1
|
Zimmermann D, Kress M, Nagy I. Established and emerging roles of protein kinases in regulating primary sensory neurons in injury-and inflammation-associated pain. Expert Opin Ther Targets 2025; 29:267-280. [PMID: 40200157 DOI: 10.1080/14728222.2025.2489540] [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: 08/26/2024] [Revised: 03/06/2025] [Accepted: 04/02/2025] [Indexed: 04/10/2025]
Abstract
INTRODUCTION Recent seminal neuroscience research has significantly increased our knowledge on cellular and molecular responses of various cells in the pain pathway to peripheral nerve injuries and inflammatory processes. Transcriptomic and epigenetic analysis of primary sensory neurons (PSNs) in animal models of peripheral injuries revealed new insights into altered gene expression profiles and epigenetic modifications, which, via increasing spinal nociceptive input, lead to the development of pain. Among the various classes of molecules involved in driving differential gene expression, protein kinases, the enzymes that catalyze the phosphorylation of molecules, are emerging to control histone modification and chromatin remodeling needed for the alteration in transcriptional activity. AREAS COVERED Here, we focused on how protein kinases contribute to transcriptomic changes and pathways of induced reprogramming within PSNs upon peripheral nerve injury and inflammation. We conducted systematic literature search across multiple databases, including PubMed, NIH ClinicalTrials.gov portal and GEOData from 1980 to 2024 and compared protein kinase expression frequencies between publicly available RNA sequencing datasets of PSNs and investigated differences in protein kinase expression levels after peripheral nerve injury. EXPERT OPINION Novel findings support a new concept that protein kinases constitute regulatory hubs of reprogramming of PSNs, which offers novel analgesic approaches.
Collapse
Affiliation(s)
- David Zimmermann
- Institute of Physiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Michaela Kress
- Institute of Physiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Istvan Nagy
- Department of Surgery and Cancer, Nociception group, Division of Anaesthetics, Pain Medicine and Intensive Care, Imperial College, London, UK
- Department of Physiology, University of Debrecen, Debrecen, Hungary
| |
Collapse
|
2
|
Guo L, Zhuo X, Lu C, Guo H, Chen Z, Wu G, Liu F, Wei X, Rong X, Li P. The N-terminal fragment of histone deacetylase 4 (1-669aa) promotes chondrocyte apoptosis via the p53-dependent endoplasmic reticulum stress pathway. J Cell Mol Med 2024; 28:e70135. [PMID: 39428562 PMCID: PMC11491302 DOI: 10.1111/jcmm.70135] [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] [Received: 01/19/2024] [Revised: 09/12/2024] [Accepted: 09/20/2024] [Indexed: 10/22/2024] Open
Abstract
Exogenous administration of the histone deacetylation 4 (HDAC4) protein can effectively delay osteoarthritis (OA) progression. However, HDAC4 is unstable and easily degrades into N-terminal (HDAC4-NT) and C-terminal fragments, and the HDAC4-NT can exert biological effects, but little is known about its role in chondrocytes and cartilage. Thus, the roles of HDAC4-NT fragments (1-289aa, 1-326aa and 1-669aa) in chondrocytes and cartilage were evaluated via real-time cell analysis (RTCA), safranin O staining, Sirius Red staining and nanoindentation. Molecular mechanisms were profiled via whole-transcriptome sequencing (RNA-seq) and verified in vitro and in vivo by a live cell real-time monitoring system, flow cytometry, western blotting and immunohistochemistry. The results showed that 1-669aa induced chondrocyte death and cartilage injury significantly, and the differentially expressed genes (DEGs) were enriched mainly in the apoptotic term and p53 signalling pathway. The validation experiments showed that 1-669aa induced chondrocyte apoptosis via the endoplasmic reticulum stress (ERS) pathway, and up-regulated p53 expression was essential for this process. Thus, we concluded that the HDAC4-NT fragment 1-669aa induces chondrocyte apoptosis via the p53-dependent ERS pathway, suggesting that in addition to overexpressing HDAC4, preventing it from degradation may be a new strategy for the treatment of OA.
Collapse
Affiliation(s)
- Li Guo
- Department of Orthopedics, Shanxi Key Laboratory of Bone and Soft Tissue Injury RepairSecond Hospital of Shanxi Medical UniversityTaiyuanShanxiChina
| | - Xuhao Zhuo
- Department of Orthopedics, Shanxi Key Laboratory of Bone and Soft Tissue Injury RepairSecond Hospital of Shanxi Medical UniversityTaiyuanShanxiChina
| | - Chengyang Lu
- Department of Orthopedics, Shanxi Key Laboratory of Bone and Soft Tissue Injury RepairSecond Hospital of Shanxi Medical UniversityTaiyuanShanxiChina
| | - Hua Guo
- Department of Orthopedics, Shanxi Key Laboratory of Bone and Soft Tissue Injury RepairSecond Hospital of Shanxi Medical UniversityTaiyuanShanxiChina
- Department of OrthopedicsPeople's Hospital of XinzhouXinzhouShanxiChina
| | - Zhi Chen
- Department of Orthopedics, Shanxi Key Laboratory of Bone and Soft Tissue Injury RepairSecond Hospital of Shanxi Medical UniversityTaiyuanShanxiChina
| | - Gaige Wu
- Department of Orthopedics, Shanxi Key Laboratory of Bone and Soft Tissue Injury RepairSecond Hospital of Shanxi Medical UniversityTaiyuanShanxiChina
| | - Fengrui Liu
- Department of Orthopedics, Shanxi Key Laboratory of Bone and Soft Tissue Injury RepairSecond Hospital of Shanxi Medical UniversityTaiyuanShanxiChina
| | - Xiaochun Wei
- Department of Orthopedics, Shanxi Key Laboratory of Bone and Soft Tissue Injury RepairSecond Hospital of Shanxi Medical UniversityTaiyuanShanxiChina
| | - Xueqin Rong
- Department of Pain Medicine CenterCentral Hospital of SanyaSanyaHainanChina
| | - Pengcui Li
- Department of Orthopedics, Shanxi Key Laboratory of Bone and Soft Tissue Injury RepairSecond Hospital of Shanxi Medical UniversityTaiyuanShanxiChina
| |
Collapse
|
3
|
Li L, Xie K, Xie H, Wang L, Li Z, Lu Q, Feng J. AURKB promotes colorectal cancer progression by triggering the phosphorylation of histone H3 at serine 10 to activate CCNE1 expression. Aging (Albany NY) 2024; 16:8019-8030. [PMID: 38713155 PMCID: PMC11132018 DOI: 10.18632/aging.205801] [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: 09/27/2023] [Accepted: 02/13/2024] [Indexed: 05/08/2024]
Abstract
Aurora kinase B (AURKB) initiates the phosphorylation of serine 10 on histone H3 (pH3S10), a crucial process for chromosome condensation and cytokinesis in mammalian mitosis. Nonetheless, the precise mechanisms through which AURKB regulates the cell cycle and contributes to tumorigenesis as an oncogenic factor in colorectal cancer (CRC) remain unclear. Here, we report that AURKB was highly expressed and positively correlated with Ki-67 expression in CRC. The abundant expression of AURKB promotes the growth of CRC cells and xenograft tumors in animal model. AURKB knockdown substantially suppressed CRC proliferation and triggered cell cycle arrest in G2/M phase. Interestingly, cyclin E1 (CCNE1) was discovered as a direct downstream target of AURKB and functioned synergistically with AURKB to promote CRC cell proliferation. Mechanically, AURKB activated CCNE1 expression by triggering pH3S10 in the promoter region of CCNE1. Furthermore, it was showed that the inhibitor specific for AURKB (AZD1152) can suppress CCNE1 expression in CRC cells and inhibit tumor cell growth. To conclude, this research demonstrates that AURKB accelerated the tumorigenesis of CRC through its potential to epigenetically activate CCNE1 expression, suggesting AURKB as a promising therapeutic target in CRC.
Collapse
Affiliation(s)
- Ling Li
- Department of Gastrointestinal Surgery, The First People’s Hospital of Changzhou, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Ke Xie
- Department of Gastrointestinal Surgery, The First People’s Hospital of Changzhou, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Honghu Xie
- Department of Gastrointestinal Surgery, The First People’s Hospital of Changzhou, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Lei Wang
- Department of Gastrointestinal Surgery, The First People’s Hospital of Changzhou, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Zhong Li
- Department of Gastrointestinal Surgery, The First People’s Hospital of Changzhou, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Qicheng Lu
- Department of Gastrointestinal Surgery, The First People’s Hospital of Changzhou, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Jin Feng
- Department of Gastrointestinal Surgery, The First People’s Hospital of Changzhou, The Third Affiliated Hospital of Soochow University, Changzhou, China
| |
Collapse
|
4
|
Cuttini E, Goi C, Pellarin E, Vida R, Brancolini C. HDAC4 in cancer: A multitasking platform to drive not only epigenetic modifications. Front Mol Biosci 2023; 10:1116660. [PMID: 36762207 PMCID: PMC9902726 DOI: 10.3389/fmolb.2023.1116660] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 01/09/2023] [Indexed: 01/25/2023] Open
Abstract
Controlling access to genomic information and maintaining its stability are key aspects of cell life. Histone acetylation is a reversible epigenetic modification that allows access to DNA and the assembly of protein complexes that regulate mainly transcription but also other activities. Enzymes known as histone deacetylases (HDACs) are involved in the removal of the acetyl-group or in some cases of small hydrophobic moieties from histones but also from the non-histone substrate. The main achievement of HDACs on histones is to repress transcription and promote the formation of more compact chromatin. There are 18 different HDACs encoded in the human genome. Here we will discuss HDAC4, a member of the class IIa family, and its possible contribution to cancer development.
