1
|
Lodovichi S, Nepomuceno TC, Woods NT, Rix U, Koomen JM, Pellicioli A, Galli A, Monteiro ANA. SART1 modulates poly-(ADP-ribose) chain accumulation and PARP1 chromatin localization. iScience 2024; 27:111252. [PMID: 39569366 PMCID: PMC11576398 DOI: 10.1016/j.isci.2024.111252] [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: 03/15/2023] [Revised: 02/23/2024] [Accepted: 10/22/2024] [Indexed: 11/22/2024] Open
Abstract
PARP1 inhibitors (PARPis) are used for treatment of cancers with mutations in BRCA1 or BRCA2 that are deficient in homologous recombination. The identification of modulators of PARP1 activity is critical to understand and overcome resistance to PARPis. We integrated data from three omics-scale screens to discover new regulators of PARP1 activity. We identified SART1 and show that its silencing leads to an increase in poly-ADP ribosylation and chromatin-bound PARP1. SART1 is recruited to chromatin following DNA damage and limits PARP1 chromatin retention and activity. The SART1 N-terminus is sufficient to regulate the accumulation of PAR chains and PARP1 on chromatin, an activity dependent on the RGG/RG box. Silencing of SART1 leads to an increased sensitivity of cells to DNA damage induced by IR, irrespective of BRCA1 status and to PARPis only in absence of BRCA1. These results suggest that SART1 could be clinically utilized to improve PARPi efficacy.
Collapse
Affiliation(s)
- Samuele Lodovichi
- Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
- Yeast Genetics and Genomics, Laboratory of Functional Genetics and Genomics, Institute of Clinical Physiology, CNR, 56125 Pisa, Italy
- Dipartimento di Bioscienze, Università degli Studi di Milano, 20131 Milano, Italy
| | - Thales C Nepomuceno
- Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
| | - Nicholas T Woods
- Gastrointestinal Cancer Program, Eppley Institute for Research in Cancer, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Uwe Rix
- Department of Drug Discovery, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
| | - John M Koomen
- Molecular Oncology and Molecular Medicine Program, H. Lee Moffitt Cancer Center, Tampa, FL 33612, USA
| | - Achille Pellicioli
- Dipartimento di Bioscienze, Università degli Studi di Milano, 20131 Milano, Italy
| | - Alvaro Galli
- Yeast Genetics and Genomics, Laboratory of Functional Genetics and Genomics, Institute of Clinical Physiology, CNR, 56125 Pisa, Italy
| | - Alvaro N A Monteiro
- Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
| |
Collapse
|
2
|
Schou KB, Mandacaru S, Tahir M, Tom N, Nilsson AS, Andersen JS, Tiberti M, Papaleo E, Bartek J. Exploring the structural landscape of DNA maintenance proteins. Nat Commun 2024; 15:7748. [PMID: 39237506 PMCID: PMC11377751 DOI: 10.1038/s41467-024-49983-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2023] [Accepted: 06/25/2024] [Indexed: 09/07/2024] Open
Abstract
Evolutionary annotation of genome maintenance (GM) proteins has conventionally been established by remote relationships within protein sequence databases. However, often no significant relationship can be established. Highly sensitive approaches to attain remote homologies based on iterative profile-to-profile methods have been developed. Still, these methods have not been systematically applied in the evolutionary annotation of GM proteins. Here, by applying profile-to-profile models, we systematically survey the repertoire of GM proteins from bacteria to man. We identify multiple GM protein candidates and annotate domains in numerous established GM proteins, among other PARP, OB-fold, Macro, TUDOR, SAP, BRCT, KU, MYB (SANT), and nuclease domains. We experimentally validate OB-fold and MIS18 (Yippee) domains in SPIDR and FAM72 protein families, respectively. Our results indicate that, surprisingly, despite the immense interest and long-term research efforts, the repertoire of genome stability caretakers is still not fully appreciated.
Collapse
Affiliation(s)
- Kenneth Bødkter Schou
- Genome Integrity, Danish Cancer Institute, Danish Cancer Society, Strandboulevarden 49, 2100, Copenhagen, Denmark.
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Campusvej 55, 5230, Odense M, Denmark.
- Division of Genome Biology, Department of Medical Biochemistry and Biophysics, Science for Laboratory, Karolinska Institute, Solna, 171 77, Sweden.
| | - Samuel Mandacaru
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Campusvej 55, 5230, Odense M, Denmark
| | - Muhammad Tahir
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Campusvej 55, 5230, Odense M, Denmark
| | - Nikola Tom
- Lipidomics Core Facility, Danish Cancer Institute (DCI), DK-2100, Copenhagen, Denmark
| | - Ann-Sofie Nilsson
- Division of Genome Biology, Department of Medical Biochemistry and Biophysics, Science for Laboratory, Karolinska Institute, Solna, 171 77, Sweden
| | - Jens S Andersen
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Campusvej 55, 5230, Odense M, Denmark
| | - Matteo Tiberti
- Cancer Structural Biology, Danish Cancer Society Research Center, Strandboulevarden 49, 2100, Copenhagen, Denmark
| | - Elena Papaleo
- Cancer Structural Biology, Danish Cancer Society Research Center, Strandboulevarden 49, 2100, Copenhagen, Denmark
- Cancer Systems Biology, Section for Bioinformatics, Department of Health and Technology, Technical University of Denmark, 2800, Lyngby, Denmark
| | - Jiri Bartek
- Genome Integrity, Danish Cancer Institute, Danish Cancer Society, Strandboulevarden 49, 2100, Copenhagen, Denmark.
- Division of Genome Biology, Department of Medical Biochemistry and Biophysics, Science for Laboratory, Karolinska Institute, Solna, 171 77, Sweden.
| |
Collapse
|
3
|
Ma R, Xu X. Deciphering the role of post-translational modifications in fanconi anemia proteins and their influence on tumorigenesis. Cancer Gene Ther 2024; 31:1113-1123. [PMID: 38879655 DOI: 10.1038/s41417-024-00797-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 06/01/2024] [Accepted: 06/04/2024] [Indexed: 08/17/2024]
Abstract
Fanconi anemia (FA) is an autosomal or X-linked human disease, characterized by bone marrow failure, cancer susceptibility and various developmental abnormalities. So far, at least 22 FA genes (FANCA-W) have been identified. Germline inactivation of any one of these FA genes causes FA symptoms. Proteins encoded by FA genes are involved in the Fanconi anemia pathway, which is known for its roles in DNA inter-strand crosslinks (ICLs) repair. Besides, its roles in genome maintenance upon replication stress has also been reported. Post-translational modifications (PTMs) of FA proteins, particularly phosphorylation and ubiquitination, emerge as critical determinants in the activation of the FA pathway during ICL repair or replication stress response. Consequent inactivation of the FA pathway engenders heightened chromosomal instability, thereby constituting a genetic susceptibility conducive to cancer predisposition and the exacerbation of tumorigenesis. In this review, we have combined recent structural analysis of FA proteins and summarized knowledge on the functions of different PTMs in regulating FA pathways, and discuss potential contributions stemming from mutations at PTMs to the genesis and progression of tumorigenesis.
Collapse
Affiliation(s)
- Rui Ma
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Sciences, Hubei University, Wuhan, Hubei, 430062, China
| | - Xinlin Xu
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Sciences, Hubei University, Wuhan, Hubei, 430062, China.
| |
Collapse
|
4
|
Liu X, Mao X, Zhu C, Liu H, Fang Y, Fu T, Fan L, Liu M, Xiong Z, Tang H, Hu P, Le A. COMMD10 inhibited DNA damage to promote the progression of gastric cancer. J Cancer Res Clin Oncol 2024; 150:305. [PMID: 38871970 PMCID: PMC11176250 DOI: 10.1007/s00432-024-05817-z] [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: 02/15/2024] [Accepted: 05/22/2024] [Indexed: 06/15/2024]
Abstract
PURPOSE The copper metabolism MURR1 domain 10 (COMMD10) plays a role in a variety of tumors. Here, we investigated its role in gastric cancer (GC). METHODS Online prediction tools, quantitative real-time PCR, western blotting and immunohistochemistry were used to evaluate the expression of COMMD10 in GC. The effect of COMMD10 knockdown was investigated in the GC cell lines and in in vivo xenograft tumor experiments. Western blotting and immunofluorescence were used to explore the relationships between COMMD10 and DNA damage. RESULTS The expression of COMMD10 was upregulated in GC compared to that in para-cancerous tissue and correlated with a higher clinical TNM stage (P = 0.044) and tumor size (P = 0.0366). High COMMD10 expression predicted poor prognosis in GC. Knockdown of COMMD10 resulted in the suppression of cell proliferation, migration, and invasion, accompanied by cell cycle arrest and an elevation in apoptosis rate. Moreover, the protein expression of COMMD10 was decreased in cisplatin-induced DNA-damaged GC cells. Suppression of COMMD10 impeded DNA damage repair, intensified DNA damage, and activated ATM-p53 signaling pathway in GC. Conversely, restoration of COMMD10 levels suppressed DNA damage and activation of the ATM-p53 signaling cascade. Additionally, knockdown of COMMD10 significantly restrained the growth of GC xenograft tumors while inhibiting DNA repair, augmenting DNA damage, and activating the ATM-p53 signaling pathway in xenograft tumor tissue. CONCLUSION COMMD10 is involved in DNA damage repair and maintains genomic stability in GC; knockdown of COMMD10 impedes the development of GC by exacerbating DNA damage, suggesting that COMMD10 may be new target for GC therapy.
Collapse
Affiliation(s)
- Xiaohua Liu
- Department of Blood Transfusion, Key Laboratory of Jiangxi Province for Transfusion Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, 1519 Dongyue Avenue, Nanchang, Jiangxi, People's Republic of China
| | - Xiaocheng Mao
- Department of Blood Transfusion, Key Laboratory of Jiangxi Province for Transfusion Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, 1519 Dongyue Avenue, Nanchang, Jiangxi, People's Republic of China
| | - Chao Zhu
- Department of Blood Transfusion, Key Laboratory of Jiangxi Province for Transfusion Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, 1519 Dongyue Avenue, Nanchang, Jiangxi, People's Republic of China
| | - Hongfei Liu
- Department of Blood Transfusion, Key Laboratory of Jiangxi Province for Transfusion Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, 1519 Dongyue Avenue, Nanchang, Jiangxi, People's Republic of China
| | - Yangyang Fang
- Department of Blood Transfusion, Key Laboratory of Jiangxi Province for Transfusion Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, 1519 Dongyue Avenue, Nanchang, Jiangxi, People's Republic of China
| | - Tianmei Fu
- Department of Blood Transfusion, Key Laboratory of Jiangxi Province for Transfusion Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, 1519 Dongyue Avenue, Nanchang, Jiangxi, People's Republic of China
| | - Linwei Fan
- Department of Blood Transfusion, Key Laboratory of Jiangxi Province for Transfusion Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, 1519 Dongyue Avenue, Nanchang, Jiangxi, People's Republic of China
| | - Mengwei Liu
- Department of Blood Transfusion, Key Laboratory of Jiangxi Province for Transfusion Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, 1519 Dongyue Avenue, Nanchang, Jiangxi, People's Republic of China
| | - Ziqing Xiong
- Department of Blood Transfusion, Key Laboratory of Jiangxi Province for Transfusion Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, 1519 Dongyue Avenue, Nanchang, Jiangxi, People's Republic of China
| | - Hong Tang
- Department of Blood Transfusion, Key Laboratory of Jiangxi Province for Transfusion Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, 1519 Dongyue Avenue, Nanchang, Jiangxi, People's Republic of China
| | - Piaoping Hu
- Department of Blood Transfusion, Key Laboratory of Jiangxi Province for Transfusion Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, 1519 Dongyue Avenue, Nanchang, Jiangxi, People's Republic of China.
| | - Aiping Le
- Department of Blood Transfusion, Key Laboratory of Jiangxi Province for Transfusion Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, 1519 Dongyue Avenue, Nanchang, Jiangxi, People's Republic of China.
| |
Collapse
|
5
|
Lorković ZJ, Klingenbrunner M, Cho CH, Berger F. Identification of plants' functional counterpart of the metazoan mediator of DNA Damage checkpoint 1. EMBO Rep 2024; 25:1936-1961. [PMID: 38438802 PMCID: PMC11014961 DOI: 10.1038/s44319-024-00107-8] [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: 01/04/2024] [Revised: 02/13/2024] [Accepted: 02/19/2024] [Indexed: 03/06/2024] Open
Abstract
Induction of DNA damage triggers rapid phosphorylation of the histone H2A.X (γH2A.X). In animals, mediator of DNA damage checkpoint 1 (MDC1) binds γH2A.X through a tandem BRCA1 carboxyl-terminal (tBRCT) domain and mediates recruitment of downstream effectors of DNA damage response (DDR). However, readers of this modification in plants have remained elusive. We show that from the Arabidopsis BRCT domain proteome, BCP1-4 proteins with tBRCT domains are involved in DDR. Through its tBRCT domain BCP4 binds γH2A.X in vitro and localizes to DNA damage-induced foci in an H2A.X-dependent manner. BCP4 also contains a domain that interacts directly with NBS1 and thus acts as a functional counterpart of MDC1. We also show that BCP1, that contains two tBRCT domains, co-localizes with γH2A.X but it does not bind γH2A.X suggesting functional similarity with human PAXIP1. A phylogenetic analysis supports that PAXIP1 and MDC1 in metazoa and their plant counterparts evolved independently from common ancestors with tBRCT domains. Collectively, our study reveals missing components and provides mechanistic and evolutionary insights into plant DDR.
Collapse
Affiliation(s)
- Zdravko J Lorković
- Gregor Mendel Institute (GMI), Austrian Academy of Sciences, Vienna Biocenter (VBC), Vienna, Austria.
| | - Michael Klingenbrunner
- Gregor Mendel Institute (GMI), Austrian Academy of Sciences, Vienna Biocenter (VBC), Vienna, Austria
- Department of Molecular Life Sciences, University of Zurich, Zurich, Switzerland
| | - Chung Hyun Cho
- Gregor Mendel Institute (GMI), Austrian Academy of Sciences, Vienna Biocenter (VBC), Vienna, Austria
| | - Frédéric Berger
- Gregor Mendel Institute (GMI), Austrian Academy of Sciences, Vienna Biocenter (VBC), Vienna, Austria.
| |
Collapse
|
6
|
Ezine E, Lebbe C, Dumaz N. Unmasking the tumourigenic role of SIN1/MAPKAP1 in the mTOR complex 2. Clin Transl Med 2023; 13:e1464. [PMID: 37877351 PMCID: PMC10599286 DOI: 10.1002/ctm2.1464] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 10/09/2023] [Accepted: 10/16/2023] [Indexed: 10/26/2023] Open
Abstract
BACKGROUND Although the PI3K/AKT/mTOR pathway is one of the most altered pathways in human tumours, therapies targeting this pathway have shown numerous adverse effects due to positive feedback paradoxically activating upstream signaling nodes. The somewhat limited clinical efficacy of these inhibitors calls for the development of novel and more effective approaches for targeting the PI3K pathway for therapeutic benefit in cancer. MAIN BODY Recent studies have shown the central role of mTOR complex 2 (mTORC2) as a pro-tumourigenic factor of the PI3K/AKT/mTOR pathway in a number of cancers. SIN1/MAPKAP1 is a major partner of mTORC2, acting as a scaffold and responsible for the substrate specificity of the mTOR catalytic subunit. Its overexpression promotes the proliferation, invasion and metastasis of certain cancers whereas its inhibition decreases tumour growth in vitro and in vivo. It is also involved in epithelial-mesenchymal transition, stress response and lipogenesis. Moreover, the numerous interactions of SIN1 inside or outside mTORC2 connect it with other signaling pathways, which are often disrupted in human tumours such as Hippo, WNT, Notch and MAPK. CONCLUSION Therefore, SIN1's fundamental characteristics and numerous connexions with oncogenic pathways make it a particularly interesting therapeutic target. This review is an opportunity to highlight the tumourigenic role of SIN1 across many solid cancers and demonstrates the importance of targeting SIN1 with a specific therapy.
