1
|
Howe J, Barbar EJ. Dynamic interactions of dimeric hub proteins underlie their diverse functions and structures: A comparative analysis of 14-3-3 and LC8. J Biol Chem 2025; 301:108416. [PMID: 40107617 PMCID: PMC12017986 DOI: 10.1016/j.jbc.2025.108416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 02/06/2025] [Accepted: 02/08/2025] [Indexed: 03/22/2025] Open
Abstract
Hub proteins interact with a host of client proteins and regulate multiple cellular functions. Dynamic hubs have a single binding interface for one client at a time resulting in competition among clients with the highest affinity. Dynamic dimeric hubs with two identical sites bind either two different client proteins or two chains of the same client to form homogenous complexes and could also form heterogeneous mixtures of interconverting complexes. Here, we review the interactions of the dimeric hubs 14-3-3 and LC8. 14-3-3 is a phosphoserine/threonine binding protein involved in structuring client proteins and regulating their phosphorylation. LC8 is involved in promoting the dimerization of client peptides and the rigidification of their disordered regions. Both 14-3-3 and LC8 are essential genes, with 14-3-3 playing a crucial role in apoptosis and cell cycle regulation, while LC8 is critical for the assembly of proteins involved in transport, DNA repair, and transcription. Interestingly, both protein dimers can dissociate by phosphorylation, which results in their interactome-wide changes. Their interactions are also regulated by the phosphorylation of their clients. Both form heterogeneous complexes with various functions including phase separation, signaling, and viral hijacking where they restrict the conformational heterogeneity of their dimeric clients that bind nucleic acids. This comparative analysis highlights the importance of dynamic protein-protein interactions in the diversity of functions of 14-3-3 and LC8 and how small differences in structures of interfaces explain why 14-3-3 is primarily involved in the regulation of phosphorylation states while LC8 is primarily involved in the regulation of assembly of large dynamic complexes.
Collapse
Affiliation(s)
- Jesse Howe
- Oregon State University, Department of Biochemistry and Biophysics, Corvallis, Oregon, USA
| | - Elisar J Barbar
- Oregon State University, Department of Biochemistry and Biophysics, Corvallis, Oregon, USA.
| |
Collapse
|
2
|
Katz LS, Visser EJ, Plitzko KF, Pennings MAM, Cossar PJ, Tse IL, Kaiser M, Brunsveld L, Ottmann C, Scott DK. Molecular glues of the regulatory ChREBP/14-3-3 complex protect beta cells from glucolipotoxicity. Nat Commun 2025; 16:2110. [PMID: 40025013 PMCID: PMC11873037 DOI: 10.1038/s41467-025-57241-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Accepted: 02/17/2025] [Indexed: 03/04/2025] Open
Abstract
The Carbohydrate Response Element Binding Protein (ChREBP) is a glucose-responsive transcription factor (TF) with two major splice isoforms (α and β). In chronic hyperglycemia and glucolipotoxicity, ChREBPα-mediated ChREBPβ expression surges, leading to insulin-secreting β-cell dedifferentiation and death. 14-3-3 binding to ChREBPα results in cytoplasmic retention and suppression of transcriptional activity. Thus, small molecule-mediated stabilization of this protein-protein interaction (PPI) may be of therapeutic value. Here, we show that structure-based optimizations of a 'molecular glue' compound led to potent ChREBPα/14-3-3 PPI stabilizers with cellular activity. In primary human β-cells, the most active compound retained ChREBPα in the cytoplasm, and efficiently protected β-cells from glucolipotoxicity while maintaining β-cell identity. This study may thus not only provide the basis for the development of a unique class of compounds for the treatment of Type 2 Diabetes but also showcases an alternative 'molecular glue' approach for achieving small molecule control of notoriously difficult to target TFs.
Collapse
Affiliation(s)
- Liora S Katz
- Diabetes, Obesity and Metabolism Institute and Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Emira J Visser
- Laboratory of Chemical Biology, Department of Biomedical Engineering and Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - Kathrin F Plitzko
- Chemical Biology, Center of Medical Biotechnology, Faculty of Biology, University of Duisburg-Essen, Essen, Germany
| | - Marloes A M Pennings
- Laboratory of Chemical Biology, Department of Biomedical Engineering and Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - Peter J Cossar
- Laboratory of Chemical Biology, Department of Biomedical Engineering and Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - Isabelle L Tse
- Diabetes, Obesity and Metabolism Institute and Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Markus Kaiser
- Chemical Biology, Center of Medical Biotechnology, Faculty of Biology, University of Duisburg-Essen, Essen, Germany.
| | - Luc Brunsveld
- Laboratory of Chemical Biology, Department of Biomedical Engineering and Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, The Netherlands.
| | - Christian Ottmann
- Laboratory of Chemical Biology, Department of Biomedical Engineering and Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, The Netherlands.
| | - Donald K Scott
- Diabetes, Obesity and Metabolism Institute and Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
3
|
Shimamura MI, Satoh K. Challenges and Revisions in Diagnostic Criteria: Advancing Early Detection of Prion Diseases. Int J Mol Sci 2025; 26:2037. [PMID: 40076658 PMCID: PMC11900056 DOI: 10.3390/ijms26052037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Revised: 02/21/2025] [Accepted: 02/25/2025] [Indexed: 03/14/2025] Open
Abstract
Prion diseases are fatal neurological disorders characterized by abnormal protein accumulation in the brain, leading to neurodegeneration, dementia, and ataxia. Sporadic Creutzfeldt-Jakob disease (sCJD), the most common form, accounts for 80-90% of cases and progresses rapidly, with most patients surviving <6 months to a year after symptom onset, indicating the importance of early diagnosis. The disease is classified into six subtypes based on PRNP gene polymorphisms, with differences in protein degradation patterns contributing to the diversity of clinical symptoms. However, diagnosis remains challenging because of the variability in clinical presentation and disease duration. Traditional diagnostic criteria established by the World Health Organization (WHO) rely on clinical findings, electroencephalogram, and cerebrospinal fluid tests, such as the 14-3-3 protein assay. However, these criteria require pathological confirmation, often delaying diagnosis. The recently proposed Hermann's criteria represent a significant advancement by incorporating newer biomarkers, including magnetic resonance imaging, real-time quaking-induced conversion assay, tau protein, and neurofilament light chain. These criteria improve diagnostic sensitivity and specificity but have a slightly higher risk of false positives. This review compares the effectiveness of these biomarkers with the WHO criteria and highlights the importance of early diagnosis for improving patient care.
Collapse
Affiliation(s)
- Mika Inada Shimamura
- Biomedical Research Support Center, Nagasaki University, 1-12-4 Sakamoto, Nagasaki 852-8523, Japan;
| | - Katsuya Satoh
- Unit of Medical and Dental Sciences, Department of Health Sciences, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8501, Japan
- Leading Medical Research Core Unit, Department of Brain Research Unit, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8523, Japan
| |
Collapse
|
4
|
Tian Y, Li L, Wu L, Xu Q, Li Y, Pan H, Bing T, Bai X, Finko AV, Li Z, Bian J. Recent Developments in 14-3-3 Stabilizers for Regulating Protein-Protein Interactions: An Update. J Med Chem 2025; 68:2124-2146. [PMID: 39902774 DOI: 10.1021/acs.jmedchem.4c01936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2025]
Abstract
14-3-3 proteins play a crucial role in the regulation of protein-protein interactions, impacting various cellular processes and disease mechanisms. Recent advancements have led to the development of stabilizers that enhance the binding of 14-3-3 proteins to clients, presenting promising therapeutic potentials. This perspective provides an updated overview of the latest developments in the field of 14-3-3 stabilizers, with a focus on their design, synthesis, and biological evaluation. We discuss the structural basis for the interaction between 14-3-3 proteins and their ligands, highlighting key modifications that enhance binding affinity and selectivity. Additionally, we explore the therapeutic applications of 14-3-3 stabilizers across major therapeutic areas such as cancer, metabolic disorders, and neurodegenerative diseases. By summarizing recent research findings and technological advancements, this perspective aims to shed light on the current state of 14-3-3 stabilizer developments and outline future directions for optimizing these compounds as effective therapeutic agents.