Collapse
Affiliation(s)
- Emma Cuttini
- Scuola Superiore Universitaria di Toppo Wassermann, Università degli Studi di Udine, Udine, Italy
| | - Camilla Goi
- Scuola Superiore Universitaria di Toppo Wassermann, Università degli Studi di Udine, Udine, Italy
| | - Ester Pellarin
- Scuola Superiore Universitaria di Toppo Wassermann, Università degli Studi di Udine, Udine, Italy
| | - Riccardo Vida
- Scuola Superiore Universitaria di Toppo Wassermann, Università degli Studi di Udine, Udine, Italy
| | - Claudio Brancolini
- Scuola Superiore Universitaria di Toppo Wassermann, Università degli Studi di Udine, Udine, Italy,Laboratory of Epigenomics, Department of Medicine, Università degli Studi di Udine, Udine, Italy,*Correspondence: Claudio Brancolini,
| |
Collapse
|
5
|
Yang C, Croteau S, Hardy P. Histone deacetylase (HDAC) 9: versatile biological functions and emerging roles in human cancer. Cell Oncol (Dordr) 2021; 44:997-1017. [PMID: 34318404 PMCID: PMC8516780 DOI: 10.1007/s13402-021-00626-9] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Accepted: 07/02/2021] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND HDAC9 (histone deacetylase 9) belongs to the class IIa family of histone deacetylases. This enzyme can shuttle freely between the nucleus and cytoplasm and promotes tissue-specific transcriptional regulation by interacting with histone and non-histone substrates. HDAC9 plays an essential role in diverse physiological processes including cardiac muscle development, bone formation, adipocyte differentiation and innate immunity. HDAC9 inhibition or activation is therefore a promising avenue for therapeutic intervention in several diseases. HDAC9 overexpression is also common in cancer cells, where HDAC9 alters the expression and activity of numerous relevant proteins involved in carcinogenesis. CONCLUSIONS This review summarizes the most recent discoveries regarding HDAC9 as a crucial regulator of specific physiological systems and, more importantly, highlights the diverse spectrum of HDAC9-mediated posttranslational modifications and their contributions to cancer pathogenesis. HDAC9 is a potential novel therapeutic target, and the restoration of aberrant expression patterns observed among HDAC9 target genes and their related signaling pathways may provide opportunities to the design of novel anticancer therapeutic strategies.
Collapse
Affiliation(s)
- Chun Yang
- Research Center of CHU Sainte-Justine, University of Montréal, 3175 Côte-Sainte-Catherine, Room 2.17.004, Montréal, Québec H3T 1C5 Canada
| | - Stéphane Croteau
- Departments of Medicine, Pediatrics, Pharmacology and Physiology, University of Montréal, Montréal, QC Canada
| | - Pierre Hardy
- Research Center of CHU Sainte-Justine, University of Montréal, 3175 Côte-Sainte-Catherine, Room 2.17.004, Montréal, Québec H3T 1C5 Canada
- Departments of Medicine, Pediatrics, Pharmacology and Physiology, University of Montréal, Montréal, QC Canada
| |
Collapse
|
6
|
Guttzeit S, Backs J. Post-translational modifications talk and crosstalk to class IIa histone deacetylases. J Mol Cell Cardiol 2021; 162:53-61. [PMID: 34416247 DOI: 10.1016/j.yjmcc.2021.08.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 07/08/2021] [Accepted: 08/12/2021] [Indexed: 12/30/2022]
Abstract
Epigenetic modifications, such as histone or DNA modifications are key regulators of gene transcription and changes are often associated with maladaptive processes underlying cardiovascular disease. Epigenetic regulators therefore likely play a crucial role in cardiomyocyte homeostasis and facilitate the cellular adaption to various internal and external stimuli, responding to different intercellular and extracellular cues. Class IIa histone deacetylases are a class of epigenetic regulators that possess a myriad of post-transcriptional modification sites that modulate their activity in response to oxidative stress, altered catecholamine signalling or changes in the cellular metabolism. This review summaries the known reversible, post-translational modifications (PTMs) of class IIa histone deacetylases (HDACs) that ultimately drive transcriptional changes in homeostasis and disease. We also highlight the idea of a crosstalk of various PTMs on class IIa HDACs potentially leading to compensatory or synergistic effects on the class IIa HDAC-regulated cell behavior.
Collapse
Affiliation(s)
- Sebastian Guttzeit
- Institute of Experimental Cardiology, Heidelberg University, Heidelberg, Germany; DZHK (German Center for Cardiovascular Research), Partner Site Heidelberg/Mannheim, Heidelberg, Germany
| | - Johannes Backs
- Institute of Experimental Cardiology, Heidelberg University, Heidelberg, Germany; DZHK (German Center for Cardiovascular Research), Partner Site Heidelberg/Mannheim, Heidelberg, Germany.
| |
Collapse
|
7
|
Regulation of histone deacetylase activities and functions by phosphorylation and its physiological relevance. Cell Mol Life Sci 2020; 78:427-445. [PMID: 32683534 DOI: 10.1007/s00018-020-03599-4] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 07/02/2020] [Accepted: 07/09/2020] [Indexed: 12/31/2022]
Abstract
Histone deacetylases (HDACs) are conserved enzymes that regulate many cellular processes by catalyzing the removal of acetyl groups from lysine residues on histones and non-histone proteins. As appropriate for proteins that occupy such an essential biological role, HDAC activities and functions are in turn highly regulated. Overwhelming evidence suggests that the dysregulation of HDACs plays a major role in many human diseases. The regulation of HDACs is achieved by multiple different mechanisms, including posttranslational modifications. One of the most common posttranslational modifications on HDACs is reversible phosphorylation. Many HDAC phosphorylations are context-dependent, occurring in specific tissues or as a consequence of certain stimuli. Additionally, whereas phosphorylation can regulate some HDACs in a non-specific manner, many HDAC phosphorylations result in specific consequences. Although some of these modifications support normal HDAC function, aberrations can contribute to disease development. Here we review and critically evaluate how reversible phosphorylation activates or deactivates HDACs and, thereby, regulates their many functions under various cellular and physiological contexts.
Collapse
|
8
|
Song B, Greco TM, Lum KK, Taber CE, Cristea IM. The DNA Sensor cGAS is Decorated by Acetylation and Phosphorylation Modifications in the Context of Immune Signaling. Mol Cell Proteomics 2020; 19:1193-1208. [PMID: 32345711 PMCID: PMC7338091 DOI: 10.1074/mcp.ra120.001981] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 04/23/2020] [Indexed: 12/25/2022] Open
Abstract
The cyclic GMP-AMP synthase (cGAS) protein is a pattern-recognition receptor of the mammalian innate immune system that is recognized as a main cytosolic sensor of pathogenic or damaged DNA. cGAS DNA binding initiates catalytic production of the second messenger, cyclic GMP-AMP, which activates the STING-TBK1-IRF3 signaling axis to induce cytokine expression. Post-translational modification (PTM) has started to be recognized as a critical component of cGAS regulation, yet the extent of these modifications remains unclear. Here, we report the identification and functional analysis of cGAS phosphorylations and acetylations in several cell types under basal and immune-stimulated conditions. cGAS was enriched by immunoaffinity purification from human primary fibroblasts prior to and after infection with herpes simplex virus type 1 (HSV-1), as well as from immune-stimulated STING-HEK293T cells. Six phosphorylations and eight acetylations were detected, of which eight PTMs were not previously documented. PTMs were validated by parallel reaction monitoring (PRM) mass spectrometry in fibroblasts, HEK293T cells, and THP-1 macrophage-like cells. Primary sequence and structural analysis of cGAS highlighted a subset of PTM sites with elevated surface accessibility and high evolutionary sequence conservation. To assess the functional relevance of each PTM, we generated a series of single-point cGAS mutations. Stable cell lines were constructed to express cGAS with amino acid substitutions that prevented phosphorylation (Ser-to-Ala) and acetylation (Lys-to-Arg) or that mimicked the modification state (Ser-to-Asp and Lys-to-Gln). cGAS-dependent apoptotic and immune signaling activities were then assessed for each mutation. Our results show that acetyl-mimic mutations at Lys384 and Lys414 inhibit the ability of cGAS to induce apoptosis. In contrast, the Lys198 acetyl-mimic mutation increased cGAS-dependent interferon signaling when compared with the unmodified charge-mimic. Moreover, targeted PRM quantification showed that Lys198 acetylation is decreased upon infections with two herpesviruses-HSV-1 and human cytomegalovirus (HCMV), highlighting this residue as a regulatory point during virus infection.