Collapse
Affiliation(s)
- Emilien Ezine
- INSERMU976Team 1Human Immunology Pathophysiology & Immunotherapy (HIPI)ParisFrance
- Département de DermatologieHôpital Saint LouisAP‐HPParisFrance
| | - Céleste Lebbe
- INSERMU976Team 1Human Immunology Pathophysiology & Immunotherapy (HIPI)ParisFrance
- Département de DermatologieHôpital Saint LouisAP‐HPParisFrance
- Université Paris CitéInstitut de Recherche Saint Louis (IRSL)ParisFrance
| | - Nicolas Dumaz
- INSERMU976Team 1Human Immunology Pathophysiology & Immunotherapy (HIPI)ParisFrance
- Université Paris CitéInstitut de Recherche Saint Louis (IRSL)ParisFrance
| |
Collapse
|
7
|
Mota MBS, Woods NT, Carvalho MA, Monteiro ANA, Mesquita RD. Evolution of the triplet BRCT domain. DNA Repair (Amst) 2023; 129:103532. [PMID: 37453244 DOI: 10.1016/j.dnarep.2023.103532] [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: 02/06/2023] [Revised: 06/28/2023] [Accepted: 07/01/2023] [Indexed: 07/18/2023]
Abstract
Organisms have evolved a complex system, called the DNA damage response (DDR), which maintains genome integrity. The DDR is responsible for identifying and repairing a variety of lesions and alterations in DNA. DDR proteins coordinate DNA damage detection, cell cycle arrest, and repair, with many of these events regulated by protein phosphorylation. In the human proteome, 23 proteins contain the BRCT (BRCA1 C-Terminus domain) domain, a modular signaling domain that can bind phosphopeptides and mediate protein-protein interactions. BRCTs can be found as functional single units, tandem (tBRCT), triplet (tpBRCT), and quartet. Here we examine the evolution of the tpBRCT architecture present in TOPBP1 (DNA topoisomerase II binding protein 1) and ECT2 (epithelial cell transforming 2), and their respective interaction partners RAD9 (Cell cycle checkpoint control protein RAD9) and CYK-4 (Rac GTPase-activating protein 1), with a focus on the conservation of the phosphopeptide-binding residues. The pair TOPBP1-RAD9 arose with the Eukaryotes and ECT2-CYK-4 with the Eumetazoans. Triplet structural and functional characteristics were conserved in almost all organisms. The first unit of the triplet (BRCT0) is different from the other two BRCTs but conserved between orthologs for both TOPBP1 and ECT2. BRCT domain evolution simulations suggest a trend to retain the singlet or towards two or three BRCT copies per protein consistent with functional tBRCT and tpBRCT architectures. Our results shed light on the emergence of the function and architecture of multiple BRCT domain organizations and provide information about the evolution of the BRCT triplet. Knowledge of BRCT domain evolution can improve the understanding of DNA damage response mechanisms and signal transduction in DDR.
Collapse
Affiliation(s)
- M B S Mota
- Departamento de Bioquímica, Instituto de Química, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - N T Woods
- Eppley Institute, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - M A Carvalho
- Instituto Federal de Educação, Ciência e Tecnologia do Rio de Janeiro, RJ, Brazil; Instituto Nacional de Câncer, Rio de Janeiro, RJ, Brazil
| | - A N A Monteiro
- Cancer Epidemiology Program, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
| | - R D Mesquita
- Departamento de Bioquímica, Instituto de Química, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil.
| |
Collapse
|
8
|
Fu AB, Xiang SF, He QJ, Ying MD. Kelch-like proteins in the gastrointestinal tumors. Acta Pharmacol Sin 2023; 44:931-939. [PMID: 36266566 PMCID: PMC10104798 DOI: 10.1038/s41401-022-01007-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2022] [Accepted: 09/22/2022] [Indexed: 11/08/2022]
Abstract
Gastrointestinal tumors have become a worldwide health problem with high morbidity and poor clinical outcomes. Chemotherapy and surgery, the main treatment methods, are still far from meeting the treatment needs of patients, and targeted therapy is in urgent need of development. Recently, emerging evidence suggests that kelch-like (KLHL) proteins play essential roles in maintaining proteostasis and are involved in the progression of various cancers, functioning as adaptors in the E3 ligase complex and promoting the specific degradation of substrates. Therefore, KLHL proteins should be taken into consideration for targeted therapy strategy discovery. This review summarizes the current knowledge of KLHL proteins in gastrointestinal tumors and discusses the potential of KLHL proteins as potential drug targets and prognostic biomarkers.
Collapse
Affiliation(s)
- An-Bo Fu
- Institute of Pharmacology and Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
- Institute of Gastroenterology, Zhejiang University, Hangzhou, 310002, China
- Department of Gastroenterology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310002, China
| | - Sen-Feng Xiang
- Institute of Pharmacology and Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China.
| | - Qiao-Jun He
- Institute of Pharmacology and Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China.
- Cancer Center, Zhejiang University, Hangzhou, 310058, China.
| | - Mei-Dan Ying
- Institute of Pharmacology and Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China.
- Cancer Center, Zhejiang University, Hangzhou, 310058, China.
| |
Collapse
|
9
|
Yuan Q, Chu Y, Li X, Shi Y, Chen Y, Zhao J, Lu J, Liu K, Guo Y. CAFrgDB: a database for cancer-associated fibroblasts related genes and their functions in cancer. Cancer Gene Ther 2023:10.1038/s41417-023-00603-4. [PMID: 36922546 DOI: 10.1038/s41417-023-00603-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 02/03/2023] [Accepted: 02/23/2023] [Indexed: 03/17/2023]
Abstract
As one of the most essential components of the tumor microenvironment (TME), cancer-associated fibroblasts (CAFs) interact extensively with cancer cells and other stromal cells to remodel TME and participate in the pathogenesis of cancer, which earmarked themselves as new promising targets for cancer therapy. Numerous studies have highlighted the heterogeneity and versatility of CAFs in most cancer types. Thus, the identification and appropriate use of CAF-related genes (CAFGenes) in the context of specific cancer types will provide critical insights into disease mechanisms and CAF-related therapeutic targets. In this study, we collected and curated 5421 CAFGenes identified from small- or large-scale experiments, encompassing 4982 responsors that directly or indirectly participate in cancer malignant behaviors managed by CAFs, 1069 secretions that are secreted by CAFs and 281 regulators that contribute in modulating CAFs in human and mouse, which covered 24 cancer types. For these human CAFGenes, we performed gene expression and prognostic marker-based analyses across 24 cancer types using TCGA data. Furthermore, we provided annotations for CAF-associated proteins by integrating the knowledge of protein-protein interaction(s), drug-target relations and basic annotations, from 9 public databases. CAFrgDB (CAF related Gene DataBase) is free for academic research at http://caf.zbiolab.cn and we anticipate CAFrgDB can be a useful resource for further study of CAFs.
Collapse
Affiliation(s)
- Qiang Yuan
- Department of Pathophysiology, State Key Laboratory of Esophageal Cancer Prevention and Treatment, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Yi Chu
- Department of Pathophysiology, State Key Laboratory of Esophageal Cancer Prevention and Treatment, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Xiaoyu Li
- Department of Pathophysiology, State Key Laboratory of Esophageal Cancer Prevention and Treatment, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Yunshu Shi
- Department of Pathophysiology, State Key Laboratory of Esophageal Cancer Prevention and Treatment, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Yingying Chen
- Department of Pathophysiology, State Key Laboratory of Esophageal Cancer Prevention and Treatment, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Jimin Zhao
- Department of Pathophysiology, State Key Laboratory of Esophageal Cancer Prevention and Treatment, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Jing Lu
- Department of Pathophysiology, State Key Laboratory of Esophageal Cancer Prevention and Treatment, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Kangdong Liu
- Department of Pathophysiology, State Key Laboratory of Esophageal Cancer Prevention and Treatment, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China. .,China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan, 450001, China.
| | - Yaping Guo
- Department of Pathophysiology, State Key Laboratory of Esophageal Cancer Prevention and Treatment, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China.
| |
Collapse
|
10
|
Law HCH, Noe D, Woods NT. Interactome Profiling of DNA Damage Response (DDR) Mediators with Immunoprecipitation-Mass Spectrometry. Methods Mol Biol 2023; 2701:185-197. [PMID: 37574483 DOI: 10.1007/978-1-0716-3373-1_12] [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] [Indexed: 08/15/2023]
Abstract
Immunoprecipitation-mass spectrometry (IP-MS) is a versatile tool to probe for global protein-protein interactions (PPIs) in biological samples. Such interactions coordinate complex biological processes, such as the DNA damage response (DDR). Induction of DNA damage activates signaling networks where posttranslational modifications cause PPI that facilitate DNA repair and cell cycle coordination. Protein interactome profiling of DDR sensors, transducers, and effectors has the potential to identify novel DDR mechanisms that could advance our understanding and treatment of diseases associated with DDR defects, such as cancer. The protocol described here is a routine PPI analysis procedure that can be performed on samples stimulated with DNA damage. All processes and reagents are optimized for maximum sensitivity on the interactome and minimal contamination for the mass spectrometer.
Collapse
Affiliation(s)
- Henry C-H Law
- Eppley Institute for Research in Cancer and Allied Diseases, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA
| | - Dragana Noe
- Mass Spectrometry and Proteomics Core Facility, University of Nebraska Medical Center, Omaha, NE, USA
- Current address: Precision Biomarker Laboratories, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Nicholas T Woods
- Eppley Institute for Research in Cancer and Allied Diseases, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA.
| |
Collapse
|
11
|
Bouyahya A, El Allam A, Aboulaghras S, Bakrim S, El Menyiy N, Alshahrani MM, Al Awadh AA, Benali T, Lee LH, El Omari N, Goh KW, Ming LC, Mubarak MS. Targeting mTOR as a Cancer Therapy: Recent Advances in Natural Bioactive Compounds and Immunotherapy. Cancers (Basel) 2022; 14:5520. [PMID: 36428613 PMCID: PMC9688668 DOI: 10.3390/cancers14225520] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 11/01/2022] [Accepted: 11/02/2022] [Indexed: 11/12/2022] Open
Abstract
The mammalian target of rapamycin (mTOR) is a highly conserved serine/threonine-protein kinase, which regulates many biological processes related to metabolism, cancer, immune function, and aging. It is an essential protein kinase that belongs to the phosphoinositide-3-kinase (PI3K) family and has two known signaling complexes, mTOR complex 1 (mTORC1) and mTOR complex 2 (mTORC2). Even though mTOR signaling plays a critical role in promoting mitochondria-related protein synthesis, suppressing the catabolic process of autophagy, contributing to lipid metabolism, engaging in ribosome formation, and acting as a critical regulator of mRNA translation, it remains one of the significant signaling systems involved in the tumor process, particularly in apoptosis, cell cycle, and cancer cell proliferation. Therefore, the mTOR signaling system could be suggested as a cancer biomarker, and its targeting is important in anti-tumor therapy research. Indeed, its dysregulation is involved in different types of cancers such as colon, neck, cervical, head, lung, breast, reproductive, and bone cancers, as well as nasopharyngeal carcinoma. Moreover, recent investigations showed that targeting mTOR could be considered as cancer therapy. Accordingly, this review presents an overview of recent developments associated with the mTOR signaling pathway and its molecular involvement in various human cancer types. It also summarizes the research progress of different mTOR inhibitors, including natural and synthetised compounds and their main mechanisms, as well as the rational combinations with immunotherapies.
Collapse
Affiliation(s)
- Abdelhakim Bouyahya
- Laboratory of Human Pathologies Biology, Department of Biology, Faculty of Sciences, Faculty of Medicine and Pharmacy, Mohammed V University in Rabat, Rabat 10106, Morocco
| | - Aicha El Allam
- Department of Immunology, Yale University School of Medicine, 333 Cedars Street, TAC S610, New Haven, CT 06519, USA
| | - Sara Aboulaghras
- Physiology and Physiopathology Team, Faculty of Sciences, Genomic of Human Pathologies Research, Mohammed V University in Rabat, Rabat 10106, Morocco
| | - Saad Bakrim
- Geo-Bio-Environment Engineering and Innovation Laboratory, Molecular Engineering, Biotechnologies and Innovation Team, Polydisciplinary Faculty of Taroudant, Ibn Zohr University, Agadir 80000, Morocco
| | - Naoual El Menyiy
- Laboratory of Pharmacology, National Agency of Medicinal and Aromatic Plants, Taounate 34025, Morocco
| | - Mohammed Merae Alshahrani
- Department of Clinical Laboratory Sciences, Faculty of Applied Medical Sciences, Najran University, 1988, Najran 61441, Saudi Arabia
| | - Ahmed Abdullah Al Awadh
- Department of Clinical Laboratory Sciences, Faculty of Applied Medical Sciences, Najran University, 1988, Najran 61441, Saudi Arabia
| | - Taoufiq Benali
- Environment and Health Team, Polydisciplinary Faculty of Safi, Cadi Ayyad University, Sidi Bouzid B.P. 4162, Morocco
| | - Learn-Han Lee
- Novel Bacteria and Drug Discovery Research Group (NBDD), Microbiome and Bioresource Research Strength (MBRS), Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway 47500, Malaysia
| | - Nasreddine El Omari
- Laboratory of Histology, Embryology, and Cytogenetic, Faculty of Medicine and Pharmacy, Mohammed V University, Rabat 10100, Morocco
| | - Khang Wen Goh
- Faculty of Data Science and Information Technology, INTI International University, Nilai 71800, Malaysia
| | - Long Chiau Ming
- Pengiran Anak Puteri Rashidah Sa’adatul Bolkiah Institute of Health Sciences, Universiti Brunei Darussalam, Gadong BE1410, Brunei
| | | |
Collapse
|
12
|
Zhang Y, He Q. The role of SELENBP1 and its epigenetic regulation in carcinogenic progression. Front Genet 2022; 13:1027726. [PMID: 36386843 PMCID: PMC9663989 DOI: 10.3389/fgene.2022.1027726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Accepted: 10/10/2022] [Indexed: 01/24/2023] Open
Abstract
The initiation and progression of cancer is modulated through diverse genetic and epigenetic modifications. The epigenetic machinery regulates gene expression through intertwined DNA methylation, histone modifications, and miRNAs without affecting their genome sequences. SELENBP1 belongs to selenium-binding proteins and functions as a tumor suppressor. Its expression is significantly downregulated and correlates with carcinogenic progression and poor survival in various cancers. The role of SELENBP1 in carcinogenesis has not been fully elucidated, and its epigenetic regulation remains poorly understood. In this review, we summarize recent findings on the function and regulatory mechanisms of SELENBP1 during carcinogenic progression, with an emphasis on epigenetic mechanisms. We also discuss the potential cancer treatment targeting epigenetic modification of SELENBP1, either alone or in combination with selenium-containing compounds or dietary selenium.
Collapse
|
13
|
Yan S, Zhao J, Kemp M, Sobol RW. Editorial: Mechanistic studies of genome integrity, environmental health, and cancer etiology. Front Cell Dev Biol 2022; 10:1026326. [PMID: 36247007 PMCID: PMC9554606 DOI: 10.3389/fcell.2022.1026326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 09/07/2022] [Indexed: 01/31/2023] Open
Affiliation(s)
- Shan Yan
- Department of Biological Sciences, University of North Carolina at Charlotte, Charlotte, NC, United States,Correspondence: Shan Yan, ; Jianjun Zhao, ; Michael Kemp, ; Robert W. Sobol,
| | - Jianjun Zhao
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States,Correspondence: Shan Yan, ; Jianjun Zhao, ; Michael Kemp, ; Robert W. Sobol,
| | - Michael Kemp
- Department of Pharmacology and Toxicology, Wright State University, Dayton, OH, United States,Correspondence: Shan Yan, ; Jianjun Zhao, ; Michael Kemp, ; Robert W. Sobol,
| | - Robert W. Sobol
- Mitchell Cancer Institute and Department of Pharmacology, University of South Alabama, Mobile, AL, United States,Correspondence: Shan Yan, ; Jianjun Zhao, ; Michael Kemp, ; Robert W. Sobol,
| |
Collapse
|
14
|
Maitland MER, Lajoie GA, Shaw GS, Schild-Poulter C. Structural and Functional Insights into GID/CTLH E3 Ligase Complexes. Int J Mol Sci 2022; 23:5863. [PMID: 35682545 PMCID: PMC9180843 DOI: 10.3390/ijms23115863] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 05/18/2022] [Accepted: 05/20/2022] [Indexed: 11/29/2022] Open
Abstract
Multi-subunit E3 ligases facilitate ubiquitin transfer by coordinating various substrate receptor subunits with a single catalytic center. Small molecules inducing targeted protein degradation have exploited such complexes, proving successful as therapeutics against previously undruggable targets. The C-terminal to LisH (CTLH) complex, also called the glucose-induced degradation deficient (GID) complex, is a multi-subunit E3 ligase complex highly conserved from Saccharomyces cerevisiae to humans, with roles in fundamental pathways controlling homeostasis and development in several species. However, we are only beginning to understand its mechanistic basis. Here, we review the literature of the CTLH complex from all organisms and place previous findings on individual subunits into context with recent breakthroughs on its structure and function.