Collapse
Affiliation(s)
- Yucheng Tian
- Jiangsu Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Longjing Li
- Jiangsu Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Liuyi Wu
- Jiangsu Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Qianqian Xu
- Jiangsu Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Yaojie Li
- Jiangsu Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Huawei Pan
- ICE Bioscience, Bldg 15, Yd 18, Kechuang 13th St, Etown, Tongzhou Dist, Beijing 100176, China
| | - Tiejun Bing
- ICE Bioscience, Bldg 15, Yd 18, Kechuang 13th St, Etown, Tongzhou Dist, Beijing 100176, China
| | - Xiumei Bai
- Department of Chemistry, Lomonosov Moscow State University (MSU), Moscow 119991, Russia
| | - Alexander V Finko
- Department of Chemistry, Lomonosov Moscow State University (MSU), Moscow 119991, Russia
| | - Zhiyu Li
- Jiangsu Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Jinlei Bian
- Jiangsu Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| |
Collapse
|
5
|
Zhang L, Gao M, Wu Y, Liu H, Zhuang X, Zhou Y, Song Q, Bi S, Zhang W, Cui Y. MST1 interactomes profiling across cell death in esophageal squamous cell carcinoma. MEDICAL REVIEW (2021) 2024; 4:531-543. [PMID: 39664081 PMCID: PMC11629308 DOI: 10.1515/mr-2024-0031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Accepted: 05/27/2024] [Indexed: 12/13/2024]
Abstract
Objectives Resistance to apoptosis in esophageal squamous cell carcinoma (ESCC) constitutes a significant impediment to treatment efficacy. Exploring alternative cell death pathways and their regulatory factors beyond apoptosis is crucial for overcoming drug resistance and enhancing therapeutic outcomes in ESCC. Methods Mammalian Ste 20-like kinase 1 (MST1) is implicated in regulating various cell deaths, including apoptosis, autophagy, and pyroptosis. Employing enhanced ascorbate peroxidase 2 (APEX2) proximity labeling coupled with immunoprecipitation-mass spectrometry (IP-MS), we elucidated the interactomes of MST1 across these three cell death paradigms. Results Proteomic profiling unveiled the functional roles and subcellular localization of MST1 and its interacting proteins during normal proliferation and various cell death processes. Notably, MST1 exhibited an expanded interactome during cell death compared to normal proliferation and chromosome remodeling functions consistently. In apoptosis, there was a notable increase of mitosis-associated proteins such as INCENP, ANLN, KIF23, SHCBP1 and SUPT16H, which interacted with MST1, alongside decreased expression of the pre-apoptotic protein STK3. During autophagy, the bindings of DNA repair-related proteins CBX8 and m6A reader YTHDC1 to MST1 were enhanced. In pyroptosis, LRRFIP2 and FLII which can inhibit pyroptosis increasingly binding to MST1. Conclusions Our findings delineate potential mechanisms through which MST1 and its interactomes regulate cell death, paving the way for further investigation to validate and consolidate these observations.
Collapse
Affiliation(s)
- Li Zhang
- Cancer Institute, Shenzhen-Peking University-the Hong Kong University of Science and Technology Medical Center, Shenzhen, Guangdong, China
| | - Mingwei Gao
- Department of Oncology, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
| | - Yueguang Wu
- Cancer Institute, Shenzhen-Peking University-the Hong Kong University of Science and Technology Medical Center, Shenzhen, Guangdong, China
| | - Huijuan Liu
- Institute of Cancer Research, Shenzhen Bay Laboratory, Shenzhen, Guangdong, China
| | - Xuehan Zhuang
- Cancer Institute, Shenzhen-Peking University-the Hong Kong University of Science and Technology Medical Center, Shenzhen, Guangdong, China
| | - Yan Zhou
- Institute of Cancer Research, Shenzhen Bay Laboratory, Shenzhen, Guangdong, China
| | - Qiqin Song
- Cancer Institute, Shenzhen-Peking University-the Hong Kong University of Science and Technology Medical Center, Shenzhen, Guangdong, China
| | - Shanshan Bi
- Cancer Institute, Shenzhen-Peking University-the Hong Kong University of Science and Technology Medical Center, Shenzhen, Guangdong, China
| | - Weimin Zhang
- Cancer Institute, Shenzhen-Peking University-the Hong Kong University of Science and Technology Medical Center, Shenzhen, Guangdong, China
| | - Yongping Cui
- Cancer Institute, Shenzhen-Peking University-the Hong Kong University of Science and Technology Medical Center, Shenzhen, Guangdong, China
| |
Collapse
|
6
|
Tilwani S, Gandhi K, Dalal SN. 14-3-3ε conditional knockout mice exhibit defects in the development of the epidermis. FEBS Lett 2024; 598:3005-3020. [PMID: 39511902 DOI: 10.1002/1873-3468.15051] [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: 02/27/2024] [Revised: 10/15/2024] [Accepted: 10/17/2024] [Indexed: 11/15/2024]
Abstract
The epidermis is a stratified epithelium that functions as the first line of defense against pathogenic invasion and acts as a barrier preventing water loss. In this study, we aimed to decipher the role of 14-3-3ε in the development of the epidermis. We report that loss of 14-3-3ε in the epidermis of juvenile and adult mice reduces cell division in the basal layer and increases the percentage of cells with multiple centrosomes, leading to a reduction in the thickness of the basal and stratified layers. We also demonstrate a decrease in the expression of differentiation markers, although no gross morphological defects in the skin or adverse effects on the survival of the mice were observed. These results suggest that loss of 14-3-3ε in the epidermis may lead to defects in proliferation and differentiation.
Collapse
Affiliation(s)
- Sarika Tilwani
- Cell and Tumor Biology, Advanced Centre for Treatment Research and Education in Cancer (ACTREC), Tata Memorial Centre, Navi Mumbai, India
- Homi Bhabha National Institute, Mumbai, India
| | - Karan Gandhi
- Cell and Tumor Biology, Advanced Centre for Treatment Research and Education in Cancer (ACTREC), Tata Memorial Centre, Navi Mumbai, India
| | - Sorab N Dalal
- Cell and Tumor Biology, Advanced Centre for Treatment Research and Education in Cancer (ACTREC), Tata Memorial Centre, Navi Mumbai, India
- Homi Bhabha National Institute, Mumbai, India
| |
Collapse
|
7
|
Yan J, Yang A, Tu S. The relationship between keratin 18 and epithelial-derived tumors: as a diagnostic marker, prognostic marker, and its role in tumorigenesis. Front Oncol 2024; 14:1445978. [PMID: 39502314 PMCID: PMC11534658 DOI: 10.3389/fonc.2024.1445978] [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: 06/08/2024] [Accepted: 09/25/2024] [Indexed: 11/08/2024] Open
Abstract
As a structural protein, keratin is mainly expressed in epithelial cells and skin appendages to provide mechanical support and external resistance. The keratin family has a total of 54 members, which are divided into type I and type II. Two types of keratins connect to each other to form keratin intermediate filaments and participate in the construction of the cytoskeleton. K18 is a non-hair keratin, which is widely expressed in simple epithelial tissues with its partner, K8. Compared with mechanical support, K8/K18 pairs play more important roles in biological regulation, such as mediating anti-apoptosis, regulating cell cycle progression, and transmitting signals. Mutations in K18 can cause a variety of non-neoplastic diseases of the visceral epithelium. In addition, the expression levels of K18 are frequently altered in various epithelial-derived tumors, especially adenocarcinomas, which suggests that K18 may be involved in tumorigenesis. Due to the specific expression pattern of K18 in tumor tissues and its serum level reflecting tumor cell death, apply K18 to diagnose tumors and predict its prognosis have the potential to be simple and effective alternative methods. However, these potential roles of K18 in tumors have not been fully summarized. In this review, we focus on the relationship between K18 and epithelial-derived tumors, discuss the value of K18 as a diagnostic and prognostic marker, and summarize the interactions of K18 with various related proteins in tumorigenesis, with examples of simple epithelial tumors such as lung, breast, liver, and gastrointestinal cancers.
