Optimal flowering time is critical to agricultural productivity. Despite this, flowering regulation in the Fabaceae (legume) family is not fully understood. For example, FLC and CO control Arabidopsis flowering, but do not regulate flowering in the temperate legume Medicago. Little is known about the genetic roles of the two plant ING genes. They encode proteins with conserved ING and PHD finger domains predicted to function as epigenetic readers. Previously, using CRISPR-Cas9 knock outs, we reported that Medicago MtING2 promotes flowering and growth. However, surprisingly, Mting2 PHD finger mutants flowered similarly to wild type. Additionally, MtING1 did not regulate flowering because Mting1 mutants flowered like wild type. METHODS To further dissect the combined genetic function of MtING1 and MtING2 and their PHD fingers, we cross-pollinated Mting1 and Mting2 single mutants to create two double mutants: The Mting1-7 Mting2-2 double knockout mutant and the Mting1-1 Mting2-11 double PHD finger mutant. Mutant phenotypes were assessed in floral-inductive conditions. We used fluorescence confocal microscopy and in vitro protein biophysical analysis to investigate the subcellular localization and oligomerization of the proteins. We carried out gene expression analysis by RNA-seq and RT-qPCR to determine how the two genes affect transcript accumulation to influence growth and flowering. RESULTS The Mting double knockout mutants displayed a striking, non-flowering, highly dwarfed phenotype indicating overlapping and complementary functions. Conversely Mting double PHD finger mutants showed only mild dwarfing and weak delays to flowering, indicating that the PHD fingers did not have a major impact on MtING function. MtING proteins localised to the nucleus, consistent with their predicted roles as histone readers, but did not interact in solution. Large changes to gene expression were seen in the Mting2-2 single mutant and the double knockout mutant, with key flowering genes downregulated and predicted floral repressors elevated. Furthermore, the MtINGs promoted the expression of Medicago homologs of target genes of the Arabidopsis NuA4 HAT complex. CONCLUSIONS Our findings demonstrate the key combined function the MtING genes play in regulation of global gene expression, flowering time and wider development and implicate an important role in epigenetic regulation via HAT complexes.
Collapse
Affiliation(s)
- Matthew Mayo-Smith
- School of Biological Sciences, Faculty of Science, University of Auckland, Auckland Mail Centre, Private Bag 92019, Auckland, 1142, New Zealand
| | - Axel Poulet
- Department of Molecular, Cellular and Developmental Biology, Faculty of Arts and Sciences, Yale University, New Haven, CT, USA
| | - Lulu Zhang
- School of Biological Sciences, Faculty of Science, University of Auckland, Auckland Mail Centre, Private Bag 92019, Auckland, 1142, New Zealand
| | - Yongyan Peng
- The New Zealand Institute for Plant and Food Research Limited (Plant & Food Research) Mt Albert, Auckland Mail Centre, Private Bag 92169, Auckland, 1142, New Zealand
| | - David Goldstone
- School of Biological Sciences, Faculty of Science, University of Auckland, Auckland Mail Centre, Private Bag 92019, Auckland, 1142, New Zealand
| | - Joanna Putterill
- School of Biological Sciences, Faculty of Science, University of Auckland, Auckland Mail Centre, Private Bag 92019, Auckland, 1142, New Zealand.
| |
Collapse
|
3
|
Lang V, Barones L, Hu SM, Hashemi F, Blote K, Riabowol K, Fink D. Loss of ING3 in the Prostate Leads to Activation of DNA Damage Repair Markers. Cancers (Basel) 2025; 17:1037. [PMID: 40149370 PMCID: PMC11940784 DOI: 10.3390/cancers17061037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2025] [Revised: 03/05/2025] [Accepted: 03/17/2025] [Indexed: 03/29/2025] Open
Abstract
BACKGROUND/OBJECTIVES The inhibitor of growth family member 3 (ING3) acts as an epigenetic reader through physical interactions with histone-modifying enzymes and subsequent chromatin remodelling processes. It is involved in various cellular functions, such as cell cycle control, cell growth, and apoptosis. Although ING3 was assigned tumour suppressor candidate status in some types of cancers, including prostate cancer, some studies suggest it acts to promote growth. To address these contradictory reports regarding its role in the initiation and progression of prostate cancer, we specifically addressed the question of whether ablation of ING3 in the mouse prostate is sufficient to initiate malignant transformation of the prostate and support its (candidate) tumour suppressor status. METHODS To generate the prostate-specific Ing3 knockout mouse, paternal inheritance of the PB-Cre4 transgene was used, while for the generation of a global knockout control, a female mouse harbouring the PB-Cre4 transgene was utilized. To determine the recombination efficiency of the Cre-LoxP system in the prostate at the Ing3 locus, a duplex probe-based digital PCR assay capable of counting undisrupted Ing3 copies was designed. The impact of DNA recombination on the protein level was investigated by immunohistochemical staining of prostate tissue samples. RESULTS In the prostate-specific knockout, digital PCR analysis revealed mosaic gene deletion. We found recombination efficiencies in the anterior, dorsolateral, and ventral prostate lobes ranging from approximately 15 to 30%. ING3 staining in the prostate was faint with no detectable differences in signal intensity between the knockout specimen and wild-type controls. This low ING3 expression in the prostate is consistent with observations of X-gal staining of an Ing3-LacZ reporter allele. Immunohistochemistry showed increased expression of DNA-damage-associated markers γH2AX and 53BP1. However, no gross anatomical abnormalities or prostate intraepithelial neoplasia (PIN) lesions in the prostate of tissue-specific knockout animals compared to wild-type controls were observed. CONCLUSIONS Altogether, our data provide evidence that disruption of ING3 expression in prostate cells does not lead to malignant transformation and challenges the idea that ING3 acts primarily in a tumour-suppressive manner. Furthermore, this work supports the crucial role of ING3 in maintaining genomic stability, and we confirmed the embryonic lethal phenotype of homozygous Ing3 null mice that is rescued by ectopic expression of ING3.
Collapse
Affiliation(s)
- Viktor Lang
- Institute of Laboratory Animal Science, University of Veterinary Medicine Vienna, 1210 Vienna, Austria
- Departments of Biochemistry & Molecular Biology and Oncology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Lisa Barones
- Institute of Laboratory Animal Science, University of Veterinary Medicine Vienna, 1210 Vienna, Austria
| | - ShiTing Misaki Hu
- Institute of Laboratory Animal Science, University of Veterinary Medicine Vienna, 1210 Vienna, Austria
| | - Fatemeh Hashemi
- Departments of Biochemistry & Molecular Biology and Oncology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Karen Blote
- Departments of Biochemistry & Molecular Biology and Oncology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Karl Riabowol
- Departments of Biochemistry & Molecular Biology and Oncology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Dieter Fink
- Institute of Laboratory Animal Science, University of Veterinary Medicine Vienna, 1210 Vienna, Austria
- British Columbia Cancer Research Centre, Vancouver, BC V5Z 1L3, Canada
| |
Collapse
|
4
|
Xu Z, Xu H, Shi J, Liu R, Li X, Liu S, Wei W. Inhibitor of Growth Proteins: Epigenetic Regulators Shaping Neurobiology. Biomolecules 2025; 15:281. [PMID: 40001584 PMCID: PMC11852745 DOI: 10.3390/biom15020281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 02/06/2025] [Accepted: 02/12/2025] [Indexed: 02/16/2025] Open
Abstract
The inhibitor of growth (ING) family of proteins is emerging as a pivotal regulator of epigenetic modifications within the nervous system. These proteins are involved in various cellular processes, including apoptosis, cell cycle control, and DNA repair, through interactions with chromatin-modifying complexes. Recent studies underscore the dual role of ING proteins in both tumor suppression and neuronal differentiation, development, and neuroprotection. This review summarizes the epigenetic functions of ING proteins in neurobiology, with a focus on their involvement in neural development and their relevance to neuro-oncological diseases. We explore the mechanisms by which ING proteins influence chromatin state and gene expression, highlighting their interactions with histone acetyltransferases, deacetylases, histone methyltransferases, DNA modification enzymes, and non-coding RNAs. A deeper understanding of the role of ING proteins in epigenetic regulation in the nervous system may pave the way for novel therapeutic strategies targeting neurological disorders.
Collapse
Affiliation(s)
- Ziyue Xu
- Brain Research Center, Zhongnan Hospital of Wuhan University, Wuhan 430071, China; (Z.X.); (H.X.); (J.S.); (R.L.); (X.L.)
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Hongyu Xu
- Brain Research Center, Zhongnan Hospital of Wuhan University, Wuhan 430071, China; (Z.X.); (H.X.); (J.S.); (R.L.); (X.L.)
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Jichun Shi
- Brain Research Center, Zhongnan Hospital of Wuhan University, Wuhan 430071, China; (Z.X.); (H.X.); (J.S.); (R.L.); (X.L.)
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Runming Liu
- Brain Research Center, Zhongnan Hospital of Wuhan University, Wuhan 430071, China; (Z.X.); (H.X.); (J.S.); (R.L.); (X.L.)
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Xiang Li
- Brain Research Center, Zhongnan Hospital of Wuhan University, Wuhan 430071, China; (Z.X.); (H.X.); (J.S.); (R.L.); (X.L.)
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
- Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan 430071, China
- Medical Research Institute, Wuhan University, Wuhan 430071, China
- Sino-Italian Ascula Brain Science Joint Laboratory, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Sha Liu
- Brain Research Center, Zhongnan Hospital of Wuhan University, Wuhan 430071, China; (Z.X.); (H.X.); (J.S.); (R.L.); (X.L.)
- Department of General Practice, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Wei Wei
- Brain Research Center, Zhongnan Hospital of Wuhan University, Wuhan 430071, China; (Z.X.); (H.X.); (J.S.); (R.L.); (X.L.)
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| |
Collapse
|
5
|
Al Shueili B, Dantas A, Mahe E, Chu TH, Yang Y, Labit E, Kutluberk E, Lasaleta N, Masson A, Omairi H, Ito K, Krawetz RJ, Midha R, Cairncross G, Riabowol K. Knockout of the ING5 epigenetic regulator confirms roles in stem cell maintenance and tumor suppression in vivo. PLoS One 2025; 20:e0313255. [PMID: 39787145 PMCID: PMC11717183 DOI: 10.1371/journal.pone.0313255] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Accepted: 10/21/2024] [Indexed: 01/12/2025] Open
Abstract
INhibitor of Growth (ING1-5) proteins are epigenetic readers that target histone acetyltransferase (HAT) or histone deacetylase (HDAC) complexes to the H3K4Me3 mark of active transcription. ING5 targets Moz/Morf and HBO1 HAT complexes that alter acetylation of H3 and H4 core histones, affecting gene expression. Previous experiments in vitro indicated that ING5 functions to maintain stem cell character in normal and in cancer stem cells. Here we find that CRISPR/Cas9 ING5 knockout (KO) mice are sub-fertile but show no decrease in lifespan or ability to heal wounds despite indications of depleted stem cell pools in several tissues. ING5 KO mouse embryo fibroblasts accumulate in G2 of the cell cycle, have high levels of abnormal nuclei and show high basal levels of the γH2AX indicator of DNA damage. KO animals also develop severe dermatitis at a 5-fold higher rate that wild-type littermates. Consistent with ING5 serving a tumor suppressive role, ING5 KO mice developed germinal centre diffuse large B-cell lymphomas at a rate 6-fold higher than control mice at 18 months of age. These data suggest that ING5 functions in vivo to maintain stem cell character in multiple organs, that reduction of stem cell populations is not limiting for murine lifespan and that like a subset of other ING family members, ING5 functions as a tumor suppressor in hematopoietic cells in vivo.
Collapse
Affiliation(s)
- Buthaina Al Shueili
- Robson DNA Sciences Centre, Calgary, Canada
- Arnie Charbonneau Cancer Institute, Calgary, Canada
- Departments of Biochemistry & Molecular Biology, University of Calgary, Calgary, Canada
| | - Arthur Dantas
- Robson DNA Sciences Centre, Calgary, Canada
- Arnie Charbonneau Cancer Institute, Calgary, Canada
| | - Etienne Mahe
- Arnie Charbonneau Cancer Institute, Calgary, Canada
- Departments of Pathology and Laboratory Medicine, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Tak Ho Chu
- Hotchkiss Brain Institute, University of Calgary, Calgary, Canada
- Departments of Clinical Neurosciences, University of Calgary, Calgary, Canada
| | - Yang Yang
- Robson DNA Sciences Centre, Calgary, Canada
- Arnie Charbonneau Cancer Institute, Calgary, Canada
- Departments of Alberta Children’s Hospital Research Institute, University of Calgary, Calgary, Canada
| | - Elodie Labit
- Departments of Comparative Biology and Experimental Medicine, University of Calgary, Calgary, Canada
| | - Eren Kutluberk
- Departments of Comparative Biology and Experimental Medicine, University of Calgary, Calgary, Canada
| | - Nicolas Lasaleta
- Hotchkiss Brain Institute, University of Calgary, Calgary, Canada
- Departments of Clinical Neurosciences, University of Calgary, Calgary, Canada
| | - Anand Masson
- Departments of McCaig Institute for Bone & Joint Health, University of Calgary, Calgary, Canada
- Departments of Cell Biology and Anatomy, University of Calgary, Calgary, Canada
| | - Hiba Omairi
- Arnie Charbonneau Cancer Institute, Calgary, Canada
- Departments of Biochemistry & Molecular Biology, University of Calgary, Calgary, Canada
| | - Kenichi Ito
- Departments of Biochemistry & Molecular Biology, University of Calgary, Calgary, Canada
| | - Roman J. Krawetz
- Departments of McCaig Institute for Bone & Joint Health, University of Calgary, Calgary, Canada
- Departments of Cell Biology and Anatomy, University of Calgary, Calgary, Canada
- Departments of Surgery, University of Calgary, Calgary, Canada
| | - Rajiv Midha
- Hotchkiss Brain Institute, University of Calgary, Calgary, Canada
- Departments of Clinical Neurosciences, University of Calgary, Calgary, Canada
| | - Gregory Cairncross
- Arnie Charbonneau Cancer Institute, Calgary, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, Canada
- Departments of Oncology, University of Calgary, Calgary, Canada
| | - Karl Riabowol
- Robson DNA Sciences Centre, Calgary, Canada
- Arnie Charbonneau Cancer Institute, Calgary, Canada
- Departments of Biochemistry & Molecular Biology, University of Calgary, Calgary, Canada
- Departments of Alberta Children’s Hospital Research Institute, University of Calgary, Calgary, Canada
- Departments of Oncology, University of Calgary, Calgary, Canada
| |
Collapse
|
6
|
Xia A, Wang X, Huang Y, Yang Q, Ye M, Wang Y, Jiang C, Duan K. The ING protein Fng2 associated with RPD3 HDAC complex for the regulation of fungal development and pathogenesis in wheat head blight fungus. Int J Biol Macromol 2024; 268:131938. [PMID: 38692539 DOI: 10.1016/j.ijbiomac.2024.131938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 04/25/2024] [Accepted: 04/26/2024] [Indexed: 05/03/2024]
Abstract
ING proteins display a high level of evolutionary conservation across various species, and play a crucial role in modulating histone acetylation levels, thus regulating various important biological processes in yeast and humans. Filamentous fungi possess distinct biological characteristics that differentiate them from yeasts and humans, and the specific roles of ING proteins in filamentous fungi remain largely unexplored. In this study, an ING protein, Fng2, orthologous to the yeast Pho23, has been identified in the wheat head blight fungus Fusarium graminearum. The deletion of the FNG2 gene resulted in defects in vegetative growth, conidiation, sexual reproduction, plant infection, and deoxynivalenol (DON) biosynthesis. Acting as a global regulator, Fng2 exerts negative control over histone H4 acetylation and governs the expression of over 4000 genes. Moreover, almost half of the differentially expressed genes in the fng3 mutant were found to be co-regulated by Fng2, emphasizing the functional association between these two ING proteins. Notably, the fng2 fng3 double mutant exhibits significantly increased H4 acetylation and severe defects in both fungal development and pathogenesis. Furthermore, Fng2 localizes within the nucleus and associates with the FgRpd3 histone deacetylase (HDAC) to modulate gene expression. Overall, Fng2's interaction with FgRpd3, along with its functional association with Fng3, underscores its crucial involvement in governing gene expression, thereby significantly influencing fungal growth, asexual and sexual development, pathogenicity, and secondary metabolism.