Collapse
Affiliation(s)
- Bokai Song
- Department of Molecular Biology, Princeton University, Princeton, New Jersey
| | - Todd M Greco
- Department of Molecular Biology, Princeton University, Princeton, New Jersey
| | - Krystal K Lum
- Department of Molecular Biology, Princeton University, Princeton, New Jersey
| | - Caroline E Taber
- Department of Molecular Biology, Princeton University, Princeton, New Jersey
| | - Ileana M Cristea
- Department of Molecular Biology, Princeton University, Princeton, New Jersey.
| |
Collapse
|
9
|
Song H, Zhou Y, Peng A, Liu J, Wu X, Chen W, Liu Z. Aurora-B Promotes Osteosarcoma Cell Growth and Metastasis Through Activation of the NPM1/ERK/NF-κβ/MMPs Axis. Cancer Manag Res 2020; 12:4817-4827. [PMID: 32606971 PMCID: PMC7320907 DOI: 10.2147/cmar.s252847] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2020] [Accepted: 05/10/2020] [Indexed: 12/14/2022] Open
Abstract
Purpose Osteosarcoma (OS) is the most common primary malignant tumor of the bone in young adolescents and children. We explored the underlying mechanism of Aurora-B in promoting OS cell proliferation and metastasis. Patient and Methods Bioinformatics was employed to predict the substrate of Aurora-B. IHC and Western blot were used to confirm the correlation between Aurora-B and NPM1. ERK/NF-κβ pathway-related proteins were detected by Western blot and immunofluorescence (IF). CCK8, wound healing, transwell, and Tunel assays were used to identify the cell proliferation, migration and apoptosis potential. Spontaneous metastasis xenografts were established to confirm the role of Aurora-B and NPM1. Results Aurora-B promotes NPM1 phosphorylation on Ser125. The phosphorylation of NPM1Ser125 induced by Aurora-B activates the ERK/NF-κβ signaling. Further study revealed that Aurora-B promotes proliferation, migration and inhibits apoptosis via phosphorylating NPM1 in vitro and in vivo. Conclusion Aurora-B promotes OS malignancy via phosphorylating NPM1Ser125 and activating ERK/NF-κβ signaling.
Collapse
Affiliation(s)
- Honghai Song
- Department of Orthopedic Surgery, The First Affiliated Hospital of Nanchang University, Nanchang 330006, People's Republic of China.,Institute of Spinal and Spinal Cord Diseases, Nanchang University, Nanchang 330031, People's Republic of China
| | - Yang Zhou
- Department of Orthopedic Surgery, The First Affiliated Hospital of Nanchang University, Nanchang 330006, People's Republic of China
| | - Aifen Peng
- College of Humanities, Jiangxi University of Traditional Chinese Medicine, Nanchang, Jiangxi 330004, People's Republic of China
| | - Jiaming Liu
- Department of Orthopedic Surgery, The First Affiliated Hospital of Nanchang University, Nanchang 330006, People's Republic of China.,Institute of Spinal and Spinal Cord Diseases, Nanchang University, Nanchang 330031, People's Republic of China.,Jiangxi Institute of Respiratory Disease, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, People's Republic of China
| | - Xin Wu
- Department of Orthopedic Surgery, The First Affiliated Hospital of Nanchang University, Nanchang 330006, People's Republic of China
| | - Wenzhao Chen
- Department of Orthopedic Surgery, The First Affiliated Hospital of Nanchang University, Nanchang 330006, People's Republic of China
| | - Zhili Liu
- Department of Orthopedic Surgery, The First Affiliated Hospital of Nanchang University, Nanchang 330006, People's Republic of China.,Institute of Spinal and Spinal Cord Diseases, Nanchang University, Nanchang 330031, People's Republic of China.,Division of Science and Technology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, People's Republic of China
| |
Collapse
|
10
|
Zhang J, Lin X, Wu L, Huang JJ, Jiang WQ, Kipps TJ, Zhang S. Aurora B induces epithelial-mesenchymal transition by stabilizing Snail1 to promote basal-like breast cancer metastasis. Oncogene 2020; 39:2550-2567. [PMID: 31996785 DOI: 10.1038/s41388-020-1165-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 01/08/2020] [Accepted: 01/15/2020] [Indexed: 01/06/2023]
Abstract
Aurora B is a serine/threonine kinase that has been implicated in regulating cell proliferation in distinct cancers, including breast cancer. Here we show that Aurora B expression is elevated in basal-like breast cancer (BLBC) compared with other breast cancer subtypes. This high level of expression seems to correlate with poor metastasis-free survival and relapse-free survival in affected patients. Mechanistically, we show that elevated Aurora B expression in breast cancer cells activates AKT/GSK3β to stabilize Snail1 protein, a master regulator of epithelial-mesenchymal transition (EMT), leading to EMT induction in a kinase-dependent manner. Conversely, Aurora B knock down by short-hairpin RNAs (shRNAs) suppresses AKT/GSK3β/Snail1 signaling, reverses EMT and reduces breast cancer metastatic potential in vitro and in vivo. Finally, we identified a specific OCT4 phosphorylation site (T343) responsible for mediating Aurora B-induced AKT/GSK3β/Snail1 signaling and EMT that could be attenuated by Aurora B kinase inhibitor treatment. These findings support that Aurora B induces EMT to promote breast cancer metastasis via OCT4/AKT/GSK3β/Snail1 signaling. Pharmacologic Aurora B inhibition might be a potential effective treatment for breast cancer patients with metastatic disease.
Collapse
Affiliation(s)
- Jianchao Zhang
- Guangdong Key Laboratory for Genome Stability and Human Disease Prevention, Department of Pharmacology, Base for International Science and Technology Cooperation: Carson Cancer Stem Cell Vaccines R&D Center, International Cancer Center, Shenzhen University Health Science Center, Shenzhen, 518055, China
| | - Xinxin Lin
- Guangdong Key Laboratory for Genome Stability and Human Disease Prevention, Department of Pharmacology, Base for International Science and Technology Cooperation: Carson Cancer Stem Cell Vaccines R&D Center, International Cancer Center, Shenzhen University Health Science Center, Shenzhen, 518055, China
| | - Liufeng Wu
- Guangdong Key Laboratory for Genome Stability and Human Disease Prevention, Department of Pharmacology, Base for International Science and Technology Cooperation: Carson Cancer Stem Cell Vaccines R&D Center, International Cancer Center, Shenzhen University Health Science Center, Shenzhen, 518055, China
| | - Jia-Jia Huang
- State Key Laboratory of Oncology in South China, Department of Medical Oncology, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Wen-Qi Jiang
- State Key Laboratory of Oncology in South China, Department of Medical Oncology, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Thomas J Kipps
- Moores UCSD Cancer Center, University of California, 9310 Athena Circle, La Jolla, San Diego, CA, 92093, USA
| | - Suping Zhang
- Guangdong Key Laboratory for Genome Stability and Human Disease Prevention, Department of Pharmacology, Base for International Science and Technology Cooperation: Carson Cancer Stem Cell Vaccines R&D Center, International Cancer Center, Shenzhen University Health Science Center, Shenzhen, 518055, China. .,Moores UCSD Cancer Center, University of California, 9310 Athena Circle, La Jolla, San Diego, CA, 92093, USA.
| |
Collapse
|
11
|
AURKB promotes gastric cancer progression via activation of CCND1 expression. Aging (Albany NY) 2020; 12:1304-1321. [PMID: 31982864 PMCID: PMC7053608 DOI: 10.18632/aging.102684] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Accepted: 12/25/2019] [Indexed: 01/21/2023]
Abstract
Aurora kinase B (AURKB) triggers the phosphorylation of serine 10 on histone H3 (H3S10ph), which is important for chromosome condensation and cytokinesis during mitosis in mammals. However, how exactly AURKB controls cell cycle and contributes to tumorigenesis as an oncoprotein under pathological conditions remains largely unknown. Here, we report that AURKB promotes gastric cancer cell proliferation in vitro and in vivo. Silencing AURKB expression inhibits gastric cell proliferation and arrests the cell cycle in G2/M phase. We demonstrate that cyclin D1 (CCND1) is a direct downstream target of AURKB that plays a key role in gastric cancer cell proliferation. AURKB is able to activate the expression of CCND1 through mediating H3S10ph in the promoter of the CCND1 gene. Furthermore, we show that AZD1152, a specific inhibitor of AURKB, can suppress the expression of CCND1 in the gastric cancer cells and inhibit cell proliferation in vitro and in vivo. Importantly, we found that high AURKB and CCND1 expression levels are correlated with shorter overall survival of gastric cancer patients. This study demonstrates that AURKB promotes gastric tumorigenesis potentially through epigenetically activating CCND1 expression, suggesting AURKB as a promising therapeutic target in gastric cancer.