Collapse
Affiliation(s)
- Jiazhi Yan
- Queen Mary School, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Aiwei Yang
- Queen Mary School, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Shuo Tu
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| |
Collapse
|
8
|
Han Z, Wang R, Chi P, Zhang Z, Min L, Jiao H, Ou G, Zhou D, Qin D, Xu C, Gao Z, Qi Q, Li J, Lu Y, Wang X, Chen J, Yu X, Hu H, Li L, Deng D. The subcortical maternal complex modulates the cell cycle during early mammalian embryogenesis via 14-3-3. Nat Commun 2024; 15:8887. [PMID: 39406751 PMCID: PMC11480350 DOI: 10.1038/s41467-024-53277-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2024] [Accepted: 10/07/2024] [Indexed: 10/19/2024] Open
Abstract
The subcortical maternal complex (SCMC) is essential for safeguarding female fertility in mammals. Assembled in oocytes, the SCMC maintains the cleavage of early embryos, but the underlying mechanism remains unclear. Here, we report that 14-3-3, a multifunctional protein, is a component of the SCMC. By resolving the structure of the 14-3-3-containing SCMC, we discover that phosphorylation of TLE6 contributes to the recruitment of 14-3-3. Mechanistically, during maternal-to-embryo transition, the SCMC stabilizes 14-3-3 protein and contributes to the proper control of CDC25B, thus ensuring the activation of the maturation-promoting factor and mitotic entry in mouse zygotes. Notably, the SCMC establishes a conserved molecular link with 14-3-3 and CDC25B in human oocytes/embryos. This study discloses the molecular mechanism through which the SCMC regulates the cell cycle in early embryos and elucidates the function of the SCMC in mammalian early embryogenesis.
Collapse
Affiliation(s)
- Zhuo Han
- Key Laboratory of Birth Defects and Related Disease of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Rui Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, Key Laboratory of Organ Regeneration and Reconstruction, UCAS/IOZ/CAS, Beijing, China
- Beijing Institute of Stem Cell and Regenerative Medicine, Beijing, China
- Department of Reproductive Medicine, the First People's Hospital of Yunnan Province, Kunming, China
| | - Pengliang Chi
- Key Laboratory of Birth Defects and Related Disease of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Zihan Zhang
- Key Laboratory of Birth Defects and Related Disease of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Ling Min
- Key Laboratory of Birth Defects and Related Disease of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Haizhan Jiao
- Kobilka Institute of Innovative Drug Discovery, School of Medicine, The Chinese University of Hong Kong (Shenzhen), Shenzhen, China
| | - Guojin Ou
- Key Laboratory of Birth Defects and Related Disease of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, China
- Clinical laboratory, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Dan Zhou
- State Key Laboratory of Stem Cell and Reproductive Biology, Key Laboratory of Organ Regeneration and Reconstruction, UCAS/IOZ/CAS, Beijing, China
- Beijing Institute of Stem Cell and Regenerative Medicine, Beijing, China
| | - Dandan Qin
- State Key Laboratory of Stem Cell and Reproductive Biology, Key Laboratory of Organ Regeneration and Reconstruction, UCAS/IOZ/CAS, Beijing, China
- Beijing Institute of Stem Cell and Regenerative Medicine, Beijing, China
| | - Chengpeng Xu
- State Key Laboratory of Stem Cell and Reproductive Biology, Key Laboratory of Organ Regeneration and Reconstruction, UCAS/IOZ/CAS, Beijing, China
- Beijing Institute of Stem Cell and Regenerative Medicine, Beijing, China
| | - Zheng Gao
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Qianqian Qi
- Clinical laboratory, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Jialu Li
- Key Laboratory of Birth Defects and Related Disease of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Yuechao Lu
- Key Laboratory of Birth Defects and Related Disease of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, China
- Department of Reproductive Medicine, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Xiang Wang
- Key Laboratory of Birth Defects and Related Disease of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, China
- NHC key Laboratory of Chronobiology, Sichuan University, Chengdu, China
- Development and Related Diseases of Women and Children Key Laboratory of Sichuan Province, Sichuan University, Chengdu, China
| | - Jing Chen
- Laboratory of Pediatric Surgery, Department of Pediatric Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Xingjiang Yu
- State Key Laboratory of Stem Cell and Reproductive Biology, Key Laboratory of Organ Regeneration and Reconstruction, UCAS/IOZ/CAS, Beijing, China
- Beijing Institute of Stem Cell and Regenerative Medicine, Beijing, China
| | - Hongli Hu
- Kobilka Institute of Innovative Drug Discovery, School of Medicine, The Chinese University of Hong Kong (Shenzhen), Shenzhen, China
| | - Lei Li
- State Key Laboratory of Stem Cell and Reproductive Biology, Key Laboratory of Organ Regeneration and Reconstruction, UCAS/IOZ/CAS, Beijing, China.
- Beijing Institute of Stem Cell and Regenerative Medicine, Beijing, China.
| | - Dong Deng
- Key Laboratory of Birth Defects and Related Disease of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, China.
- NHC key Laboratory of Chronobiology, Sichuan University, Chengdu, China.
- Development and Related Diseases of Women and Children Key Laboratory of Sichuan Province, Sichuan University, Chengdu, China.
| |
Collapse
|
9
|
Wang SH, Hsieh YY, Ong KH, Lai HY, Tsai HH, Sun DP, Huang SKH, Tian YF, Wu HC, Chan TC, Joseph K, Chang IW. The clinicopathological significance and prognostic impact of 14-3-3σ/stratifin expression on patients with surgically resectable intrahepatic cholangiocarcinoma. Asian J Surg 2024:S1015-9584(24)01873-6. [PMID: 39232956 DOI: 10.1016/j.asjsur.2024.08.133] [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: 05/01/2024] [Accepted: 08/20/2024] [Indexed: 09/06/2024] Open
Abstract
INTRODUCTION Intrahepatic cholangiocarcinoma (iCCA) is the second most common primary liver cancer after hepatocellular carcinoma. Through data mining of publicly available iCCA transcriptomic datasets from the Gene Expression Omnibus, we identified SFN as the most significantly up-regulated gene in iCCA compared to normal tissue, focusing on the Gene Ontology term "cell proliferation" (GO:0008283). SFN encodes the 14-3-3σ protein, also known as stratifin, which plays crucial roles in various cellular processes. MATERIALS AND METHODS Immunohistochemistry was used to assess stratifin expression in 182 patients with localized iCCAs undergoing surgical resection. Patients were divided into low and high expression groups, and the association between stratifin expression and clinicopathological features was analyzed. Univariate and multivariate survival analyses were performed to assess overall survival (OS), disease-specific survival (DSS), local recurrence-free survival (LRFS), and metastasis-free survival (MeFS). RESULTS Elevated stratifin expression in iCCAs was significantly associated with the absence of hepatitis, positive surgical margins, advanced primary tumor stages, and higher histological grades (all p ≤ 0.011). Survival analyses demonstrated a significant negative association between stratifin expression and all prognostic indicators, including OS, DSS, LRFS, and MeFS (all p ≤ 0.0004). Multivariate analysis revealed that stratifin overexpression was significantly correlated with poorer outcomes in terms of DSS, LRFS, and MeFS (all p < 0.001). CONCLUSIONS These findings suggest that stratifin may play a crucial role in iCCA oncogenesis and tumor progression, serving as a potential novel prognostic biomarker.