Collapse
Affiliation(s)
- Aliang Xia
- State Key Laboratory for Crop Stress Resistance and High-Efficiency Production, College of Plant Protection, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Xuan Wang
- State Key Laboratory for Crop Stress Resistance and High-Efficiency Production, College of Plant Protection, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Yingao Huang
- State Key Laboratory for Crop Stress Resistance and High-Efficiency Production, College of Plant Protection, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Qing Yang
- State Key Laboratory for Crop Stress Resistance and High-Efficiency Production, College of Plant Protection, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Meng Ye
- State Key Laboratory for Crop Stress Resistance and High-Efficiency Production, College of Plant Protection, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Yankun Wang
- State Key Laboratory for Crop Stress Resistance and High-Efficiency Production, College of Plant Protection, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Cong Jiang
- State Key Laboratory for Crop Stress Resistance and High-Efficiency Production, College of Plant Protection, Northwest A&F University, Yangling, Shaanxi 712100, China.
| | - Kaili Duan
- State Key Laboratory for Crop Stress Resistance and High-Efficiency Production, College of Plant Protection, Northwest A&F University, Yangling, Shaanxi 712100, China.
| |
Collapse
|
7
|
Gao L, Zhou Y, Cao L, Cao Y, Zhang H, Zhang M, Yin H, Ai S. Photoelectrochemical sensor for histone deacetylase Sirt1 detection based on Z-scheme heterojunction of CuS-BiVO 4 photoactive material and the cyclic etching of MnO 2 by NADH. Talanta 2024; 268:125307. [PMID: 37866306 DOI: 10.1016/j.talanta.2023.125307] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 10/09/2023] [Accepted: 10/12/2023] [Indexed: 10/24/2023]
Abstract
A novel photoelectrochemical (PEC) biosensor was constructed for histone deacetylase Sirt1 detection based on the Z-Scheme heterojunction of CuS-BiVO4 and reduced nicotinamide adenine dinucleotide (NADH) induced cyclic etching of MnO2 triggered by Sirt1 enzyme catalytic histone deacetylation event. Based on the Z-Scheme heterojunction, the photoactivity of the CuS-BiVO4 was improved greatly due to the highly effective separation of the photogenerated electron-hole pairs. In the presence of MnO2 nanosheets on the CuS-BiVO4/ITO electrode surface, the photocurrent decreased due to the inhibition effect of MnO2. However, this inhibition effect was eliminated by the incubation of MnO2/CuS-BiVO4/ITO with NADH, where NADH was produced in the deacetylation process of acetylated peptide catalyzed by Sirt1 with NAD+. The formed NADH etched MnO2, resulting in an increased photocurrent. In this process, NADH was oxidized to produce NAD+, which further involved the deacetylation process. Based on this cycle, the photocurrent of the biosensor was improved greatly and the sensitive and selective detection of Sirt1 was achieved. The biosensor presented a wide linear range from 0.005 to 10 nM with the low detection limit of 3.38 pM (S/N = 3). In addition, the applicability of the developed method was evaluated by investigating the effect of sodium butyrate and perfluorohexane sulfonate on Sirt1 activity.
Collapse
Affiliation(s)
- Lanlan Gao
- College of Chemistry and Material Science, Key Laboratory of Agricultural Film Application of Ministry of Agriculture and Rural Affairs, Food Safety Analysis and Test Engineering Technology Research Center of Shandong Province, Shandong Agricultural University, 271018, Taian, Shandong, China
| | - Yunlei Zhou
- College of Chemistry and Material Science, Key Laboratory of Agricultural Film Application of Ministry of Agriculture and Rural Affairs, Food Safety Analysis and Test Engineering Technology Research Center of Shandong Province, Shandong Agricultural University, 271018, Taian, Shandong, China.
| | - Lulu Cao
- College of Chemistry and Material Science, Key Laboratory of Agricultural Film Application of Ministry of Agriculture and Rural Affairs, Food Safety Analysis and Test Engineering Technology Research Center of Shandong Province, Shandong Agricultural University, 271018, Taian, Shandong, China
| | - Yaoyuan Cao
- College of Chemistry and Material Science, Key Laboratory of Agricultural Film Application of Ministry of Agriculture and Rural Affairs, Food Safety Analysis and Test Engineering Technology Research Center of Shandong Province, Shandong Agricultural University, 271018, Taian, Shandong, China
| | - Haowei Zhang
- College of Chemistry and Material Science, Key Laboratory of Agricultural Film Application of Ministry of Agriculture and Rural Affairs, Food Safety Analysis and Test Engineering Technology Research Center of Shandong Province, Shandong Agricultural University, 271018, Taian, Shandong, China
| | - Miao Zhang
- College of Chemistry and Material Science, Key Laboratory of Agricultural Film Application of Ministry of Agriculture and Rural Affairs, Food Safety Analysis and Test Engineering Technology Research Center of Shandong Province, Shandong Agricultural University, 271018, Taian, Shandong, China
| | - Huanshun Yin
- College of Chemistry and Material Science, Key Laboratory of Agricultural Film Application of Ministry of Agriculture and Rural Affairs, Food Safety Analysis and Test Engineering Technology Research Center of Shandong Province, Shandong Agricultural University, 271018, Taian, Shandong, China
| | - Shiyun Ai
- College of Chemistry and Material Science, Key Laboratory of Agricultural Film Application of Ministry of Agriculture and Rural Affairs, Food Safety Analysis and Test Engineering Technology Research Center of Shandong Province, Shandong Agricultural University, 271018, Taian, Shandong, China
| |
Collapse
|
8
|
Yang Y, Ma B, Djamshidi M, Zhang Q, Sarkar A, Chanda A, Tran U, Soh J, Sandall C, Chen HM, MacDonald JA, Bonni S, Sensen CW, Zheng J, Riabowol K. ING1 inhibits Twist1 expression to block EMT and is antagonized by the HDAC inhibitor vorinostat. Eur J Cell Biol 2023; 102:151341. [PMID: 37459799 DOI: 10.1016/j.ejcb.2023.151341] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 06/28/2023] [Accepted: 07/06/2023] [Indexed: 09/22/2023] Open
Abstract
ING1 is a chromatin targeting subunit of the Sin3a histone deacetylase (HDAC) complex that alters chromatin structure to subsequently regulate gene expression. We find that ING1 knockdown increases expression of Twist1, Zeb 1&2, Snai1, Bmi1 and TSHZ1 drivers of EMT, promoting EMT and cell motility. ING1 expression had the opposite effect, promoting epithelial cell morphology and inhibiting basal and TGF-β-induced motility in 3D organoid cultures. ING1 binds the Twist1 promoter and Twist1 was largely responsible for the ability of ING1 to reduce cell migration. Consistent with ING1 inhibiting Twist1 expression in vivo, an inverse relationship between ING1 and Twist1 levels was seen in breast cancer samples from The Cancer Genome Atlas (TCGA). The HDAC inhibitor vorinostat is approved for treatment of multiple myeloma and cutaneous T cell lymphoma and is in clinical trials for solid tumours as adjuvant therapy. One molecular target of vorinostat is INhibitor of Growth 2 (ING2), that together with ING1 serve as targeting subunits of the Sin3a HDAC complex. Treatment with sublethal (LD25-LD50) levels of vorinostat promoted breast cancer cell migration several-fold, which increased further upon ING1 knockout. These observations indicate that correct targeting of the Sin3a HDAC complex, and HDAC activity in general decreases luminal and basal breast cancer cell motility, suggesting that use of HDAC inhibitors as adjuvant therapies in breast cancers that are prone to metastasize may not be optimal and requires further investigation.
Collapse
Affiliation(s)
- Yang Yang
- Arnie Charbonneau Cancer Institute, Departments of Biochemistry and Molecular Biology and Oncology, University of Calgary, Calgary, Alberta, Canada; Department of Obstetrics and Gynecology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, PR China
| | - Biao Ma
- Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, PR China
| | - Mahbod Djamshidi
- Arnie Charbonneau Cancer Institute, Departments of Biochemistry and Molecular Biology and Oncology, University of Calgary, Calgary, Alberta, Canada
| | - Qingrun Zhang
- Arnie Charbonneau Cancer Institute, Departments of Biochemistry and Molecular Biology and Oncology, University of Calgary, Calgary, Alberta, Canada
| | - Anusi Sarkar
- Arnie Charbonneau Cancer Institute, Departments of Biochemistry and Molecular Biology and Oncology, University of Calgary, Calgary, Alberta, Canada
| | - Ayan Chanda
- Arnie Charbonneau Cancer Institute, Departments of Biochemistry and Molecular Biology and Oncology, University of Calgary, Calgary, Alberta, Canada
| | - Uyen Tran
- Arnie Charbonneau Cancer Institute, Departments of Biochemistry and Molecular Biology and Oncology, University of Calgary, Calgary, Alberta, Canada
| | - Jung Soh
- Arnie Charbonneau Cancer Institute, Departments of Biochemistry and Molecular Biology and Oncology, University of Calgary, Calgary, Alberta, Canada
| | - Christina Sandall
- Libin Cardiovascular Institute of Alberta, Department of Biochemistry and Molecular Biology, University of Calgary, Calgary, Alberta, Canada
| | - Huey-Miin Chen
- Libin Cardiovascular Institute of Alberta, Department of Biochemistry and Molecular Biology, University of Calgary, Calgary, Alberta, Canada
| | - Justin A MacDonald
- Libin Cardiovascular Institute of Alberta, Department of Biochemistry and Molecular Biology, University of Calgary, Calgary, Alberta, Canada
| | - Shirin Bonni
- Arnie Charbonneau Cancer Institute, Departments of Biochemistry and Molecular Biology and Oncology, University of Calgary, Calgary, Alberta, Canada
| | | | - Jianhua Zheng
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, PR China
| | - Karl Riabowol
- Arnie Charbonneau Cancer Institute, Departments of Biochemistry and Molecular Biology and Oncology, University of Calgary, Calgary, Alberta, Canada.
| |
Collapse
|
9
|
Šimková A, Civáňová Křížová K, Voříšková K, Vetešník L, Bystrý V, Demko M. Transcriptome Profile Analyses of Head Kidney in Roach ( Rutilus rutilus), Common Bream ( Abramis brama) and Their Hybrids: Does Infection by Monogenean Parasites in Freshwater Fish Reveal Differences in Fish Vigour among Parental Species and Their Hybrids? BIOLOGY 2023; 12:1199. [PMID: 37759598 PMCID: PMC10525477 DOI: 10.3390/biology12091199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 08/23/2023] [Accepted: 08/28/2023] [Indexed: 09/29/2023]
Abstract
Hybrid generations usually face either a heterosis advantage or a breakdown, that can be expressed by the level of parasite infection in hybrid hosts. Hybrids are less infected by parasites than parental species (especially F1 generations) or more infected than parental species (especially post-F1 generations). We performed the experiment with blood-feeding gill parasite Paradiplozoon homoion (Monogenea) infecting leuciscid species, Abramis brama and Rutilus rutilus, their F1 generation and two backcross generations. Backcross generations tended to be more parasitized than parental lines and the F1 generation. The number of differentially expressed genes (DEGs) was lower in F1 hybrids and higher in backcross hybrids when compared to each of the parental lines. The main groups of DEGs were shared among lines; however, A. brama and R. rutilus differed in some of the top gene ontology (GO) terms. DEG analyses revealed the role of heme binding and erythrocyte differentiation after infection by blood-feeding P. homoion. Two backcross generations shared some of the top GO terms, representing mostly downregulated genes associated with P. homoion infection. KEGG analysis revealed the importance of disease-associated pathways; the majority of them were shared by two backcross generations. Our study revealed the most pronounced DEGs associated with blood-feeding monogeneans in backcross hybrids, potentially (but not exclusively) explainable by hybrid breakdown. The lower DEGs reported in F1 hybrids being less parasitized than backcross hybrids is in line with the hybrid advantage.
Collapse
Affiliation(s)
- Andrea Šimková
- Department of Botany and Zoology, Faculty of Science, Masaryk University, Kotlářská 2, 611 37 Brno, Czech Republic; (K.C.K.); (K.V.)
| | - Kristína Civáňová Křížová
- Department of Botany and Zoology, Faculty of Science, Masaryk University, Kotlářská 2, 611 37 Brno, Czech Republic; (K.C.K.); (K.V.)
| | - Kristýna Voříšková
- Department of Botany and Zoology, Faculty of Science, Masaryk University, Kotlářská 2, 611 37 Brno, Czech Republic; (K.C.K.); (K.V.)
| | - Lukáš Vetešník
- Department of Botany and Zoology, Faculty of Science, Masaryk University, Kotlářská 2, 611 37 Brno, Czech Republic; (K.C.K.); (K.V.)
- Institute of Vertebrate Biology of the Czech Academy of Sciences, Květná 8, 603 65 Brno, Czech Republic; (L.V.)
| | - Vojtěch Bystrý
- Central European Institute of Technology, Masaryk University, 625 00 Brno, Czech Republic; (V.B.); (M.D.)
| | - Martin Demko
- Department of Botany and Zoology, Faculty of Science, Masaryk University, Kotlářská 2, 611 37 Brno, Czech Republic; (K.C.K.); (K.V.)
- Central European Institute of Technology, Masaryk University, 625 00 Brno, Czech Republic; (V.B.); (M.D.)
| |
Collapse
|
10
|
Ferreras-Gutiérrez M, Chaves-Arquero B, González-Magaña A, Merino N, Amusategui-Mateu I, Huecas S, Medrano FJ, Blanco FJ. Structural analysis of ING3 protein and histone H3 binding. Int J Biol Macromol 2023; 242:124724. [PMID: 37148949 DOI: 10.1016/j.ijbiomac.2023.124724] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 04/21/2023] [Accepted: 04/30/2023] [Indexed: 05/08/2023]
Abstract
Proteins belonging to the ING family regulate the transcriptional state of chromatin by recruiting remodeling complexes to sites with histone H3 trimethylated at Lysine 4 (H3K4me3). This modification is recognized by the Plant HomeoDomain (PHD) present at the C-terminal region of the five ING proteins. ING3 facilitates acetylation of histones H2A and H4 by the NuA4-Tip60 MYST histone acetyl transferase complex, and it has been proposed to be an oncoprotein. The crystal structure of the N-terminal domain of ING3 shows that it forms homodimers with an antiparallel coiled-coil fold. The crystal structure of the PHD is similar to those of its four homologs. These structures explain the possible deleterious effects of ING3 mutations detected in tumors. The PHD binds histone H3K4me3 with low-micromolar, and binds the non-methylated histone with a 54-fold reduced affinity. Our structure explains the impact of site directed mutagenesis experiments on histone recognition. These structural features could not be confirmed for the full-length protein as solubility was insufficient for structural studies, but the structure of its folded domains suggest a conserved structural organization for the ING proteins as homodimers and bivalent readers of the histone H3K4me3 mark.