Collapse
|
12
|
Bose A, Sudevan S, Rao VJ, Shima H, Trivedi AK, Igarashi K, Kundu TK. Haploinsufficient tumor suppressor Tip60 negatively regulates oncogenic Aurora B kinase. J Biosci 2019; 44:147. [PMID: 31894128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
The Aurora kinases represent a group of serine/threonine kinases which are crucial regulators of mitosis. Dysregulated Aurora kinase B (AurkB) expression, stemming from genomic amplification, increased gene transcription or overexpression of its allosteric activators, is capable of initiating and sustaining malignant phenotypes. Although AurkB level in cells is well-orchestrated, studies that relate to its stability or activity, independent of mitosis, are lacking. We report that AurkB undergoes acetylation in vitro by lysine acetyltransferases (KATs) belonging to different families, namely by p300 and Tip60. The haploinsufficient tumor suppressor Tip60 acetylates two highly conserved lysine residues within the kinase domain of AurkB which not only impinges the protein stability but also its kinase activity. These results signify a probable outcome on the increase in "overall activity" of AurkB upon Tip60 downregulation, as observed under cancerous conditions. The present work, therefore, uncovers an important functional interplay between AurkB and Tip60, frailty of which may be an initial event in carcinogenesis.
Collapse
Affiliation(s)
- Arnab Bose
- Transcription and Disease Laboratory, Molecular Biology and Genetics Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Bangalore, Karnataka, India
| | | | | | | | | | | | | |
Collapse
|
13
|
|
14
|
Federspiel JD, Greco TM, Lum KK, Cristea IM. Hdac4 Interactions in Huntington's Disease Viewed Through the Prism of Multiomics. Mol Cell Proteomics 2019; 18:S92-S113. [PMID: 31040226 PMCID: PMC6692770 DOI: 10.1074/mcp.ra118.001253] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2018] [Revised: 03/27/2019] [Indexed: 12/29/2022] Open
Abstract
Huntington's disease (HD) is a monogenic disorder, driven by the expansion of a trinucleotide (CAG) repeat within the huntingtin (Htt) gene and culminating in neuronal degeneration in the brain, predominantly in the striatum and cortex. Histone deacetylase 4 (Hdac4) was previously found to contribute to the disease progression, providing a potential therapeutic target. Hdac4 knockdown reduced accumulation of misfolded Htt protein and improved HD phenotypes. However, the underlying mechanism remains unclear, given its independence on deacetylase activity and the predominant cytoplasmic Hdac4 localization in the brain. Here, we undertook a multiomics approach to uncover the function of Hdac4 in the context of HD pathogenesis. We characterized the interactome of endogenous Hdac4 in brains of HD mouse models. Alterations in interactions were investigated in response to Htt polyQ length, comparing mice with normal (Q20) and disease (Q140) Htt, at both pre- and post-symptomatic ages (2 and 10 months, respectively). Parallel analyses for Hdac5, a related class IIa Hdac, highlighted the unique interaction network established by Hdac4. To validate and distinguish interactions specifically enhanced in an HD-vulnerable brain region, we next characterized endogenous Hdac4 interactions in dissected striata from this HD mouse series. Hdac4 associations were polyQ-dependent in the striatum, but not in the whole brain, particularly in symptomatic mice. Hdac5 interactions did not exhibit polyQ dependence. To identify which Hdac4 interactions and functions could participate in HD pathogenesis, we integrated our interactome with proteome and transcriptome data sets generated from the striata. We discovered an overlap in enriched functional classes with the Hdac4 interactome, particularly in vesicular trafficking and synaptic functions, and we further validated the Hdac4 interaction with the Wiskott-Aldrich Syndrome Protein and SCAR Homolog (WASH) complex. This study expands the knowledge of Hdac4 regulation and functions in HD, adding to the understanding of the molecular underpinning of HD phenotypes.
Collapse
Affiliation(s)
- Joel D Federspiel
- Department of Molecular Biology, Princeton University, Washington Road, Princeton, NJ 08544
| | - Todd M Greco
- Department of Molecular Biology, Princeton University, Washington Road, Princeton, NJ 08544
| | - Krystal K Lum
- Department of Molecular Biology, Princeton University, Washington Road, Princeton, NJ 08544
| | - Ileana M Cristea
- Department of Molecular Biology, Princeton University, Washington Road, Princeton, NJ 08544.
| |
Collapse
|
15
|
Compounds targeting class II histone deacetylases do not cause panHDACI-associated impairment of megakaryocyte differentiation. Exp Hematol 2019; 72:36-46. [PMID: 30611870 DOI: 10.1016/j.exphem.2018.12.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Revised: 12/27/2018] [Accepted: 12/28/2018] [Indexed: 12/25/2022]
Abstract
Histone deacetylase inhibitors (HDACIs) have demonstrated effectiveness against lymphomas and myelomas in clinical practice. However, common to all currently approved broad-acting HDACIs (panHDACIs) is dose-limiting thrombocytopenia, which has prevented wider use in cancer therapy. Using CD34+ hematopoietic stem cells (HSCs), we show that megakaryocyte (MK) cell maturation and differentiation are impaired by panHDACIs, correlating to clinical thrombocytopenia. Importantly, we demonstrate that inhibitors of class II histone deacetylases (HDACs), including LMK235 and tubacin at clinically relevant concentrations, do not affect MK maturation. Furthermore, we show that HDACI-induced impairment of MK differentiation is associated with reduction of protein levels of the transcription factor GATA-1, but not tubulin hyperacetylation. Finally, we report that panHDACIs trigger a rapid loss of GATA-1 protein via a proteasome-dependent pathway. Our data support the notion that specifically targeting class II HDACs in cancer treatment is a potential strategy that would offer a safer alternative than current panHDACIs.
Collapse
|
16
|
Tang A, Gao K, Chu L, Zhang R, Yang J, Zheng J. Aurora kinases: novel therapy targets in cancers. Oncotarget 2017; 8:23937-23954. [PMID: 28147341 PMCID: PMC5410356 DOI: 10.18632/oncotarget.14893] [Citation(s) in RCA: 257] [Impact Index Per Article: 32.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Accepted: 01/17/2017] [Indexed: 02/06/2023] Open
Abstract
Aurora kinases, a family of serine/threonine kinases, consisting of Aurora A (AURKA), Aurora B (AURKB) and Aurora C (AURKC), are essential kinases for cell division via regulating mitosis especially the process of chromosomal segregation. Besides regulating mitosis, Aurora kinases have been implicated in regulating meiosis. The deletion of Aurora kinases could lead to failure of cell division and impair the embryonic development. Overexpression or gene amplification of Aurora kinases has been clarified in a number of cancers. And a growing number of studies have demonstrated that inhibition of Aurora kinases could potentiate the effect of chemotherapies. For the past decades, a series of Aurora kinases inhibitors (AKIs) developed effectively repress the progression and growth of many cancers both in vivo and in vitro, suggesting that Aurora kinases could be a novel therapeutic target. In this review, we'll first briefly present the structure, localization and physiological functions of Aurora kinases in mitosis, then describe the oncogenic role of Aurora kinases in tumorigenesis, we shall finally discuss the outcomes of AKIs combination with conventional therapy.
Collapse
Affiliation(s)
- Anqun Tang
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Jiangsu, China
| | - Keyu Gao
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Jiangsu, China
| | - Laili Chu
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Jiangsu, China
| | - Rui Zhang
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Jiangsu, China
| | - Jing Yang
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Jiangsu, China
| | - Junnian Zheng
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Jiangsu, China.,Department of Oncology, The First Affiliated Hospital, Xuzhou Medical University, Xuzhou, Jiangsu, China
| |
Collapse
|
17
|
Budayeva HG, Cristea IM. Human Sirtuin 2 Localization, Transient Interactions, and Impact on the Proteome Point to Its Role in Intracellular Trafficking. Mol Cell Proteomics 2016; 15:3107-3125. [PMID: 27503897 DOI: 10.1074/mcp.m116.061333] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Indexed: 11/06/2022] Open
Abstract
Human sirtuin 2 (SIRT2) is an NAD+-dependent deacetylase that primarily functions in the cytoplasm, where it can regulate α-tubulin acetylation levels. SIRT2 is linked to cancer progression, neurodegeneration, and infection with bacteria or viruses. However, the current knowledge about its interactions and the means through which it exerts its functions has remained limited. Here, we aimed to gain a better understanding of its cellular functions by characterizing SIRT2 subcellular localization, the identity and relative stability of its protein interactions, and its impact on the proteome of primary human fibroblasts. To assess the relative stability of SIRT2 interactions, we used immunoaffinity purification in conjunction with both label-free and metabolic labeling quantitative mass spectrometry. In addition to the expected associations with cytoskeleton proteins, including its known substrate TUBA1A, our results reveal that SIRT2 specifically interacts with proteins functioning in membrane trafficking, secretory processes, and transcriptional regulation. By quantifying their relative stability, we found most interactions to be transient, indicating a dynamic SIRT2 environment. We discover that SIRT2 localizes to the ER-Golgi intermediate compartment (ERGIC), and that this recruitment requires an intact ER-Golgi trafficking pathway. Further expanding these findings, we used microscopy and interaction assays to establish the interaction and coregulation of SIRT2 with liprin-β1 scaffolding protein (PPFiBP1), a protein with roles in focal adhesions disassembly. As SIRT2 functions may be accomplished via interactions, enzymatic activity, and transcriptional regulation, we next assessed the impact of SIRT2 levels on the cellular proteome. SIRT2 knockdown led to changes in the levels of proteins functioning in membrane trafficking, including some of its interaction partners. Altogether, our study expands the knowledge of SIRT2 cytoplasmic functions to define a previously unrecognized involvement in intracellular trafficking pathways, which may contribute to its roles in cellular homeostasis and human diseases.