Collapse
Affiliation(s)
- Su-Hong Wang
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Chi Mei Medical Center, Tainan, Taiwan
| | - Yao-Yu Hsieh
- Division of Hematology and Oncology, Shuang Ho Hospital, Taipei Medical University, Taipei, Taiwan; Division of Hematology and Oncology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Khaa Hoo Ong
- Department of Surgery, Division of Gastroenterology and General Surgery, Chi Mei Medical Center, Tainan, Taiwan; Department of Medical Technology, Chung Hwa University of Medical Technology, Tainan, Taiwan; Institute of Biomedical Sciences, National Sun Yat-sen University, Kaohsiung, Taiwan
| | - Hong-Yue Lai
- Department of Pharmacology, School of Medicine, College of Medicine, China Medical University, Taichung, Taiwan
| | - Hsin-Hwa Tsai
- Department of Laboratory Medicine, China Medical University Hospital, Taichung, Taiwan
| | - Ding-Ping Sun
- Department of Surgery, Division of Gastroenterology and General Surgery, Chi Mei Medical Center, Tainan, Taiwan
| | - Steven Kuan-Hua Huang
- Department of Surgery, Division of Urology, Chi Mei Medical Center, Tainan, Taiwan; Department of Medical Science Industries, College of Health Sciences, Chang Jung Christian University, Tainan, Taiwan
| | - Yu-Feng Tian
- Department of Surgery, Division of Colon and Rectal Surgery, Chi Mei Medical Center, Tainan, Taiwan
| | - Hung-Chang Wu
- Department of Internal Medicine, Division of Hematology and Oncology, Chi Mei Medical Center, Tainan, Taiwan; College of Pharmacy and Science, Chia Nan University, Tainan, Taiwan
| | - Ti-Chun Chan
- Department of Medical Research, Chi Mei Medical Center, Tainan, Taiwan; National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan
| | | | - I-Wei Chang
- Department of Pathology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; Department of Clinical Pathology, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan; Department of Pathology, Taipei Medical University Hospital, Taipei, Taiwan; Department of Pathology, Shuang Ho Hospital, Taipei Medical University, Taipei, Taiwan.
| |
Collapse
|
10
|
McMenemy CM, Guo D, Quinn JA, Greenhalgh DA. 14-3-3σ/Stratifin and p21 limit AKT-related malignant progression in skin carcinogenesis following MDM2-associated p53 loss. Mol Carcinog 2024; 63:1768-1782. [PMID: 38869281 DOI: 10.1002/mc.23771] [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: 03/05/2024] [Revised: 05/20/2024] [Accepted: 05/28/2024] [Indexed: 06/14/2024]
Abstract
To study mechanisms driving/inhibiting skin carcinogenesis, stage-specific expression of 14-3-3σ (Stratifin) was analyzed in skin carcinogenesis driven by activated rasHa/fos expression (HK1.ras/fos) and ablation of PTEN-mediated AKT regulation (K14.creP/Δ5PTENflx/flx). Consistent with 14-3-3σ roles in epidermal differentiation, HK1.ras hyperplasia and papillomas displayed elevated 14-3-3σ expression in supra-basal keratinocytes, paralleled by supra-basal p-MDM2166 activation and sporadic p-AKT473 expression. In bi-genic HK1.fos/Δ5PTENflx/flx hyperplasia, basal-layer 14-3-3σ expression appeared, and alongside p53/p21, was associated with keratinocyte differentiation and keratoacanthoma etiology. Tri-genic HK1.ras/fos-Δ5PTENflx/flx hyperplasia/papillomas initially displayed increased basal-layer 14-3-3σ, suggesting attempts to maintain supra-basal p-MDM2166 and protect basal-layer p53. However, HK1.ras/fos-Δ5PTENflx/flx papillomas exhibited increasing basal-layer p-MDM2166 activation that reduced p53, which coincided with malignant conversion. Despite p53 loss, 14-3-3σ expression persisted in well-differentiated squamous cell carcinomas (wdSCCs) and alongside elevated p21, limited malignant progression via inhibiting p-AKT1473 expression; until 14-3-3σ/p21 loss facilitated progression to aggressive SCC exhibiting uniform p-AKT1473. Analysis of TPA-promoted HK1.ras-Δ5PTENflx/flx mouse skin, demonstrated early loss of 14-3-3σ/p53/p21 in hyperplasia and papillomas, with increased p-MDM2166/p-AKT1473 that resulted in rapid malignant conversion and progression to poorly differentiated SCC. In 2D/3D cultures, membranous 14-3-3σ expression observed in normal HaCaT and SP1ras61 papilloma keratinocytes was unexpectedly detected in malignant T52ras61/v-fos SCC cells cultured in monolayers, but not invasive 3D-cells. Collectively, these data suggest 14-3-3σ/Stratifin exerts suppressive roles in papillomatogenesis via MDM2/p53-dependent mechanisms; while persistent p53-independent expression in early wdSCC may involve p21-mediated AKT1 inhibition to limit malignant progression.
Collapse
Affiliation(s)
- Carol M McMenemy
- Section of Dermatology and Molecular Carcinogenesis, School of Medicine, Dentistry and Nursing, College of Medical, Veterinary and Life Sciences, Glasgow University, Glasgow, Scotland
| | - Dajiang Guo
- Section of Dermatology and Molecular Carcinogenesis, School of Medicine, Dentistry and Nursing, College of Medical, Veterinary and Life Sciences, Glasgow University, Glasgow, Scotland
| | - Jean A Quinn
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, Scotland
| | - David A Greenhalgh
- Section of Dermatology and Molecular Carcinogenesis, School of Medicine, Dentistry and Nursing, College of Medical, Veterinary and Life Sciences, Glasgow University, Glasgow, Scotland
| |
Collapse
|
11
|
Zhang T, Zhu Y, Wang X, Chong D, Wang H, Bu D, Zhao M, Fang L, Li C. The characterization of protein lactylation in relation to cardiac metabolic reprogramming in neonatal mouse hearts. J Genet Genomics 2024; 51:735-748. [PMID: 38479452 DOI: 10.1016/j.jgg.2024.02.009] [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: 01/06/2024] [Revised: 02/18/2024] [Accepted: 02/28/2024] [Indexed: 04/23/2024]
Abstract
In mammals, the neonatal heart can regenerate upon injury within a short time after birth, while adults lose this ability. Metabolic reprogramming has been demonstrated to be critical for cardiomyocyte proliferation in the neonatal heart. Here, we reveal that cardiac metabolic reprogramming could be regulated by altering global protein lactylation. By performing 4D label-free proteomics and lysine lactylation (Kla) omics analyses in mouse hearts at postnatal days 1, 5, and 7, 2297 Kla sites from 980 proteins are identified, among which 1262 Kla sites from 409 proteins are quantified. Functional clustering analysis reveals that the proteins with altered Kla sites are mainly involved in metabolic processes. The expression and Kla levels of proteins in glycolysis show a positive correlation while a negative correlation in fatty acid oxidation. Furthermore, we verify the Kla levels of several differentially modified proteins, including ACAT1, ACADL, ACADVL, PFKM, PKM, and NPM1. Overall, our study reports a comprehensive Kla map in the neonatal mouse heart, which will help to understand the regulatory network of metabolic reprogramming and cardiac regeneration.
Collapse
Affiliation(s)
- Tongyu Zhang
- Ministry of Education Key Laboratory of Model Animals for Disease Study, Model Animal Research Center, Medical School of Nanjing University, National Resource Center for Mutant Mice, Nanjing, Jiangsu 210093, China
| | - Yingxi Zhu
- State Key Laboratory of Reproductive Medicine and Offspring Health, China International Joint Research Center on Environment and Human Health, Center for Global Health, School of Public Health, Gusu School, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Xiaochen Wang
- Ministry of Education Key Laboratory of Model Animals for Disease Study, Model Animal Research Center, Medical School of Nanjing University, National Resource Center for Mutant Mice, Nanjing, Jiangsu 210093, China
| | - Danyang Chong
- Ministry of Education Key Laboratory of Model Animals for Disease Study, Model Animal Research Center, Medical School of Nanjing University, National Resource Center for Mutant Mice, Nanjing, Jiangsu 210093, China; State Key Laboratory of Reproductive Medicine and Offspring Health, China International Joint Research Center on Environment and Human Health, Center for Global Health, School of Public Health, Gusu School, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Haiquan Wang
- Ministry of Education Key Laboratory of Model Animals for Disease Study, Model Animal Research Center, Medical School of Nanjing University, National Resource Center for Mutant Mice, Nanjing, Jiangsu 210093, China
| | - Dandan Bu
- Ministry of Education Key Laboratory of Model Animals for Disease Study, Model Animal Research Center, Medical School of Nanjing University, National Resource Center for Mutant Mice, Nanjing, Jiangsu 210093, China
| | - Mengfei Zhao
- Ministry of Education Key Laboratory of Model Animals for Disease Study, Model Animal Research Center, Medical School of Nanjing University, National Resource Center for Mutant Mice, Nanjing, Jiangsu 210093, China
| | - Lei Fang
- Ministry of Education Key Laboratory of Model Animals for Disease Study, Model Animal Research Center, Medical School of Nanjing University, National Resource Center for Mutant Mice, Nanjing, Jiangsu 210093, China.