Collapse
Affiliation(s)
| | - Belén Chaves-Arquero
- Centro de Investigaciones Biológicas Margarita Salas (CIB), CSIC, Madrid 28040, Spain
| | - Amaia González-Magaña
- Instituto Biofisika and Departamento de Bioquímica y Biología Molecular (CSIC, UPV/EHU), University of the Basque Country, 48940 Leioa, Spain
| | - Nekane Merino
- CIC bioGUNE, Parque Tecnológico de Bizkaia, 48160 Derio, Spain
| | | | - Sonia Huecas
- Centro de Investigaciones Biológicas Margarita Salas (CIB), CSIC, Madrid 28040, Spain
| | - Francisco J Medrano
- Centro de Investigaciones Biológicas Margarita Salas (CIB), CSIC, Madrid 28040, Spain
| | - Francisco J Blanco
- Centro de Investigaciones Biológicas Margarita Salas (CIB), CSIC, Madrid 28040, Spain.
| |
Collapse
|
11
|
Arasawa T, Hiwasa T, Kagaya A, Maruyama T, Uesato M, Kano M, Kobayashi S, Takizawa H, Iwase K, Nomura F, Matsushita K, Matsubara H. Analysis of patients with colorectal cancer shows a specific increase in serum anti-ING1 autoantibody levels. BMC Cancer 2023; 23:356. [PMID: 37072777 PMCID: PMC10111810 DOI: 10.1186/s12885-023-10845-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 04/13/2023] [Indexed: 04/20/2023] Open
Abstract
Colorectal cancer (CRC) is the third most prevalent cancer in the world, yet the sensitivity and specificity of biomarkers for CRC diagnosis are insufficient. In the present study, we performed a protein microarray screening method to identify antibody markers for CRC. Inhibitor of growth family 1 (ING1) was identified as a candidate tumor antigen for CRC using protein microarrays (ProtoArray). Subsequent amplified luminescence proximity homogeneous assay-linked immunosorbent assay using recombinant ING1 protein showed that the serum levels of anti-ING1 antibodies were increased not only in patients with CRC but also in those with esophageal cancer (EC), gastric cancer (GC), breast cancer (BrC), and pancreatic cancer (PC) compared with those of healthy donors (HDs). Antibodies against the ING1 amino acids between 239 and 253 were present at significantly higher levels in patients with CRC than in those with EC, GC, BrC, or PC. Anti-ING1 antibody levels were significantly higher in the patients with CRC at any stages than in the HDs. Immunohistochemical staining revealed higher expression of ING1 protein in CRC cells than in the adjacent normal tissues. In luciferase reporter assays using a CRC cell line, ING1 augmented p53-mediated NOXA promoter activity but attenuated p53-stimulated Bax, p21, and PUMA promoter activities. Consequently, serum anti-ING1 antibodies can be used for sensitive and specific diagnoses of CRC.
Collapse
Affiliation(s)
- Takahiro Arasawa
- Department of Frontier Surgery, Graduate School of Medicine, Chiba University, Chiba, 260-8670, Japan
- Department of Biochemistry and Genetics, Graduate School of Medicine, Chiba University, Chiba, 260-8670, Japan
| | - Takaki Hiwasa
- Department of Biochemistry and Genetics, Graduate School of Medicine, Chiba University, Chiba, 260-8670, Japan.
- Department of Neurological Surgery, Graduate School of Medicine, Chiba University, Inohana 1-8-1, Chuo-Ku, Chiba, 260-8670, Japan.
| | - Akiko Kagaya
- Department of Frontier Surgery, Graduate School of Medicine, Chiba University, Chiba, 260-8670, Japan
- Clinical Research Center, Chiba University Hospital, Chiba, 260-8677, Japan
| | - Tetsuro Maruyama
- Department of Frontier Surgery, Graduate School of Medicine, Chiba University, Chiba, 260-8670, Japan
| | - Masaya Uesato
- Department of Frontier Surgery, Graduate School of Medicine, Chiba University, Chiba, 260-8670, Japan
| | - Masayuki Kano
- Department of Frontier Surgery, Graduate School of Medicine, Chiba University, Chiba, 260-8670, Japan
| | - Sohei Kobayashi
- Department of Frontier Surgery, Graduate School of Medicine, Chiba University, Chiba, 260-8670, Japan
- Department of Laboratory Medicine & Division of Clinical Genetics, Chiba University Hospital, Chiba, 260-8677, Japan
- Department of Medical Technology & Sciences, School of Health Sciences at Narita, International University of Health and Welfare, Chiba, 286-8686, Japan
| | - Hirotaka Takizawa
- Port Square Kashiwado Clinic, Kashiwado Memorial Foundation, Chiba, 260-0025, Japan
| | - Katsuro Iwase
- Department of Biochemistry and Genetics, Graduate School of Medicine, Chiba University, Chiba, 260-8670, Japan
| | - Fumio Nomura
- Division of Clinical Genetics, Chiba Foundation for Health Promotion & Disease Prevention, Chiba, 261-0002, Japan
| | - Kazuyuki Matsushita
- Department of Laboratory Medicine & Division of Clinical Genetics, Chiba University Hospital, Chiba, 260-8677, Japan
| | - Hisahiro Matsubara
- Department of Frontier Surgery, Graduate School of Medicine, Chiba University, Chiba, 260-8670, Japan
| |
Collapse
|
12
|
Azemin WA, Alias N, Ali AM, Shamsir MS. In silico analysis prediction of HepTH1-5 as a potential therapeutic agent by targeting tumour suppressor protein networks. J Biomol Struct Dyn 2023; 41:1141-1167. [PMID: 34935583 DOI: 10.1080/07391102.2021.2017349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Many studies reported that the activation of tumour suppressor protein, p53 induced the human hepcidin expression. However, its expression decreased when p53 was silenced in human hepatoma cells. Contrary to Tilapia hepcidin TH1-5, HepTH1-5 was previously reported to trigger the p53 activation through the molecular docking approach. The INhibitor of Growth (ING) family members are also shown to directly interact with p53 and promote cell cycle arrest, senescence, apoptosis and participate in DNA replication and DNA damage responses to suppress the tumour initiation and progression. However, the interrelation between INGs and HepTH1-5 remains unknown. Therefore, this study aims to identify the mechanism and their protein interactions using in silico approaches. The finding revealed that HepTH1-5 and its ligands had interacted mostly on hotspot residues of ING proteins which involved in histone modifications via acetylation, phosphorylation, and methylation. This proves that HepTH1-5 might implicate in an apoptosis signalling pathway and preserve the protein structure and function of INGs by reducing the perturbation of histone binding upon oxidative stress response. This study would provide theoretical guidance for the design and experimental studies to decipher the role of HepTH1-5 as a potential therapeutic agent for cancer therapy. Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Wan-Atirah Azemin
- Faculty of Bioresources and Food Industry, School of Agriculture Science and Biotechnology, Universiti Sultan Zainal Abidin, Besut, Malaysia.,Faculty of Science, Bioinformatics Research Group (BIRG), Department of Biosciences, Universiti Teknologi Malaysia, Skudai, Malaysia
| | - Nadiawati Alias
- Faculty of Bioresources and Food Industry, School of Agriculture Science and Biotechnology, Universiti Sultan Zainal Abidin, Besut, Malaysia
| | - Abdul Manaf Ali
- Faculty of Bioresources and Food Industry, School of Agriculture Science and Biotechnology, Universiti Sultan Zainal Abidin, Besut, Malaysia
| | - Mohd Shahir Shamsir
- Faculty of Science, Bioinformatics Research Group (BIRG), Department of Biosciences, Universiti Teknologi Malaysia, Skudai, Malaysia.,Faculty of Applied Sciences and Technology, Universiti Tun Hussein Onn Malaysia, Pagoh Higher Education Hub, Muar, Malaysia
| |
Collapse
|
13
|
Li Z, Xu S, Chen L, Huang S, Kuerban X, Li T. Prognostic significance of ING3 expression in patients with cancer: A systematic review and meta-analysis. Front Oncol 2023; 13:1090860. [PMID: 36845697 PMCID: PMC9948604 DOI: 10.3389/fonc.2023.1090860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Accepted: 01/19/2023] [Indexed: 02/11/2023] Open
Abstract
Background It has been reported that ING3 inhibits the progression of various cancers. However, some studies have shown that it promotes the development of prostate cancer. The purpose of this study was to investigate whether ING3 expression is associated with the prognosis of patients with cancer. Materials and methods PubMed, Cochrane Database, Embase, Medline, ScienceDirect, Scopus and Web of Science were searched until September 2022. The hazard ratio (HR)/odds ratio (OR) and 95% confidence interval (95% CI) were calculated using Stata 17 software. We used the Newcastle-Ottawa Scale (NOS) to assess the risk of bias. Result Seven studies involving 2371 patients with five types of cancer were included. The results showed that high expression of ING3 was negatively associated with a more advanced TNM stage (III-IV vs. I-II) (OR=0.61, 95% CI: 0.43-0.86), lymph node metastasis (OR=0.67, 95% CI: 0.49-0.90) and disease-free survival (HR=0.63, 95% CI: 0.37-0.88). However, ING3 expression was not associated with overall survival (HR=0.77, 95% CI: 0.41-1.12), tumor size (OR=0.67, 95% CI: 0.33-1.37), tumor differentiation (OR=0.86, 95% CI: 0.36-2.09) and gender (OR=1.14, 95% CI: 0.78-1.66). Conclusion This study showed that the expression of ING3 was associated with better prognosis, suggesting that ING3 may be a potential biomarker for cancer prognosis. Systematic review registration https://www.crd.york.ac.uk/prospero/, identifier (CRD42022306354).
Collapse
Affiliation(s)
- Zehan Li
- The Department of Surgery, the First Dongguan Affiliated Hospital of Guangdong Medical University, Guangdong, China
| | - Shengchao Xu
- The Department of Surgery, Guangzhou Medical University, Guangdong, China
| | - Lin Chen
- The Department of Surgery, the First Dongguan Affiliated Hospital of Guangdong Medical University, Guangdong, China
| | - Shuqi Huang
- The Department of Surgery, the First Dongguan Affiliated Hospital of Guangdong Medical University, Guangdong, China
| | - Xieyida Kuerban
- The Department of Surgery, the First Dongguan Affiliated Hospital of Guangdong Medical University, Guangdong, China
| | - Tianyu Li
- The Department of Surgery, the First Dongguan Affiliated Hospital of Guangdong Medical University, Guangdong, China,*Correspondence: Tianyu Li,
| |
Collapse
|
14
|
Gül N, Yıldız A. An in silico study of how histone tail conformation affects the binding affinity of ING family proteins. PeerJ 2022; 10:e14029. [PMID: 36199288 PMCID: PMC9528904 DOI: 10.7717/peerj.14029] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 08/16/2022] [Indexed: 01/19/2023] Open
Abstract
Background Due to its intrinsically disordered nature, the histone tail is conformationally heterogenic. Therefore, it provides specific binding sites for different binding proteins or factors through reversible post-translational modifications (PTMs). For instance, experimental studies stated that the ING family binds with the histone tail that has methylation on the lysine in position 4. However, numerous complexes featuring a methylated fourth lysine residue of the histone tail can be found in the UniProt database. So the question arose if other factors like the conformation of the histone tail affect the binding affinity. Methods The crystal structure of the PHD finger domain from the proteins ING1, ING2, ING4, and ING5 are docked to four histone H3 tails with two different conformations using Haddock 2.4 and ClusPro. The best four models for each combination are selected and a two-sample t-test is performed to compare the binding affinities of helical conformations vs. linear conformations using Prodigy. The protein-protein interactions are examined using LigPlot. Results The linear histone conformations in predicted INGs-histone H3 complexes exhibit statistically significant higher binding affinity than their helical counterparts (confidence level of 99%). The outputs of predicted models generated by the molecular docking programs Haddock 2.4 and ClusPro are comparable, and the obtained protein-protein interaction patterns are consistent with experimentally confirmed binding patterns. Conclusion The results show that the conformation of the histone tail is significantly affecting the binding affinity of the docking protein. Herewith, this in silico study demonstrated in detail the binding preference of the ING protein family to histone H3 tail. Further research on the effect of certain PTMs on the final tail conformation and the interaction between those factors seem to be promising for a better understanding of epigenetics.
Collapse
Affiliation(s)
- Nadir Gül
- Faculty of Natural Sciences, Turkish-German University, Istanbul, Turkey
| | - Ahmet Yıldız
- Faculty of Engineering, Turkish-German University, Istanbul, Turkey
| |
Collapse
|
15
|
Taheri M, Hussen BM, Najafi S, Abak A, Ghafouri-Fard S, Samsami M, Baniahmad A. Molecular mechanisms of inhibitor of growth (ING) family members in health and malignancy. Cancer Cell Int 2022; 22:272. [PMID: 36056353 PMCID: PMC9438315 DOI: 10.1186/s12935-022-02693-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 08/22/2022] [Indexed: 11/10/2022] Open
Abstract
ING genes belong to family of tumor suppressor genes with regulatory functions on cell proliferation, apoptosis, and cellular senescence. These include a family of proteins with 5 members (ING1-5), which are downregulated in human malignancies and/or affected by pathogenic mutations. ING proteins are highly evolutionarily conserved proteins containing several domains through which bind to chromatin structures by exerting their effects as readers of histone modification marks, and also binding to proteins like p53 involved in biological processes such as cell cycle regulation. Further, they are known as subunits of histone acetylation as well as deacetylation complexes and so exert their regulatory roles through epigenetic mechanisms. Playing role in restriction of proliferative but also invasive potentials of normal cells, INGs are particularly involved in cancer development and progression. However, additional studies and experimental confirmation are required for these models. This paper highlights the potential impact that INGs may have on the development of human cancer and explores what new information has recently arise on the functions of ING genes.
Collapse
Affiliation(s)
- Mohammad Taheri
- Institute of Human Genetics, Jena University Hospital, Jena, Germany.,Urology and Nephrology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Bashdar Mahmud Hussen
- Department of Pharmacognosy, College of Pharmacy, Hawler Medical University, Kurdistan Region, Erbil, Iraq.,Center of Research and Strategic Studies, Lebanese French University, Erbil, Kurdistan Region, Iraq
| | - Sajad Najafi
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Atefe Abak
- Skull Base Research Center, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Soudeh Ghafouri-Fard
- Department of Medical Genetics, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Majid Samsami
- Cancer Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Aria Baniahmad
- Institute of Human Genetics, Jena University Hospital, Jena, Germany.
| |
Collapse
|
16
|
Xu H, Ye M, Xia A, Jiang H, Huang P, Liu H, Hou R, Wang Q, Li D, Xu JR, Jiang C. The Fng3 ING protein regulates H3 acetylation and H4 deacetylation by interacting with two distinct histone-modifying complexes. THE NEW PHYTOLOGIST 2022; 235:2350-2364. [PMID: 35653584 DOI: 10.1111/nph.18294] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Accepted: 05/30/2022] [Indexed: 05/26/2023]
Abstract
The steady-state level of histone acetylation is maintained by histone acetyltransferase (HAT) and histone deacetylase (HDAC) complexes. INhibitor of Growth (ING) proteins are key components of the HAT or HDAC complexes but their relationship with other components and roles in phytopathogenic fungi are not well-characterized. Here, the FNG3 ING gene was functionally characterized in the wheat head blight fungus Fusarium graminearum. Deletion of FNG3 results in defects in fungal development and pathogenesis. Unlike other ING proteins that are specifically associated with distinct complexes, Fng3 was associated with both NuA3 HAT and FgRpd3 HDAC complexes to regulate H3 acetylation and H4 deacetylation. Whereas FgNto1 mediates the FgSas3-Fng3 interaction in the NuA3 complex, Fng3 interacted with the C-terminal region of FgRpd3 that is present in Rpd3 orthologs from filamentous fungi but absent in yeast Rpd3. The intrinsically disordered regions in the C-terminal tail of FgRpd3 underwent phase separation, which was important for its interaction with Fng3. Furthermore, the ING domain of Fng3 is responsible for its specificities in protein-protein interactions and functions. Taken together, Fng3 is involved in the dynamic regulation of histone acetylation by interacting with two histone modification complexes, and is important for fungal development and pathogenicity.