Collapse
Affiliation(s)
- Hanna G Budayeva
- From the ‡Department of Molecular Biology, Princeton University, Princeton, New Jersey, 08544
| | - Ileana M Cristea
- From the ‡Department of Molecular Biology, Princeton University, Princeton, New Jersey, 08544
| |
Collapse
|
18
|
Patrick R, Horin C, Kobe B, Cao KAL, Bodén M. Prediction of kinase-specific phosphorylation sites through an integrative model of protein context and sequence. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2016; 1864:1599-608. [PMID: 27507704 DOI: 10.1016/j.bbapap.2016.08.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Revised: 07/08/2016] [Accepted: 08/03/2016] [Indexed: 01/17/2023]
Abstract
Identifying kinase substrates and the specific phosphorylation sites they regulate is an important factor in understanding protein function regulation and signalling pathways. Computational prediction of kinase targets - assigning kinases to putative substrates, and selecting from protein sequence the sites that kinases can phosphorylate - requires the consideration of both the cellular context that kinases operate in, as well as their binding affinity. This consideration enables investigation of how phosphorylation influences a range of biological processes. We report here a novel probabilistic model for classifying kinase-specific phosphorylation sites from sequence across three model organisms: human, mouse and yeast. The model incorporates position-specific amino acid frequencies, and counts of co-occurring amino acids from kinase binding sites. We show how this model can be seamlessly integrated with protein interactions and cell-cycle abundance profiles. When evaluating the prediction accuracy of our method, PhosphoPICK, on an independent hold-out set of kinase-specific phosphorylation sites, it achieved an average specificity of 97%, with 32% sensitivity. We compared PhosphoPICK's ability, through cross-validation, to predict kinase-specific phosphorylation sites with alternative methods, and show that at high levels of specificity PhosphoPICK obtains greater sensitivity for most comparisons made. We investigated the relationship between kinase-specific phosphorylation sites and nuclear localisation signals. We show that kinases PKA, Akt1 and AurB have an over-representation of predicted binding sites at particular positions downstream from predicted nuclear localisation signals, demonstrating an important role for these kinases in regulating the nuclear import of proteins. PhosphoPICK is freely available as a web-service at http://bioinf.scmb.uq.edu.au/phosphopick.
Collapse
Affiliation(s)
- Ralph Patrick
- School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia 4072, Australia.
| | - Coralie Horin
- Polytech Nice-Sophia, Université Nice Sophia-Antipolis, Nice 06103, France
| | - Bostjan Kobe
- School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia 4072, Australia; Institute for Molecular Bioscience, The University of Queensland, St Lucia 4072, Australia; Australian Infectious Diseases Research Centre, The University of Queensland, St Lucia 4072, Australia
| | - Kim-Anh Lê Cao
- The University of Queensland Diamantina Institute, Translational Research Institute, Woolloongabba, QLD 4102, Australia
| | - Mikael Bodén
- School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia 4072, Australia; Institute for Molecular Bioscience, The University of Queensland, St Lucia 4072, Australia
| |
Collapse
|
19
|
Luo Y, Jacobs EY, Greco TM, Mohammed KD, Tong T, Keegan S, Binley JM, Cristea IM, Fenyö D, Rout MP, Chait BT, Muesing MA. HIV-host interactome revealed directly from infected cells. Nat Microbiol 2016; 1:16068. [PMID: 27375898 PMCID: PMC4928716 DOI: 10.1038/nmicrobiol.2016.68] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2015] [Accepted: 04/16/2016] [Indexed: 01/27/2023]
Abstract
Although genetically compact, HIV-1 commandeers vast arrays of cellular machinery to sustain and protect it during cycles of viral outgrowth. Transposon-mediated saturation linker scanning mutagenesis was used to isolate fully replication-competent viruses harbouring a potent foreign epitope tag. Using these viral isolates, we performed differential isotopic labelling and affinity-capture mass spectrometric analyses on samples obtained from cultures of human lymphocytes to classify the vicinal interactomes of the viral Env and Vif proteins as they occur during natural infection. Importantly, interacting proteins were recovered without bias, regardless of their potential for positive, negative or neutral impact on viral replication. We identified specific host associations made with trimerized Env during its biosynthesis, at virological synapses, with innate immune effectors (such as HLA-E) and with certain cellular signalling pathways (for example, Notch1). We also defined Vif associations with host proteins involved in the control of nuclear transcription and nucleoside biosynthesis as well as those interacting stably or transiently with the cytoplasmic protein degradation apparatus. Our approach is broadly applicable to elucidating pathogen-host interactomes, providing high-certainty identification of interactors by their direct access during cycling infection. Understanding the pathophysiological consequences of these associations is likely to provide strategic targets for antiviral intervention.
Collapse
Affiliation(s)
- Yang Luo
- Aaron Diamond AIDS Research Center, 455 1st Avenue, New York, New York 10016, USA
| | - Erica Y. Jacobs
- Laboratory of Mass Spectrometry and Gaseous Ion Chemistry, The Rockefeller University, 1230 York Avenue, New York, New York 10065, USA
| | - Todd M. Greco
- Department of Molecular Biology, Lewis Thomas Laboratory, Princeton University, Princeton, New Jersey 08540, USA
| | - Kevin D. Mohammed
- Aaron Diamond AIDS Research Center, 455 1st Avenue, New York, New York 10016, USA
| | - Tommy Tong
- San Diego Biomedical Research Institute, 10865 Road to the Cure, San Diego, California 92121, USA
| | - Sarah Keegan
- Department of Biochemistry, New York University Langone Medical Center, 227 East 30th Street, New York, New York 10016, USA
| | - James M. Binley
- San Diego Biomedical Research Institute, 10865 Road to the Cure, San Diego, California 92121, USA
| | - Ileana M. Cristea
- Department of Molecular Biology, Lewis Thomas Laboratory, Princeton University, Princeton, New Jersey 08540, USA
| | - David Fenyö
- Department of Biochemistry, New York University Langone Medical Center, 227 East 30th Street, New York, New York 10016, USA
| | - Michael P. Rout
- Laboratory of Cellular and Structural Biology, The Rockefeller University, 1230 York Avenue, New York, New York 10065, USA
| | - Brian T. Chait
- Laboratory of Mass Spectrometry and Gaseous Ion Chemistry, The Rockefeller University, 1230 York Avenue, New York, New York 10065, USA
| | - Mark A. Muesing
- Aaron Diamond AIDS Research Center, 455 1st Avenue, New York, New York 10016, USA
| |
Collapse
|
20
|
Di Giorgio E, Brancolini C. Regulation of class IIa HDAC activities: it is not only matter of subcellular localization. Epigenomics 2016; 8:251-69. [DOI: 10.2217/epi.15.106] [Citation(s) in RCA: 86] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
In response to environmental cues, enzymes that influence the functions of proteins, through reversible post-translational modifications supervise the coordination of cell behavior like orchestral conductors. Class IIa histone deacetylases (HDACs) belong to this category. Even though in vertebrates these deacetylases have discarded the core enzymatic activity, class IIa HDACs can assemble into multiprotein complexes devoted to transcriptional reprogramming, including but not limited to epigenetic changes. Class IIa HDACs are subjected to variegated and interconnected layers of regulation, which reflect the wide range of biological responses under the scrutiny of this gene family. Here, we discuss about the key mechanisms that fine tune class IIa HDACs activities.
Collapse
Affiliation(s)
- Eros Di Giorgio
- Department of Medical & Biological Sciences, Università degli Studi di Udine., P.le Kolbe 4 - 33100 Udine, Italy
| | - Claudio Brancolini
- Department of Medical & Biological Sciences, Università degli Studi di Udine., P.le Kolbe 4 - 33100 Udine, Italy
| |
Collapse
|
21
|
de Castro IJ, Gokhan E, Vagnarelli P. Resetting a functional G1 nucleus after mitosis. Chromosoma 2016; 125:607-19. [PMID: 26728621 PMCID: PMC5023730 DOI: 10.1007/s00412-015-0561-6] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Accepted: 11/13/2015] [Indexed: 12/21/2022]
Abstract
The maintenance of the correct cellular information goes beyond the simple transmission of an intact genetic code from one generation to the next. Epigenetic changes, topological cues and correct protein-protein interactions need to be re-established after each cell division to allow the next cell cycle to resume in the correct regulated manner. This process begins with mitotic exit and re-sets all the changes that occurred during mitosis thus restoring a functional G1 nucleus in preparation for the next cell cycle. Mitotic exit is triggered by inactivation of mitotic kinases and the reversal of their phosphorylation activities on many cellular components, from nuclear lamina to transcription factors and chromatin itself. To reverse all these phosphorylations, phosphatases act during mitotic exit in a timely and spatially controlled manner directing the events that lead to a functional G1 nucleus. In this review, we will summarise the recent developments on the control of phosphatases and their known substrates during mitotic exit, and the key steps that control the restoration of chromatin status, nuclear envelope reassembly and nuclear body re-organisation. Although pivotal work has been conducted in this area in yeast, due to differences between the mitotic exit network between yeast and vertebrates, we will mainly concentrate on the vertebrate system.