| | - Chaojun Li
- Ministry of Education Key Laboratory of Model Animals for Disease Study, Model Animal Research Center, Medical School of Nanjing University, National Resource Center for Mutant Mice, Nanjing, Jiangsu 210093, China; State Key Laboratory of Reproductive Medicine and Offspring Health, China International Joint Research Center on Environment and Human Health, Center for Global Health, School of Public Health, Gusu School, Nanjing Medical University, Nanjing, Jiangsu 211166, China.
| |
Collapse
|
12
|
Becker A, Filipp M, Lantz C, Glinton K, Thorp EB. HIF-1α is Required to Differentiate the Neonatal Macrophage Secretome from Adults. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.24.591000. [PMID: 38712137 PMCID: PMC11071477 DOI: 10.1101/2024.04.24.591000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2024]
Abstract
The immune response to stress diverges with age, with neonatal macrophages implicated in tissue regeneration versus tissue scarring and maladaptive inflammation in adults. Integral to the macrophage stress response is the recognition of hypoxia and pathogen-associated molecular patterns (PAMPs), which are often coupled. The age-specific, cell-intrinsic nature of this stress response remains vague. To uncover age-defined divergences in macrophage crosstalk potential after exposure to hypoxia and PAMPs, we interrogated the secreted proteomes of neonatal versus adult macrophages via non-biased mass spectrometry. Through this approach, we newly identified age-specific signatures in the secretomes of neonatal versus adult macrophages in response to hypoxia and the prototypical PAMP, lipopolysaccharide (LPS). Neonatal macrophages polarized to an anti-inflammatory, regenerative phenotype protective against apoptosis and oxidative stress, dependent on hypoxia inducible transcription factor-1α ( HIF-1α). In contrast, adult macrophages adopted a pro-inflammatory, glycolytic phenotypic signature consistent with pathogen killing. Taken together, these data uncover fundamental age and HIF-1α dependent macrophage programs that may be targeted to calibrate the innate immune response during stress and inflammation.
Collapse
|
13
|
Varlı M, Bhosle SR, Kim E, Yang Y, Taş İ, Zhou R, Pulat S, Gamage CDB, Park SY, Ha HH, Kim H. Usnic Acid Targets 14-3-3 Proteins and Suppresses Cancer Progression by Blocking Substrate Interaction. JACS AU 2024; 4:1521-1537. [PMID: 38665668 PMCID: PMC11040559 DOI: 10.1021/jacsau.3c00774] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 03/07/2024] [Accepted: 03/19/2024] [Indexed: 04/28/2024]
Abstract
The anticancer therapeutic effects of usnic acid (UA), a lichen secondary metabolite, have been demonstrated in vitro and in vivo. However, the mechanism underlying the anticancer effect of UA remains to be clarified. In this study, the target protein of UA was identified using a UA-linker-Affi-Gel molecule, which showed that UA binds to the 14-3-3 protein. UA binds to 14-3-3, causing the degradation of proteasomal and autophagosomal proteins. The interaction of UA with 14-3-3 isoforms modulated cell invasion, cell cycle progression, aerobic glycolysis, mitochondrial biogenesis, and the Akt/mTOR, JNK, STAT3, NF-κB, and AP-1 signaling pathways in colorectal cancer. A peptide inhibitor of 14-3-3 blocked or regressed the activity of UA and inhibited its effects. The results suggest that UA binds to 14-3-3 isoforms and suppresses cancer progression by affecting 14-3-3 targets and phosphorylated proteins.
Collapse
Affiliation(s)
- Mücahit Varlı
- College
of Pharmacy, Sunchon National University, 255 Jungang-ro, Sunchon, Jeonnam 57922, Republic of Korea
| | - Suresh R. Bhosle
- College
of Pharmacy, Sunchon National University, 255 Jungang-ro, Sunchon, Jeonnam 57922, Republic of Korea
| | - Eunae Kim
- College
of Pharmacy, Chosun University, 146 Chosundae-gil, Gwangju 61452, Republic of Korea
| | - Yi Yang
- College
of Pharmacy, Sunchon National University, 255 Jungang-ro, Sunchon, Jeonnam 57922, Republic of Korea
| | - İsa Taş
- College
of Pharmacy, Sunchon National University, 255 Jungang-ro, Sunchon, Jeonnam 57922, Republic of Korea
| | - Rui Zhou
- College
of Pharmacy, Sunchon National University, 255 Jungang-ro, Sunchon, Jeonnam 57922, Republic of Korea
| | - Sultan Pulat
- College
of Pharmacy, Sunchon National University, 255 Jungang-ro, Sunchon, Jeonnam 57922, Republic of Korea
| | - Chathurika D. B. Gamage
- College
of Pharmacy, Sunchon National University, 255 Jungang-ro, Sunchon, Jeonnam 57922, Republic of Korea
| | - So-Yeon Park
- College
of Pharmacy, Sunchon National University, 255 Jungang-ro, Sunchon, Jeonnam 57922, Republic of Korea
| | - Hyung-Ho Ha
- College
of Pharmacy, Sunchon National University, 255 Jungang-ro, Sunchon, Jeonnam 57922, Republic of Korea
| | - Hangun Kim
- College
of Pharmacy, Sunchon National University, 255 Jungang-ro, Sunchon, Jeonnam 57922, Republic of Korea
| |
Collapse
|
14
|
Abdi G, Jain M, Patil N, Upadhyay B, Vyas N, Dwivedi M, Kaushal RS. 14-3-3 proteins-a moonlight protein complex with therapeutic potential in neurological disorder: in-depth review with Alzheimer's disease. Front Mol Biosci 2024; 11:1286536. [PMID: 38375509 PMCID: PMC10876095 DOI: 10.3389/fmolb.2024.1286536] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 01/05/2024] [Indexed: 02/21/2024] Open
Abstract
Alzheimer's disease (AD) affects millions of people worldwide and is a gradually worsening neurodegenerative condition. The accumulation of abnormal proteins, such as tau and beta-amyloid, in the brain is a hallmark of AD pathology. 14-3-3 proteins have been implicated in AD pathology in several ways. One proposed mechanism is that 14-3-3 proteins interact with tau protein and modulate its phosphorylation, aggregation, and toxicity. Tau is a protein associated with microtubules, playing a role in maintaining the structural integrity of neuronal cytoskeleton. However, in the context of Alzheimer's disease (AD), an abnormal increase in its phosphorylation occurs. This leads to the aggregation of tau into neurofibrillary tangles, which is a distinctive feature of this condition. Studies have shown that 14-3-3 proteins can bind to phosphorylated tau and regulate its function and stability. In addition, 14-3-3 proteins have been shown to interact with beta-amyloid (Aβ), the primary component of amyloid plaques in AD. 14-3-3 proteins can regulate the clearance of Aβ through the lysosomal degradation pathway by interacting with the lysosomal membrane protein LAMP2A. Dysfunction of lysosomal degradation pathway is thought to contribute to the accumulation of Aβ in the brain and the progression of AD. Furthermore, 14-3-3 proteins have been found to be downregulated in the brains of AD patients, suggesting that their dysregulation may contribute to AD pathology. For example, decreased levels of 14-3-3 proteins in cerebrospinal fluid have been suggested as a biomarker for AD. Overall, these findings suggest that 14-3-3 proteins may play an important role in AD pathology and may represent a potential therapeutic target for the disease. However, further research is needed to fully understand the mechanisms underlying the involvement of 14-3-3 proteins in AD and to explore their potential as a therapeutic target.