Collapse
Affiliation(s)
- Huaijian Xu
- State Key Laboratory of Crop Stress Biology for Arid Areas and NWAFU-Purdue Joint Research Center, College of Plant Protection, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Meng Ye
- State Key Laboratory of Crop Stress Biology for Arid Areas and NWAFU-Purdue Joint Research Center, College of Plant Protection, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Aliang Xia
- State Key Laboratory of Crop Stress Biology for Arid Areas and NWAFU-Purdue Joint Research Center, College of Plant Protection, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Hang Jiang
- Institution of Plant Protection, Shandong Academy of Agricultural Sciences, Jinan, Shandong, 250100, China
| | - Panpan Huang
- State Key Laboratory of Crop Stress Biology for Arid Areas and NWAFU-Purdue Joint Research Center, College of Plant Protection, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Huiquan Liu
- State Key Laboratory of Crop Stress Biology for Arid Areas and NWAFU-Purdue Joint Research Center, College of Plant Protection, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Rui Hou
- College of Forestry, Guizhou University, Guiyang, 550025, China
| | - Qinhu Wang
- State Key Laboratory of Crop Stress Biology for Arid Areas and NWAFU-Purdue Joint Research Center, College of Plant Protection, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Dongao Li
- State Key Laboratory of Crop Stress Biology for Arid Areas and NWAFU-Purdue Joint Research Center, College of Plant Protection, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Jin-Rong Xu
- Department of Botany and Plant Pathology, Purdue University, West Lafayette, IN, 47907, USA
| | - Cong Jiang
- State Key Laboratory of Crop Stress Biology for Arid Areas and NWAFU-Purdue Joint Research Center, College of Plant Protection, Northwest A&F University, Yangling, Shaanxi, 712100, China
| |
Collapse
|
17
|
Inhibitor of Growth Factors Regulate Cellular Senescence. Cancers (Basel) 2022; 14:cancers14133107. [PMID: 35804879 PMCID: PMC9264871 DOI: 10.3390/cancers14133107] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 06/20/2022] [Accepted: 06/21/2022] [Indexed: 12/26/2022] Open
Abstract
Simple Summary Five members of the Inhibitor of Growth (ING) family share a highly conserved plant homeodomian with affinity to the specific histone modification H3K4me3. Since some ING family members are preferentially associated with histone acetyltransferaseactivity while other members with histone deacetlyse activity, the ING family membres are epigenetic regulators. Interestingly, ING members can regulate the induction cellular senescence in both primray untransformed human cells as well as human cancer cells. We discuss here the up-to-date knowledge about their regulatory activity within the cellular senescent program. Abstract The Inhibitor of Growth (ING) proteins are a group of tumor suppressors with five conserved genes. A common motif of ING factors is the conserved plant homeodomain (PHD), with which they bind to chromatin as readers of the histone mark trimethylated histone H3 (H3K4me3). These genes often produce several protein products through alternative splicing events. Interestingly, ING1 and ING2 participate in the establishment of the repressive mSIN3a-HDAC complexes, whereas ING3, ING4, and ING5 are associated with the activating HAT protein complexes. In addition to the modulation of chromatin’s structure, they regulate cell cycle transition, cellular senescence, repair of DNA damage, apoptosis, and angiogenic pathways. They also have fundamental effects on regulating cellular senescence in cancer cells. In the current review, we explain their role in cellular senescence based on the evidence obtained from cell line and animal studies, particularly in the context of cancer.
Collapse
|
18
|
Cheng F, Zhao S, Li J, Niu Y, Huang H, Yang J, Ma S, Liu J, Sun P. Enhanced effect of recombinant adenoviruses co‐expression of
ING4
and
OSM
on anti‐tumour activity of laryngeal cancer. J Cell Mol Med 2022; 26:1556-1566. [PMID: 35075768 PMCID: PMC8899183 DOI: 10.1111/jcmm.17192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2021] [Revised: 01/02/2022] [Accepted: 01/08/2022] [Indexed: 11/29/2022] Open
Abstract
The inhibitor of growth family member 4 (ING4) is one of the ING family genes, serves as a repressor of angiogenesis or tumour growth and suppresses loss of contact inhibition. Oncostatin M (OSM) is a multifunctional cytokine that belongs to the interleukin (IL)‐6 subfamily with several biological activities. However, the role of recombinant adenoviruses co‐expressing ING4 and OSM (Ad‐ING4‐OSM) in anti‐tumour activity of laryngeal cancer has not yet been identified. Recombinant Ad‐ING4‐OSM was used to evaluate their combined effect on enhanced anti‐tumour activity in Hep‐2 cells of laryngeal cancer in vivo. Moreover, in vitro function assays of co‐expression of Ad‐ING4‐OSM were performed to explore impact of co‐expression of Ad‐ING4‐OSM on biological phenotype of laryngeal cancer cell line, that is Hep‐2 cells. In vitro, Ad‐ING4‐OSM significantly inhibited the growth, enhanced apoptosis, altered cell cycle with G1 and G2/M phase arrest, and upregulated the expression of P21, P27, P53 and downregulated survivin in laryngeal cancer Hep‐2 cells. Furthermore, in vivo functional experiments of co‐expressing of Ad‐ING4‐OSM demonstrated that solid tumours in the nude mouse model were significantly suppressed, and the co‐expressing Ad‐ING4‐OSM showed a significant upregulation expression of P21, P53, Bax and Caspase‐3 and a downregulation of Cox‐2, Bcl‐2 and CD34. This study for the first time demonstrated the clinical value and the role of co‐expressing Ad‐ING4‐OSM in biological function of laryngeal cancer. This work suggested that co‐expressing Ad‐ING4‐OSM might serve as a potential therapeutic target for laryngeal cancer patients.
Collapse
Affiliation(s)
- Fuwei Cheng
- Department of Otolaryngology The First Affiliated Hospital of Soochow University Suzhou China
| | - Shuangping Zhao
- Department of Otolaryngology The First Affiliated Hospital of Soochow University Suzhou China
| | - Jiachen Li
- Department of Otolaryngology The First Affiliated Hospital of Soochow University Suzhou China
| | - Yuyu Niu
- Department of Otolaryngology The First Affiliated Hospital of Soochow University Suzhou China
| | - Haiping Huang
- Department of Otolaryngology The First Affiliated Hospital of Soochow University Suzhou China
| | - Jicheng Yang
- Cell and Molecular Biology Institute College of Medicine Soochow University Suzhou China
| | - Shiyin Ma
- Department of Otolaryngology The First Affiliated Hospital of Bengbu Medical College Bengbu China
| | - Jisheng Liu
- Department of Otolaryngology The First Affiliated Hospital of Soochow University Suzhou China
| | - Peng Sun
- Department of Otolaryngology The First Affiliated Hospital of Soochow University Suzhou China
| |
Collapse
|
19
|
Rusu AD, Cornhill ZE, Coutiño BC, Uribe MC, Lourdusamy A, Markus Z, May ST, Rahman R, Georgiou M. CG7379 and ING1 suppress cancer cell invasion by maintaining cell-cell junction integrity. Open Biol 2021; 11:210077. [PMID: 34493070 PMCID: PMC8424350 DOI: 10.1098/rsob.210077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Approximately 90% of cancer-related deaths can be attributed to a tumour's ability to spread. We have identified CG7379, the fly orthologue of human ING1, as a potent invasion suppressor. ING1 is a type II tumour suppressor with well-established roles in the transcriptional regulation of genes that control cell proliferation, response to DNA damage, oncogene-induced senescence and apoptosis. Recent work suggests a possible role for ING1 in cancer cell invasion and metastasis, but the molecular mechanism underlying this observation is lacking. Our results show that reduced expression of CG7379 promotes invasion in vivo in Drosophila, reduces the junctional localization of several adherens and septate junction components, and severely disrupts cell-cell junction architecture. Similarly, ING1 knockdown significantly enhances invasion in vitro and disrupts E-cadherin distribution at cell-cell junctions. A transcriptome analysis reveals that loss of ING1 affects the expression of several junctional and cytoskeletal modulators, confirming ING1 as an invasion suppressor and a key regulator of cell-cell junction integrity.
Collapse
Affiliation(s)
- Alexandra D. Rusu
- School of Life Sciences, University of Nottingham, Nottingham NG7 2UH, UK,Leicester Institute for Structural and Chemical Biology, Department of Molecular and Cell Biology, University of Leicester, Leicester LE1 9HN, UK
| | - Zoe E. Cornhill
- School of Life Sciences, University of Nottingham, Nottingham NG7 2UH, UK
| | - Brenda Canales Coutiño
- School of Life Sciences, University of Nottingham, Nottingham NG7 2UH, UK,Department of Cell and Developmental Biology, University College London, Gower Street, London WC1E 6BT, UK
| | | | - Anbarasu Lourdusamy
- School of Medicine, Biodiscovery Institute, University of Nottingham, Nottingham NG7 2RD, UK
| | - Zsuzsa Markus
- School of Life Sciences, University of Nottingham, Nottingham NG7 2UH, UK
| | - Sean T. May
- School of Biosciences, University of Nottingham, Sutton Bonington, Leicestershire LE12 5RD, UK
| | - Ruman Rahman
- School of Medicine, Biodiscovery Institute, University of Nottingham, Nottingham NG7 2RD, UK
| | - Marios Georgiou
- School of Life Sciences, University of Nottingham, Nottingham NG7 2UH, UK
| |
Collapse
|
20
|
Antithetic hTERT Regulation by Androgens in Prostate Cancer Cells: hTERT Inhibition Is Mediated by the ING1 and ING2 Tumor Suppressors. Cancers (Basel) 2021; 13:cancers13164025. [PMID: 34439179 PMCID: PMC8391603 DOI: 10.3390/cancers13164025] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 08/02/2021] [Accepted: 08/05/2021] [Indexed: 02/06/2023] Open
Abstract
Simple Summary The expression of the catalytic subunit of the human telomerase reverse transcriptase subunit (hTERT) is hormonally controlled. Androgen treatment suppresses the hTERT expression at a transcriptional level in prostate cancer cells. Here, we identified the responsive promoter element that mediates the androgen receptor induced transrepression of hTERT. The negative androgen response element (nARE) is identified as 62 bp located in the core promoter of hTERT. Chromatin immunoprecipitations indicate an androgen-dependent recruitment of the androgen receptor (AR) ING1 and ING2 to the hTERT promoter. Interestingly, the androgen-induced transrepression is mediated by the class II tumor suppressors inhibitor of growth 1 and 2, namely ING1 and ING2, respectively. Abstract The human telomerase is a key factor during tumorigenesis in prostate cancer (PCa). The androgen receptor (AR) is a key drug target controlling PCa growth and regulates hTERT expression, but is described to either inhibit or to activate. Here, we reveal that androgens repress and activate hTERT expression in a concentration-dependent manner. Physiological low androgen levels activate, while, notably, supraphysiological androgen levels (SAL), used in bipolar androgen therapy (BAT), repress hTERT expression. We confirmed the SAL-mediated gene repression of hTERT in PCa cell lines, native human PCa samples derived from patients treated ex vivo, as well as in cancer spheroids derived from androgen-dependent or castration resistant PCa (CRPC) cells. Interestingly, chromatin immuno-precipitation (ChIP) combined with functional assays revealed a positive (pARE) and a negative androgen response element (nARE). The nARE was narrowed down to 63 bp in the hTERT core promoter region. AR and tumor suppressors, inhibitor of growth 1 and 2 (ING1 and ING2, respectively), are androgen-dependently recruited. Mechanistically, knockdown indicates that ING1 and ING2 mediate AR-regulated transrepression. Thus, our data suggest an oppositional, biphasic function of AR to control the hTERT expression, while the inhibition of hTERT by androgens is mediated by the AR co-repressors ING1 and ING2.
Collapse
|
21
|
Shatnawi A, Abu Rabe DI, Frigo DE. Roles of the tumor suppressor inhibitor of growth family member 4 (ING4) in cancer. Adv Cancer Res 2021; 152:225-262. [PMID: 34353439 DOI: 10.1016/bs.acr.2021.05.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Inhibitor of growth family member 4 (ING4) is best known as a tumor suppressor that is frequently downregulated, deleted, or mutated in many cancers. ING4 regulates a broad array of tumor-related processes including proliferation, apoptosis, migration, autophagy, invasion, angiogenesis, DNA repair and chromatin remodeling. ING4 alters local chromatin structure by functioning as an epigenetic reader of H3K4 trimethylation histone marks (H3K4Me3) and regulating gene transcription through directing histone acetyltransferase (HAT) and histone deacetylase (HDAC) protein complexes. ING4 may serve as a useful prognostic biomarker for many cancer types and help guide treatment decisions. This review provides an overview of ING4's central functions in gene expression and summarizes current literature on the role of ING4 in cancer and its possible use in therapy.
Collapse
Affiliation(s)
- Aymen Shatnawi
- Department of Pharmaceutical and Administrative Sciences, University of Charleston School of Pharmacy, Charleston, WV, United States.
| | - Dina I Abu Rabe
- Integrated Bioscience Program, North Carolina Central University, Durham, NC, United States
| | - Daniel E Frigo
- Department of Cancer Systems Imaging, University of Texas MD Anderson Cancer Center, Houston, TX, United States; Department of Genitourinary Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| |
Collapse
|
22
|
Huang YJ, Cao ZF, Wang J, Yang J, Wei YJ, Tang YC, Cheng YX, Zhou J, Zhang ZX. Why MUC16 mutations lead to a better prognosis: A study based on The Cancer Genome Atlas gastric cancer cohort. World J Clin Cases 2021; 9:4143-4158. [PMID: 34141777 PMCID: PMC8173414 DOI: 10.12998/wjcc.v9.i17.4143] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 01/25/2021] [Accepted: 03/05/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND MUC16, encoding cancer antigen 125, is a frequently mutated gene in gastric cancer. In addition, MUC16 mutations seem to result in a better prognosis in gastric cancer. However, the mechanisms that lead to a better prognosis by MUC16 mutations have not yet been clarified. AIM To delve deeper into the underlying mechanisms that explain why MUC16 mutations signal a better prognosis in gastric cancer. METHODS We used multi-omics data, including mRNA, simple nucleotide variation, copy number variation and methylation data from The Cancer Genome Atlas, to explore the relationship between MUC16 mutations and prognosis. Cox regression and random survival forest algorithms were applied to search for hub genes. Gene set enrichment analysis was used to elucidate the molecular mechanisms. Single-sample gene set enrichment analysis and "EpiDISH" were used to assess immune cells infiltration, and "ESTIMATE" for analysis of the tumor microenvironment. RESULTS Our study found that compared to the wild-type group, the mutation group had a better prognosis. Additional analysis indicated that the MUC16 mutations appear to activate the DNA repair and p53 pathways to act as an anti-tumor agent. We also identified a key gene, NPY1R (neuropeptide Y receptor Y1), which was significantly more highly expressed in the MUC16 mutations group than in the MUC16 wild-type group. The high expression of NPY1R predicted a poorer prognosis, which was also confirmed in a separate Gene Expression Omnibus cohort. Further susceptibility analysis revealed that NPY1R might be a potential drug target for gastric cancer. Furthermore, in the analysis of the tumor microenvironment, we found that immune cells in the mutation group exhibited higher anti-tumor effects. In addition, the tumor mutation burden and cancer stem cells index were also higher in the mutation group than in the wild-type group. CONCLUSION We speculated that the MUC16 mutations might activate the p53 pathway and DNA repair pathway: alternatively, the tumor microenvironment may be involved.