Collapse
Affiliation(s)
- Ines J de Castro
- College of Health and Life Science, Research Institute of Environment Health and Society, Brunel University London, Uxbridge, UB8 3PH, UK
| | - Ezgi Gokhan
- College of Health and Life Science, Research Institute of Environment Health and Society, Brunel University London, Uxbridge, UB8 3PH, UK
| | - Paola Vagnarelli
- College of Health and Life Science, Research Institute of Environment Health and Society, Brunel University London, Uxbridge, UB8 3PH, UK.
| |
Collapse
|
22
|
Greco TM, Guise AJ, Cristea IM. Determining the Composition and Stability of Protein Complexes Using an Integrated Label-Free and Stable Isotope Labeling Strategy. Methods Mol Biol 2016; 1410:39-63. [PMID: 26867737 PMCID: PMC4916643 DOI: 10.1007/978-1-4939-3524-6_3] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
In biological systems, proteins catalyze the fundamental reactions that underlie all cellular functions, including metabolic processes and cell survival and death pathways. These biochemical reactions are rarely accomplished alone. Rather, they involve a concerted effect from many proteins that may operate in a directed signaling pathway and/or may physically associate in a complex to achieve a specific enzymatic activity. Therefore, defining the composition and regulation of protein complexes is critical for understanding cellular functions. In this chapter, we describe an approach that uses quantitative mass spectrometry (MS) to assess the specificity and the relative stability of protein interactions. Isolation of protein complexes from mammalian cells is performed by rapid immunoaffinity purification, and followed by in-solution digestion and high-resolution mass spectrometry analysis. We employ complementary quantitative MS workflows to assess the specificity of protein interactions using label-free MS and statistical analysis, and the relative stability of the interactions using a metabolic labeling technique. For each candidate protein interaction, scores from the two workflows can be correlated to minimize nonspecific background and profile protein complex composition and relative stability.
Collapse
Affiliation(s)
- Todd M Greco
- Department of Molecular Biology, Princeton University, 210 Lewis Thomas Laboratory, Washington Road, Princeton, NJ, 08544, USA
| | - Amanda J Guise
- Department of Molecular Biology, Princeton University, 210 Lewis Thomas Laboratory, Washington Road, Princeton, NJ, 08544, USA
| | - Ileana M Cristea
- Department of Molecular Biology, Princeton University, 210 Lewis Thomas Laboratory, Washington Road, Princeton, NJ, 08544, USA.
| |
Collapse
|
23
|
Guise AJ, Cristea IM. Approaches for Studying the Subcellular Localization, Interactions, and Regulation of Histone Deacetylase 5 (HDAC5). Methods Mol Biol 2016; 1436:47-84. [PMID: 27246208 PMCID: PMC5644287 DOI: 10.1007/978-1-4939-3667-0_5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
As a member of the class IIa family of histone deacetylases, the histone deacetylase 5 (HDAC5) is known to undergo nuclear-cytoplasmic shuttling and to be a critical transcriptional regulator. Its misregulation has been linked to prominent human diseases, including cardiac diseases and tumorigenesis. In this chapter, we describe several experimental methods that have proven effective for studying the functions and regulatory features of HDAC5. We present methods for assessing the subcellular localization, protein interactions, posttranslational modifications (PTMs), and activity of HDAC5 from the standpoint of investigating either the endogenous protein or tagged protein forms in human cells. Specifically, given that at the heart of HDAC5 regulation lie its dynamic localization, interactions, and PTMs, we present methods for assessing HDAC5 localization in fixed and live cells, for isolating HDAC5-containing protein complexes to identify its interactions and modifications, and for determining how these PTMs map to predicted HDAC5 structural motifs. Lastly, we provide examples of approaches for studying HDAC5 functions with a focus on its regulation during cell-cycle progression. These methods can readily be adapted for the study of other HDACs or non-HDAC-proteins of interest. Individually, these techniques capture temporal and spatial snapshots of HDAC5 functions; yet together, these approaches provide powerful tools for investigating both the regulation and regulatory roles of HDAC5 in different cell contexts relevant to health and disease.
Collapse
Affiliation(s)
- Amanda J Guise
- Department of Molecular Biology, Princeton University, 210 Lewis Thomas Laboratory, Princeton, NJ, 08544, USA
| | - Ileana M Cristea
- Department of Molecular Biology, Princeton University, 210 Lewis Thomas Laboratory, Princeton, NJ, 08544, USA.
| |
Collapse
|
24
|
Diner BA, Li T, Greco TM, Crow MS, Fuesler JA, Wang J, Cristea IM. The functional interactome of PYHIN immune regulators reveals IFIX is a sensor of viral DNA. Mol Syst Biol 2015; 11:787. [PMID: 25665578 PMCID: PMC4358659 DOI: 10.15252/msb.20145808] [Citation(s) in RCA: 75] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
The human PYHIN proteins, AIM2, IFI16, IFIX, and MNDA, are critical regulators of immune response, transcription, apoptosis, and cell cycle. However, their protein interactions and underlying mechanisms remain largely uncharacterized. Here, we provide the interaction network for all PYHIN proteins and define a function in sensing of viral DNA for the previously uncharacterized IFIX protein. By designing a cell-based inducible system and integrating microscopy, immunoaffinity capture, quantitative mass spectrometry, and bioinformatics, we identify over 300 PYHIN interactions reflective of diverse functions, including DNA damage response, transcription regulation, intracellular signaling, and antiviral response. In view of the IFIX interaction with antiviral factors, including nuclear PML bodies, we further characterize IFIX and demonstrate its function in restricting herpesvirus replication. We discover that IFIX detects viral DNA in both the nucleus and cytoplasm, binding foreign DNA via its HIN domain in a sequence-non-specific manner. Furthermore, IFIX contributes to the induction of interferon response. Our results highlight the value of integrative proteomics in deducing protein function and establish IFIX as an antiviral DNA sensor important for mounting immune responses.
Collapse
Affiliation(s)
- Benjamin A Diner
- Department of Molecular Biology, Lewis Thomas Laboratory, Princeton University, Princeton, NJ, USA
| | - Tuo Li
- Department of Molecular Biology, Lewis Thomas Laboratory, Princeton University, Princeton, NJ, USA
| | - Todd M Greco
- Department of Molecular Biology, Lewis Thomas Laboratory, Princeton University, Princeton, NJ, USA
| | - Marni S Crow
- Department of Molecular Biology, Lewis Thomas Laboratory, Princeton University, Princeton, NJ, USA
| | - John A Fuesler
- Department of Molecular Biology, Lewis Thomas Laboratory, Princeton University, Princeton, NJ, USA
| | - Jennifer Wang
- Department of Molecular Biology, Lewis Thomas Laboratory, Princeton University, Princeton, NJ, USA
| | - Ileana M Cristea
- Department of Molecular Biology, Lewis Thomas Laboratory, Princeton University, Princeton, NJ, USA
| |
Collapse
|
25
|
Mathias RA, Guise AJ, Cristea IM. Post-translational modifications regulate class IIa histone deacetylase (HDAC) function in health and disease. Mol Cell Proteomics 2015; 14:456-70. [PMID: 25616866 DOI: 10.1074/mcp.o114.046565] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Class IIa histone deacetylases (HDACs4, -5, -7, and -9) modulate the physiology of the human cardiovascular, musculoskeletal, nervous, and immune systems. The regulatory capacity of this family of enzymes stems from their ability to shuttle between nuclear and cytoplasmic compartments in response to signal-driven post-translational modification. Here, we review the current knowledge of modifications that control spatial and temporal histone deacetylase functions by regulating subcellular localization, transcriptional functions, and cell cycle-dependent activity, ultimately impacting on human disease. We discuss the contribution of these modifications to cardiac and vascular hypertrophy, myoblast differentiation, neuronal cell survival, and neurodegenerative disorders.