Collapse
Affiliation(s)
- Gholamareza Abdi
- Department of Biotechnology, Persian Gulf Research Institute, Persian Gulf University, Bushehr, Iran
| | - Mukul Jain
- Cell and Developmental Biology Laboratory, Research and Development Cell, Parul University, Vadodara, Gujarat, India
- Department of Life Sciences, Parul Institute of Applied Sciences, Parul University, Vadodara, Gujarat, India
| | - Nil Patil
- Cell and Developmental Biology Laboratory, Research and Development Cell, Parul University, Vadodara, Gujarat, India
- Department of Life Sciences, Parul Institute of Applied Sciences, Parul University, Vadodara, Gujarat, India
| | - Bindiya Upadhyay
- Department of Life Sciences, Parul Institute of Applied Sciences, Parul University, Vadodara, Gujarat, India
| | - Nigam Vyas
- Department of Life Sciences, Parul Institute of Applied Sciences, Parul University, Vadodara, Gujarat, India
- Biophysics and Structural Biology Laboratory, Research and Development Cell, Parul University, Vadodara, Gujarat, India
| | - Manish Dwivedi
- Amity Institute of Biotechnology, Amity University, Lucknow, Uttar Pradesh, India
| | - Radhey Shyam Kaushal
- Department of Life Sciences, Parul Institute of Applied Sciences, Parul University, Vadodara, Gujarat, India
- Biophysics and Structural Biology Laboratory, Research and Development Cell, Parul University, Vadodara, Gujarat, India
| |
Collapse
|
15
|
Jiang W, Jiang Y, Luo Y, Qiao W, Yang T. Facilitating the development of molecular glues: Opportunities from serendipity and rational design. Eur J Med Chem 2024; 263:115950. [PMID: 37984298 DOI: 10.1016/j.ejmech.2023.115950] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 11/09/2023] [Accepted: 11/09/2023] [Indexed: 11/22/2023]
Abstract
Molecular glues can specifically induce interactions between two or more proteins to modulate biological functions and have been proven to be a powerful therapeutic modality in drug discovery. It plays a variety of vital roles in several biological processes, such as complex stabilization, interactome modulation and transporter inhibition, thus enabling challenging therapeutic targets to be druggable. Most known molecular glues were identified serendipitously, such as IMiDs, auxin, and rapamycin. In recent years, more rational strategies were explored with the development of chemical biology and a deep understanding of the interaction between molecular glues and proteins, which led to the rational discovery of several molecular glues. Thus, in this review, we aim to highlight the discovery strategies of molecular glues from three aspects: serendipitous discovery, screening methods and rational design principles. We expect that this review will provide a reasonable reference and insights for the discovery of molecular glues.
Collapse
Affiliation(s)
- Weiqing Jiang
- Laboratory of Human Diseases and Immunotherapies, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China; Institute of Immunology and Inflammation, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yunhan Jiang
- Laboratory of Human Diseases and Immunotherapies, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China; Institute of Immunology and Inflammation, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, 610041, China; Cardiovascular Surgery Research Laboratory, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Youfu Luo
- Laboratory of Human Diseases and Immunotherapies, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China.
| | - Wenliang Qiao
- Lung Cancer Center, Laboratory of Lung Cancer, West China Hospital, Sichuan University, Chengdu, 610041, China.
| | - Tao Yang
- Laboratory of Human Diseases and Immunotherapies, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China; Institute of Immunology and Inflammation, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
16
|
Wang J, Cheng X, Mei X, Wu H, Yu Q, Xiao M. The effect of Par3 on the cellular junctions and biological functions of odontoblast-lineage cells. Odontology 2024; 112:125-137. [PMID: 37493885 DOI: 10.1007/s10266-023-00838-5] [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: 12/12/2022] [Accepted: 04/17/2023] [Indexed: 07/27/2023]
Abstract
Perfect intercellular junctions are key for odontoblast barrier function. However, whether Partitioning defective-3 (Par3) is expressed in odontoblasts and its potential effects on odontoblast junctions are unknown. Herein, we investigated the effect of Par3 on cellular junctions and the biological behavior of odontoblast-lineage cells (OLCs). Whole-transcriptome sequencing was used to analyze the effects of Par3 on OLCs and the underlying molecular mechanism. Par3 was detected under physiological and inflammatory conditions in OLCs. To investigate the regulatory effect of Par3 on junctions between mouse OLCs, the effects of Par3 downregulation on the proliferation, migration, cycle and apoptosis of OLCs were detected by 5-ethyl-2'-deoxyuridine (EdU) and Transwell assays and flow cytometry. Western blotting and alizarin red S and alkaline phosphatase (ALP) staining were used to observe the effect of Par3 downregulation on OLC mineralization. Whole-transcriptome sequencing was used to investigate the biological role of Par3 in OLCs and potential molecular mechanisms. Par3 was located along the odontoblast layer in the rat pulp tissue and in the cytoplasm of OLCs. Par3 expression was downregulated under inflammatory conditions. The OLC junctions were discontinuous, and total Zona occluden-1 (ZO-1) expression and expression of ZO-1 at the membrane in OLCs were reduced after Par3 silencing (P < 0.05). Expression of a junction-related protein (ZO-1) was downregulated after the downregulation of Par3 (P < 0.05), and ZO-1 moved from the cell membrane to the cytoplasm. OLC proliferation and migration were enhanced, but apoptosis and mineralization were inhibited in shPar3-transfected cells (P < 0.05). Sequencing identified 2996 differentially expressed genes (DEGs), which were mainly enriched in the response to stimuli and binding. Downregulation of Par3 could overactivate the PI3k-AKT pathway by promoting AKT phosphorylation (P < 0.05). Downregulation of Par3 may disrupt junctions between OLCs by affecting ZO-1 expression and distribution and promote OLC proliferation and migration but inhibit OLC mineralization. Par3 may interact with 14-3-3 proteins for PI3K-AKT pathway activation to affect OLC junctions and function.
Collapse
Affiliation(s)
- Jueyu Wang
- State Key Laboratory of Oral & Maxillofacial reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Key Laboratory of Stomatology, Department of Operative Dentistry and Endodontics, School of Stomatology, Air Force Medical University, 145 West Chang-le Road, Xi'an, China
| | - Xiaogang Cheng
- State Key Laboratory of Oral & Maxillofacial reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Key Laboratory of Stomatology, Department of Operative Dentistry and Endodontics, School of Stomatology, Air Force Medical University, 145 West Chang-le Road, Xi'an, China
| | - Xiaohan Mei
- State Key Laboratory of Oral & Maxillofacial reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Key Laboratory of Stomatology, Department of Operative Dentistry and Endodontics, School of Stomatology, Air Force Medical University, 145 West Chang-le Road, Xi'an, China
| | - Haoze Wu
- State Key Laboratory of Oral & Maxillofacial reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Key Laboratory of Stomatology, Department of Operative Dentistry and Endodontics, School of Stomatology, Air Force Medical University, 145 West Chang-le Road, Xi'an, China
| | - Qing Yu
- State Key Laboratory of Oral & Maxillofacial reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Key Laboratory of Stomatology, Department of Operative Dentistry and Endodontics, School of Stomatology, Air Force Medical University, 145 West Chang-le Road, Xi'an, China
| | - Min Xiao
- State Key Laboratory of Oral & Maxillofacial reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Key Laboratory of Stomatology, Department of Operative Dentistry and Endodontics, School of Stomatology, Air Force Medical University, 145 West Chang-le Road, Xi'an, China.
| |
Collapse
|
17
|
Gabizon R, Tivon B, Reddi RN, van den Oetelaar MCM, Amartely H, Cossar PJ, Ottmann C, London N. A simple method for developing lysine targeted covalent protein reagents. Nat Commun 2023; 14:7933. [PMID: 38040731 PMCID: PMC10692228 DOI: 10.1038/s41467-023-42632-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Accepted: 10/16/2023] [Indexed: 12/03/2023] Open
Abstract
Peptide-based covalent probes can target shallow protein surfaces not typically addressable using small molecules, yet there is a need for versatile approaches to convert native peptide sequences into covalent binders that can target a broad range of residues. Here we report protein-based thio-methacrylate esters-electrophiles that can be installed easily on unprotected peptides and proteins via cysteine side chains, and react efficiently and selectively with cysteine and lysine side chains on the target. Methacrylate phosphopeptides derived from 14-3-3-binding proteins irreversibly label 14-3-3σ via either lysine or cysteine residues, depending on the position of the electrophile. Methacrylate peptides targeting a conserved lysine residue exhibit pan-isoform binding of 14-3-3 proteins both in lysates and in extracellular media. Finally, we apply this approach to develop protein-based covalent binders. A methacrylate-modified variant of the colicin E9 immunity protein irreversibly binds to the E9 DNAse, resulting in significantly higher thermal stability relative to the non-covalent complex. Our approach offers a simple and versatile route to convert peptides and proteins into potent covalent binders.