Collapse
Affiliation(s)
- Matthew E. R. Maitland
- Robarts Research Institute, Schulich School of Medicine & Dentistry, Western University, London, ON N6A 5B7, Canada;
- Department of Biochemistry, Schulich School of Medicine & Dentistry, Western University, London, ON N6G 2V4, Canada; (G.A.L.); (G.S.S.)
| | - Gilles A. Lajoie
- Department of Biochemistry, Schulich School of Medicine & Dentistry, Western University, London, ON N6G 2V4, Canada; (G.A.L.); (G.S.S.)
| | - Gary S. Shaw
- Department of Biochemistry, Schulich School of Medicine & Dentistry, Western University, London, ON N6G 2V4, Canada; (G.A.L.); (G.S.S.)
| | - Caroline Schild-Poulter
- Robarts Research Institute, Schulich School of Medicine & Dentistry, Western University, London, ON N6A 5B7, Canada;
- Department of Biochemistry, Schulich School of Medicine & Dentistry, Western University, London, ON N6G 2V4, Canada; (G.A.L.); (G.S.S.)
| |
Collapse
|
15
|
Kristofova M, Ori A, Wang ZQ. Multifaceted Microcephaly-Related Gene MCPH1. Cells 2022; 11:cells11020275. [PMID: 35053391 PMCID: PMC8774270 DOI: 10.3390/cells11020275] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Revised: 01/11/2022] [Accepted: 01/12/2022] [Indexed: 12/19/2022] Open
Abstract
MCPH1, or BRIT1, is often mutated in human primary microcephaly type 1, a neurodevelopmental disorder characterized by a smaller brain size at birth, due to its dysfunction in regulating the proliferation and self-renewal of neuroprogenitor cells. In the last 20 years or so, genetic and cellular studies have identified MCPH1 as a multifaceted protein in various cellular functions, including DNA damage signaling and repair, the regulation of chromosome condensation, cell-cycle progression, centrosome activity and the metabolism. Yet, genetic and animal model studies have revealed an unpredicted essential function of MPCH1 in gonad development and tumorigenesis, although the underlying mechanism remains elusive. These studies have begun to shed light on the role of MPCH1 in controlling various pathobiological processes of the disorder. Here, we summarize the biological functions of MCPH1, and lessons learnt from cellular and mouse models of MCPH1.
Collapse
Affiliation(s)
- Martina Kristofova
- Leibniz Institute on Aging—Fritz Lipmann Institute (FLI), Beutenbergstrasse 11, 07745 Jena, Germany; (M.K.); (A.O.)
| | - Alessandro Ori
- Leibniz Institute on Aging—Fritz Lipmann Institute (FLI), Beutenbergstrasse 11, 07745 Jena, Germany; (M.K.); (A.O.)
| | - Zhao-Qi Wang
- Leibniz Institute on Aging—Fritz Lipmann Institute (FLI), Beutenbergstrasse 11, 07745 Jena, Germany; (M.K.); (A.O.)
- Faculty of Biological Sciences, Friedrich-Schiller University of Jena, Bachstrasse 18k, 07743 Jena, Germany
- Correspondence: ; Tel.: +49-3641-656415; Fax: +49-3641-656335
| |
Collapse
|
16
|
Kim M, Park J, Bouhaddou M, Kim K, Rojc A, Modak M, Soucheray M, McGregor MJ, O'Leary P, Wolf D, Stevenson E, Foo TK, Mitchell D, Herrington KA, Muñoz DP, Tutuncuoglu B, Chen KH, Zheng F, Kreisberg JF, Diolaiti ME, Gordan JD, Coppé JP, Swaney DL, Xia B, van 't Veer L, Ashworth A, Ideker T, Krogan NJ. A protein interaction landscape of breast cancer. Science 2021; 374:eabf3066. [PMID: 34591612 PMCID: PMC9040556 DOI: 10.1126/science.abf3066] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Minkyu Kim
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, CA, USA.,The J. David Gladstone Institute of Data Science and Biotechnology, San Francisco, CA, USA.,Quantitative Biosciences Institute, University of California, San Francisco, CA, USA.,The Cancer Cell Map Initiative, San Francisco and La Jolla, CA, USA
| | - Jisoo Park
- The Cancer Cell Map Initiative, San Francisco and La Jolla, CA, USA.,Department of Medicine, University of California, San Diego, CA, USA
| | - Mehdi Bouhaddou
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, CA, USA.,The J. David Gladstone Institute of Data Science and Biotechnology, San Francisco, CA, USA.,Quantitative Biosciences Institute, University of California, San Francisco, CA, USA.,The Cancer Cell Map Initiative, San Francisco and La Jolla, CA, USA
| | - Kyumin Kim
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, CA, USA.,The J. David Gladstone Institute of Data Science and Biotechnology, San Francisco, CA, USA.,Quantitative Biosciences Institute, University of California, San Francisco, CA, USA.,The Cancer Cell Map Initiative, San Francisco and La Jolla, CA, USA
| | - Ajda Rojc
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, CA, USA.,The J. David Gladstone Institute of Data Science and Biotechnology, San Francisco, CA, USA.,Quantitative Biosciences Institute, University of California, San Francisco, CA, USA.,The Cancer Cell Map Initiative, San Francisco and La Jolla, CA, USA
| | - Maya Modak
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, CA, USA.,The J. David Gladstone Institute of Data Science and Biotechnology, San Francisco, CA, USA.,Quantitative Biosciences Institute, University of California, San Francisco, CA, USA.,The Cancer Cell Map Initiative, San Francisco and La Jolla, CA, USA
| | - Margaret Soucheray
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, CA, USA.,The J. David Gladstone Institute of Data Science and Biotechnology, San Francisco, CA, USA.,Quantitative Biosciences Institute, University of California, San Francisco, CA, USA.,The Cancer Cell Map Initiative, San Francisco and La Jolla, CA, USA
| | - Michael J McGregor
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, CA, USA.,The J. David Gladstone Institute of Data Science and Biotechnology, San Francisco, CA, USA.,Quantitative Biosciences Institute, University of California, San Francisco, CA, USA.,The Cancer Cell Map Initiative, San Francisco and La Jolla, CA, USA
| | - Patrick O'Leary
- The Cancer Cell Map Initiative, San Francisco and La Jolla, CA, USA.,Department of Medicine, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, USA
| | - Denise Wolf
- The Cancer Cell Map Initiative, San Francisco and La Jolla, CA, USA.,Department of Medicine, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, USA
| | - Erica Stevenson
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, CA, USA.,The J. David Gladstone Institute of Data Science and Biotechnology, San Francisco, CA, USA.,Quantitative Biosciences Institute, University of California, San Francisco, CA, USA.,The Cancer Cell Map Initiative, San Francisco and La Jolla, CA, USA
| | - Tzeh Keong Foo
- Department of Radiation Oncology, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, USA
| | - Dominique Mitchell
- Quantitative Biosciences Institute, University of California, San Francisco, CA, USA.,Department of Medicine, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, USA.,Division of Hematology/Oncology, University of California, San Francisco, CA, USA
| | - Kari A Herrington
- Department of Biochemistry and Biophysics, Center for Advanced Light Microscopy, University of California, San Francisco, CA, USA
| | - Denise P Muñoz
- The Cancer Cell Map Initiative, San Francisco and La Jolla, CA, USA.,Department of Medicine, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, USA
| | - Beril Tutuncuoglu
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, CA, USA.,The J. David Gladstone Institute of Data Science and Biotechnology, San Francisco, CA, USA.,Quantitative Biosciences Institute, University of California, San Francisco, CA, USA.,The Cancer Cell Map Initiative, San Francisco and La Jolla, CA, USA
| | - Kuei-Ho Chen
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, CA, USA.,The J. David Gladstone Institute of Data Science and Biotechnology, San Francisco, CA, USA.,Quantitative Biosciences Institute, University of California, San Francisco, CA, USA.,The Cancer Cell Map Initiative, San Francisco and La Jolla, CA, USA
| | - Fan Zheng
- The Cancer Cell Map Initiative, San Francisco and La Jolla, CA, USA.,Department of Medicine, University of California, San Diego, CA, USA
| | - Jason F Kreisberg
- The Cancer Cell Map Initiative, San Francisco and La Jolla, CA, USA.,Department of Medicine, University of California, San Diego, CA, USA
| | - Morgan E Diolaiti
- The Cancer Cell Map Initiative, San Francisco and La Jolla, CA, USA.,Department of Medicine, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, USA
| | - John D Gordan
- Quantitative Biosciences Institute, University of California, San Francisco, CA, USA.,Department of Medicine, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, USA.,Division of Hematology/Oncology, University of California, San Francisco, CA, USA
| | - Jean-Philippe Coppé
- The Cancer Cell Map Initiative, San Francisco and La Jolla, CA, USA.,Department of Medicine, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, USA
| | - Danielle L Swaney
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, CA, USA.,The J. David Gladstone Institute of Data Science and Biotechnology, San Francisco, CA, USA.,Quantitative Biosciences Institute, University of California, San Francisco, CA, USA.,The Cancer Cell Map Initiative, San Francisco and La Jolla, CA, USA
| | - Bing Xia
- Department of Radiation Oncology, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, USA
| | - Laura van 't Veer
- The Cancer Cell Map Initiative, San Francisco and La Jolla, CA, USA.,Department of Medicine, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, USA
| | - Alan Ashworth
- The Cancer Cell Map Initiative, San Francisco and La Jolla, CA, USA.,Department of Medicine, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, USA
| | - Trey Ideker
- The Cancer Cell Map Initiative, San Francisco and La Jolla, CA, USA.,Department of Medicine, University of California, San Diego, CA, USA.,Department of Bioengineering, University of California, San Diego, CA, USA
| | - Nevan J Krogan
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, CA, USA.,The J. David Gladstone Institute of Data Science and Biotechnology, San Francisco, CA, USA.,Quantitative Biosciences Institute, University of California, San Francisco, CA, USA.,The Cancer Cell Map Initiative, San Francisco and La Jolla, CA, USA
| |
Collapse
|
17
|
Zhu X, Jia W, Yan Y, Huang Y, Wang B. NOP14 regulates the growth, migration, and invasion of colorectal cancer cells by modulating the NRIP1/GSK-3β/β-catenin signaling pathway. Eur J Histochem 2021; 65. [PMID: 34218653 PMCID: PMC8273630 DOI: 10.4081/ejh.2021.3246] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Accepted: 05/25/2021] [Indexed: 12/24/2022] Open
Abstract
Colorectal cancer (CRC) is the third most common cancer diagnosed worldwide. Recently, nucleolar complex protein 14 (NOP14) has been discovered to play a critical role in cancer development and progression, but the mechanisms of action of NOP14 in colorectal cancer remain to be elucidated. In this study, we used collected colorectal cancer tissues and cultured colorectal cancer cell lines (SW480, HT29, HCT116, DLD1, Lovo), and measured the mRNA and protein expression levels of NOP14 in colorectal cancer cells using qPCR and Western blotting. GFP-NOP14 was constructed and siRNA fragments against NOP14 were synthesized to investigate the importance of NOP14 for the development of colorectal cells. Transwell migration assays were used to measure cell invasion and migration, CCK-8 kits were used to measure cell activity, and flow cytometry was applied to the observation of apoptosis. We found that both the mRNA and protein levels of NOP14 were significantly upregulated in CRC tissues and cell lines. Overexpression of GFP-NOP14 markedly promoted the growth, migration, and invasion of the CRC cells HT19 and SW480, while genetic knockdown of NOP14 inhibited these behaviors. Overexpression of NOP14 promoted the expression of NRIP1 and phosphorylated inactivation of GSK-3β, leading to the upregulation of β-catenin. Genetic knockdown of NOP14 had the opposite effect on NRIP1/GSK-3/β-catenin signals. NOP14 therefore appears to be overexpressed in clinical samples and cell lines of colorectal cancer, and promotes the proliferation, growth, and metastasis of colorectal cancer cells by modulating the NRIP1/GSK-3β/β-catenin signaling pathway.
Collapse
Affiliation(s)
- Xuanjin Zhu
- Department of General Surgery, Guangzhou Red Cross Hospital, Medical College of Jinan University, Guangzhou.
| | - Weilu Jia
- School of Clinical Medicine, Guizhou Medical University, Guiyang.
| | - Yong Yan
- Department of General Surgery, Guangzhou Red Cross Hospital, Medical College of Jinan University, Guangzhou .
| | - Yong Huang
- Department of General Surgery, Guangzhou Red Cross Hospital, Medical College of Jinan University, Guangzhou .
| | - Bailin Wang
- Department of General Surgery, Guangzhou Red Cross Hospital, Medical College of Jinan University, Guangzhou .
| |
Collapse
|
18
|
Suraweera A, Duijf PHG, Jekimovs C, Schrobback K, Liu C, Adams MN, O’Byrne KJ, Richard DJ. COMMD1, from the Repair of DNA Double Strand Breaks, to a Novel Anti-Cancer Therapeutic Target. Cancers (Basel) 2021; 13:830. [PMID: 33669398 PMCID: PMC7920454 DOI: 10.3390/cancers13040830] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 02/10/2021] [Accepted: 02/12/2021] [Indexed: 12/24/2022] Open
Abstract
Lung cancer has the highest incidence and mortality among all cancers, with non-small cell lung cancer (NSCLC) accounting for 85-90% of all lung cancers. Here we investigated the function of COMMD1 in the repair of DNA double strand breaks (DSBs) and as a prognostic and therapeutic target in NSCLC. COMMD1 function in DSB repair was investigated using reporter assays in COMMD1-siRNA-depleted cells. The role of COMMD1 in NSCLC was investigated using bioinformatic analysis, qRT-PCR and immunoblotting of control and NSCLC cells, tissue microarrays, cell viability and cell cycle experiments. DNA repair assays demonstrated that COMMD1 is required for the efficient repair of DSBs and reporter assays showed that COMMD1 functions in both non-homologous-end-joining and homologous recombination. Bioinformatic analysis showed that COMMD1 is upregulated in NSCLC, with high levels of COMMD1 associated with poor patient prognosis. COMMD1 mRNA and protein were upregulated across a panel of NSCLC cell lines and siRNA-mediated depletion of COMMD1 decreased cell proliferation and reduced cell viability of NSCLC, with enhanced death after exposure to DNA damaging-agents. Bioinformatic analyses demonstrated that COMMD1 levels positively correlate with the gene ontology DNA repair gene set enrichment signature in NSCLC. Taken together, COMMD1 functions in DSB repair, is a prognostic maker in NSCLC and is potentially a novel anti-cancer therapeutic target for NSCLC.
Collapse
Affiliation(s)
- Amila Suraweera
- School of Biomedical Sciences, Centre for Genomics and Personalised Health, Translational Research Institute, Queensland University of Technology (QUT), 37 Kent Street, Woolloongabba, QLD 4102, Australia; (P.H.G.D.); (C.J.); (K.S.); (M.N.A.); (K.J.O.)
- Princess Alexandra Hospital, 199 Ipswich Road, Woolloongabba, QLD 4102, Australia
| | - Pascal H. G. Duijf
- School of Biomedical Sciences, Centre for Genomics and Personalised Health, Translational Research Institute, Queensland University of Technology (QUT), 37 Kent Street, Woolloongabba, QLD 4102, Australia; (P.H.G.D.); (C.J.); (K.S.); (M.N.A.); (K.J.O.)