Collapse
Affiliation(s)
- Yu-Jie Huang
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu Province, China
| | - Zhi-Fei Cao
- Department of Pathology, The Second Affiliated Hospital of Soochow University, Suzhou 215004, Jiangsu Province, China
| | - Jie Wang
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu Province, China
| | - Jian Yang
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu Province, China
| | - Yi-Jun Wei
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu Province, China
| | - Yu-Chen Tang
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu Province, China
| | - Yin-Xiang Cheng
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu Province, China
| | - Jian Zhou
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu Province, China
| | - Zi-Xiang Zhang
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu Province, China
| |
Collapse
|
23
|
ING2 tumor suppressive protein translocates into mitochondria and is involved in cellular metabolism homeostasis. Oncogene 2021; 40:4111-4123. [PMID: 34017078 DOI: 10.1038/s41388-021-01832-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 04/25/2021] [Accepted: 05/05/2021] [Indexed: 02/04/2023]
Abstract
ING2 (Inhibitor of Growth 2) is a tumor suppressor gene that has been implicated in critical biological functions (cell-cycle regulation, replicative senescence, DNA repair and DNA replication), most of which are recognized hallmarks of tumorigenesis occurring in the cell nucleus. As its close homolog ING1 has been recently observed in the mitochondrial compartment, we hypothesized that ING2 could also translocate into the mitochondria and be involved in new biological functions. In the present study, we demonstrate that ING2 is imported in the inner mitochondrial fraction in a redox-sensitive manner in human cells and that this mechanism is modulated by 14-3-3η protein expression. Remarkably, ING2 is necessary to maintain mitochondrial ultrastructure integrity without interfering with mitochondrial networks or polarization. We observed an interaction between ING2 and mtDNA under basal conditions. This interaction appears to be mediated by TFAM, a critical regulator of mtDNA integrity. The loss of mitochondrial ING2 does not impair mtDNA repair, replication or transcription but leads to a decrease in mitochondrial ROS production, suggesting a detrimental impact on OXPHOS activity. We finally show using multiple models that ING2 is involved in mitochondrial respiration and that its loss confers a protection against mitochondrial respiratory chain inhibition in vitro. Consequently, we propose a new tumor suppressor role for ING2 protein in the mitochondria as a metabolic shift gatekeeper during tumorigenesis.
Collapse
|
24
|
Yang D, Su Z, Wei G, Long F, Zhu YC, Ni T, Liu X, Zhu YZ. H3K4 Methyltransferase Smyd3 Mediates Vascular Smooth Muscle Cell Proliferation, Migration, and Neointima Formation. Arterioscler Thromb Vasc Biol 2021; 41:1901-1914. [PMID: 33827259 DOI: 10.1161/atvbaha.121.314689] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
[Figure: see text].
Collapse
MESH Headings
- Animals
- Carotid Arteries/enzymology
- Carotid Arteries/pathology
- Carotid Artery Injuries/enzymology
- Carotid Artery Injuries/genetics
- Carotid Artery Injuries/pathology
- Carotid Stenosis/enzymology
- Carotid Stenosis/genetics
- Carotid Stenosis/pathology
- Cell Movement
- Cell Proliferation
- Cells, Cultured
- Disease Models, Animal
- Histone-Lysine N-Methyltransferase/genetics
- Histone-Lysine N-Methyltransferase/metabolism
- Male
- Mice, Inbred C57BL
- Mice, Knockout
- Muscle, Smooth, Vascular/enzymology
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/enzymology
- Myocytes, Smooth Muscle/pathology
- Neointima
- Rats
- Signal Transduction
- Vascular Remodeling
- Mice
Collapse
Affiliation(s)
- Di Yang
- Pharmacophenomics Laboratory, Human Phenome Institute, Fudan University, Shanghai, 201203 P.R. China (D.Y., Z.H.S., F.L., X.H.L.)
- State Key Laboratory of Quality Research in Chinese Medicine and School of Pharmacy, Macau University of Science and Technology, Macau, China (D.Y., Y.Z.Z.)
| | - Zhenghua Su
- Pharmacophenomics Laboratory, Human Phenome Institute, Fudan University, Shanghai, 201203 P.R. China (D.Y., Z.H.S., F.L., X.H.L.)
| | - Gang Wei
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center of Genetics and Development, Human Phenome Institute, School of Life Sciences, Fudan University, Shanghai, 200438 P.R. China (G.W., T.N.)
| | - Fen Long
- Pharmacophenomics Laboratory, Human Phenome Institute, Fudan University, Shanghai, 201203 P.R. China (D.Y., Z.H.S., F.L., X.H.L.)
| | - Yi-Chun Zhu
- Shanghai Key Laboratory of Bioactive Small Molecules and Research Center on Aging and Medicine, Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China (Y.C.Z.)
| | - Ting Ni
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center of Genetics and Development, Human Phenome Institute, School of Life Sciences, Fudan University, Shanghai, 200438 P.R. China (G.W., T.N.)
| | - Xinhua Liu
- Pharmacophenomics Laboratory, Human Phenome Institute, Fudan University, Shanghai, 201203 P.R. China (D.Y., Z.H.S., F.L., X.H.L.)
| | - Yi Zhun Zhu
- State Key Laboratory of Quality Research in Chinese Medicine and School of Pharmacy, Macau University of Science and Technology, Macau, China (D.Y., Y.Z.Z.)
| |
Collapse
|
25
|
Fng1 is involved in crosstalk between histone acetylation and methylation. Curr Genet 2021; 67:535-538. [PMID: 33641041 DOI: 10.1007/s00294-021-01167-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 02/18/2021] [Accepted: 02/19/2021] [Indexed: 10/22/2022]
Abstract
The histone modifications usually form complicated networks to regulate accessibility of DNA and transcription. Identification of proteins that are involved in the crosstalk among different histone modifications will help to better understand the epigenetic regulatory network in eukaryotes. The Inhibitor of Growth (ING) proteins represent a tumor suppressor family were first linked to histone modification in yeast and their functions in epigenetic regulation were further characterized. This review summarizes the crosstalk of histone modification in fungi and describes recently achieved mechanistic insights into the role of Fng1 (an ING protein in filamentous ascomycetes) in this process. We conclude that Fng1 is involved in crosstalk among histone acetylation, deacetylation and methylation.
Collapse
|
26
|
Zhou R, Rotte A, Li G, Chen X, Chen G, Bhandaru M. Nuclear localization of ING3 is required to suppress melanoma cell migration, invasion and angiogenesis. Biochem Biophys Res Commun 2020; 527:418-424. [PMID: 32334834 DOI: 10.1016/j.bbrc.2020.04.056] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2020] [Accepted: 04/12/2020] [Indexed: 02/08/2023]
Abstract
Inhibitor of growth family member 3 (ING3), a tumor suppressor, plays crucial roles in cell cycle regulation, apoptosis and transcription. Previous studies suggest important roles of nuclear ING3, however, the nuclear localization sequence (NLS) of ING3 is not defined and its biological functions remain to be elucidated. In this study, various ING3 mutants were generated to identify its NLS. The NLS of ING3 was determined as KKFK between 164 and 167 amino acids. More intriguingly, replacement of Lysine 164 residue of ING3 with alanine (K164A) resulted in retention of ING3 in the cytoplasm. Overexpression of ING3 led to inhibition of melanoma cell migration, invasion, and angiogenesis respectively, however, this inhibition was abrogated in cells with overexpression of ING3-K164A mutant. In conclusion, this study identified the NLS of ING3 and demonstrated the significance of ING3 nuclear localization for tumor suppressive functions of ING3, and future studies await to elucidate the role of ING3 (K164) post-modificaton in its nuclear transportation and cancer development.
Collapse
Affiliation(s)
- Ruiyao Zhou
- Department of General Surgery, The Third Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China.
| | - Anand Rotte
- Department of Dermatology and Skin Science, Jack Bell Research Centre, Vancouver Coastal Health Research Institute, University of British Columbia, Vancouver, British Columbia, Canada.
| | - Gang Li
- Department of Dermatology and Skin Science, Jack Bell Research Centre, Vancouver Coastal Health Research Institute, University of British Columbia, Vancouver, British Columbia, Canada.
| | - Xiaolei Chen
- Department of General Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China.
| | - Guangdi Chen
- Department of Dermatology and Skin Science, Jack Bell Research Centre, Vancouver Coastal Health Research Institute, University of British Columbia, Vancouver, British Columbia, Canada; Bioelectromagnetics Laboratory, Department of Public Health, Zhejiang University School of Medicine, China.
| | - Madhuri Bhandaru
- Department of Dermatology and Skin Science, Jack Bell Research Centre, Vancouver Coastal Health Research Institute, University of British Columbia, Vancouver, British Columbia, Canada.
| |
Collapse
|
27
|
Cheng S, Li C, Xie W, Miao Y, Guo J, Wang J, Zhang Y. Integrated analysis of DNA methylation and mRNA expression profiles to identify key genes involved in the regrowth of clinically non-functioning pituitary adenoma. Aging (Albany NY) 2020; 12:2408-2427. [PMID: 32015217 PMCID: PMC7041752 DOI: 10.18632/aging.102751] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Accepted: 01/07/2020] [Indexed: 12/15/2022]
Abstract
Tumour regrowth is a key characteristic of clinically non-functioning pituitary adenoma (NFPA). No applicable prognosis evaluation method is available for post-operative patients. We aimed to identify DNA methylation biomarkers that can facilitate prognosis evaluation. Genome-wide DNA methylation and mRNA microarray analyses were performed for tumour samples from 71 NFPA patients. Differentially expressed genes and methylated genes were identified based on the regrowth vs non-regrowth grouping. There were 139 genes that showed alterations in methylation status and expression level, and only 13 genes showed a negative correlation. The progression-free analysis found that FAM90A1, ETS2, STAT6, MYT1L, ING2 and KCNK1 are related to tumour regrowth. A prognosis-prediction model was built based on all 13 genes from integrated analysis, and the 6-gene model achieved the best area under the receiver operating characteristic curves (AUC) of 0.820, compared with 0.785 and 0.568 for the 13-gene and 7-gene models, respectively. Our prognostic biomarkers were validated by pyrosequencing and RT-PCR. FAM90A1 and ING2 was found to be independent prognostic factors of tumour regrowth with univariate Cox regression. The DNA methylation and expression levels of FAM90A1 and ING2 are associated with tumour regrowth, and may serve as biomarkers for predicting the prognosis of patients with NFPA.
Collapse
Affiliation(s)
- Sen Cheng
- Beijing Neurosurgical Institute, Capital Medical University, Beijing 100070, China
| | - Chuzhong Li
- Beijing Neurosurgical Institute, Beijing Tiantan Hospital Affiliated to Capital Medical University, Beijing Institute for Brain Disorders Brain Tumour Center, China National Clinical Research Center for Neurological Diseases, Key Laboratory of Central Nervous System Injury Research, Beijing 100070, China
| | - Weiyan Xie
- Beijing Neurosurgical Institute, Capital Medical University, Beijing 100070, China
| | - Yazhou Miao
- Beijing Neurosurgical Institute, Capital Medical University, Beijing 100070, China
| | - Jing Guo
- Beijing Neurosurgical Institute, Capital Medical University, Beijing 100070, China
| | - Jichao Wang
- People's Hospital of Xin Jiang Uygur Autonomous Region, Urumqi 830001, China
| | - Yazhuo Zhang
- Beijing Neurosurgical Institute, Beijing Tiantan Hospital Affiliated to Capital Medical University, Beijing Institute for Brain Disorders Brain Tumour Center, China National Clinical Research Center for Neurological Diseases, Key Laboratory of Central Nervous System Injury Research, Beijing 100070, China
| |
Collapse
|
28
|
Barlak N, Capik O, Sanli F, Kilic A, Aytatli A, Yazici A, Ortucu S, Ittmann M, Karatas OF. ING5 inhibits cancer aggressiveness by inhibiting Akt and activating p53 in prostate cancer. Cell Biol Int 2020; 44:242-252. [PMID: 31475765 DOI: 10.1002/cbin.11227] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Accepted: 08/22/2019] [Indexed: 02/06/2023]
Abstract
Prostate cancer (PCa) is one of the most common types of cancer in men. In several recent studies, chromosomal deletions in the q arm of chromosome 2, where ING5 resides within, have been identified in various cancer types including PCa. In this study, we investigate the role of ING5 as a tumor suppressor in PCa. We examined the expression level of ING5 in tissue samples and cell lines using quantitative real-time polymerase chain reaction and western blot analysis. We tested the in vitro tumor suppressor potential of ING5 in PC3 and LNCaP cells stably overexpressing it using cell viability, colony formation, migration, invasion, and apoptosis assays. We then investigated the effects of ING5 on the Akt and p53 signaling using western blot analysis. We show that ING5 is significantly downregulated in PCa tumor tissue samples and cell lines compared with the corresponding controls. In vitro assays demonstrate that ING5 effectively suppresses proliferative, clonogenic, migratory, and invasive potential and induce apoptosis in PCa cells. ING5 may potentially exert its anti-tumor potential by inhibiting AKT and inducing p53 signaling pathways. Our findings demonstrate that ING5 possesses tumor suppressor roles in vitro, pointing its importance during the prostatic carcinogenesis processes.
Collapse
Affiliation(s)
- Neslisah Barlak
- Department of Molecular Biology and Genetics, Erzurum Technical University, Erzurum, 25250, Turkey
| | - Ozel Capik
- Department of Molecular Biology and Genetics, Erzurum Technical University, Erzurum, 25250, Turkey
| | - Fatma Sanli
- Department of Molecular Biology and Genetics, Erzurum Technical University, Erzurum, 25250, Turkey
| | - Ahsen Kilic
- Department of Molecular Biology and Genetics, Erzurum Technical University, Erzurum, 25250, Turkey
| | - Abdulmelik Aytatli
- Department of Molecular Biology and Genetics, Erzurum Technical University, Erzurum, 25250, Turkey
| | - Aysenur Yazici
- Department of Molecular Biology and Genetics, Erzurum Technical University, Erzurum, 25250, Turkey
| | - Serkan Ortucu
- Department of Molecular Biology and Genetics, Erzurum Technical University, Erzurum, 25250, Turkey
| | - Michael Ittmann
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, Texas, 77030, USA.,Michael E. DeBakey VAMC, Houston, Texas, 77030, USA
| | - Omer Faruk Karatas
- Department of Molecular Biology and Genetics, Erzurum Technical University, Erzurum, 25250, Turkey
| |
Collapse
|
29
|
Espinosa-Cores L, Bouza-Morcillo L, Barrero-Gil J, Jiménez-Suárez V, Lázaro A, Piqueras R, Jarillo JA, Piñeiro M. Insights Into the Function of the NuA4 Complex in Plants. FRONTIERS IN PLANT SCIENCE 2020; 11:125. [PMID: 32153620 PMCID: PMC7047200 DOI: 10.3389/fpls.2020.00125] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Accepted: 01/28/2020] [Indexed: 05/14/2023]
Abstract
Chromatin remodeling plays a key role in the establishment and maintenance of gene expression patterns essential for plant development and responses to environmental factors. Post-translational modification of histones, including acetylation, is one of the most relevant chromatin remodeling mechanisms that operate in eukaryotic cells. Histone acetylation is an evolutionarily conserved chromatin signature commonly associated with transcriptional activation. Histone acetylation levels are tightly regulated through the antagonistic activity of histone acetyltransferases (HATs) and histone deacetylases (HDACs). In plants, different families of HATs are present, including the MYST family, which comprises homologs of the catalytic subunit of the Nucleosome Acetyltransferase of H4 (NuA4) complex in yeast. This complex mediates acetylation of histones H4, H2A, and H2A.Z, and is involved in transcriptional regulation, heterochromatin silencing, cell cycle progression, and DNA repair in yeast. In Arabidopsis and, other plant species, homologs for most of the yeast NuA4 subunits are present and although the existence of this complex has not been demonstrated yet, compelling evidence supports the notion that this type of HAT complex functions from mosses to angiosperms. Recent proteomic studies show that several Arabidopsis homologs of NuA4 components, including the assembly platform proteins and the catalytic subunit, are associated in vivo with additional members of this complex suggesting that a NuA4-like HAT complex is present in plants. Furthermore, the functional characterization of some Arabidopsis NuA4 subunits has uncovered the involvement of these proteins in the regulation of different plant biological processes. Interestingly, for most of the mutant plants deficient in subunits of this complex characterized so far, conspicuous defects in flowering time are observed, suggesting a role for NuA4 in the control of this plant developmental program. Moreover, the participation of Arabidopsis NuA4 homologs in other developmental processes, such as gametophyte development, as well as in cell proliferation and stress and hormone responses, has also been reported. In this review, we summarize the current state of knowledge on plant putative NuA4 subunits and discuss the latest progress concerning the function of this chromatin modifying complex.