Collapse
Affiliation(s)
- Ronen Gabizon
- Department of Chemical and Structural Biology, The Weizmann Institute of Science, Rehovot, 7610001, Israel
| | - Barr Tivon
- Department of Chemical and Structural Biology, The Weizmann Institute of Science, Rehovot, 7610001, Israel
| | - Rambabu N Reddi
- Department of Chemical and Structural Biology, The Weizmann Institute of Science, Rehovot, 7610001, Israel
| | - Maxime C M van den Oetelaar
- Laboratory of Chemical Biology, Department of Biomedical Engineering, Institute for Complex Molecular Systems, Eindhoven University of Technology, P.O. Box 513, 5600MB, Eindhoven, The Netherlands
| | - Hadar Amartely
- Wolfson Centre for Applied Structural Biology, The Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem, 9190401, Israel
| | - Peter J Cossar
- Laboratory of Chemical Biology, Department of Biomedical Engineering, Institute for Complex Molecular Systems, Eindhoven University of Technology, P.O. Box 513, 5600MB, Eindhoven, The Netherlands
| | - Christian Ottmann
- Laboratory of Chemical Biology, Department of Biomedical Engineering, Institute for Complex Molecular Systems, Eindhoven University of Technology, P.O. Box 513, 5600MB, Eindhoven, The Netherlands
| | - Nir London
- Department of Chemical and Structural Biology, The Weizmann Institute of Science, Rehovot, 7610001, Israel.
| |
Collapse
|
18
|
Swami S, Mughees M, Mangangcha IR, Kauser S, Wajid S. Secretome analysis of breast cancer cells to identify potential target proteins of Ipomoea turpethum extract-loaded nanoparticles in the tumor microenvironment. Front Cell Dev Biol 2023; 11:1247632. [PMID: 37900279 PMCID: PMC10602817 DOI: 10.3389/fcell.2023.1247632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Accepted: 09/18/2023] [Indexed: 10/31/2023] Open
Abstract
Background: Breast cancer is the leading cause of frequent malignancy and morbidity among women across the globe, with an increment of 0.5% incidences every year. The deleterious effects of traditional treatment on off-target surrounding cells make it difficult to win the battle against breast cancer. Hence, an advancement in the therapeutic approach is crucial. Nanotechnology is one of the emerging methods for precise, targeted, and efficient drug delivery in cells. The previous study has demonstrated the cytotoxic effect of Ipomoea turpethum extract on breast cancer cells delivered via NIPAAM-VP-AA nanoparticles (NVA-IT). Manipulating the tumor microenvironment (TME) to inhibit cancer progression, invasion, and metastasis seems to be very insightful for researchers these days. With the help of secretome analysis of breast cancer cells after treatment with NVA-IT, we have tried to find out the possible TME manipulation achieved to favor a better prognosis of the disease. Method: MCF-7 and MDA MB-231 cells were treated with the IC50 value of NVA-IT, and the medium was separated from the cells after 24 h of the treatment. Nano LCMS/MS analysis was performed to identify the secretory proteins in the media. Further bioinformatics tools like GENT2, GSCA, GeneCodis 4, and STRING were used to identify the key proteins and their interactions. Result: From the nano LCMS/MS analysis, 70 differentially expressed secretory proteins in MCF-7 and 191 in MDA MB-231 were identified in the cell's media. Fifteen key target proteins were filtered using bioinformatics analysis, and the interaction of proteins involved in vesicular trafficking, cell cycle checkpoints, and oxidative stress-related proteins was prominent. Conclusion: This study concluded that I. turpethum extract-loaded NIPAAM-VP-AA nanoparticles alter the secretory proteins constituting the TME to cease cancer cell growth and metastasis.
Collapse
Affiliation(s)
- Sanskriti Swami
- Department of Biotechnology, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi, India
| | - Mohd Mughees
- Department of Biotechnology, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi, India
| | | | - Sana Kauser
- Department of Biotechnology, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi, India
| | - Saima Wajid
- Department of Biotechnology, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi, India
| |
Collapse
|
19
|
Kongsamut S, Eishingdrelo H. Modulating GPCR and 14-3-3 protein interactions: Prospects for CNS drug discovery. Drug Discov Today 2023; 28:103641. [PMID: 37236523 PMCID: PMC10524340 DOI: 10.1016/j.drudis.2023.103641] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Revised: 04/29/2023] [Accepted: 05/19/2023] [Indexed: 05/28/2023]
Abstract
The activation of G-protein-coupled receptors (GPCRs) triggers a series of protein-protein interaction events that subsequently induce a chain of reactions, including alteration of receptor structures, phosphorylation, recruitment of associated proteins, protein trafficking and gene expression. Multiple GPCR signaling transduction pathways are evident - two well-studied pathways are the GPCR-mediated G-protein and β-arrestin pathways. Recently, ligand-induced interactions between GPCRs and 14-3-3 proteins have been demonstrated. This linking of GPCRs to 14-3-3 protein signal hubs opens up a whole new realm of signal transduction possibilities. 14-3-3 proteins play a key part in GPCR trafficking and signal transduction. GPCR-mediated 14-3-3 protein signaling can be harnessed for the study of GPCR function and therapeutics.
Collapse
Affiliation(s)
- Sathapana Kongsamut
- Research Institute for Scientists Emeriti, Drew University, 36 Madison Avenue, Madison, NJ 07940, USA
| | | |
Collapse
|
20
|
Rial SA, Shishani R, Cummings BP, Lim GE. Is 14-3-3 the Combination to Unlock New Pathways to Improve Metabolic Homeostasis and β-Cell Function? Diabetes 2023; 72:1045-1054. [PMID: 37471599 PMCID: PMC10382651 DOI: 10.2337/db23-0094] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 05/10/2023] [Indexed: 07/22/2023]
Abstract
Since their discovery nearly five decades ago, molecular scaffolds belonging to the 14-3-3 protein family have been recognized as pleiotropic regulators of diverse cellular and physiological functions. With their ability to bind to proteins harboring specific serine and threonine phosphorylation motifs, 14-3-3 proteins can interact with and influence the function of docking proteins, enzymes, transcription factors, and transporters that have essential roles in metabolism and glucose homeostasis. Here, we will discuss the regulatory functions of 14-3-3 proteins that will be of great interest to the fields of metabolism, pancreatic β-cell biology, and diabetes. We first describe how 14-3-3 proteins play a central role in glucose and lipid homeostasis by modulating key pathways of glucose uptake, glycolysis, oxidative phosphorylation, and adipogenesis. This is followed by a discussion of the contributions of 14-3-3 proteins to calcium-dependent exocytosis and how this relates to insulin secretion from β-cells. As 14-3-3 proteins are major modulators of apoptosis and cell cycle progression, we will explore if 14-3-3 proteins represent a viable target for promoting β-cell regeneration and discuss the feasibility of targeting 14-3-3 proteins to treat metabolic diseases such as diabetes. ARTICLE HIGHLIGHTS 14-3-3 proteins are ubiquitously expressed scaffolds with multiple roles in glucose homeostasis and metabolism. 14-3-3ζ regulates adipogenesis via distinct mechanisms and is required for postnatal adiposity and adipocyte function. 14-3-3ζ controls glucose-stimulated insulin secretion from pancreatic β-cells by regulating mitochondrial function and ATP synthesis as well as facilitating cross talk between β-cells and α-cells.