- Centre for Data Science, Queensland University of Technology (QUT), Brisbane, QLD 4000, Australia
- University of Queensland Diamantina Institute, University of Queensland, Brisbane, QLD 4102, Australia
| | - Christian Jekimovs
- School of Biomedical Sciences, Centre for Genomics and Personalised Health, Translational Research Institute, Queensland University of Technology (QUT), 37 Kent Street, Woolloongabba, QLD 4102, Australia; (P.H.G.D.); (C.J.); (K.S.); (M.N.A.); (K.J.O.)
| | - Karsten Schrobback
- School of Biomedical Sciences, Centre for Genomics and Personalised Health, Translational Research Institute, Queensland University of Technology (QUT), 37 Kent Street, Woolloongabba, QLD 4102, Australia; (P.H.G.D.); (C.J.); (K.S.); (M.N.A.); (K.J.O.)
| | - Cheng Liu
- QIMR Berghofer Medical Research Institute, 300 Herston Road, Herston, QLD 4006, Australia;
- Envoi Specialist Pathologists, 5/38 Bishop Street, Kelvin Grove, QLD 4059, Australia
| | - Mark N. Adams
- School of Biomedical Sciences, Centre for Genomics and Personalised Health, Translational Research Institute, Queensland University of Technology (QUT), 37 Kent Street, Woolloongabba, QLD 4102, Australia; (P.H.G.D.); (C.J.); (K.S.); (M.N.A.); (K.J.O.)
- Princess Alexandra Hospital, 199 Ipswich Road, Woolloongabba, QLD 4102, Australia
| | - Kenneth J. O’Byrne
- School of Biomedical Sciences, Centre for Genomics and Personalised Health, Translational Research Institute, Queensland University of Technology (QUT), 37 Kent Street, Woolloongabba, QLD 4102, Australia; (P.H.G.D.); (C.J.); (K.S.); (M.N.A.); (K.J.O.)
- Princess Alexandra Hospital, 199 Ipswich Road, Woolloongabba, QLD 4102, Australia
| | - Derek J. Richard
- School of Biomedical Sciences, Centre for Genomics and Personalised Health, Translational Research Institute, Queensland University of Technology (QUT), 37 Kent Street, Woolloongabba, QLD 4102, Australia; (P.H.G.D.); (C.J.); (K.S.); (M.N.A.); (K.J.O.)
- Princess Alexandra Hospital, 199 Ipswich Road, Woolloongabba, QLD 4102, Australia
| |
Collapse
|
19
|
Jamin SP, Hikmet F, Mathieu R, Jégou B, Lindskog C, Chalmel F, Primig M. Combined RNA/tissue profiling identifies novel Cancer/testis genes. Mol Oncol 2021; 15:3003-3023. [PMID: 33426787 PMCID: PMC8564638 DOI: 10.1002/1878-0261.12900] [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: 07/20/2020] [Revised: 11/19/2020] [Accepted: 12/24/2020] [Indexed: 11/14/2022] Open
Abstract
Cancer/Testis (CT) genes are induced in germ cells, repressed in somatic cells, and derepressed in somatic tumors, where these genes can contribute to cancer progression. CT gene identification requires data obtained using standardized protocols and technologies. This is a challenge because data for germ cells, gonads, normal somatic tissues, and a wide range of cancer samples stem from multiple sources and were generated over substantial periods of time. We carried out a GeneChip‐based RNA profiling analysis using our own data for testis and enriched germ cells, data for somatic cancers from the Expression Project for Oncology, and data for normal somatic tissues from the Gene Omnibus Repository. We identified 478 candidate loci that include known CT genes, numerous genes associated with oncogenic processes, and novel candidates that are not referenced in the Cancer/Testis Database (www.cta.lncc.br). We complemented RNA expression data at the protein level for SPESP1, GALNTL5, PDCL2, and C11orf42 using cancer tissue microarrays covering malignant tumors of breast, uterus, thyroid, and kidney, as well as published RNA profiling and immunohistochemical data provided by the Human Protein Atlas (www.proteinatlas.org). We report that combined RNA/tissue profiling identifies novel CT genes that may be of clinical interest as therapeutical targets or biomarkers. Our findings also highlight the challenges of detecting truly germ cell‐specific mRNAs and the proteins they encode in highly heterogenous testicular, somatic, and tumor tissues.
Collapse
Affiliation(s)
- Soazik P Jamin
- Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S, Univ Rennes, France
| | - Feria Hikmet
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Rudbeck Laboratory, Uppsala University, Sweden
| | - Romain Mathieu
- Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S, Univ Rennes, France.,Department of Urology, University Hospital, Rennes, France
| | - Bernard Jégou
- Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S, Univ Rennes, France
| | - Cecilia Lindskog
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Rudbeck Laboratory, Uppsala University, Sweden
| | - Frédéric Chalmel
- Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S, Univ Rennes, France
| | - Michael Primig
- Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S, Univ Rennes, France
| |
Collapse
|
20
|
Zhao Z, Zhang G, Li W. MT2A Promotes Oxaliplatin Resistance in Colorectal Cancer Cells. Cell Biochem Biophys 2020; 78:475-482. [PMID: 32638210 DOI: 10.1007/s12013-020-00930-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Accepted: 06/24/2020] [Indexed: 12/20/2022]
Abstract
We aimed to understand the molecular mechanism underlying the incidence of Oxaliplatin resistance in colorectal cancer. The Oxaliplatin-resistant (OR) HT29 colorectal cell line was established by long-term exposure to Oxaliplatin. Cell viability and proliferation were determined by the 3-(4,5-dimethyl thiazol-2-yl)-2,5-diphenyltetrazolium bromide and direct counting assays, respectively. Transcript level of metallothionein 2A (MT2A) was measured by real-time polymerase chain reaction. Protein levels of MT2A, BRCA1-associated RING domain 1 (BARD1), BRCA1, and β-actin were quantified by immunoblotting. Direct interaction between MT2A with BARD1 and BRCA1 was analyzed by co-immunoprecipitation. Colocalization between of MT2A and BARD1 was determined by immunofluorescence. MT2A was upregulated in OR cells at both transcript and protein levels. Knockdown of MT2A in HT29 OR cells improved sensitivity to Oxaliplatin, while ectopic overexpression of MT2A conferred HT29 cells relative resistance to Oxaliplatin. We further demonstrated that MT2A interacted with and positively regulated BARD1/BRCA1 in colorectal cancer cells. BARD1 overexpression partially restored the compromised Oxaliplatin resistance elicited by MT2A deficiency in terms of both cell proliferation and viability. Our data highlighted the critical contributions of MT2A-BARD1/BRCA1 in Oxaliplatin resistance in colorectal cancer cells.
Collapse
Affiliation(s)
- Zhicheng Zhao
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Guojing Zhang
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Weidong Li
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, 300052, China.
| |
Collapse
|
21
|
Prabhu D, Rajamanikandan S, Anusha SB, Chowdary MS, Veerapandiyan M, Jeyakanthan J. In silico Functional Annotation and Characterization of Hypothetical Proteins from Serratia marcescens FGI94. BIOL BULL+ 2020; 47:319-331. [PMID: 32834707 PMCID: PMC7394047 DOI: 10.1134/s1062359020300019] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2019] [Revised: 09/28/2019] [Accepted: 09/30/2019] [Indexed: 01/16/2023]
Abstract
Serratia marcescens, rod-shaped Gram-negative bacteria is classified as an opportunistic pathogen in the family Enterobacteriaceae. It causes a wide variety of infections in humans, including urinary, respiratory, ocular lens and ear infections, osteomyelitis, endocarditis, meningitis and septicemia. Unfortunately, over the past decade, antibiotic resistance has become a serious health care issue; the effective means to control and dissemination of S. marcescens resistance is the need of hour. The whole genome sequencing of S. marcescens FGI94 strain contains 4434 functional proteins, among which 690 (15.56%) proteins were classified under hypothetical. In the present study, we applied the power of various bioinformatics tools on the basis of protein family comparison, motifs, functional properties of amino acids and genome context to assign the possible functions for the HPs. The pseudo sequences (protein sequence that contain ≤100 amino acid residues) are eliminated from the study. Although we have successfully predicted the function for 483 proteins, we were able to infer the high level of confidence only for 108 proteins. The predicted HPs were classified into various classes such as enzymes, transporters, binding proteins, cell division, cell regulatory and other proteins. The outcome of the study could be helpful to understand the molecular mechanism in bacterial pathogenesis and also provide an insight into the identification of potential targets for drug and vaccine development.
Collapse
Affiliation(s)
- D Prabhu
- Department of Bioinformatics, Alagappa University, Science Campus, 630004 Karaikudi, Tamil Nadu India
| | - S Rajamanikandan
- ICAR-National Institute of Veterinary Epidemiology and Disease Informatics, 560064 Yelahanka, Bengaluru India
| | - S Baby Anusha
- Department of Bioinformatics, Sathyabama University, 600119 Chennai, Tamil Nadu India
| | - M Sushma Chowdary
- Department of Bioinformatics, Sathyabama University, 600119 Chennai, Tamil Nadu India
| | - M Veerapandiyan
- Department of Bioinformatics, Alagappa University, Science Campus, 630004 Karaikudi, Tamil Nadu India
| | - J Jeyakanthan
- Department of Bioinformatics, Alagappa University, Science Campus, 630004 Karaikudi, Tamil Nadu India
| |
Collapse
|
22
|
L ARP7 Is a BRCA1 Ubiquitinase Substrate and Regulates Genome Stability and Tumorigenesis. Cell Rep 2020; 32:107974. [DOI: 10.1016/j.celrep.2020.107974] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Revised: 05/05/2020] [Accepted: 07/08/2020] [Indexed: 12/13/2022] Open
|
23
|
Guo Y, Ning W, Jiang P, Lin S, Wang C, Tan X, Yao L, Peng D, Xue Y. GPS-PBS: A Deep Learning Framework to Predict Phosphorylation Sites that Specifically Interact with Phosphoprotein-Binding Domains. Cells 2020; 9:cells9051266. [PMID: 32443803 PMCID: PMC7290655 DOI: 10.3390/cells9051266] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 05/17/2020] [Accepted: 05/18/2020] [Indexed: 12/13/2022] Open
Abstract
Protein phosphorylation is essential for regulating cellular activities by modifying substrates at specific residues, which frequently interact with proteins containing phosphoprotein-binding domains (PPBDs) to propagate the phosphorylation signaling into downstream pathways. Although massive phosphorylation sites (p-sites) have been reported, most of their interacting PPBDs are unknown. Here, we collected 4458 known PPBD-specific binding p-sites (PBSs), considerably improved our previously developed group-based prediction system (GPS) algorithm, and implemented a deep learning plus transfer learning strategy for model training. Then, we developed a new online service named GPS-PBS, which can hierarchically predict PBSs of 122 single PPBD clusters belonging to two groups and 16 families. By comparison, GPS-PBS achieved a highly competitive accuracy against other existing tools. Using GPS-PBS, we predicted 371,018 mammalian p-sites that potentially interact with at least one PPBD, and revealed that various PPBD-containing proteins (PPCPs) and protein kinases (PKs) can simultaneously regulate the same p-sites to orchestrate important pathways, such as the PI3K-Akt signaling pathway. Taken together, we anticipate GPS-PBS can be a great help for further dissecting phosphorylation signaling networks.
Collapse
Affiliation(s)
- Yaping Guo
- Key Laboratory of Molecular Biophysics of Ministry of Education, Hubei Bioinformatics and Molecular Imaging Key Laboratory, Center for Artificial Intelligence Biology, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
| | - Wanshan Ning
- Key Laboratory of Molecular Biophysics of Ministry of Education, Hubei Bioinformatics and Molecular Imaging Key Laboratory, Center for Artificial Intelligence Biology, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
| | - Peiran Jiang
- Key Laboratory of Molecular Biophysics of Ministry of Education, Hubei Bioinformatics and Molecular Imaging Key Laboratory, Center for Artificial Intelligence Biology, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
| | - Shaofeng Lin
- Key Laboratory of Molecular Biophysics of Ministry of Education, Hubei Bioinformatics and Molecular Imaging Key Laboratory, Center for Artificial Intelligence Biology, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
| | - Chenwei Wang
- Key Laboratory of Molecular Biophysics of Ministry of Education, Hubei Bioinformatics and Molecular Imaging Key Laboratory, Center for Artificial Intelligence Biology, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
| | - Xiaodan Tan
- Key Laboratory of Molecular Biophysics of Ministry of Education, Hubei Bioinformatics and Molecular Imaging Key Laboratory, Center for Artificial Intelligence Biology, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
| | - Lan Yao
- Key Laboratory of Molecular Biophysics of Ministry of Education, Hubei Bioinformatics and Molecular Imaging Key Laboratory, Center for Artificial Intelligence Biology, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
| | - Di Peng
- Key Laboratory of Molecular Biophysics of Ministry of Education, Hubei Bioinformatics and Molecular Imaging Key Laboratory, Center for Artificial Intelligence Biology, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
| | - Yu Xue
- Key Laboratory of Molecular Biophysics of Ministry of Education, Hubei Bioinformatics and Molecular Imaging Key Laboratory, Center for Artificial Intelligence Biology, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
| |
Collapse
|
24
|
Network of Interactions between ZIKA Virus Non-Structural Proteins and Human Host Proteins. Cells 2020; 9:cells9010153. [PMID: 31936331 PMCID: PMC7016862 DOI: 10.3390/cells9010153] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Revised: 12/20/2019] [Accepted: 01/01/2020] [Indexed: 12/16/2022] Open
Abstract
The Zika virus (ZIKV) is a mosquito-borne Flavivirus and can be transmitted through an infected mosquito bite or through human-to-human interaction by sexual activity, blood transfusion, breastfeeding, or perinatal exposure. After the 2015-2016 outbreak in Brazil, a strong link between ZIKV infection and microcephaly emerged. ZIKV specifically targets human neural progenitor cells, suggesting that proteins encoded by ZIKV bind and inactivate host cell proteins, leading to microcephaly. Here, we present a systematic annotation of interactions between human proteins and the seven non-structural ZIKV proteins corresponding to a Brazilian isolate. The interaction network was generated by combining tandem-affinity purification followed by mass spectrometry with yeast two-hybrid screens. We identified 150 human proteins, involved in distinct biological processes, as interactors to ZIKV non-structural proteins. Our interacting network is composed of proteins that have been previously associated with microcephaly in human genetic disorders and/or animal models. Further, we show that the protein inhibitor of activated STAT1 (PIAS1) interacts with NS5 and modulates its stability. This study builds on previously published interacting networks of ZIKV and genes related to autosomal recessive primary microcephaly to generate a catalog of human cellular targets of ZIKV proteins implicated in processes related to microcephaly in humans. Collectively, these data can be used as a resource for future characterization of ZIKV infection biology and help create a basis for the discovery of drugs that may disrupt the interaction and reduce the health damage to the fetus.
Collapse
|
25
|
Functional Impacts of the BRCA1-mTORC2 Interaction in Breast Cancer. Int J Mol Sci 2019; 20:ijms20235876. [PMID: 31771139 PMCID: PMC6928641 DOI: 10.3390/ijms20235876] [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: 10/28/2019] [Revised: 11/20/2019] [Accepted: 11/21/2019] [Indexed: 12/26/2022] Open
Abstract
Deleterious mutations in Breast Cancer 1 (BRCA1) are associated with an increased risk of breast and ovarian cancer. Mutations in the tandem BRCA1 C-terminal (tBRCT) protein domain disrupt critical protein interactions required for the faithful repair of DNA through homologous recombination, which contributes to oncogenesis. Our studies have identified RICTOR, PRR5, and SIN1 subunits of the mammalian target of rapamycin complex 2 (mTORC2) as interacting partners with the tBRCT domain of BRCA1 leading to the disruption of the mTORC2 complex. However, the interplay between mTORC2 signaling and BRCA1 function in the DNA damage response (DDR) remains to be determined. In this study, we used protein interaction assays to determine the binary interactions between the tBRCT domain and mTORC2 subunits, evaluated the impact of mTOR inhibition on the transcriptional function of the tBRCT, evaluated the impact of mTOR signaling on BRCA1 recruitment to DNA damage-induced foci and determined the breast cancer cell line response to mTOR inhibition dependent upon BRCA1 expression and mutation. This study determined that PRR5, RICTOR, and SIN1 could each independently interact with the BRCA1 tBRCT. Inhibition of mTORC1, but not mTORC1/2, increases BRCA1 transcriptional activation activity. Treatment with pan-mTOR inhibitor PP242 diminishes DNA damage-induced γH2AX and BRCA1 foci formation. Breast cancer cells lacking expression of functional BRCA1 are more sensitive to mTOR inhibitors. These data suggest that mTOR signaling is required for BRCA1 response to DNA damage and breast cancer cells lacking BRCA1 are more sensitive to pan-mTOR inhibition. This work suggests chemotherapeutic strategies using mTOR inhibitors could be tailored for patients that lack functional BRCA1.