Collapse
|
30
|
Li X, Zhang Q, Zhang M, Luo Y, Fu Y. Downregulation of nuclear ING3 expression and translocalization to cytoplasm promotes tumorigenesis and progression in head and neck squamous cell carcinoma (HNSCC). Histol Histopathol 2019; 35:681-690. [PMID: 31886514 DOI: 10.14670/hh-18-197] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
ING3 (inhibitor of growth gene 3) is a member of the ING gene family, and is considered as a candidate tumor suppressor gene. In order to explore the roles of ING3 in tumorigenesis and cancer progression of head and neck squamous cell carcinoma (HNSCC), ING3 expression was assessed in 173 cases of HNSCC by immunohistochemistry. The expression of ING3 was also compared to clinicopathological variables, and the expression of several tumorigenic markers. Nuclear expression of ING3 in HNSCC was significantly lower than that in dysplasia and normal epithelium, and was negatively correlated with a poor-differentiated status, T staging and TNM staging. In contrast, cytoplasmic expression of ING3 was significantly increased in HNSCC, and was statistically associated with lymph node metastasis and 14-3-3η expression. In addition, nuclear expression of ING3 was positively correlated with the expression of p300, p21 and acetylated p53. In conclusion, decreases in nuclear ING3 may play important roles in tumorigenesis, progression and tumor differentiation in HNSCC. Increases in cytoplasmic ING3 may be due to 14-3-3η binding and may also be involved in malignant progression. Nuclear ING3 may modulate the transactivation of target genes, promoting apoptosis through interactions with p300 and p21. Moreover, ING3 may interact with p300 to upregulate the level of acetylation of p53, and promote p53-mediated cell cycle arrest, senescence and/or apoptosis. Therefore, ING3 may be a potential tumor suppressor and a possible therapeutic target in HNSCC.
Collapse
Affiliation(s)
- Xiaohan Li
- Department of Pathology, Shengjing Hospital of China Medical University, Shenyang, China.
| | - Qun Zhang
- Department of Pathology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Mingming Zhang
- Department of Pathology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yusong Luo
- Department of Pathology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yaping Fu
- Department of Pathology, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
31
|
Fink D, Yau T, Nabbi A, Wagner B, Wagner C, Hu SM, Lang V, Handschuh S, Riabowol K, Rülicke T. Loss of Ing3 Expression Results in Growth Retardation and Embryonic Death. Cancers (Basel) 2019; 12:cancers12010080. [PMID: 31905726 PMCID: PMC7017303 DOI: 10.3390/cancers12010080] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 12/19/2019] [Accepted: 12/24/2019] [Indexed: 12/29/2022] Open
Abstract
The ING3 candidate tumour suppressor belongs to a family of histone modifying proteins involved in regulating cell proliferation, senescence, apoptosis, chromatin remodeling, and DNA repair. It is a stoichiometric member of the minimal NuA4 histone acetyl transferase (HAT) complex consisting of EAF6, EPC1, ING3, and TIP60. This complex is responsible for the transcription of an essential cascade of genes involved in embryonic development and in tumour suppression. ING3 has been linked to head and neck and hepatocellular cancers, although its status as a tumour suppressor has not been well established. Recent studies suggest a pro-metastasis role in prostate cancer progression. Here, we describe a transgenic mouse strain with insertional mutation of an UbC-mCherry expression cassette into the endogenous Ing3 locus, resulting in the disruption of ING3 protein expression. Homozygous mutants are embryonically lethal, display growth retardation, and severe developmental disorders. At embryonic day (E) 10.5, the last time point viable homozygous embryos were found, they were approximately half the size of heterozygous mice that develop normally. µCT analysis revealed a developmental defect in neural tube closure, resulting in the failure of formation of closed primary brain vesicles in homozygous mid-gestation embryos. This is consistent with high ING3 expression levels in the embryonic brains of heterozygous and wild type mice and its lack in homozygous mutant embryos that show a lack of ectodermal differentiation. Our data provide direct evidence that ING3 is an essential factor for normal embryonic development and that it plays a fundamental role in prenatal brain formation.
Collapse
Affiliation(s)
- Dieter Fink
- Institute of Laboratory Animal Science, University of Veterinary Medicine Vienna, 1210 Vienna, Austria; (T.Y.); (B.W.); (S.M.H.); (V.L.); (T.R.)
- Correspondence: ; Tel.: +43-(0)-1-25077-2820
| | - Tienyin Yau
- Institute of Laboratory Animal Science, University of Veterinary Medicine Vienna, 1210 Vienna, Austria; (T.Y.); (B.W.); (S.M.H.); (V.L.); (T.R.)
| | - Arash Nabbi
- Departments of Biochemistry & Molecular Biology and Oncology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada; (A.N.); (K.R.)
| | - Bettina Wagner
- Institute of Laboratory Animal Science, University of Veterinary Medicine Vienna, 1210 Vienna, Austria; (T.Y.); (B.W.); (S.M.H.); (V.L.); (T.R.)
| | - Christine Wagner
- Division of Immunology, Allergy and Infectious Diseases (DIAID), Department of Dermatology, Medical University of Vienna, 1090 Vienna, Austria;
| | - Shiting Misaki Hu
- Institute of Laboratory Animal Science, University of Veterinary Medicine Vienna, 1210 Vienna, Austria; (T.Y.); (B.W.); (S.M.H.); (V.L.); (T.R.)
| | - Viktor Lang
- Institute of Laboratory Animal Science, University of Veterinary Medicine Vienna, 1210 Vienna, Austria; (T.Y.); (B.W.); (S.M.H.); (V.L.); (T.R.)
| | - Stephan Handschuh
- VetImaging, VetCore Facility for Research, University of Veterinary Medicine Vienna, 1210 Vienna, Austria;
| | - Karl Riabowol
- Departments of Biochemistry & Molecular Biology and Oncology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada; (A.N.); (K.R.)
| | - Thomas Rülicke
- Institute of Laboratory Animal Science, University of Veterinary Medicine Vienna, 1210 Vienna, Austria; (T.Y.); (B.W.); (S.M.H.); (V.L.); (T.R.)
| |
Collapse
|
32
|
Archambeau J, Blondel A, Pedeux R. Focus-ING on DNA Integrity: Implication of ING Proteins in Cell Cycle Regulation and DNA Repair Modulation. Cancers (Basel) 2019; 12:cancers12010058. [PMID: 31878273 PMCID: PMC7017203 DOI: 10.3390/cancers12010058] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 12/20/2019] [Accepted: 12/21/2019] [Indexed: 12/16/2022] Open
Abstract
The ING family of tumor suppressor genes is composed of five members (ING1-5) involved in cell cycle regulation, DNA damage response, apoptosis and senescence. All ING proteins belong to various HAT or HDAC complexes and participate in chromatin remodeling that is essential for genomic stability and signaling pathways. The gatekeeper functions of the INGs are well described by their role in the negative regulation of the cell cycle, notably by modulating the stability of p53 or the p300 HAT activity. However, the caretaker functions are described only for ING1, ING2 and ING3. This is due to their involvement in DNA repair such as ING1 that participates not only in NERs after UV-induced damage, but also in DSB repair in which ING2 and ING3 are required for accumulation of ATM, 53BP1 and BRCA1 near the lesion and for the subsequent repair. This review summarizes evidence of the critical roles of ING proteins in cell cycle regulation and DNA repair to maintain genomic stability.
Collapse
|
33
|
Dantas A, Al Shueili B, Yang Y, Nabbi A, Fink D, Riabowol K. Biological Functions of the ING Proteins. Cancers (Basel) 2019; 11:E1817. [PMID: 31752342 PMCID: PMC6896041 DOI: 10.3390/cancers11111817] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Accepted: 11/07/2019] [Indexed: 02/06/2023] Open
Abstract
The proteins belonging to the inhibitor of growth (ING) family of proteins serve as epigenetic readers of the H3K4Me3 histone mark of active gene transcription and target histone acetyltransferase (HAT) or histone deacetylase (HDAC) protein complexes, in order to alter local chromatin structure. These multidomain adaptor proteins interact with numerous other proteins to facilitate their localization and the regulation of numerous biochemical pathways that impinge upon biological functions. Knockout of some of the ING genes in murine models by various groups has verified their status as tumor suppressors, with ING1 knockout resulting in the formation of large clear-cell B-lymphomas and ING2 knockout increasing the frequency of ameloblastomas, among other phenotypic effects. ING4 knockout strongly affects innate immunity and angiogenesis, and INGs1, ING2, and ING4 have been reported to affect apoptosis in different cellular models. Although ING3 and ING5 knockouts have yet to be published, preliminary reports indicate that ING3 knockout results in embryonic lethality and that ING5 knockout may have postpartum effects on stem cell maintenance. In this review, we compile the known information on the domains of the INGs and the effects of altering ING protein expression, to better understand the functions of this adaptor protein family and its possible uses for targeted cancer therapy.
Collapse
Affiliation(s)
- Arthur Dantas
- Arnie Charbonneau Cancer Institute, Departments of Biochemistry and Molecular Biology and Oncology, University of Calgary, 374 HMRB, 3330 Hospital Dr. NW, Calgary, AB T2N 4N1, Canada; (A.D.); (B.A.S.); (Y.Y.)
| | - Buthaina Al Shueili
- Arnie Charbonneau Cancer Institute, Departments of Biochemistry and Molecular Biology and Oncology, University of Calgary, 374 HMRB, 3330 Hospital Dr. NW, Calgary, AB T2N 4N1, Canada; (A.D.); (B.A.S.); (Y.Y.)
| | - Yang Yang
- Arnie Charbonneau Cancer Institute, Departments of Biochemistry and Molecular Biology and Oncology, University of Calgary, 374 HMRB, 3330 Hospital Dr. NW, Calgary, AB T2N 4N1, Canada; (A.D.); (B.A.S.); (Y.Y.)
| | - Arash Nabbi
- Princess Margaret Cancer Centre, Toronto, ON M5G 2M9, Canada
| | - Dieter Fink
- Institute of Laboratory Animal Science, Department for Biomedical Sciences, University of Veterinary Medicine Vienna, 1210 Vienna, Austria;
| | - Karl Riabowol
- Arnie Charbonneau Cancer Institute, Departments of Biochemistry and Molecular Biology and Oncology, University of Calgary, 374 HMRB, 3330 Hospital Dr. NW, Calgary, AB T2N 4N1, Canada; (A.D.); (B.A.S.); (Y.Y.)
| |
Collapse
|
34
|
Ormaza G, Rodríguez JA, Ibáñez de Opakua A, Merino N, Villate M, Gorroño I, Rábano M, Palmero I, Vilaseca M, Kypta R, Vivanco MDM, Rojas AL, Blanco FJ. The Tumor Suppressor ING5 Is a Dimeric, Bivalent Recognition Molecule of the Histone H3K4me3 Mark. J Mol Biol 2019; 431:2298-2319. [PMID: 31026448 DOI: 10.1016/j.jmb.2019.04.018] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Revised: 04/10/2019] [Accepted: 04/10/2019] [Indexed: 10/26/2022]
Abstract
The INhibitor of Growth (ING) family of tumor suppressors regulates the transcriptional state of chromatin by recruiting remodeling complexes to sites with histone H3 trimethylated at lysine 4 (H3K4me3). This modification is recognized by the plant homeodomain (PHD) present at the C-terminus of the five ING proteins. ING5 facilitates histone H3 acetylation by the HBO1 complex, and also H4 acetylation by the MOZ/MORF complex. We show that ING5 forms homodimers through its N-terminal domain, which folds independently into an elongated coiled-coil structure. The central region of ING5, which contains the nuclear localization sequence, is flexible and disordered, but it binds dsDNA with micromolar affinity. NMR analysis of the full-length protein reveals that the two PHD fingers of the dimer are chemically equivalent and independent of the rest of the molecule, and they bind H3K4me3 in the same way as the isolated PHD. We have observed that ING5 can form heterodimers with the highly homologous ING4, and that two of three primary tumor-associated mutants in the N-terminal domain strongly destabilize the coiled-coil structure. They also affect cell proliferation and cell cycle phase distribution, suggesting a driver role in cancer progression.
Collapse
Affiliation(s)
- Georgina Ormaza
- CIC bioGUNE, Parque Tecnológico de Bizkaia, 48160 Derio, Spain
| | | | | | - Nekane Merino
- CIC bioGUNE, Parque Tecnológico de Bizkaia, 48160 Derio, Spain
| | - Maider Villate
- CIC bioGUNE, Parque Tecnológico de Bizkaia, 48160 Derio, Spain
| | - Irantzu Gorroño
- CIC bioGUNE, Parque Tecnológico de Bizkaia, 48160 Derio, Spain
| | - Miriam Rábano
- CIC bioGUNE, Parque Tecnológico de Bizkaia, 48160 Derio, Spain
| | - Ignacio Palmero
- Instituto de Investigaciones Biomédicas "Alberto Sols", CSIC-UAM, 28029 Madrid, Spain
| | - Marta Vilaseca
- Institute for Research in Biomedicine, 08028 Barcelona, Spain
| | - Robert Kypta
- CIC bioGUNE, Parque Tecnológico de Bizkaia, 48160 Derio, Spain; Department of Surgery and Cancer, Imperial College London, London, W12 0NN, UK
| | | | - Adriana L Rojas
- CIC bioGUNE, Parque Tecnológico de Bizkaia, 48160 Derio, Spain
| | - Francisco J Blanco
- CIC bioGUNE, Parque Tecnológico de Bizkaia, 48160 Derio, Spain; IKERBASQUE, Basque Foundation for Science, 48011 Bilbao, Spain.
| |
Collapse
|
35
|
Gao J, Yang Y, Qiu R, Zhang K, Teng X, Liu R, Wang Y. Proteomic analysis of the OGT interactome: novel links to epithelial-mesenchymal transition and metastasis of cervical cancer. Carcinogenesis 2019; 39:1222-1234. [PMID: 30052810 PMCID: PMC6175026 DOI: 10.1093/carcin/bgy097] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Accepted: 07/22/2018] [Indexed: 12/19/2022] Open
Abstract
The role of O-GlcNAc transferase (OGT) in gene regulation and tumor invasion is poorly understood. Here, we have identified several previously undiscovered OGT-interacting proteins, including the PRMT5/WDR77 complex, the PRC2 complex, the ten-eleven translocation (TET) family, the CRL4B complex and the nucleosome remodeling and deacetylase (NuRD) complex. Genome-wide analysis of target genes responsive to OGT resulted in identification of a cohort of genes including SNAI1 and ING4 that are critically involved in cell epithelial–mesenchymal transition and invasion/metastasis. We have demonstrated that OGT promotes carcinogenesis and metastasis of cervical cancer cells. OGT’s expression is significantly upregulated in cervical cancer, and low OGT level is correlated with improved prognosis. Our study has thus revealed a mechanistic link between OGT and tumor progression, providing potential prognostic indicators and targets for cancer therapy.