Collapse
Affiliation(s)
- Rommel A Mathias
- From the ‡Department of Molecular Biology, Princeton University, Princeton, New Jersey, 08544; §Department of Biochemistry, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, 3086, Australia
| | - Amanda J Guise
- From the ‡Department of Molecular Biology, Princeton University, Princeton, New Jersey, 08544
| | - Ileana M Cristea
- From the ‡Department of Molecular Biology, Princeton University, Princeton, New Jersey, 08544;
| |
Collapse
|
26
|
Colarossi L, Memeo L, Colarossi C, Aiello E, Iuppa A, Espina V, Liotta L, Mueller C. Inhibition of histone deacetylase 4 increases cytotoxicity of docetaxel in gastric cancer cells. Proteomics Clin Appl 2014; 8:924-931. [DOI: 10.1002/prca.201400058] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023]
Affiliation(s)
- Lorenzo Colarossi
- Center for Applied Proteomics and Molecular Medicine; George Mason University; Manassas VA USA
- Fondazione IOM; Viagrande Catania Italy
| | - Lorenzo Memeo
- Department of Experimental Oncology; Mediterranean Institute of Oncology; Viagrande Catania Italy
- IOM Ricerca srl; Viagrande Catania Italy
| | - Cristina Colarossi
- Department of Experimental Oncology; Mediterranean Institute of Oncology; Viagrande Catania Italy
| | - Eleonora Aiello
- Department of Experimental Oncology; Mediterranean Institute of Oncology; Viagrande Catania Italy
| | - Antonio Iuppa
- Department of Experimental Oncology; Mediterranean Institute of Oncology; Viagrande Catania Italy
| | - Virginia Espina
- Center for Applied Proteomics and Molecular Medicine; George Mason University; Manassas VA USA
| | - Lance Liotta
- Center for Applied Proteomics and Molecular Medicine; George Mason University; Manassas VA USA
| | - Claudius Mueller
- Center for Applied Proteomics and Molecular Medicine; George Mason University; Manassas VA USA
| |
Collapse
|
27
|
Guise AJ, Mathias RA, Rowland EA, Yu F, Cristea IM. Probing phosphorylation-dependent protein interactions within functional domains of histone deacetylase 5 (HDAC5). Proteomics 2014; 14:2156-66. [PMID: 24920159 PMCID: PMC4184959 DOI: 10.1002/pmic.201400092] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2014] [Revised: 04/14/2014] [Accepted: 06/04/2014] [Indexed: 01/26/2023]
Abstract
Class IIa histone deacetylases (HDACs) are critical transcriptional regulators, shuttling between nuclear and cytoplasmic cellular compartments. Within the nucleus, these HDACs repress transcription as components of multiprotein complexes, such as the nuclear corepressor and beclin-6 corepressor (BCoR) complexes. Cytoplasmic relocalization relieves this transcriptional repressive function. Class IIa HDAC shuttling is controlled, in part, by phosphorylations flanking the nuclear localization signal (NLS). Furthermore, we have reported that phosphorylation within the NLS by the kinase Aurora B modulates the localization and function of the class IIa HDAC5 during mitosis. While we identified numerous additional HDAC5 phosphorylations, their regulatory functions remain unknown. Here, we studied phosphorylation sites within functional HDAC5 domains, including the deacetylation domain (DAC, Ser755), nuclear export signal (NES, Ser1108), and an acidic domain (AD, Ser611). We have generated phosphomutant cell lines to investigate how absence of phosphorylation at these sites impacts HDAC5 localization, enzymatic activity, and protein interactions. Combining molecular biology and quantitative MS, we have defined the interactions and HDAC5-containing complexes mediated by site-specific phosphorylation and quantified selected changes using parallel reaction monitoring. These results expand the current understanding of HDAC regulation, and the functions of this critical family of proteins within human cells.
Collapse
Affiliation(s)
- Amanda J. Guise
- Princeton University, Department of Molecular Biology, Princeton, NJ 08544
| | - Rommel A. Mathias
- Princeton University, Department of Molecular Biology, Princeton, NJ 08544
| | | | - Fang Yu
- Princeton University, Department of Molecular Biology, Princeton, NJ 08544
| | - Ileana M. Cristea
- Princeton University, Department of Molecular Biology, Princeton, NJ 08544
| |
Collapse
|
28
|
Bøgebo R, Horn H, Olsen JV, Gammeltoft S, Jensen LJ, Hansen JL, Christensen GL. Predicting kinase activity in angiotensin receptor phosphoproteomes based on sequence-motifs and interactions. PLoS One 2014; 9:e94672. [PMID: 24722691 PMCID: PMC3983226 DOI: 10.1371/journal.pone.0094672] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2013] [Accepted: 03/17/2014] [Indexed: 01/14/2023] Open
Abstract
Recent progress in the understanding of seven-transmembrane receptor (7TMR) signalling has promoted the development of a new generation of pathway selective ligands. The angiotensin II type I receptor (AT1aR) is one of the most studied 7TMRs with respect to selective activation of the β-arrestin dependent signalling. Two complimentary global phosphoproteomics studies have analyzed the complex signalling induced by the AT1aR. Here we integrate the data sets from these studies and perform a joint analysis using a novel method for prediction of differential kinase activity from phosphoproteomics data. The method builds upon NetworKIN, which applies sophisticated linear motif analysis in combination with contextual network modelling to predict kinase-substrate associations with high accuracy and sensitivity. These predictions form the basis for subsequently nonparametric statistical analysis to identify likely activated kinases. This suggested that AT1aR-dependent signalling activates 48 of the 285 kinases detected in HEK293 cells. Of these, Aurora B, CLK3 and PKG1 have not previously been described in the pathway whereas others, such as PKA, PKB and PKC, are well known. In summary, we have developed a new method for kinase-centric analysis of phosphoproteomes to pinpoint differential kinase activity in large-scale data sets.
Collapse
Affiliation(s)
- Rikke Bøgebo
- Department of Clinical Biochemistry, Glostrup Research Institute, Glostrup Hospital, Glostrup, Denmark
| | - Heiko Horn
- Department of Disease Systems Biology, Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, Copenhagen, Denmark
| | - Jesper V. Olsen
- Department of Proteomics, Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, Copenhagen, Denmark
| | - Steen Gammeltoft
- Department of Clinical Biochemistry, Glostrup Research Institute, Glostrup Hospital, Glostrup, Denmark
| | - Lars J. Jensen
- Department of Disease Systems Biology, Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, Copenhagen, Denmark
| | - Jakob L. Hansen
- Department of Biomedical Sciences and The Danish National Research Foundation Centre for Cardiac Arrhythmia, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
- * E-mail: (JLH); (GLC)
| | - Gitte L. Christensen
- Department of Clinical Biochemistry, Glostrup Research Institute, Glostrup Hospital, Glostrup, Denmark
- Cellular and Metabolic Research Section, Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
- * E-mail: (JLH); (GLC)
| |
Collapse
|
29
|
Amin NM, Greco TM, Kuchenbrod LM, Rigney MM, Chung MI, Wallingford JB, Cristea IM, Conlon FL. Proteomic profiling of cardiac tissue by isolation of nuclei tagged in specific cell types (INTACT). Development 2014; 141:962-73. [PMID: 24496632 DOI: 10.1242/dev.098327] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The proper dissection of the molecular mechanisms governing the specification and differentiation of specific cell types requires isolation of pure cell populations from heterogeneous tissues and whole organisms. Here, we describe a method for purification of nuclei from defined cell or tissue types in vertebrate embryos using INTACT (isolation of nuclei tagged in specific cell types). This method, previously developed in plants, flies and worms, utilizes in vivo tagging of the nuclear envelope with biotin and the subsequent affinity purification of the labeled nuclei. In this study we successfully purified nuclei of cardiac and skeletal muscle from Xenopus using this strategy. We went on to demonstrate the utility of this approach by coupling the INTACT approach with liquid chromatography-tandem mass spectrometry (LC-MS/MS) proteomic methodologies to profile proteins expressed in the nuclei of developing hearts. From these studies we have identified the Xenopus orthologs of 12 human proteins encoded by genes, which when mutated in human lead to congenital heart disease. Thus, by combining these technologies we are able to identify tissue-specific proteins that are expressed and required for normal vertebrate organ development.
Collapse
Affiliation(s)
- Nirav M Amin
- University of North Carolina McAllister Heart Institute, UNC-Chapel Hill, Chapel Hill, NC 27599-3280, USA
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Miteva YV, Cristea IM. A proteomic perspective of Sirtuin 6 (SIRT6) phosphorylation and interactions and their dependence on its catalytic activity. Mol Cell Proteomics 2013; 13:168-83. [PMID: 24163442 DOI: 10.1074/mcp.m113.032847] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Sirtuin 6 (SIRT6), a member of the mammalian sirtuin family, is a nuclear deacetylase with substrate-specific NAD(+)-dependent activity. SIRT6 has emerged as a critical regulator of diverse processes, including DNA repair, gene expression, telomere maintenance, and metabolism. However, our knowledge regarding its interactions and regulation remains limited. Here, we present a comprehensive proteomics-based analysis of SIRT6 protein interactions and their dependence on SIRT6 catalytic activity. We also identify evolutionarily conserved SIRT6 phosphorylations, including four within a proline-rich disordered region, and show that the conserved S338 phosphorylation can modulate selected SIRT6 interactions. By integrating molecular biology tools, microscopy, immunoaffinity purifications, label-free quantitative mass spectrometry, and bioinformatic analyses, we have established the first large-scale SIRT6 interaction network. Relative protein abundances and gene ontology functional assessment highlighted proteins involved in transcription regulation, chromatin organization, nuclear transport, telomerase function, and RNA processing. Independent immunoisolations under increased stringency distinguished the most stable SIRT6 interactions. One prominent interaction with Ras-GTPase-activating protein-binding protein 1 (G3BP1) was further validated by microscopy, reciprocal purifications, and isolations in different cell types and of endogenous SIRT6. Interestingly, a subset of specific interactions, including G3BP1, were significantly reduced or abolished in isolations of catalytically deficient SIRT6 mutant, revealing previously unknown interplay between SIRT6 activity and its associations. Overall, our study reveals putative means of regulation of SIRT6 functions via interactions and modifications, providing an important resource for future studies on the molecular mechanisms underlying sirtuin functions.