Collapse
|
26
|
Kurdekar V, Giridharan S, Subbarao J, Nijaguna MB, Periasamy J, Boggaram S, Shivange AV, Sadasivam G, Padigaru M, Potluri V, Venkitaraman AR, Bharatham K. Structure-Guided Synthesis and Evaluation of Small-Molecule Inhibitors Targeting Protein-Protein Interactions of BRCA1 tBRCT Domain. ChemMedChem 2019; 14:1620-1632. [PMID: 31334915 DOI: 10.1002/cmdc.201900300] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2019] [Revised: 07/20/2019] [Indexed: 01/09/2023]
Abstract
The tandem BRCT domains (tBRCT) of BRCA1 engage phosphoserine-containing motifs in target proteins to propagate intracellular signals initiated by DNA damage, thereby controlling cell cycle arrest and DNA repair. Recently, we identified Bractoppin, the first small-molecule inhibitor of the BRCA1 tBRCT domain, which selectively interrupts BRCA1-mediated cellular responses evoked by DNA damage. Here, we combine structure-guided chemical elaboration, protein mutagenesis and cellular assays to define the structural features responsible for Bractoppin's activity. Bractoppin fails to bind mutant forms of BRCA1 tBRCT bearing K1702A, a key residue mediating phosphopeptide recognition, or F1662R or L1701K that adjoin the pSer-recognition site. However, the M1775R mutation, which engages the Phe residue in the consensus phosphopeptide motif pSer-X-X-Phe, does not affect Bractoppin binding, confirming a binding mode distinct from the substrate phosphopeptide binding. We explored these structural features through structure-guided chemical elaboration and characterized structure-activity relationships (SARs) in biochemical assays. Two analogues, CCBT2088 and CCBT2103 were effective in abrogating BRCA1 foci formation and inhibiting G2 arrest induced by irradiation of cells. Collectively, our findings reveal structural features underlying the activity of a novel inhibitor of phosphopeptide recognition by the BRCA1 tBRCT domain, providing fresh insights to guide the development of inhibitors that target protein-protein interactions.
Collapse
Affiliation(s)
- Vadiraj Kurdekar
- Center for Chemical Biology & Therapeutics, InStem, Bellary Road, Bangalore, Karnataka, 560065, India
| | - Saranya Giridharan
- Center for Chemical Biology & Therapeutics, InStem, Bellary Road, Bangalore, Karnataka, 560065, India
| | - Jasti Subbarao
- Center for Chemical Biology & Therapeutics, InStem, Bellary Road, Bangalore, Karnataka, 560065, India
| | - Mamatha B Nijaguna
- Center for Chemical Biology & Therapeutics, InStem, Bellary Road, Bangalore, Karnataka, 560065, India
| | - Jayaprakash Periasamy
- Center for Chemical Biology & Therapeutics, InStem, Bellary Road, Bangalore, Karnataka, 560065, India
| | - Sanjana Boggaram
- Center for Chemical Biology & Therapeutics, InStem, Bellary Road, Bangalore, Karnataka, 560065, India
| | - Amol V Shivange
- Center for Chemical Biology & Therapeutics, InStem, Bellary Road, Bangalore, Karnataka, 560065, India
| | - Gayathri Sadasivam
- Center for Chemical Biology & Therapeutics, InStem, Bellary Road, Bangalore, Karnataka, 560065, India
| | - Muralidhara Padigaru
- Center for Chemical Biology & Therapeutics, InStem, Bellary Road, Bangalore, Karnataka, 560065, India
| | - Vijay Potluri
- Center for Chemical Biology & Therapeutics, InStem, Bellary Road, Bangalore, Karnataka, 560065, India
| | - Ashok R Venkitaraman
- Center for Chemical Biology & Therapeutics, InStem, Bellary Road, Bangalore, Karnataka, 560065, India.,Medical Research Council Cancer Unit, University of Cambridge, Hills Road, Cambridge, CB2 0XZ, UK
| | - Kavitha Bharatham
- Center for Chemical Biology & Therapeutics, InStem, Bellary Road, Bangalore, Karnataka, 560065, India
| |
Collapse
|
27
|
Pillon MC, Lo YH, Stanley RE. IT'S 2 for the price of 1: Multifaceted ITS2 processing machines in RNA and DNA maintenance. DNA Repair (Amst) 2019; 81:102653. [PMID: 31324529 PMCID: PMC6764878 DOI: 10.1016/j.dnarep.2019.102653] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Cells utilize sophisticated RNA processing machines to ensure the quality of RNA. Many RNA processing machines have been further implicated in regulating the DNA damage response signifying a strong link between RNA processing and genome maintenance. One of the most intricate and highly regulated RNA processing pathways is the processing of the precursor ribosomal RNA (pre-rRNA), which is paramount for the production of ribosomes. Removal of the Internal Transcribed Spacer 2 (ITS2), located between the 5.8S and 25S rRNA, is one of the most complex steps of ribosome assembly. Processing of the ITS2 is initiated by the newly discovered endoribonuclease Las1, which cleaves at the C2 site within the ITS2, generating products that are further processed by the polynucleotide kinase Grc3, the 5'→3' exonuclease Rat1, and the 3'→5' RNA exosome complex. In addition to their defined roles in ITS2 processing, these critical cellular machines participate in other stages of ribosome assembly, turnover of numerous cellular RNAs, and genome maintenance. Here we summarize recent work defining the molecular mechanisms of ITS2 processing by these essential RNA processing machines and highlight their emerging roles in transcription termination, heterochromatin function, telomere maintenance, and DNA repair.
Collapse
Affiliation(s)
- Monica C Pillon
- Signal Transduction Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, 111 T. W. Alexander Drive, Research Triangle Park, NC 27709, USA
| | - Yu-Hua Lo
- Signal Transduction Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, 111 T. W. Alexander Drive, Research Triangle Park, NC 27709, USA
| | - Robin E Stanley
- Signal Transduction Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, 111 T. W. Alexander Drive, Research Triangle Park, NC 27709, USA.
| |
Collapse
|
28
|
Lagundžin D, Hu WF, Law HCH, Krieger KL, Qiao F, Clement EJ, Drincic AT, Nedić O, Naldrett MJ, Alvarez S, Woods NT. Delineating the role of FANCA in glucose-stimulated insulin secretion in β cells through its protein interactome. PLoS One 2019; 14:e0220568. [PMID: 31461451 PMCID: PMC6713327 DOI: 10.1371/journal.pone.0220568] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Accepted: 07/18/2019] [Indexed: 12/31/2022] Open
Abstract
Hyperinsulinemia affects 72% of Fanconi anemia (FA) patients and an additional 25% experience lowered glucose tolerance or frank diabetes. The underlying molecular mechanisms contributing to the dysfunction of FA pancreas β cells is unknown. Therefore, we sought to evaluate the functional role of FANCA, the most commonly mutated gene in FA, in glucose-stimulated insulin secretion (GSIS). This study reveals that FANCA or FANCB knockdown impairs GSIS in human pancreas β cell line EndoC-βH3. To identify potential pathways by which FANCA might regulate GSIS, we employed a proteomics approach to identify FANCA protein interactions in EndoC-βH3 differentially regulated in response to elevated glucose levels. Glucose-dependent changes in the FANCA interaction network were observed, including increased association with other FA family proteins, suggesting an activation of the DNA damage response in response to elevated glucose levels. Reactive oxygen species increase in response to glucose stimulation and are necessary for GSIS in EndoC-βH3 cells. Glucose-induced activation of the DNA damage response was also observed as an increase in the DNA damage foci marker γ-H2AX and dependent upon the presence of reactive oxygen species. These results illuminate the role of FANCA in GSIS and its protein interactions regulated by glucose stimulation that may explain the prevalence of β cell-specific endocrinopathies in FA patients.
Collapse
Affiliation(s)
- Dragana Lagundžin
- Eppley Institute for Research in Cancer and Allied Diseases, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
- Mass Spectrometry and Proteomics Core Facility, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Wen-Feng Hu
- Eppley Institute for Research in Cancer and Allied Diseases, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Henry C. H. Law
- Eppley Institute for Research in Cancer and Allied Diseases, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Kimiko L. Krieger
- Eppley Institute for Research in Cancer and Allied Diseases, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Fangfang Qiao
- Eppley Institute for Research in Cancer and Allied Diseases, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Emalie J. Clement
- Eppley Institute for Research in Cancer and Allied Diseases, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Andjela T. Drincic
- Department of Internal Medicine: Diabetes, Endocrinology and Metabolism, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Olgica Nedić
- Institute for the Application of Nuclear Energy, University of Belgrade, Banatska, Belgrade, Serbia
| | - Michael J. Naldrett
- Proteomics & Metabolomics Facility, Nebraska Center for Biotechnology, University of Nebraska–Lincoln, Nebraska, United States of America
| | - Sophie Alvarez
- Proteomics & Metabolomics Facility, Nebraska Center for Biotechnology, University of Nebraska–Lincoln, Nebraska, United States of America
| | - Nicholas T. Woods
- Eppley Institute for Research in Cancer and Allied Diseases, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
- * E-mail:
| |
Collapse
|
29
|
Bluhm A, Viceconte N, Li F, Rane G, Ritz S, Wang S, Levin M, Shi Y, Kappei D, Butter F. ZBTB10 binds the telomeric variant repeat TTGGGG and interacts with TRF2. Nucleic Acids Res 2019; 47:1896-1907. [PMID: 30629181 PMCID: PMC6393293 DOI: 10.1093/nar/gky1289] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Revised: 11/27/2018] [Accepted: 12/14/2018] [Indexed: 12/18/2022] Open
Abstract
Telomeres are nucleoprotein structures at the ends of linear chromosomes and present an essential feature for genome integrity. Vertebrate telomeres usually consist of hexameric TTAGGG repeats, however, in cells that use the alternative lengthening of telomeres (ALT) mechanism, variant repeat sequences are interspersed throughout telomeres. Previously, it was shown that NR2C/F transcription factors bind to TCAGGG variant repeats and contribute to telomere maintenance in ALT cells. While specific binders to other variant repeat sequences have been lacking to date, we here identify ZBTB10 as the first TTGGGG-binding protein and demonstrate direct binding via the two zinc fingers with affinity in the nanomolar range. Concomitantly, ZBTB10 co-localizes with a subset of telomeres in ALT-positive U2OS cells and interacts with TRF2/RAP1 via the N-terminal region of TRF2. Our data establishes ZBTB10 as a novel variant repeat binding protein at ALT telomeres.
Collapse
Affiliation(s)
- Alina Bluhm
- Quantitative Proteomics, Institute of Molecular Biology, 55128 Mainz, Germany
| | - Nikenza Viceconte
- Quantitative Proteomics, Institute of Molecular Biology, 55128 Mainz, Germany
| | - Fudong Li
- Hefei National Laboratory for Physical Sciences at Microscale and School of Life Sciences, University of Science and Technology of China, 230027 Hefei, Anhui, China
| | - Grishma Rane
- Cancer Science Institute of Singapore, National University of Singapore, Singapore 117599
| | - Sandra Ritz
- Microscopy Core Facility, Institute of Molecular Biology, 55128 Mainz, Germany
| | - Suman Wang
- Hefei National Laboratory for Physical Sciences at Microscale and School of Life Sciences, University of Science and Technology of China, 230027 Hefei, Anhui, China
| | - Michal Levin
- Quantitative Proteomics, Institute of Molecular Biology, 55128 Mainz, Germany
| | - Yunyu Shi
- Hefei National Laboratory for Physical Sciences at Microscale and School of Life Sciences, University of Science and Technology of China, 230027 Hefei, Anhui, China
| | - Dennis Kappei
- Cancer Science Institute of Singapore, National University of Singapore, Singapore 117599
| | - Falk Butter
- Quantitative Proteomics, Institute of Molecular Biology, 55128 Mainz, Germany
| |
Collapse
|
30
|
Wan B, Wu J, Meng X, Lei M, Zhao X. Molecular Basis for Control of Diverse Genome Stability Factors by the Multi-BRCT Scaffold Rtt107. Mol Cell 2019; 75:238-251.e5. [PMID: 31348879 PMCID: PMC6745058 DOI: 10.1016/j.molcel.2019.05.035] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Revised: 04/11/2019] [Accepted: 05/30/2019] [Indexed: 01/08/2023]
Abstract
BRCT domains support myriad protein-protein interactions involved in genome maintenance. Although di-BRCT recognition of phospho-proteins is well known to support the genotoxic response, whether multi-BRCT domains can acquire distinct structures and functions is unclear. Here we present the tetra-BRCT structures from the conserved yeast protein Rtt107 in free and ligand-bound forms. The four BRCT repeats fold into a tetrahedral structure that recognizes unmodified ligands using a bi-partite mechanism, suggesting repeat origami enabling function acquisition. Functional studies show that Rtt107 binding of partner proteins of diverse activities promotes genome replication and stability in both distinct and concerted manners. A unified theme is that tetra- and di-BRCT domains of Rtt107 collaborate to recruit partner proteins to chromatin. Our work thus illustrates how a master regulator uses two types of BRCT domains to recognize distinct genome factors and direct them to chromatin for constitutive genome protection.
Collapse
Affiliation(s)
- Bingbing Wan
- Molecular Biology Department, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Jian Wu
- Shanghai Institute of Precision Medicine, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200125, China
| | - Xiangzhou Meng
- Molecular Biology Department, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Ming Lei
- Shanghai Institute of Precision Medicine, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200125, China; Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai 200125, China; National Facility for Protein Science in Shanghai, Zhangjiang Lab, Shanghai 201210, China; Shanghai Science Research Center, Chinese Academy of Sciences, Shanghai 201204, China.
| | - Xiaolan Zhao
- Molecular Biology Department, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA.
| |
Collapse
|
31
|
Hu WF, Krieger KL, Lagundžin D, Li X, Cheung RS, Taniguchi T, Johnson KR, Bessho T, Monteiro ANA, Woods NT. CTDP1 regulates breast cancer survival and DNA repair through BRCT-specific interactions with FANCI. Cell Death Discov 2019; 5:105. [PMID: 31240132 PMCID: PMC6584691 DOI: 10.1038/s41420-019-0185-3] [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: 04/22/2019] [Revised: 05/22/2019] [Accepted: 05/30/2019] [Indexed: 12/20/2022] Open
Abstract
BRCA1 C-terminal domains are found in a specialized group of 23 proteins that function in the DNA damage response to protect genomic integrity. C-terminal domain phosphatase 1 (CTDP1) is the only phosphatase with a BRCA1 C-terminal domain in the human proteome, yet direct participation in the DNA damage response has not been reported. Examination of the CTDP1 BRCA1 C-terminal domain-specific protein interaction network revealed 103 high confidence interactions enriched in DNA damage response proteins, including FANCA and FANCI that are central to the Fanconi anemia DNA repair pathway necessary for the resolution of DNA interstrand crosslink damage. CTDP1 expression promotes DNA damage-induced FANCA and FANCD2 foci formation and enhances homologous recombination repair efficiency. CTDP1 was found to regulate multiple aspects of FANCI activity, including chromatin localization, interaction with γ-H2AX, and SQ motif phosphorylations. Knockdown of CTDP1 increases MCF-10A sensitivity to DNA interstrand crosslinks and double-strand breaks, but not ultraviolet radiation. In addition, CTDP1 knockdown impairs in vitro and in vivo growth of breast cancer cell lines. These results elucidate the molecular functions of CTDP1 in Fanconi anemia interstrand crosslink repair and identify this protein as a potential target for breast cancer therapy.