Collapse
Affiliation(s)
- Jie Gao
- 2011 Collaborative Innovation Center of Tianjin for Medical Epigenetics, Tianjin Key Laboratory of Cellular and Molecular Immunology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Yang Yang
- 2011 Collaborative Innovation Center of Tianjin for Medical Epigenetics, Tianjin Key Laboratory of Cellular and Molecular Immunology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Rongfang Qiu
- 2011 Collaborative Innovation Center of Tianjin for Medical Epigenetics, Tianjin Key Laboratory of Cellular and Molecular Immunology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Kai Zhang
- 2011 Collaborative Innovation Center of Tianjin for Medical Epigenetics, Tianjin Key Laboratory of Cellular and Molecular Immunology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Xu Teng
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Ruiqiong Liu
- Cancer Center, The Second Hospital of Shandong University, Jinan, China
| | - Yan Wang
- 2011 Collaborative Innovation Center of Tianjin for Medical Epigenetics, Tianjin Key Laboratory of Cellular and Molecular Immunology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China.,Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| |
Collapse
|
36
|
Mouche A, Archambeau J, Ricordel C, Chaillot L, Bigot N, Guillaudeux T, Grenon M, Pedeux R. ING3 is required for ATM signaling and DNA repair in response to DNA double strand breaks. Cell Death Differ 2019; 26:2344-2357. [PMID: 30804473 DOI: 10.1038/s41418-019-0305-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Revised: 12/21/2018] [Accepted: 01/28/2019] [Indexed: 01/16/2023] Open
Abstract
Inhibitor of Growth 3 (ING3) is a candidate tumor suppressor gene whose expression is lost in tumors such as hepatocellular carcinoma, head and neck squamous cell carcinoma and melanoma. In the present study, we show that ING3-depleted human cells and yeast cells deleted for its ortholog YNG2 are sensitive to DNA damage suggesting a conserved role in response to such stress. In human cells, ING3 is recruited to DNA double strand breaks and is required for ATM activation. Remarkably, in response to doxorubicin, ATM activation is dependent on ING3 but not on TIP60, whose recruitment to DNA breaks also depends on ING3. These events lead to ATM-mediated phosphorylation of NBS1 and the subsequent recruitment of RNF8, RNF168, 53BP1, and BRCA1, which are major mediators of the DNA damage response. Accordingly, upon genotoxic stress, DNA repair by non-homologous end joining (NHEJ) or homologous recombination (HR) were impaired in absence of ING3. Finally, immunoglobulin class switch recombination (CSR), a physiological mechanism requiring NHEJ repair, was impaired in the absence of ING3. Since deregulation of DNA double strand break repair is associated with genomic instability, we propose a novel function of ING3 as a caretaker tumor suppressor involved in the DNA damage signaling and repair.
Collapse
Affiliation(s)
- Audrey Mouche
- INSERM U1242, Chemistry Oncogenesis Stress and Signaling, CLCC Eugène Marquis, Rennes, France.,Université de Rennes 1, Rennes, France.,INSERM U1236, MICMAC, Rennes, France
| | - Jérôme Archambeau
- INSERM U1242, Chemistry Oncogenesis Stress and Signaling, CLCC Eugène Marquis, Rennes, France.,Université de Rennes 1, Rennes, France
| | - Charles Ricordel
- INSERM U1242, Chemistry Oncogenesis Stress and Signaling, CLCC Eugène Marquis, Rennes, France.,Université de Rennes 1, Rennes, France
| | - Laura Chaillot
- INSERM U1242, Chemistry Oncogenesis Stress and Signaling, CLCC Eugène Marquis, Rennes, France.,Université de Rennes 1, Rennes, France.,UMS Biosit, SFR Biologie-Santé, Rennes, France
| | - Nicolas Bigot
- Université de Rennes 1, Rennes, France.,INSERM U1236, MICMAC, Rennes, France.,Genome Damage and Stability Centre, University of Sussex, Falmer, Brighton, BN1 9RQ, UK
| | - Thierry Guillaudeux
- INSERM U1242, Chemistry Oncogenesis Stress and Signaling, CLCC Eugène Marquis, Rennes, France.,Université de Rennes 1, Rennes, France.,UMS Biosit, SFR Biologie-Santé, Rennes, France
| | - Muriel Grenon
- Biochemistry, School of Natural Sciences, National University of Ireland, Galway, Ireland
| | - Rémy Pedeux
- INSERM U1242, Chemistry Oncogenesis Stress and Signaling, CLCC Eugène Marquis, Rennes, France. .,Université de Rennes 1, Rennes, France.
| |
Collapse
|
37
|
The essential role of tumor suppressor gene ING4 in various human cancers and non-neoplastic disorders. Biosci Rep 2019; 39:BSR20180773. [PMID: 30643005 PMCID: PMC6356015 DOI: 10.1042/bsr20180773] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Revised: 12/19/2018] [Accepted: 01/13/2019] [Indexed: 12/21/2022] Open
Abstract
Inhibitor of growth 4 (ING4), a member of the ING family discovered in 2003, has been shown to act as a tumor suppressor and is frequently down-regulated in various human cancers. Numerous published in vivo and in vitro studies have shown that ING4 is responsible for important cancer hallmarks such as pathologic cell cycle arrest, apoptosis, autophagy, contact inhibition, and hypoxic adaptation, and also affects tumor angiogenesis, invasion, and metastasis. These characteristics are typically associated with regulation through chromatin acetylation by binding histone H3 trimethylated at lysine 4 (H3K4me3) and through transcriptional activity of transcription factor P53 and NF-κB. In addition, emerging evidence has indicated that abnormalities in ING4 expression and function play key roles in non-neoplastic disorders. Here, we provide an overview of ING4-modulated chromosome remodeling and transcriptional function, as well as the functional consequences of different genetic variants. We also present the current understanding concerning the role of ING4 in the development of neoplastic and non-neoplastic diseases. These studies offer inspiration for pursuing novel therapeutics for various cancers.
Collapse
|
38
|
Regulat-INGs in tumors and diseases: Focus on ncRNAs. Cancer Lett 2019; 447:66-74. [PMID: 30673590 DOI: 10.1016/j.canlet.2019.01.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Revised: 12/21/2018] [Accepted: 01/08/2019] [Indexed: 12/11/2022]
Abstract
ING family genes (Inhibitor of Growth) are tumor suppressor genes that play a vital role in cell homeostasis. It has been shown that their expression is lost or diminished in many cancers and other diseases. The main mechanisms by which they are regulated in oncogenesis have not yet been fully elucidated. The involvement of non-coding RNAs (ncRNAs) and in particular microRNAs (miRNAs) in post-transcriptional gene regulation is well established. miRNAs are short sequences (18-25 nucleotides) that can bind to the 3 'UTR sequence of the targeted messenger RNA (mRNA), leading to its degradation or translational repression. Interactions between the ING family and miRNAs have been described in some cancers but also in other diseases. The involvement of miRNAs in ING family regulation opens up new fields of investigation, particularly for targeted therapies. In this review, we will summarize the regulatory mechanisms at the RNA and protein level of the ING family and focus on the interactions with ncRNAs.
Collapse
|
39
|
Inhibitor of growth 3 induces cell death by regulating cell proliferation, apoptosis and cell cycle arrest by blocking the PI3K/AKT pathway. Cancer Gene Ther 2018; 25:240-247. [DOI: 10.1038/s41417-018-0023-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Revised: 03/18/2018] [Accepted: 03/24/2018] [Indexed: 12/14/2022]
|
40
|
Ye P, Ke X, Zang X, Sun H, Dong Z, Lin J, Wang L, Liu W, Miao G, Tan Y, Tong W, Xiao H, Gao L. Up-regulated MiR-27-3p promotes the G1-S phase transition by targeting inhibitor of growth family member 5 in osteosarcoma. Biomed Pharmacother 2018; 101:219-227. [PMID: 29494959 DOI: 10.1016/j.biopha.2018.02.066] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Revised: 02/14/2018] [Accepted: 02/15/2018] [Indexed: 12/12/2022] Open
Abstract
OBJECTIVE MicroRNAs (miRNAs) play an essential role in regulating malignant progression of tumour cells by inhibiting translation or stability of messenger RNA. However, the expression pattern and regulatory mechanism of miR-27-3p in osteosarcoma remains unclear. METHODS We examined the expression of miR-27-3p in 5 osteosarcoma cell lines compared with that in 2 normal osteocyte cell lines. Osteosarcoma cells U-2OS and MG-63 were transduced to up-regulate or down-regulate the expression of miR-27-3p. The 3-(4, 5-Dimethyl-2-thiazolyl)-2, 5-diphenyl-2H-tetrazolium bromide, or MTT, assay, colony formation assays, BrdUrd labelling, immunofluorescence, anchorage-independent growth ability assay and flow cytometry analysis were used to test the effect of miR-27-3p. Luciferase assays were added to verify the direct relationship between miR-27-3p and the predicted target gene inhibitor of growth family member 5 (ING5). RESULTS The expression of miR-27-3p was significantly increased in examined osteosarcoma cell lines compared with that in normal osteocyte cell lines. Up-regulation of miR-27-3p significantly accelerated osteosarcoma cell growth via promoting G1-S transition. In addition, the opposite result was observed in miR-27-3p-down-regulated cells. Up-regulation of ING5 significantly attenuated the miR-27-3p-induced proliferation in osteosarcoma cells. CONCLUSIONS These data suggested that miR-27-3p could promote the G1-S phase transition that leads to proliferation by down-regulating the expression of ING5 in osteosarcoma.
Collapse
Affiliation(s)
- Pei Ye
- Department of Orthopedics, Nanhai Affiliated Hospital of Southern Medical University, Foshan, Guangdong, 528000, China
| | - Xueping Ke
- Department of Gastroenterology, the Liwan Hospital of the Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510175, China
| | - Xuehui Zang
- Department of Orthopedics, Nanhai Affiliated Hospital of Southern Medical University, Foshan, Guangdong, 528000, China
| | - Hui Sun
- Department of Orthopedics, Nanhai Affiliated Hospital of Southern Medical University, Foshan, Guangdong, 528000, China
| | - Zhixing Dong
- Department of Radiology, Nanhai Affiliated Hospital of Southern Medical University, Foshan, Guangdong, 528000, China
| | - Jun Lin
- Department of Orthopedics, Nanhai Affiliated Hospital of Southern Medical University, Foshan, Guangdong, 528000, China
| | - Lihui Wang
- Department of Orthopedics, Nanhai Affiliated Hospital of Southern Medical University, Foshan, Guangdong, 528000, China
| | - Wenzhou Liu
- Department of Orthopedics, Nanhai Affiliated Hospital of Southern Medical University, Foshan, Guangdong, 528000, China
| | - Guiqiang Miao
- Department of Orthopedics, Nanhai Affiliated Hospital of Southern Medical University, Foshan, Guangdong, 528000, China
| | - Yongtao Tan
- Department of Orthopedics, Nanhai Affiliated Hospital of Southern Medical University, Foshan, Guangdong, 528000, China
| | - Weilai Tong
- Department of Orthopedics, Nanhai Affiliated Hospital of Southern Medical University, Foshan, Guangdong, 528000, China
| | - Haichang Xiao
- Department of Orthopedics, Nanhai Affiliated Hospital of Southern Medical University, Foshan, Guangdong, 528000, China
| | - Lihua Gao
- Department of Orthopedics, Nanhai Affiliated Hospital of Southern Medical University, Foshan, Guangdong, 528000, China.
| |
Collapse
|
41
|
Gao Y, Ma H, Gao C, Lv Y, Chen X, Xu R, Sun M, Liu X, Lu X, Pei X, Li P. Tumor-promoting properties of miR-8084 in breast cancer through enhancing proliferation, suppressing apoptosis and inducing epithelial-mesenchymal transition. J Transl Med 2018; 16:38. [PMID: 29471858 PMCID: PMC5824560 DOI: 10.1186/s12967-018-1419-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Accepted: 02/18/2018] [Indexed: 02/06/2023] Open
Abstract
Background Breast cancer is one of the most frequent malignancies and the second leading cause of cancer-related mortality in women. MicroRNAs play a key role in breast cancer development and progression. microRNA(miR)-8084 has been observed an aberrant expression in breast cancer. However, the functions and regulatory axes of miR-8084, particularly in breast cancer, were not entirely clear. Methods miR-8084 expression in breast cancer were investigated in a GEO dataset by in silico analysis and in 42 paired tumor tissues by qPCR. The effects of deregulation of miR-8084 on breast cancer cell proliferation, migration and invasion in vitro and tumorigenicity in vivo were examined by colony-formation assay, wound healing assay, transwell assay and nude mouse subcutaneous tumor formation model. The target gene of miR-8084 were predicted by TargetScan and miRDB, and confirmed by luciferase reporter system. The roles of miR-8084 in the breast cancer cell proliferation, apoptosis and epithelial–mesenchymal transition (EMT) were investigated by MTS, FACS and associated-marker detection by western blot. Results miR-8084 is significantly up-regulated in both serum and malignant tissues from the source of breast cancer patients. miR-8084 promotes the proliferation of breast cancer cells by activating ERK1/2 and AKT. Meanwhile miR-8084 inhibits apoptosis by decreasing p53-BAX related pathway. miR-8084 also enhances migration and invasion by inducing EMT. Moreover, the tumor suppressor ING2 is a potential target of miR-8084, and miR-8084 regulatory axes contribute to pro-tumor effect, at least partially through regulating ING2. Conclusion Our results strongly suggest that miR-8084 functions as an oncogene that promotes the development and progression of breast cancer, and miR-8084 is a potential new diagnostic marker and therapeutic target of breast cancer.