Collapse
Affiliation(s)
- Yana V Miteva
- Department of Molecular Biology, Princeton University, Princeton, New Jersey 08544
| | | |
Collapse
|
31
|
Tsai YC, Greco TM, Cristea IM. Sirtuin 7 plays a role in ribosome biogenesis and protein synthesis. Mol Cell Proteomics 2013; 13:73-83. [PMID: 24113281 DOI: 10.1074/mcp.m113.031377] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
It has been shown that SIRT7 regulates rDNA transcription and that reduced SIRT7 levels inhibit tumor growth. This anti-tumor effect could be due to reduced Pol I activity and perturbed ribosome biogenesis. In this study, using pulse labeling with RNA and amino acid analogs, we found that SIRT7 knockdown efficiently suppressed both RNA and protein synthesis. Surprisingly, SIRT7 knockdown preferentially inhibited protein synthesis over rDNA transcription, whereas the levels of both were reduced to similar extents following Pol I knockdown. Using an affinity purification mass spectrometry approach and functional analyses of the resulting SIRT7 interactome, we identified and validated SIRT7 interactions with proteins involved in ribosomal biogenesis. Indeed, SIRT7 co-fractionated with monoribosomes within a sucrose gradient. Using reciprocal isolations, we determined that SIRT7 interacts specifically with mTOR and GTF3C1, a component of the Pol III transcription factor TFIIIC2 complex. Further studies found that SIRT7 knockdown triggered an increase in the levels of LC3B-II, an autophagosome marker, suggesting a link between SIRT7 and the mTOR pathway. Additionally, we provide several lines of evidence that SIRT7 plays a role in modulating Pol III function. Immunoaffinity purification of SIRT7-GFP from a nuclear fraction demonstrated specific SIRT7 interaction with five out of six components of the TFIIIC2 complex, but not with the TFIIIA or TFIIIB complex, the former of which is required for Pol III-dependent transcription of tRNA genes. ChIP assays showed SIRT7 localization to the Pol III targeting genes, and SIRT7 knockdown triggered a reduction in tRNA levels. Taken together, these data suggest that SIRT7 may regulate Pol III transcription through mTOR and the TFIIIC2 complex. We propose that SIRT7 is involved in multiple pathways involved in ribosome biogenesis, and we hypothesize that its down-regulation may contribute to an antitumor effect, partly through the inhibition of protein synthesis.
Collapse
Affiliation(s)
- Yuan-Chin Tsai
- Department of Molecular Biology, Princeton University, Princeton, New Jersey 08544
| | | | | |
Collapse
|
32
|
The functional interactome landscape of the human histone deacetylase family. Mol Syst Biol 2013; 9:672. [PMID: 23752268 PMCID: PMC3964310 DOI: 10.1038/msb.2013.26] [Citation(s) in RCA: 228] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2013] [Accepted: 04/29/2013] [Indexed: 12/22/2022] Open
Abstract
This study presents the first global protein interaction network for all 11 human HDACs in T cells and an integrative mass spectrometry approach for profiling relative interaction stability within isolated protein complexes. ![]()
T-cell lines stably expressing each of the human HDACs (1 - 11), C-terminally tagged with both EGFP and FLAG, were generated using retroviral transduction. Affinity purification coupled to mass spectrometry-based proteomics (AP-MS) was used to build the first global protein interaction network for all eleven human HDACs in T cells. An optimized label free AP-MS and computational workflow was developed for profiling relative interaction stability among isolated protein complexes. HDAC11 is a member of the “survival of motor neuron” protein complex with a functional role in mRNA splicing.
Histone deacetylases (HDACs) are a diverse family of essential transcriptional regulatory enzymes, that function through the spatial and temporal recruitment of protein complexes. As the composition and regulation of HDAC complexes are only partially characterized, we built the first global protein interaction network for all 11 human HDACs in T cells. Integrating fluorescence microscopy, immunoaffinity purifications, quantitative mass spectrometry, and bioinformatics, we identified over 200 unreported interactions for both well-characterized and lesser-studied HDACs, a subset of which were validated by orthogonal approaches. We establish HDAC11 as a member of the survival of motor neuron complex and pinpoint a functional role in mRNA splicing. We designed a complementary label-free and metabolic-labeling mass spectrometry-based proteomics strategy for profiling interaction stability among different HDAC classes, revealing that HDAC1 interactions within chromatin-remodeling complexes are largely stable, while transcription factors preferentially exist in rapid equilibrium. Overall, this study represents a valuable resource for investigating HDAC functions in health and disease, encompassing emerging themes of HDAC regulation in cell cycle and RNA processing and a deeper functional understanding of HDAC complex stability.
Collapse
|
33
|
Guise AJ, Budayeva HG, Diner BA, Cristea IM. Histone deacetylases in herpesvirus replication and virus-stimulated host defense. Viruses 2013; 5:1607-32. [PMID: 23807710 PMCID: PMC3738950 DOI: 10.3390/v5071607] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2013] [Revised: 06/17/2013] [Accepted: 06/18/2013] [Indexed: 02/06/2023] Open
Abstract
Emerging evidence highlights a critical role for protein acetylation during herpesvirus infection. As prominent modulators of protein acetylation, histone deacetylases (HDACs) are essential transcriptional and epigenetic regulators. Not surprisingly, viruses have evolved a wide array of mechanisms to subvert HDAC functions. Here, we review the mechanisms underlying HDAC regulation during herpesvirus infection. We next discuss the roles of acetylation in host defense against herpesvirus infection. Finally, we provide a perspective on the contribution of current mass spectrometry-based “omic” technologies to infectious disease research, offering a systems biology view of infection.
Collapse
Affiliation(s)
| | | | | | - Ileana M. Cristea
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +1-609-258-9417; Fax: +1-609-258-4575
| |
Collapse
|
34
|
Joshi P, Quach OL, Giguere SSB, Cristea IM. A Functional Proteomics Perspective of DBC1 as a Regulator of Transcription. JOURNAL OF PROTEOMICS & BIOINFORMATICS 2013; Suppl 2:002. [PMID: 24273392 PMCID: PMC3837576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
The past few years have seen significant advances in the use of modern proteomics approaches for biological discoveries. Among the fields impacted by proteomics is that of epigenetics, as mass spectrometry-based approaches have allowed the identification and characterization of transcriptional regulators, epigenetic marks, and the constantly evolving epigenetic landscape of a cell in health and disease states. These studies have substantially expanded our understanding of critical genes that mediate cell processes, such as differentiation, cell cycle regulation, and apoptosis. Not surprisingly, a great emphasis has been placed on defining factors that are de-regulated in cancers, in an attempt to define new and specific targets for therapeutic design. Differential gene expression observed during carcinogenesis can be induced by aberrant activities of transcription factors and chromatin remodeling enzymes. Through a series of recent mass spectrometry studies of histone deacetylases and nuclear receptors, Deleted in Breast Cancer 1 (DBC1) has emerged as a master regulator of transcriptional processes. DBC1 acts as a modulator of cellular epigenetic mechanisms and is frequently associated with human metastasis. Through its negative regulation of SIRT1 and HDAC3 deacetylation activities, DBC1 has a broad impact on gene expression, downstream cellular pathways, and associated human diseases. Here, we review the identified roles of DBC1, highlighting the critical contribution of mass spectrometry to these findings. Additionally, we provide a perspective of integrative proteomics approaches that can continue to shed light on the interplay between DBC1 and its protein targets, helping to further define its role in epigenetic modifications and to identify novel targets for cancer therapy.
Collapse
Affiliation(s)
- P Joshi
- Department of Molecular Biology, Princeton University, Washington Road, Princeton, NJ 08544, USA
| | - O L Quach
- Department of Molecular Biology, Princeton University, Washington Road, Princeton, NJ 08544, USA
| | - S S B Giguere
- Department of Molecular Biology, Princeton University, Washington Road, Princeton, NJ 08544, USA
| | - I M Cristea
- Department of Molecular Biology, Princeton University, Washington Road, Princeton, NJ 08544, USA
| |
Collapse
|
35
|
Miteva YV, Budayeva HG, Cristea IM. Proteomics-based methods for discovery, quantification, and validation of protein-protein interactions. Anal Chem 2013; 85:749-68. [PMID: 23157382 PMCID: PMC3666915 DOI: 10.1021/ac3033257] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
| | | | - Ileana M. Cristea
- Corresponding author: Ileana M. Cristea 210 Lewis Thomas Laboratory Department of Molecular Biology Princeton University Princeton, NJ 08544 Tel: 6092589417 Fax: 6092584575
| |
Collapse
|