Collapse
Affiliation(s)
- Wen-Feng Hu
- 1Eppley Institute for Research in Cancer and Allied Diseases, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198 USA.,2Center for Molecular Medicine, Xiangya Hospital, Central South University, Changsha, China
| | - Kimiko L Krieger
- 1Eppley Institute for Research in Cancer and Allied Diseases, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198 USA
| | - Dragana Lagundžin
- 1Eppley Institute for Research in Cancer and Allied Diseases, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198 USA.,3Mass Spectrometry and Proteomics Core Facility, University of Nebraska Medical Center, Omaha, NE 68198 USA
| | - Xueli Li
- 4Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612 USA
| | - Ronald S Cheung
- 5Divisions of Human Biology and Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA 98109 USA
| | - Toshiyasu Taniguchi
- 5Divisions of Human Biology and Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA 98109 USA.,6Department of Molecular Life Science, Tokai University School of Medicine, Isehara, Kanagawa Japan
| | - Keith R Johnson
- 1Eppley Institute for Research in Cancer and Allied Diseases, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198 USA
| | - Tadayoshi Bessho
- 1Eppley Institute for Research in Cancer and Allied Diseases, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198 USA
| | - Alvaro N A Monteiro
- 4Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612 USA
| | - Nicholas T Woods
- 1Eppley Institute for Research in Cancer and Allied Diseases, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198 USA
| |
Collapse
|
32
|
Huff LP, Kikuchi DS, Faidley E, Forrester SJ, Tsai MZ, Lassègue B, Griendling KK. Polymerase-δ-interacting protein 2 activates the RhoGEF epithelial cell transforming sequence 2 in vascular smooth muscle cells. Am J Physiol Cell Physiol 2019; 316:C621-C631. [PMID: 30726115 DOI: 10.1152/ajpcell.00208.2018] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Polymerase-δ-interacting protein 2 (Poldip2) controls a wide variety of cellular functions and vascular pathologies. To mediate these effects, Poldip2 interacts with numerous proteins and generates reactive oxygen species via the enzyme NADPH oxidase 4 (Nox4). We have previously shown that Poldip2 can activate the Rho family GTPase RhoA, another signaling node within the cell. In this study, we aimed to better understand how Poldip2 activates Rho family GTPases and the functions of the involved proteins in vascular smooth muscle cells (VSMCs). RhoA is activated by guanine nucleotide exchange factors. Using nucleotide-free RhoA (isolated from bacteria) to pulldown active RhoGEFs, we found that the RhoGEF epithelial cell transforming sequence 2 (Ect2) is activated by Poldip2. Ect2 is a critical RhoGEF for Poldip2-mediated RhoA activation, because siRNA against Ect2 prevented Poldip2-mediated RhoA activity (measured by rhotekin pulldowns). Surprisingly, we were unable to detect a direct interaction between Poldip2 and Ect2, as they did not coimmunoprecipitate. Nox4 is not required for Poldip2-driven Ect2 activation, as Poldip2 overexpression induced Ect2 activation in Nox4 knockout VSMCs similar to wild-type cells. However, antioxidant treatment blocked Poldip2-induced Ect2 activation. This indicates a novel reactive oxygen species-driven mechanism by which Poldip2 regulates Rho family GTPases. Finally, we examined the function of these proteins in VSMCs, using siRNA against Poldip2 or Ect2 and determined that Poldip2 and Ect2 are both essential for vascular smooth muscle cell cytokinesis and proliferation.
Collapse
Affiliation(s)
- Lauren Parker Huff
- Department of Medicine, Division of Cardiology, Emory University School of Medicine , Atlanta, Georgia
| | - Daniel Seicho Kikuchi
- Department of Medicine, Division of Cardiology, Emory University School of Medicine , Atlanta, Georgia
| | - Elizabeth Faidley
- Department of Medicine, Division of Cardiology, Emory University School of Medicine , Atlanta, Georgia
| | - Steven J Forrester
- Department of Medicine, Division of Cardiology, Emory University School of Medicine , Atlanta, Georgia
| | - Michelle Z Tsai
- Department of Medicine, Division of Cardiology, Emory University School of Medicine , Atlanta, Georgia
| | - Bernard Lassègue
- Department of Medicine, Division of Cardiology, Emory University School of Medicine , Atlanta, Georgia
| | - Kathy K Griendling
- Department of Medicine, Division of Cardiology, Emory University School of Medicine , Atlanta, Georgia
| |
Collapse
|
33
|
Li J, Fang R, Wang J, Deng L. NOP14 inhibits melanoma proliferation and metastasis by regulating Wnt/β-catenin signaling pathway. ACTA ACUST UNITED AC 2018; 52:e7952. [PMID: 30484495 PMCID: PMC6262753 DOI: 10.1590/1414-431x20187952] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Accepted: 09/27/2018] [Indexed: 12/27/2022]
Abstract
Malignant melanoma is an aggressive skin cancer with a high mortality rate. Nucleolar protein 14 (NOP14) has been implicated in cancer development. However, the role of NOP14 in malignant melanoma progression remains largely unclear. In this study, we observed that malignant melanoma tissue showed NOP14 down-regulation compared to melanocytic nevi tissues. Moreover, we observed that NOP14 expression was significantly associated with melanoma tumor thickness and lymph node metastasis. NOP14 overexpression in melanoma cells suppressed proliferation, caused G1 phase arrest, promoted apoptosis, and inhibited melanoma cell migration and invasion. Further investigations revealed that NOP14 overexpression reduced the expression levels of Wnt3a, β-catenin, and GSK-3β of the Wnt/β-catenin pathway. In summary, we demonstrated that NOP14 inhibited melanoma cell proliferation and metastasis by regulating the Wnt/β-catenin signaling pathway.
Collapse
Affiliation(s)
- Jingrong Li
- Department of Dermatology, The First Affiliated Hospital of Jinan University, Guangzhou, China.,Department of Dermatology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Ruihua Fang
- Department of Dermatology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Jianqin Wang
- Department of Dermatology, Guangzhou Institute of Dermatology, Guangzhou, China
| | - Liehua Deng
- Department of Dermatology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| |
Collapse
|
34
|
Periasamy J, Kurdekar V, Jasti S, Nijaguna MB, Boggaram S, Hurakadli MA, Raina D, Kurup LM, Chintha C, Manjunath K, Goyal A, Sadasivam G, Bharatham K, Padigaru M, Potluri V, Venkitaraman AR. Targeting Phosphopeptide Recognition by the Human BRCA1 Tandem BRCT Domain to Interrupt BRCA1-Dependent Signaling. Cell Chem Biol 2018; 25:677-690.e12. [PMID: 29606576 PMCID: PMC6015222 DOI: 10.1016/j.chembiol.2018.02.012] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Revised: 11/24/2017] [Accepted: 02/22/2018] [Indexed: 02/06/2023]
Abstract
Intracellular signals triggered by DNA breakage flow through proteins containing BRCT (BRCA1 C-terminal) domains. This family, comprising 23 conserved phosphopeptide-binding modules in man, is inaccessible to small-molecule chemical inhibitors. Here, we develop Bractoppin, a drug-like inhibitor of phosphopeptide recognition by the human BRCA1 tandem (t)BRCT domain, which selectively inhibits substrate binding with nanomolar potency in vitro. Structure-activity exploration suggests that Bractoppin engages BRCA1 tBRCT residues recognizing pSer in the consensus motif, pSer-Pro-Thr-Phe, plus an abutting hydrophobic pocket that is distinct in structurally related BRCT domains, conferring selectivity. In cells, Bractoppin inhibits substrate recognition detected by Förster resonance energy transfer, and diminishes BRCA1 recruitment to DNA breaks, in turn suppressing damage-induced G2 arrest and assembly of the recombinase, RAD51. But damage-induced MDC1 recruitment, single-stranded DNA (ssDNA) generation, and TOPBP1 recruitment remain unaffected. Thus, an inhibitor of phosphopeptide recognition selectively interrupts BRCA1 tBRCT-dependent signals evoked by DNA damage. Bractoppin selectively blocks phosphopeptide recognition by the BRCA1 tBRCT domain Bractoppin engages tBRCT residues recognizing pSer, plus an adjacent pocket Bractoppin interrupts BRCA1 tBRCT-dependent cellular signals evoked by DNA damage This work opens avenues to inhibit intracellular signaling by the tBRCT domain family
Collapse
Affiliation(s)
- Jayaprakash Periasamy
- Center for Chemical Biology & Therapeutics, InSTEM, Bellary Road, Bangalore, Karnataka 560065, India
| | - Vadiraj Kurdekar
- Center for Chemical Biology & Therapeutics, InSTEM, Bellary Road, Bangalore, Karnataka 560065, India
| | - Subbarao Jasti
- Center for Chemical Biology & Therapeutics, InSTEM, Bellary Road, Bangalore, Karnataka 560065, India
| | - Mamatha B Nijaguna
- Center for Chemical Biology & Therapeutics, InSTEM, Bellary Road, Bangalore, Karnataka 560065, India
| | - Sanjana Boggaram
- Center for Chemical Biology & Therapeutics, InSTEM, Bellary Road, Bangalore, Karnataka 560065, India
| | - Manjunath A Hurakadli
- Center for Chemical Biology & Therapeutics, InSTEM, Bellary Road, Bangalore, Karnataka 560065, India
| | - Dhruv Raina
- Center for Chemical Biology & Therapeutics, InSTEM, Bellary Road, Bangalore, Karnataka 560065, India
| | - Lokavya Meenakshi Kurup
- Center for Chemical Biology & Therapeutics, InSTEM, Bellary Road, Bangalore, Karnataka 560065, India
| | - Chetan Chintha
- Center for Chemical Biology & Therapeutics, InSTEM, Bellary Road, Bangalore, Karnataka 560065, India
| | - Kavyashree Manjunath
- Center for Chemical Biology & Therapeutics, InSTEM, Bellary Road, Bangalore, Karnataka 560065, India
| | - Aneesh Goyal
- Center for Chemical Biology & Therapeutics, InSTEM, Bellary Road, Bangalore, Karnataka 560065, India
| | - Gayathri Sadasivam
- Center for Chemical Biology & Therapeutics, InSTEM, Bellary Road, Bangalore, Karnataka 560065, India
| | - Kavitha Bharatham
- Center for Chemical Biology & Therapeutics, InSTEM, Bellary Road, Bangalore, Karnataka 560065, India
| | - Muralidhara Padigaru
- Center for Chemical Biology & Therapeutics, InSTEM, Bellary Road, Bangalore, Karnataka 560065, India
| | - Vijay Potluri
- Center for Chemical Biology & Therapeutics, InSTEM, Bellary Road, Bangalore, Karnataka 560065, India
| | - Ashok R Venkitaraman
- Center for Chemical Biology & Therapeutics, InSTEM, Bellary Road, Bangalore, Karnataka 560065, India; Medical Research Council Cancer Unit, University of Cambridge, Hills Road, Cambridge CB2 0XZ, UK.
| |
Collapse
|
35
|
Tanaka H, Phipps EA, Wei T, Wu X, Goswami C, Liu Y, Sledge GW, Mina L, Herbert BS. Altered expression of telomere-associated genes in leukocytes among BRCA1 and BRCA2 carriers. Mol Carcinog 2018; 57:567-575. [PMID: 29240257 PMCID: PMC5832588 DOI: 10.1002/mc.22773] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Revised: 11/29/2017] [Accepted: 12/07/2017] [Indexed: 12/31/2022]
Abstract
Telomere dysfunction resulting from telomere shortening and deregulation of shelterin components has been linked to the pathogenesis of age-related disorders, including cancer. Recent evidence suggests that BRCA1/2 (BRCA1 and BRCA2) tumor suppressor gene products play an important role in telomere maintenance. Although telomere shortening has been reported in BRCA1/2 carriers, the direct effects of BRCA1/2 haploinsufficiency on telomere maintenance and predisposition to cancer development are not completely understood. In this study, we assessed the telomere-associated and telomere-proximal gene expression profiles in peripheral blood leukocytes from patients with a BRCA1 or BRCA2 mutation, compared to samples from sporadic and familial breast cancer individuals. We found that 25 genes, including TINF2 gene (a negative regulator of telomere length), were significantly differentially expressed in BRCA1 carriers. Leukocyte telomere length analysis revealed that BRCA1/2 carriers had relatively shorter telomeres than healthy controls. Further, affected BRCA1/2 carriers were well differentiated from unaffected BRCA1/2 carriers by the expression of telomere-proximal genes. Our results link BRCA1/2 haploinsufficiency to changes in telomere length, telomere-associated as well as telomere-proximal gene expression. Thus, this work supports the effect of BRCA1/2 haploinsufficiency in the biology underlying telomere dysfunction in cancer development. Future studies evaluating these findings will require a large study population.
Collapse
Affiliation(s)
- Hiromi Tanaka
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN
- Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, IN
| | - Elizabeth A. Phipps
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN
| | - Ting Wei
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN
| | - Xi Wu
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN
| | - Chirayu Goswami
- Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN
| | - Yunlong Liu
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN
- Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, IN
- Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN
- Center for Medical Genomics, Indiana University School of Medicine, Indianapolis, IN
| | | | - Lida Mina
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN
| | - Brittney-Shea Herbert
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN
- Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, IN
| |
Collapse
|
36
|
Kucharski TJ, Minshall PE, Moustafa-Kamal M, Turnell AS, Teodoro JG. Reciprocal Regulation between 53BP1 and the Anaphase-Promoting Complex/Cyclosome Is Required for Genomic Stability during Mitotic Stress. Cell Rep 2017; 18:1982-1995. [PMID: 28228263 DOI: 10.1016/j.celrep.2017.01.080] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Revised: 01/09/2017] [Accepted: 01/30/2017] [Indexed: 10/20/2022] Open
Abstract
The anaphase-promoting complex/cyclosome (APC/C) is an E3 ubiquitin ligase that targets substrates for degradation to promote mitotic progression. Here, we show that the DNA damage response protein 53BP1 contains conserved KEN boxes that are required for APC/C-dependent degradation in early mitosis. Mutation of the 53BP1 KEN boxes stabilized the protein and extended mitotic duration, whereas 53BP1 knockdown resulted in a shorter and delayed mitosis. Loss of 53BP1 increased APC/C activity, and we show that 53BP1 is a direct APC/C inhibitor. Although 53BP1 function is not absolutely required for normal cell cycle progression, knockdown was highly toxic in combination with mitotic spindle poisons. Moreover, chemical inhibition of the APC/C was able to rescue the lethality of 53BP1 loss. Our findings reveal a reciprocal regulation between 53BP1 and APC/C that is required for response to mitotic stress and may contribute to the tumor-suppressor functions of 53BP1.
Collapse
Affiliation(s)
- Thomas J Kucharski
- Goodman Cancer Research Centre, McGill University, Montreal, QC H3A 1A3, Canada; Department of Biochemistry, McGill University, Montreal, QC H3G 1Y6, Canada
| | - Paul E Minshall
- School of Cancer and Genomic Sciences, College of Medical and Dental Sciences, The University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
| | - Mohamed Moustafa-Kamal
- Goodman Cancer Research Centre, McGill University, Montreal, QC H3A 1A3, Canada; Department of Biochemistry, McGill University, Montreal, QC H3G 1Y6, Canada
| | - Andrew S Turnell
- School of Cancer and Genomic Sciences, College of Medical and Dental Sciences, The University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
| | - Jose G Teodoro
- Goodman Cancer Research Centre, McGill University, Montreal, QC H3A 1A3, Canada; Department of Biochemistry, McGill University, Montreal, QC H3G 1Y6, Canada; Department of Microbiology and Immunology, Montreal, QC H3A 2B4, Canada.
| |
Collapse
|
37
|
Riera‐Romo M. COMMD1: A Multifunctional Regulatory Protein. J Cell Biochem 2017; 119:34-51. [DOI: 10.1002/jcb.26151] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Accepted: 05/19/2017] [Indexed: 12/14/2022]
Affiliation(s)
- Mario Riera‐Romo
- Department of PharmacologyInstitute of Marine SciencesHavanaCuba
| |
Collapse
|
38
|
De Gregoriis G, Ramos JA, Fernandes PV, Vignal GM, Brianese RC, Carraro DM, Monteiro AN, Struchiner CJ, Suarez-Kurtz G, Vianna-Jorge R, de Carvalho MA. DNA repair genes PAXIP1 and TP53BP1 expression is associated with breast cancer prognosis. Cancer Biol Ther 2017; 18:439-449. [PMID: 28475402 DOI: 10.1080/15384047.2017.1323590] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Despite remarkable advances in diagnosis, prognosis and treatment, advanced or recurrent breast tumors have limited therapeutic approaches. Many treatment strategies try to explore the limitations of DNA damage response (DDR) in tumor cells to selectively eliminate them. BRCT (BRCA1 C-terminal) domains are present in a superfamily of proteins involved in cell cycle checkpoints and the DDR. Tandem BRCT domains (tBRCT) represent a distinct class of these domains. We investigated the expression profile of 7 tBRCT genes (BARD1, BRCA1, LIG4, ECT2, MDC1, PAXIP1/PTIP and TP53BP1) in breast cancer specimens and observed a high correlation between PAXIP1 and TP53BP1 gene expression in tumor samples. Tumors with worse prognosis (tumor grade 3 and triple negative) showed reduced expression of tBRCT genes, notably, PAXIP1 and TP53BP1. Survival analyses data indicated that tumor status of both genes may impact prognosis. PAXIP1 and 53BP1 protein levels followed gene expression results, i.e., are intrinsically correlated, and also reduced in more advanced tumors. Evaluation of both genes in triple negative breast tumor samples which were characterized for their BRCA1 status showed that PAXIP1 is overexpressed in BRCA1 mutant tumors. Taken together our findings indicate that PAXIP1 status correlates with breast cancer staging, in a manner similar to what has been characterized for TP53BP1.