Collapse
Affiliation(s)
- Yujing Gao
- Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, China.
| | - Hongning Ma
- Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, China
| | - Chanchan Gao
- Department of Oncology, Zhongda Hospital, Southeast University, Nanjing, China
| | - Ye Lv
- Oncology Department of Cancer Hospital, General Hospital, Ningxia Medical University, Yinchuan, China
| | - XueHua Chen
- Department of Pediatrics, Ruijin Hospital and Ruijin Hospital North, Shanghai Jiao Tong University School of Medicine, 197 Ruijin Er Rd, Shanghai, 200025, People's Republic of China
| | - Rongrong Xu
- Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, China
| | - Miao Sun
- Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, China
| | - Xinrui Liu
- Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, China
| | - Xiaohong Lu
- Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, China
| | - Xiuying Pei
- Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, China.
| | - Pu Li
- Department of Pediatrics, Ruijin Hospital and Ruijin Hospital North, Shanghai Jiao Tong University School of Medicine, 197 Ruijin Er Rd, Shanghai, 200025, People's Republic of China.
| |
Collapse
|
42
|
Zhang GJ, Zhao J, Jiang ML, Zhang LC. ING5 inhibits cell proliferation and invasion in esophageal squamous cell carcinoma through regulation of the Akt/NF-κB/MMP-9 signaling pathway. Biochem Biophys Res Commun 2018; 496:387-393. [DOI: 10.1016/j.bbrc.2018.01.045] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2017] [Accepted: 01/07/2018] [Indexed: 12/20/2022]
|
43
|
Ohkouchi C, Kumamoto K, Saito M, Ishigame T, Suzuki SI, Takenoshita S, Harris CC. ING2, a tumor associated gene, enhances PAI‑1 and HSPA1A expression with HDAC1 and mSin3A through the PHD domain and C‑terminal. Mol Med Rep 2017; 16:7367-7374. [PMID: 28944862 PMCID: PMC5865867 DOI: 10.3892/mmr.2017.7553] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2017] [Accepted: 09/12/2017] [Indexed: 12/21/2022] Open
Abstract
Inhibitor of growth 2 (ING2) is involved in chromatin remodeling and it has previously been suggested that ING2 may regulate gene expression. The authors previously identified matrix metalloproteinase 13 (MMP13) as a target gene of ING2 in colorectal cancer. The aim of the present study was to identify novel genes regulated by ING2 and histone deacetylase 1 (HDAC1) and to clarify the biological significance of the ING2 structure. The present study generated the point mutant constructs of ING2 and deletion constructs consisting of partial ING2 to investigate the effect on gene expression and verify the interaction with HDAC1, mSin3A and sap30. A microarray was performed to find novel ING2/HDAC1 target genes using cell co-overexpression of ING2 and HDAC1. Plasminogen activator inhibitor-1 (PAI-1) was upregulated with overexpression of ING1b and ING2. The mutation of the PHD domain at 218 significantly attenuated the MMP13 and PAI-1 expression, whereas the mutation at 224 resulted in increased expression. Furthermore, the expression levels were slightly reduced by the mutation of the C-terminal. The lack of the PHD domain and the C-terminal in ING2 resulted in a decreased ability to induce gene expression. The C-terminal with PHD domain, which lacked the N-terminal, maintained the transactive function for regulating the target genes. In addition to MMP13 and PAI-1, eight genes [heat shock protein family A member 1A (HSPA1A), MIR7-3 host gene, chorionic somatomammotropin hormone 1, growth arrest and DNA damage inducible b, dehydrogenase/reductase 2, galectin 1, myosin light chain 1, and VGF nerve growth factor inducible] were demonstrated to be associated with ING2/HDAC1. The present study demonstrated that ING2/HDAC1 regulated PAI-1 and HSPA1A expression and the PHD domain and the C-terminal of ING2, which are binding sites of HDAC1 and mSin3A, are essential regions for the regulation of gene expression.
Collapse
Affiliation(s)
- Chiyo Ohkouchi
- Department of Thyroid and Endocrinology, Fukushima Medical University School of Medicine, Fukushima 960‑1295, Japan
| | - Kensuke Kumamoto
- Department of Coloproctology, Aizu Medical Center, Fukushima Medical University, Aizuwakamatsu, Fukushima 969‑3492, Japan
| | - Motonobu Saito
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Teruhide Ishigame
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Shin-Ichi Suzuki
- Department of Thyroid and Endocrinology, Fukushima Medical University School of Medicine, Fukushima 960‑1295, Japan
| | - Seiichi Takenoshita
- Department of Organ Regulatory Surgery, Fukushima Medical University School of Medicine, Fukushima 960‑1295, Japan
| | - Cutis C Harris
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
44
|
Sica V, Bravo-San Pedro JM, Chen G, Mariño G, Lachkar S, Izzo V, Maiuri MC, Niso-Santano M, Kroemer G. Inhibitor of growth protein 4 interacts with Beclin 1 and represses autophagy. Oncotarget 2017; 8:89527-89538. [PMID: 29163768 PMCID: PMC5685689 DOI: 10.18632/oncotarget.19033] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Accepted: 04/17/2017] [Indexed: 12/14/2022] Open
Abstract
Beclin 1 (BECN1) is a multifunctional protein that activates the pro-autophagic class III phosphatidylinositol 3-kinase (PIK3C3, best known as VPS34), yet also interacts with multiple negative regulators. Here we report that BECN1 interacts with inhibitor of growth family member 4 (ING4), a tumor suppressor protein that is best known for its capacity to interact with the tumor suppressor protein p53 (TP53) and the acetyltransferase E1A binding protein p300 (EP300). Removal of TP53 or EP300 did not affect the BECN1/ING4 interaction, which however was lost upon culture of cells in autophagy-inducing, nutrient free conditions. Depletion of ING4 stimulated the enzymatic activity of PIK3C3, as visualized by means of a red fluorescent protein-tagged short peptide (FYVE) that specifically binds to phosphatidylinositol-3-phosphate (PI3P)-containing subcellular vesicles and enhanced autophagy, as indicated by an enhanced lipidation of microtubule-associated proteins 1A/1B light chain 3 beta (LC3B) and the redistribution of a green-fluorescent protein (GFP)-LC3B fusion protein to cytoplasmic puncta. The generation of GFP-LC3B puncta stimulated by ING4 depletion was reduced by simultaneous depletion, or pharmacological inhibition, of PIK3C3/VPS34. In conclusion, ING4 acts as a negative regulator of the lipid kinase activity of the BECN1 complex, and starvation-induced autophagy is accompanied by the dissociation of the ING4/BECN1 interaction.
Collapse
Affiliation(s)
- Valentina Sica
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France.,Equipe 11 labellisée Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers, Paris, France.,Institut National de la Santé et de la Recherche Médicale, Paris, France.,Université Pierre et Marie Curie, Paris, France.,Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France
| | - José Manuel Bravo-San Pedro
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France.,Equipe 11 labellisée Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers, Paris, France.,Institut National de la Santé et de la Recherche Médicale, Paris, France.,Université Pierre et Marie Curie, Paris, France.,Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France
| | - Guo Chen
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France.,Equipe 11 labellisée Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers, Paris, France.,Institut National de la Santé et de la Recherche Médicale, Paris, France.,Université Pierre et Marie Curie, Paris, France.,Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France
| | - Guillermo Mariño
- Institut National de la Santé et de la Recherche Médicale, Paris, France.,Departamento de Biología Fundamental, Instituto de Investigación Sanitaria del Principado de Asturias, Universidad de Oviedo, Spain
| | - Sylvie Lachkar
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France.,Equipe 11 labellisée Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers, Paris, France.,Institut National de la Santé et de la Recherche Médicale, Paris, France.,Université Pierre et Marie Curie, Paris, France.,Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France
| | - Valentina Izzo
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France.,Equipe 11 labellisée Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers, Paris, France.,Institut National de la Santé et de la Recherche Médicale, Paris, France.,Université Pierre et Marie Curie, Paris, France.,Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France
| | - Maria Chiara Maiuri
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France.,Equipe 11 labellisée Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers, Paris, France.,Institut National de la Santé et de la Recherche Médicale, Paris, France.,Université Pierre et Marie Curie, Paris, France.,Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France
| | - Mireia Niso-Santano
- Institut National de la Santé et de la Recherche Médicale, Paris, France.,Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas, Cáceres, Spain.,Facultad de Enfermería y Terapia Ocupacional, Universidad de Extremadura, C.P, Cáceres, Cáceres, Spain
| | - Guido Kroemer
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France.,Equipe 11 labellisée Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers, Paris, France.,Institut National de la Santé et de la Recherche Médicale, Paris, France.,Université Pierre et Marie Curie, Paris, France.,Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France.,Pôle de Biologie, Hôpital Européen Georges Pompidou, AP-HP, Paris, France.,Department of Women's and Children's Health, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
45
|
Hamann BL, Blind RD. Nuclear phosphoinositide regulation of chromatin. J Cell Physiol 2017; 233:107-123. [PMID: 28256711 DOI: 10.1002/jcp.25886] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Accepted: 03/01/2017] [Indexed: 12/26/2022]
Abstract
Phospholipid signaling has clear connections to a wide array of cellular processes, particularly in gene expression and in controlling the chromatin biology of cells. However, most of the work elucidating how phospholipid signaling pathways contribute to cellular physiology have studied cytoplasmic membranes, while relatively little attention has been paid to the role of phospholipid signaling in the nucleus. Recent work from several labs has shown that nuclear phospholipid signaling can have important roles that are specific to this cellular compartment. This review focuses on the nuclear phospholipid functions and the activities of phospholipid signaling enzymes that regulate metazoan chromatin and gene expression. In particular, we highlight the roles that nuclear phosphoinositides play in several nuclear-driven physiological processes, such as differentiation, proliferation, and gene expression. Taken together, the recent discovery of several specifically nuclear phospholipid functions could have dramatic impact on our understanding of the fundamental mechanisms that enable tight control of cellular physiology.
Collapse
Affiliation(s)
- Bree L Hamann
- Division of Diabetes Endocrinology and Metabolism, Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Raymond D Blind
- Division of Diabetes Endocrinology and Metabolism, Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee.,Departments of Medicine, Biochemistry and Pharmacology, Division of Diabetes Endocrinology and Metabolism, The Vanderbilt Diabetes Research and Training Center and the Vanderbilt-Ingram Cancer Center, Nashville, Tennessee
| |
Collapse
|
46
|
LincRNa-p21: function and mechanism in cancer. Med Oncol 2017; 34:98. [PMID: 28425074 DOI: 10.1007/s12032-017-0959-5] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Accepted: 04/12/2017] [Indexed: 12/29/2022]
Abstract
In view of the rapid development of gene chips and high-throughput sequencing technology, noncoding RNAs (ncRNas) form a high percentage of the mammalian genome. Two major subgroups of ncRNAs that have been identified are the long ncRNAs (lncRNas) and the microRNAs. A number of studies in the past few years have showed crucial functions for lncRNas in cancer. LincRNa-p21 as a p53-dependent transcriptional target gene and a potential diagnostic marker is involved in proliferation, cell cycle, metabolism and reprogramming. In addition, more researches revealed that lincRNa-p21 is associated with cancer progression and contributed to the treatment and prognosis of cancer. In this review, we briefly summarize the function and molecular mechanisms of lincRNa-p21 in cancer and its regulation for the genes expression .
Collapse
|
47
|
Zhang R, Jin J, Shi J, Hou Y. INGs are potential drug targets for cancer. J Cancer Res Clin Oncol 2017; 143:189-197. [PMID: 27544390 DOI: 10.1007/s00432-016-2219-z] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Accepted: 08/09/2016] [Indexed: 12/11/2022]
Abstract
PURPOSE The inhibitor of growth (ING) family consists of ING1, ING2, ING3, ING4 and ING5, which function as the type II tumor suppressors. INGs regulate cell proliferation, senescence, apoptosis, differentiation, angiogenesis, DNA repair, metastasis, and invasion by multiple pathways. In addition, INGs increase cancer cell sensitivity for chemotherapy and radiotherapy, while clinical observations show that INGs are frequently lost in some types of cancers. The aim of the study was to summarize the recent progress regarding INGs regulating tumor progression. METHODS The literatures of INGs regulating tumor progression were searched and assayed. RESULTS The regulating signaling pathways of ING1, ING2, ING3 or ING4 on tumor progression were shown. The mechanisms of INGs on tumor suppression were also assayed. CONCLUSIONS This review better summarized the signaling mechanism of INGs on tumor suppression, which provides a candidate therapy strategy for cancers.
Collapse
Affiliation(s)
- Runyun Zhang
- Department of Oncology, Affiliated Wujin People's Hospital, Jiangsu University, Changzhou, 212017, People's Republic of China
- Institute of Life Sciences, Jiangsu University, Zhenjiang, 212013, People's Republic of China
| | - Jianhua Jin
- Department of Oncology, Affiliated Wujin People's Hospital, Jiangsu University, Changzhou, 212017, People's Republic of China
| | - Juanjuan Shi
- Institute of Life Sciences, Jiangsu University, Zhenjiang, 212013, People's Republic of China.
| | - Yongzhong Hou
- Department of Oncology, Affiliated Wujin People's Hospital, Jiangsu University, Changzhou, 212017, People's Republic of China.
- Institute of Life Sciences, Jiangsu University, Zhenjiang, 212013, People's Republic of China.
| |
Collapse
|
48
|
Affiliation(s)
- Gesche Tallen
- Department of Paediatrics, Faculty of Medicine, University of Calgary, Canada.,Department of Paediatric Haematology and Oncology, University Medical Centre Hamburg-Eppendorf (UKE), Germany
| | - Karl Riabowol
- Departments of Biochemistry & Molecular Biology and Oncology, Cumming School of Medicine, University of Calgary, Canada
| |
Collapse
|
49
|
Ormaza G, Medagli B, Ibáñez de Opakua A, Rodríguez JA, Merino N, Villate M, Onesti S, Blanco FJ. The tumor suppressor inhibitor of growth 4 binds double-stranded DNA through its disordered central region. FEBS Lett 2016; 591:425-432. [DOI: 10.1002/1873-3468.12514] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Revised: 11/28/2016] [Accepted: 11/30/2016] [Indexed: 12/31/2022]
Affiliation(s)
| | | | | | | | | | | | | | - Francisco J. Blanco
- CIC bioGUNE; Derio Spain
- IKERBASQUE; Basque Foundation for Science; Bilbao Spain
| |
Collapse
|
50
|
Tang L, Yao A, Tang Y, Liu J, Liu X, Qiu R. Transcriptional up-regulation of genes involved in photosynthesis of the Zn/Cd hyperaccumulator Sedum alfredii in response to zinc and cadmium. CHEMOSPHERE 2016; 164:190-200. [PMID: 27591370 DOI: 10.1016/j.chemosphere.2016.08.026] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/17/2016] [Revised: 07/25/2016] [Accepted: 08/04/2016] [Indexed: 05/09/2023]
Abstract
Zinc (Zn) and cadmium (Cd) are two closely related chemical elements with very different biological roles in photosynthesis. Zinc plays unique biochemical functions in photosynthesis. Previous studies suggested that in some Zn/Cd hyperaccumulators, many steps in photosynthesis may be Cd tolerant or even Cd stimulated. Using RNA-seq data, we found not only that Cd and Zn both up-regulated the CA1 gene, which encodes a β class carbonic anhydrase (CA) in chloroplasts, but that a large number of other Zn up-regulated genes in the photosynthetic pathway were also significantly up-regulated by Cd in leaves of the Zn/Cd hyperaccumulator Sedum alfredii. These genes also include chloroplast genes involved in transcription and translation (rps18 and rps14), electron transport and ATP synthesis (atpF and ccsA), Photosystem II (PSBI, PSBM, PSBK, PSBZ/YCF9, PSBO-1, PSBQ, LHCB1.1, LHCB1.4, LHCB2.1, LHCB4.3 and LHCB6) and Photosystem I (PSAE-1, PSAF, PSAH2, LHCA1 and LHCA4). Cadmium and Zn also up-regulated the VAR1 gene, which encodes the ATP-dependent zinc metalloprotease FTSH 5 (a member of the FtsH family), and the DAG gene, which influences chloroplast differentiation and plastid development, and the CP29 gene, which supports RNA processing in chloroplasts and has a potential role in signal-dependent co-regulation of chloroplast genes. Further morphological parameters (dry biomass, cross-sectional thickness, chloroplast size, chlorophyll content) and chlorophyll fluorescence parameters confirmed that leaf photosynthesis of S. alfredii responded to Cd much as it did to Zn, which will contribute to our understanding of the positive effects of Zn and Cd on growth of this plant.
Collapse
Affiliation(s)
- Lu Tang
- School of Environmental Science and Engineering, Sun Yat-Sen University, Guangzhou, PR China
| | - Aijun Yao
- Department of Land Resource and Environment, School of Geography and Planning, Sun Yat-Sen University, Guangzhou, PR China
| | - Yetao Tang
- School of Environmental Science and Engineering, Sun Yat-Sen University, Guangzhou, PR China; Guangdong Provincial Key Lab of Environmental Pollution Control and Remediation Technology (Sun Yat-sen University), Guangzhou, PR China.
| | - Jian Liu
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou, PR China
| | - Xi Liu
- School of Environmental Science and Engineering, Sun Yat-Sen University, Guangzhou, PR China
| | - Rongliang Qiu
- School of Environmental Science and Engineering, Sun Yat-Sen University, Guangzhou, PR China; Guangdong Provincial Key Lab of Environmental Pollution Control and Remediation Technology (Sun Yat-sen University), Guangzhou, PR China.
| |
Collapse
|