Collapse
Affiliation(s)
- Giuliana De Gregoriis
- a Programa de Pesquisa Clínica , Coordenação de Pesquisa, Instituto Nacional de Câncer , Rio de Janeiro , RJ , Brazil
| | | | | | - Giselle Maria Vignal
- c Divisão de Patologia , Instituto Nacional de Câncer , Rio de Janeiro , RJ , Brazil
| | | | - Dirce Maria Carraro
- d International Research Center, A. C. Camargo Cancer Center , São Paulo , SP , Brazil
| | - Alvaro N Monteiro
- e Cancer Epidemiology Program, H. Lee Moffitt Cancer Center and Research Institute , Tampa , FL , USA
| | | | - Guilherme Suarez-Kurtz
- a Programa de Pesquisa Clínica , Coordenação de Pesquisa, Instituto Nacional de Câncer , Rio de Janeiro , RJ , Brazil
| | - Rosane Vianna-Jorge
- a Programa de Pesquisa Clínica , Coordenação de Pesquisa, Instituto Nacional de Câncer , Rio de Janeiro , RJ , Brazil.,g Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro , Rio de Janeiro , RJ , Brazil
| | - Marcelo Alex de Carvalho
- a Programa de Pesquisa Clínica , Coordenação de Pesquisa, Instituto Nacional de Câncer , Rio de Janeiro , RJ , Brazil.,b Instituto Federal do Rio de Janeiro , Rio de Janeiro , RJ , Brazil
| |
Collapse
|
39
|
Affiliation(s)
- W Douglas Cress
- a Molecular Oncology Program , H. Lee Moffitt Cancer Center and Research Institute , Tampa , FL , USA
| |
Collapse
|
40
|
Nepomuceno TC, Fernandes VC, Gomes TT, Carvalho RS, Suarez-Kurtz G, Monteiro AN, Carvalho MA. BRCA1 recruitment to damaged DNA sites is dependent on CDK9. Cell Cycle 2017; 16:665-672. [PMID: 28278048 DOI: 10.1080/15384101.2017.1295177] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Double strand break lesions, the most toxic type of DNA damage, are repaired primarily through 2 distinct pathways: homology-directed recombination (HR) and non-homologous end-joining (NHEJ). BRCA1 and 53BP1, 2 proteins containing the BRCT modular domain, play an important role in DNA damage response (DDR) by orchestrating the decision between HR and NHEJ, but the precise mechanisms regarding both pathways are not entirely understood. Previously, our group identified a putative interaction between BRCA1 and BARD1 (BRCA1-associated RING domain 1) and the cyclin-dependent kinase (CDK9). CDK9 is a component of the positive transcription elongation complex and has been implicated in genome integrity maintenance associated with the replication stress response. Here we show that CDK9 interacts with endogenous BRCA1 and BARD1 mediated by their RING finger and BRCT domains, and describe CDK9 ionizing radiation-induced foci (IRIF) formation and its co-localization with BRCA1 in DNA damage sites. Cells lacking CDK9 are characterized by an altered γ-H2AX foci dynamics after DNA damage, a reduced efficiency in HR but not in NHEJ repair, failure to form BRCA1 and RAD51 IRIF and increased sensitivity to genotoxic agents. These data indicate that CDK9 is a player in the DDR and is consistent with its participation in HR pathway by modulating BRCA1 response.
Collapse
Affiliation(s)
- Thales C Nepomuceno
- a Programa de Pesquisa Clínica , Instituto Nacional de Câncer , Rio de Janeiro , Brazil
| | - Vanessa C Fernandes
- a Programa de Pesquisa Clínica , Instituto Nacional de Câncer , Rio de Janeiro , Brazil
| | - Thiago T Gomes
- b Instituto Federal do Rio de Janeiro - IFRJ , Rio de Janeiro , Brazil
| | - Renato S Carvalho
- c Faculdade de Farmácia - Universidade Federal do Rio de Janeiro , Rio de Janeiro , RJ , Brazil
| | | | - Alvaro N Monteiro
- d Cancer Epidemiology Program , H. Lee Moffitt Cancer Center & Research Institute , Tampa , FL , USA
| | - Marcelo A Carvalho
- a Programa de Pesquisa Clínica , Instituto Nacional de Câncer , Rio de Janeiro , Brazil.,b Instituto Federal do Rio de Janeiro - IFRJ , Rio de Janeiro , Brazil
| |
Collapse
|
41
|
Li Z, Ivanov AA, Su R, Gonzalez-Pecchi V, Qi Q, Liu S, Webber P, McMillan E, Rusnak L, Pham C, Chen X, Mo X, Revennaugh B, Zhou W, Marcus A, Harati S, Chen X, Johns MA, White MA, Moreno C, Cooper LAD, Du Y, Khuri FR, Fu H. The OncoPPi network of cancer-focused protein-protein interactions to inform biological insights and therapeutic strategies. Nat Commun 2017; 8:14356. [PMID: 28205554 PMCID: PMC5316855 DOI: 10.1038/ncomms14356] [Citation(s) in RCA: 137] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Accepted: 12/20/2016] [Indexed: 12/19/2022] Open
Abstract
As genomics advances reveal the cancer gene landscape, a daunting task is to understand how these genes contribute to dysregulated oncogenic pathways. Integration of cancer genes into networks offers opportunities to reveal protein-protein interactions (PPIs) with functional and therapeutic significance. Here, we report the generation of a cancer-focused PPI network, termed OncoPPi, and identification of >260 cancer-associated PPIs not in other large-scale interactomes. PPI hubs reveal new regulatory mechanisms for cancer genes like MYC, STK11, RASSF1 and CDK4. As example, the NSD3 (WHSC1L1)-MYC interaction suggests a new mechanism for NSD3/BRD4 chromatin complex regulation of MYC-driven tumours. Association of undruggable tumour suppressors with drug targets informs therapeutic options. Based on OncoPPi-derived STK11-CDK4 connectivity, we observe enhanced sensitivity of STK11-silenced lung cancer cells to the FDA-approved CDK4 inhibitor palbociclib. OncoPPi is a focused PPI resource that links cancer genes into a signalling network for discovery of PPI targets and network-implicated tumour vulnerabilities for therapeutic interrogation.
Collapse
Affiliation(s)
- Zenggang Li
- Department of Pharmacology and Emory Chemical Biology Discovery Center, Emory University, Atlanta, Georgia 30322, USA
| | - Andrei A Ivanov
- Department of Pharmacology and Emory Chemical Biology Discovery Center, Emory University, Atlanta, Georgia 30322, USA
| | - Rina Su
- Department of Pharmacology and Emory Chemical Biology Discovery Center, Emory University, Atlanta, Georgia 30322, USA.,Department of Dermatology, Xiangya Hospital, Central South University, Changsha 410008, China.,Department of Dermatology, Beijing Chao-yang Hospital, Capital Medical University, Beijing 100020, China
| | - Valentina Gonzalez-Pecchi
- Department of Pharmacology and Emory Chemical Biology Discovery Center, Emory University, Atlanta, Georgia 30322, USA
| | - Qi Qi
- Department of Pharmacology and Emory Chemical Biology Discovery Center, Emory University, Atlanta, Georgia 30322, USA
| | - Songlin Liu
- Department of Pharmacology and Emory Chemical Biology Discovery Center, Emory University, Atlanta, Georgia 30322, USA.,Department of Dermatology, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Philip Webber
- Department of Pharmacology and Emory Chemical Biology Discovery Center, Emory University, Atlanta, Georgia 30322, USA
| | - Elizabeth McMillan
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, Texas 75220, USA
| | - Lauren Rusnak
- Department of Pharmacology and Emory Chemical Biology Discovery Center, Emory University, Atlanta, Georgia 30322, USA
| | - Cau Pham
- Department of Pharmacology and Emory Chemical Biology Discovery Center, Emory University, Atlanta, Georgia 30322, USA
| | - Xiaoqian Chen
- Department of Pharmacology and Emory Chemical Biology Discovery Center, Emory University, Atlanta, Georgia 30322, USA.,Department of Pathophysiology, School of Basic Medicine, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xiulei Mo
- Department of Pharmacology and Emory Chemical Biology Discovery Center, Emory University, Atlanta, Georgia 30322, USA
| | - Brian Revennaugh
- Department of Pharmacology and Emory Chemical Biology Discovery Center, Emory University, Atlanta, Georgia 30322, USA
| | - Wei Zhou
- Department of Hematology &Medical Oncology, Emory University, Atlanta, Georgia 30322, USA.,Winship Cancer Institute, Emory University, Atlanta, Georgia 30322, USA
| | - Adam Marcus
- Department of Hematology &Medical Oncology, Emory University, Atlanta, Georgia 30322, USA.,Winship Cancer Institute, Emory University, Atlanta, Georgia 30322, USA
| | - Sahar Harati
- Department of Biomedical Informatics, Emory University School of Medicine, Atlanta, Georgia 30322, USA
| | - Xiang Chen
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Margaret A Johns
- Department of Pharmacology and Emory Chemical Biology Discovery Center, Emory University, Atlanta, Georgia 30322, USA
| | - Michael A White
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, Texas 75220, USA
| | - Carlos Moreno
- Winship Cancer Institute, Emory University, Atlanta, Georgia 30322, USA.,Department of Biomedical Informatics, Emory University School of Medicine, Atlanta, Georgia 30322, USA.,Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia 30322, USA
| | - Lee A D Cooper
- Winship Cancer Institute, Emory University, Atlanta, Georgia 30322, USA.,Department of Biomedical Informatics, Emory University School of Medicine, Atlanta, Georgia 30322, USA.,Department of Biomedical Engineering, Georgia Institute of Technology/Emory University School of Medicine, Atlanta, Georgia 30322, USA
| | - Yuhong Du
- Department of Pharmacology and Emory Chemical Biology Discovery Center, Emory University, Atlanta, Georgia 30322, USA.,Winship Cancer Institute, Emory University, Atlanta, Georgia 30322, USA
| | - Fadlo R Khuri
- Department of Hematology &Medical Oncology, Emory University, Atlanta, Georgia 30322, USA.,Winship Cancer Institute, Emory University, Atlanta, Georgia 30322, USA
| | - Haian Fu
- Department of Pharmacology and Emory Chemical Biology Discovery Center, Emory University, Atlanta, Georgia 30322, USA.,Department of Hematology &Medical Oncology, Emory University, Atlanta, Georgia 30322, USA.,Winship Cancer Institute, Emory University, Atlanta, Georgia 30322, USA
| |
Collapse
|
42
|
mTORC1 and -2 Coordinate Transcriptional and Translational Reprogramming in Resistance to DNA Damage and Replicative Stress in Breast Cancer Cells. Mol Cell Biol 2017; 37:MCB.00577-16. [PMID: 27956700 DOI: 10.1128/mcb.00577-16] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Accepted: 12/02/2016] [Indexed: 01/04/2023] Open
Abstract
mTOR coordinates growth signals with metabolic pathways and protein synthesis and is hyperactivated in many human cancers. mTOR exists in two complexes: mTORC1, which stimulates protein, lipid, and ribosome biosynthesis, and mTORC2, which regulates cytoskeleton functions. While mTOR is known to be involved in the DNA damage response, little is actually known regarding the functions of mTORC1 compared to mTORC2 in this regard or the respective impacts on transcriptional versus translational regulation. We show that mTORC1 and mTORC2 are both required to enact DNA damage repair and cell survival, resulting in increased cancer cell survival during DNA damage. Together mTORC1 and -2 enact coordinated transcription and translation of protective cell cycle and DNA replication, recombination, and repair genes. This coordinated transcriptional-translational response to DNA damage was not impaired by rapalog inhibition of mTORC1 or independent inhibition of mTORC1 or mTORC2 but was blocked by inhibition of mTORC1/2. Only mTORC1/2 inhibition reversed cancer cell resistance to DNA damage and replicative stress and increased tumor cell killing and tumor control by DNA damage therapies in animal models. When combined with DNA damage, inhibition of mTORC1/2 blocked transcriptional induction more strongly than translation of DNA replication, survival, and DNA damage response mRNAs.
Collapse
|
43
|
Vaclová T, Woods NT, Megías D, Gomez-Lopez S, Setién F, García Bueno JM, Macías JA, Barroso A, Urioste M, Esteller M, Monteiro ANA, Benítez J, Osorio A. Germline missense pathogenic variants in the BRCA1 BRCT domain, p.Gly1706Glu and p.Ala1708Glu, increase cellular sensitivity to PARP inhibitor olaparib by a dominant negative effect. Hum Mol Genet 2016; 25:5287-5299. [PMID: 27742776 DOI: 10.1093/hmg/ddw343] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Accepted: 10/03/2016] [Indexed: 12/29/2022] Open
Abstract
BRCA1-deficient cells show defects in DNA repair and rely on other members of the DNA repair machinery, which makes them sensitive to PARP inhibitors (PARPi). Although carrying a germline pathogenic variant in BRCA1/2 is the best determinant of response to PARPi, a significant percentage of the patients do not show sensitivity and/or display increased toxicity to the agent. Considering previously suggested mutation-specific BRCA1 haploinsufficiency, we aimed to investigate whether there are any differences in cellular response to PARPi olaparib depending on the BRCA1 mutation type. Lymphoblastoid cell lines derived from carriers of missense pathogenic variants in the BRCA1 BRCT domain (c.5117G > A, p.Gly1706Glu and c.5123C > A, p.Ala1708Glu) showed higher sensitivity to olaparib than cells with truncating variants or wild types (WT). Response to olaparib depended on a basal PARP enzymatic activity, but did not correlate with PARP1 expression. Interestingly, cellular sensitivity to the agent was associated with the level of BRCA1 recruitment into γH2AX foci, being the lowest in cells with missense variants. Since these variants lead to partially stable protein mutants, we propose a model in which the mutant protein acts in a dominant negative manner on the WT BRCA1, impairing the recruitment of BRCA1 into DNA damage sites and, consequently, increasing cellular sensitivity to PARPi. Taken together, our results indicate that carriers of different BRCA1 mutations could benefit from olaparib in a distinct way and show different toxicities to the agent, which could be especially relevant for a potential future use of PARPi as prophylactic agents in BRCA1 mutation carriers.
Collapse
Affiliation(s)
- Tereza Vaclová
- Human Genetics Group, Human Cancer Genetics Programme, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Nicholas T Woods
- Eppley Institute for Research in Cancer and Allied Diseases, Molecular and Biochemical Etiology Program, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Diego Megías
- Confocal Microscopy Core Unit, Biotechnology Programme, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Sergio Gomez-Lopez
- Human Genetics Group, Human Cancer Genetics Programme, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Fernando Setién
- Cancer Epigenetics Group, Cancer Epigenetics and Biology Program (PEBC), Bellvitge Biomedical Biomedical Research Institute (IDIBELL), Barcelona, Spain
| | | | - José Antonio Macías
- Hereditary Cancer Unit, Medical Oncology Service, Hospital Morales Meseguer, Murcia, Spain
| | - Alicia Barroso
- Human Genetics Group, Human Cancer Genetics Programme, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Miguel Urioste
- Familial Cancer Unit, Human Cancer Genetics Programme, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Manel Esteller
- Cancer Epigenetics Group, Cancer Epigenetics and Biology Program (PEBC), Bellvitge Biomedical Biomedical Research Institute (IDIBELL), Barcelona, Spain.,Department of Physiological Sciences II, School of Medicine, University of Barcelona, Barcelona, Spain.,Institucio Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
| | - Alvaro N A Monteiro
- Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida, FL, USA
| | - Javier Benítez
- Human Genetics Group, Human Cancer Genetics Programme, Spanish National Cancer Research Centre (CNIO), Madrid, Spain.,Spanish Network on Rare Diseases (CIBERER), Madrid, Spain.,Genotyping Unit (CEGEN), Human Cancer Genetics Programme, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Ana Osorio
- Human Genetics Group, Human Cancer Genetics Programme, Spanish National Cancer Research Centre (CNIO), Madrid, Spain.,Spanish Network on Rare Diseases (CIBERER), Madrid, Spain
| |
Collapse
|
44
|
Nogales V, Reinhold WC, Varma S, Martinez-Cardus A, Moutinho C, Moran S, Heyn H, Sebio A, Barnadas A, Pommier Y, Esteller M. Epigenetic inactivation of the putative DNA/RNA helicase SLFN11 in human cancer confers resistance to platinum drugs. Oncotarget 2016; 7:3084-97. [PMID: 26625211 PMCID: PMC4823092 DOI: 10.18632/oncotarget.6413] [Citation(s) in RCA: 119] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Accepted: 11/16/2015] [Indexed: 12/20/2022] Open
Abstract
Platinum-derived drugs such as cisplatin and carboplatin are among the most commonly used cancer chemotherapy drugs, but very few specific molecular and cellular markers predicting differential sensitivity to these agents in a given tumor type have been clearly identified. Epigenetic gene silencing is increasingly being recognized as a factor conferring distinct tumoral drug sensitivity, so we have used a comprehensive DNA methylation microarray platform to interrogate the widely characterized NCI60 panel of human cancer cell lines with respect to CpG methylation status and cisplatin/carboplatin sensitivity. Using this approach, we have found promoter CpG island hypermethylation-associated silencing of the putative DNA/RNA helicase Schlafen-11 (SLFN11) to be associated with increased resistance to platinum compounds. We have also experimentally validated these findings in vitro. In this setting, we also identified the BRCA1 interacting DHX9 RNA helicase (also known as RHA) as a protein partner for SLFN11, suggesting a mechanistic pathway for the observed chemoresistance effect. Most importantly, we have been able to extend these findings clinically, following the observation that those patients with ovarian and non-small cell lung cancer carrying SLFN11 hypermethylation had a poor response to both cisplatin and carboplatin treatments. Overall, these results identify SLFN11 epigenetic inactivation as a predictor of resistance to platinum drugs in human cancer.
Collapse
Affiliation(s)
- Vanesa Nogales
- Cancer Epigenetics and Biology Program (PEBC), Bellvitge Biomedical Research Institute (IDIBELL), Barcelona, Catalonia, Spain
| | - William C Reinhold
- Genomics and Bioinformatics Group, Developmental Therapeutics Branch, National Cancer Institute, Bethesda, MD, USA
| | - Sudhir Varma
- Developmental Therapeutics Branch and Laboratory of Molecular Pharmacology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Anna Martinez-Cardus
- Cancer Epigenetics and Biology Program (PEBC), Bellvitge Biomedical Research Institute (IDIBELL), Barcelona, Catalonia, Spain
| | - Catia Moutinho
- Cancer Epigenetics and Biology Program (PEBC), Bellvitge Biomedical Research Institute (IDIBELL), Barcelona, Catalonia, Spain
| | - Sebastian Moran
- Cancer Epigenetics and Biology Program (PEBC), Bellvitge Biomedical Research Institute (IDIBELL), Barcelona, Catalonia, Spain
| | - Holger Heyn
- Cancer Epigenetics and Biology Program (PEBC), Bellvitge Biomedical Research Institute (IDIBELL), Barcelona, Catalonia, Spain
| | - Ana Sebio
- Department of Medical Oncology, Hospital de la Santa Ceu i Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Catalonia, Spain
| | - Agusti Barnadas
- Department of Medical Oncology, Hospital de la Santa Ceu i Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Catalonia, Spain
| | - Yves Pommier
- Developmental Therapeutics Branch and Laboratory of Molecular Pharmacology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Manel Esteller
- Cancer Epigenetics and Biology Program (PEBC), Bellvitge Biomedical Research Institute (IDIBELL), Barcelona, Catalonia, Spain.,Department of Physiological Sciences II, School of Medicine, University of Barcelona, Barcelona, Catalonia, Spain.,Institucio Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Catalonia, Spain
| |
Collapse
|
45
|
Fedoseienko A, Wieringa HW, Wisman GBA, Duiker E, Reyners AKL, Hofker MH, van der Zee AGJ, van de Sluis B, van Vugt MATM. Nuclear COMMD1 Is Associated with Cisplatin Sensitivity in Ovarian Cancer. PLoS One 2016; 11:e0165385. [PMID: 27788210 PMCID: PMC5082896 DOI: 10.1371/journal.pone.0165385] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2016] [Accepted: 10/11/2016] [Indexed: 01/18/2023] Open
Abstract
Copper metabolism MURR1 domain 1 (COMMD1) protein is a multifunctional protein, and its expression has been correlated with patients’ survival in different types of cancer. In vitro studies revealed that COMMD1 plays a role in sensitizing cancer cell lines to cisplatin, however, the mechanism and its role in platinum sensitivity in cancer has yet to be established. We evaluated the role of COMMD1 in cisplatin sensitivity in A2780 ovarian cancer cells and the relation between COMMD1 expression and response to platinum-based therapy in advanced stage high-grade serous ovarian cancer (HGSOC) patients. We found that elevation of nuclear COMMD1 expression sensitized A2780 ovarian cancer cells to cisplatin-mediated cytotoxicity. This was accompanied by a more effective G2/M checkpoint, and decreased protein expression of the DNA repair gene BRCA1, and the apoptosis inhibitor BCL2. Furthermore, COMMD1 expression was immunohistochemically analyzed in two tissue micro-arrays (TMAs), representing a historical cohort and a randomized clinical trial-based cohort of advanced stage HGSOC tumor specimens. Expression of COMMD1 was observed in all ovarian cancer samples, however, specifically nuclear expression of COMMD1 was only observed in a subset of ovarian cancers. In our historical cohort, nuclear COMMD1 expression was associated with an improved response to chemotherapy (OR = 0.167; P = 0.038), although this association could not be confirmed in the second cohort, likely due to sample size. Taken together, these results suggest that nuclear expression of COMMD1 sensitize ovarian cancer to cisplatin, possibly by modulating the G2/M checkpoint and through controlling expression of genes involved in DNA repair and apoptosis.
Collapse
Affiliation(s)
- Alina Fedoseienko
- Department of Pediatrics, Molecular Genetics Section, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Hylke W. Wieringa
- Department of Medical Oncology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- Department of Gynecological Oncology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - G. Bea A. Wisman
- Department of Gynecological Oncology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Evelien Duiker
- Department of Pathology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Anna K. L. Reyners
- Department of Medical Oncology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Marten H. Hofker
- Department of Pediatrics, Molecular Genetics Section, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Ate G. J. van der Zee
- Department of Gynecological Oncology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Bart van de Sluis
- Department of Pediatrics, Molecular Genetics Section, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- * E-mail: (BvdS); (MATMvV)
| | - Marcel A. T. M. van Vugt
- Department of Medical Oncology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- * E-mail: (BvdS); (MATMvV)
| |
Collapse
|
46
|
Kuenzi BM, Borne AL, Li J, Haura EB, Eschrich SA, Koomen JM, Rix U, Stewart PA. APOSTL: An Interactive Galaxy Pipeline for Reproducible Analysis of Affinity Proteomics Data. J Proteome Res 2016; 15:4747-4754. [PMID: 27680298 DOI: 10.1021/acs.jproteome.6b00660] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
With continuously increasing scale and depth of coverage in affinity proteomics (AP-MS) data, the analysis and visualization is becoming more challenging. A number of tools have been developed to identify high-confidence interactions; however, a cohesive and intuitive pipeline for analysis and visualization is still needed. Here we present Automated Processing of SAINT Templated Layouts (APOSTL), a freely available Galaxy-integrated software suite and analysis pipeline for reproducible, interactive analysis of AP-MS data. APOSTL contains a number of tools woven together using Galaxy workflows, which are intuitive for the user to move from raw data to publication-quality figures within a single interface. APOSTL is an evolving software project with the potential to customize individual analyses with additional Galaxy tools and widgets using the R web application framework, Shiny. The source code, data, and documentation are freely available from GitHub ( https://github.com/bornea/APOSTL ) and other sources.
Collapse
Affiliation(s)
- Brent M Kuenzi
- Department of Drug Discovery, H. Lee Moffitt Cancer Center & Research Institute , Tampa, Florida 33612-9497, United States.,Cancer Biology Ph.D. Program, University of South Florida , Tampa, Florida 33620, United States
| | - Adam L Borne
- Department of Thoracic Oncology, H. Lee Moffitt Cancer Center & Research Institute , Tampa, Florida 33612-9497, United States
| | - Jiannong Li
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center & Research Institute , Tampa, Florida 33612-9497, United States
| | - Eric B Haura
- Department of Thoracic Oncology, H. Lee Moffitt Cancer Center & Research Institute , Tampa, Florida 33612-9497, United States
| | - Steven A Eschrich
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center & Research Institute , Tampa, Florida 33612-9497, United States
| | - John M Koomen
- Molecular Oncology, H. Lee Moffitt Cancer Center & Research Institute , Tampa, Florida 33612-9497, United States
| | - Uwe Rix
- Department of Drug Discovery, H. Lee Moffitt Cancer Center & Research Institute , Tampa, Florida 33612-9497, United States
| | - Paul A Stewart
- Department of Thoracic Oncology, H. Lee Moffitt Cancer Center & Research Institute , Tampa, Florida 33612-9497, United States
| |
Collapse
|
47
|
Sakai D, Trainor PA. Face off against ROS: Tcof1/Treacle safeguards neuroepithelial cells and progenitor neural crest cells from oxidative stress during craniofacial development. Dev Growth Differ 2016; 58:577-85. [PMID: 27481486 DOI: 10.1111/dgd.12305] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2016] [Revised: 06/09/2016] [Accepted: 06/12/2016] [Indexed: 01/10/2023]
Abstract
One-third of all congenital birth defects affect the head and face, and most craniofacial anomalies are considered to arise through defects in the development of cranial neural crest cells. Cranial neural crest cells give rise to the majority of craniofacial bones, cartilages and connective tissues. Therefore, understanding the events that control normal cranial neural crest and subsequent craniofacial development is important for elucidating the pathogenetic mechanisms of craniofacial anomalies and for the exploring potential therapeutic avenues for their prevention. Treacher Collins syndrome (TCS) is a congenital disorder characterized by severe craniofacial anomalies. An animal model of TCS, generated through mutation of Tcof1, the mouse (Mus musculus) homologue of the gene primarily mutated in association with TCS in humans, has recently revealed significant insights into the pathogenesis of TCS. Apoptotic elimination of neuroepithelial cells including neural crest cells is the primary cause of craniofacial defects in Tcof1 mutant embryos. However, our understanding of the mechanisms that induce tissue-specific apoptosis remains incomplete. In this review, we describe recent advances in our understanding of the pathogenesis TCS. Furthermore, we discuss the role of Tcof1 in normal embryonic development, the correlation between genetic and environmental factors on the severity of craniofacial abnormalities, and the prospect for prenatal prevention of craniofacial anomalies.
Collapse
Affiliation(s)
- Daisuke Sakai
- Doshisha University, Graduate School of Brain Science, HC301 1-3 Tataramiyakodani, Kyotanabe, Kyoto, 610-0394, Japan.
| | - Paul A Trainor
- Stowers Institute for Medical Research, Kansas City, Missouri, 64110, USA.,Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, Kansas, 66160, USA
| |
Collapse
|
48
|
Abstract
NOP14, which is functionally conserved among eukaryotes, has been implicated in cancer development. Here, we show that NOP14 is poorly expressed in breast cancer cells and invasive breast cancer tissues. In vivo and in vitro studies indicated that NOP14 suppressed the tumorigenesis and metastasis of breast cancer cells. Further investigations revealed that NOP14 enhanced ERα expression and inhibited the Wnt/β-catenin pathway by up-regulating NRIP1 expression. Survival analysis indicated that low NOP14 expression was significantly associated with poor overall survival (P = 0.0006) and disease-free survival (P = 0.0007), suggesting that NOP14 is a potential prognostic factor in breast cancer. Taken together, our findings reveal that NOP14 may suppress breast cancer progression and provide new insights into the development of targeted therapeutic agents for breast cancer.
Collapse
|
49
|
Hang L, Zhao X. The Rtt107 BRCT scaffold and its partner modification enzymes collaborate to promote replication. Nucleus 2016; 7:346-51. [PMID: 27385431 DOI: 10.1080/19491034.2016.1201624] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
Abstract
Faithful duplication of the entire genome during each cell cycle is key for genome maintenance. Each stage of DNA replication, including initiation, progression, and termination, is tightly regulated. Some of these regulations enable replisomes to overcome tens of thousands of template obstacles that block DNA synthesis. Previous studies have identified a large number of proteins that are dedicated to this mission, including protein modification enzymes and scaffold proteins. Protein modification enzymes can bestow fast and reversible changes on many substrates, and thus are ideal for coordinating multiple events needed to promptly overcome replication impediments. Scaffold proteins can support specific protein-protein interactions that enable protein complex formation, protein recruitment, and partner enzyme functions. Taken together with previous studies, our recent work elucidates that a group of modification and scaffold proteins form several complexes to aid replication progression and are particularly important for synthesizing large replicons. Additionally, our work reveals that the intrinsic plasticity of the replication initiation program can be used to compensate for deficient replication progression. (1).
Collapse
Affiliation(s)
- Lisa Hang
- a Molecular Biology Program, Memorial Sloan-Kettering Cancer Center , New York , NY , USA
| | - Xiaolan Zhao
- a Molecular Biology Program, Memorial Sloan-Kettering Cancer Center , New York , NY , USA
| |
Collapse
|
50
|
Jhuraney A, Woods NT, Wright G, Rix L, Kinose F, Kroeger JL, Remily-Wood E, Cress WD, Koomen JM, Brantley SG, Gray JE, Haura EB, Rix U, Monteiro AN. PAXIP1 Potentiates the Combination of WEE1 Inhibitor AZD1775 and Platinum Agents in Lung Cancer. Mol Cancer Ther 2016; 15:1669-81. [PMID: 27196765 PMCID: PMC4936941 DOI: 10.1158/1535-7163.mct-15-0182] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2015] [Accepted: 04/09/2016] [Indexed: 11/16/2022]
Abstract
The DNA damage response (DDR) involves a complex network of signaling events mediated by modular protein domains such as the BRCA1 C-terminal (BRCT) domain. Thus, proteins that interact with BRCT domains and are a part of the DDR constitute potential targets for sensitization to DNA-damaging chemotherapy agents. We performed a pharmacologic screen to evaluate 17 kinases, identified in a BRCT-mediated interaction network as targets to enhance platinum-based chemotherapy in lung cancer. Inhibition of mitotic kinase WEE1 was found to have the most effective response in combination with platinum compounds in lung cancer cell lines. In the BRCT-mediated interaction network, WEE1 was found in complex with PAXIP1, a protein containing six BRCT domains involved in transcription and in the cellular response to DNA damage. We show that PAXIP1 BRCT domains regulate WEE1-mediated phosphorylation of CDK1. Furthermore, ectopic expression of PAXIP1 promotes enhanced caspase-3-mediated apoptosis in cells treated with WEE1 inhibitor AZD1775 (formerly, MK-1775) and cisplatin compared with cells treated with AZD1775 alone. Cell lines and patient-derived xenograft models expressing both PAXIP1 and WEE1 exhibited synergistic effects of AZD1775 and cisplatin. In summary, PAXIP1 is involved in sensitizing lung cancer cells to the WEE1 inhibitor AZD1775 in combination with platinum-based treatment. We propose that WEE1 and PAXIP1 levels may be used as mechanism-based biomarkers of response when WEE1 inhibitor AZD1775 is combined with DNA-damaging agents. Mol Cancer Ther; 15(7); 1669-81. ©2016 AACR.
Collapse
Affiliation(s)
- Ankita Jhuraney
- Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida. Cancer Biology PhD Program, University of South Florida, Tampa, Florida
| | - Nicholas T Woods
- Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
| | - Gabriela Wright
- Department of Drug Discovery, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
| | - Lily Rix
- Department of Drug Discovery, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
| | - Fumi Kinose
- Department of Thoracic Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
| | - Jodi L Kroeger
- Flow Cytometry Core, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
| | - Elizabeth Remily-Wood
- Molecular Oncology Program, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
| | - W Douglas Cress
- Molecular Oncology Program, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
| | - John M Koomen
- Molecular Oncology Program, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
| | - Stephen G Brantley
- M2Gen, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
| | - Jhanelle E Gray
- Department of Thoracic Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
| | - Eric B Haura
- Department of Thoracic Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
| | - Uwe Rix
- Department of Drug Discovery, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida.
| | - Alvaro N Monteiro
- Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida.
| |
Collapse
|