Collapse
Affiliation(s)
- Sabri A. Rial
- Department of Medicine, University of Montreal, Montreal, Quebec, Canada
- Cardiometabolic Axis, University of Montreal Hospital Research Center (CRCHUM), Montreal, Quebec, Canada
| | - Rahaf Shishani
- Department of Surgery, Center for Alimentary and Metabolic Sciences, School of Medicine, University of California, Davis, Sacramento, CA
| | - Bethany P. Cummings
- Department of Surgery, Center for Alimentary and Metabolic Sciences, School of Medicine, University of California, Davis, Sacramento, CA
| | - Gareth E. Lim
- Department of Medicine, University of Montreal, Montreal, Quebec, Canada
- Cardiometabolic Axis, University of Montreal Hospital Research Center (CRCHUM), Montreal, Quebec, Canada
| |
Collapse
|
21
|
Zhang J, Chen X, Cao J, Chang C, Geng A, Wang H, Chu Q, Yan Z, Zhang Y, Liu H. Proteomic Profiling of Thigh Meat at Different Ages of Chicken for Meat Quality and Development. Foods 2023; 12:2901. [PMID: 37569170 PMCID: PMC10418907 DOI: 10.3390/foods12152901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 07/20/2023] [Accepted: 07/25/2023] [Indexed: 08/13/2023] Open
Abstract
Chicken age contributes to the meat characteristics; however, knowledge regarding the pathways and proteins associated with meat quality and muscle development are still scarce, especially in chicken thigh meat. Hence, the objective of this study was to elucidate the intricate relationship between these traits by liquid chromatography mass spectrometry at three different ages. A total of 341 differential expressed proteins (DEPs) were screened out (fold change ≥ 1.50 or ≤0.67 and p < 0.05) among 45 thigh meat samples (15 samples per age) of Beijing-You chicken (BYC), collected at the age of 150, 300, or 450 days (D150, D300, and D450), respectively. Subsequently, based on the protein interaction network and Markov cluster algorithm (MCL) analyses, 91 DEPs were divided into 26 MCL clusters, which were associated with pathways of lipid transporter activity, nutrient reservoir activity, signaling pathways of PPAR and MAPK, focal adhesion, ECM-receptor interaction, the cell cycle, oocyte meiosis, ribosomes, taurine and hypotaurine metabolism, glutathione metabolism, muscle contraction, calcium signaling, nucleic acid binding, and spliceosomes. Overall, our data suggest that the thigh meat of BYC at D450 presents the most desirable nutritional value in the term of free amino acids (FAAs) and intramuscular fat (IMF), and a series of proteins and pathways associated with meat quality and development were identified. These findings also provide comprehensive insight regarding these traits across a wide age spectrum.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Huagui Liu
- Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, Beijing 100097, China; (J.Z.); (X.C.); (J.C.); (C.C.); (A.G.); (H.W.); (Q.C.); (Z.Y.); (Y.Z.)
| |
Collapse
|
22
|
Evidence and Metabolic Implications for a New Non-Canonical Role of Cu-Zn Superoxide Dismutase. Int J Mol Sci 2023; 24:ijms24043230. [PMID: 36834640 PMCID: PMC9966940 DOI: 10.3390/ijms24043230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 01/31/2023] [Accepted: 02/01/2023] [Indexed: 02/10/2023] Open
Abstract
Copper-zinc superoxide dismutase 1 (SOD1) has long been recognized as a major redox enzyme in scavenging superoxide radicals. However, there is little information on its non-canonical role and metabolic implications. Using a protein complementation assay (PCA) and pull-down assay, we revealed novel protein-protein interactions (PPIs) between SOD1 and tyrosine 3-monooxygenase/tryptophan 5-monooxygenase activation protein zeta (YWHAZ) or epsilon (YWHAE) in this research. Through site-directed mutagenesis of SOD1, we studied the binding conditions of the two PPIs. Forming the SOD1 and YWHAE or YWHAZ protein complex enhanced enzyme activity of purified SOD1 in vitro by 40% (p < 0.05) and protein stability of over-expressed intracellular YWHAE (18%, p < 0.01) and YWHAZ (14%, p < 0.05). Functionally, these PPIs were associated with lipolysis, cell growth, and cell survival in HEK293T or HepG2 cells. In conclusion, our findings reveal two new PPIs between SOD1 and YWHAE or YWHAZ and their structural dependences, responses to redox status, mutual impacts on the enzyme function and protein degradation, and metabolic implications. Overall, our finding revealed a new unorthodox role of SOD1 and will provide novel perspectives and insights for diagnosing and treating diseases related to the protein.
Collapse
|
23
|
Garan LAW, Xiao Y, Lin WC. 14-3-3τ drives estrogen receptor loss via ERα36 induction and GATA3 inhibition in breast cancer. Proc Natl Acad Sci U S A 2022; 119:e2209211119. [PMID: 36252018 PMCID: PMC9618134 DOI: 10.1073/pnas.2209211119] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Accepted: 09/21/2022] [Indexed: 11/18/2022] Open
Abstract
About one-fourth of recurrent estrogen receptor-positive (ER+) breast cancers lose ER expression, leading to endocrine therapy failure. However, the mechanisms underlying ER loss remain to be fully explored. We now show that 14-3-3τ, up-regulated in ∼60% of breast cancer, drives the conversion of ER+ to ER- and epithelial-to-mesenchymal transition (EMT). We identify ERα36, an isoform of ERα66, as a downstream effector of 14-3-3τ. Overexpression of 14-3-3τ induces ERα36 in xenografts and tumor spheroids. The regulation is further supported by a positive correlation between ERα36 and 14-3-3τ expression in human breast cancers. ERα36 can antagonize ERα66 and inhibit ERα66 expression. Isoform-specific depletion of ERα36 blocks the ER conversion and EMT induced by 14-3-3τ overexpression in tumor spheroids, thus establishing ERα36 as a key mediator in 14-3-3τ-driven ER loss and EMT. ERα36 promoter is repressed by GATA3, which can be phosphorylated by AKT at consensus binding sites for 14-3-3. Upon AKT activation, 14-3-3τ binds phosphorylated GATA3 and facilitates the degradation of GATA3 causing GATA3 to lose transcriptional control over its target genes ERα66 and ERα36. We also demonstrate a role for the collaboration between 14-3-3τ and AKT in ERα36 induction and endocrine therapy resistance by three-dimensional spheroid and tamoxifen treatment models in MCF7 and T47D ER+ breast cancer cells. Thus, the 14-3-3τ-ERα36 regulation provides a previously unrecognized mechanism for ER loss and endocrine therapy failure.
Collapse
Affiliation(s)
- Lidija A. Wilhelms Garan
- Section of Hematology/Oncology, Department of Medicine, Baylor College of Medicine, Houston, TX 77030
- Cancer and Cell Biology Graduate Program, Baylor College of Medicine, Houston, TX 77030
| | - Yang Xiao
- Section of Hematology/Oncology, Department of Medicine, Baylor College of Medicine, Houston, TX 77030
| | - Weei-Chin Lin
- Section of Hematology/Oncology, Department of Medicine, Baylor College of Medicine, Houston, TX 77030
- Cancer and Cell Biology Graduate Program, Baylor College of Medicine, Houston, TX 77030
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030
| |
Collapse
|
24
|
Iwahori S, Umaña AC, Kalejta RF, Murata T. Serine 13 of the human cytomegalovirus viral cyclin-dependent kinase UL97 is required for regulatory protein 14-3-3 binding and UL97 stability. J Biol Chem 2022; 298:102513. [PMID: 36150501 PMCID: PMC9587022 DOI: 10.1016/j.jbc.2022.102513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 09/08/2022] [Accepted: 09/13/2022] [Indexed: 11/14/2022] Open
Abstract
The human cytomegalovirus (HCMV) UL97 protein is a conserved herpesvirus protein kinase (CHPK) and a viral cyclin-dependent kinase (v-CDK). However, mechanisms regulating its activity in the context of infection are unknown. Here, we identified several cellular regulatory 14-3-3 proteins as UL97-interacting partners that promote UL97 stability. Humans are known to encode seven isoforms of 14-3-3 proteins (β, ε, η, γ, σ, θ, and ζ) that bind phosphoserines or phosphothreonines to impact protein structure, stability, activity, and localization. Our proteomic analysis of UL97 identified 49 interacting partners, including 14-3-3 isoforms β, η, and γ. Furthermore, coimmunoprecipitation with Western blotting assays demonstrated that UL97 interaction with 14-3-3 isoforms β, ε, η, γ, and θ occurs in a kinase activity-dependent manner. Using mutational analysis, we determined the serine residue at amino acid 13 of UL97 is crucial for 14-3-3 interaction. We demonstrate UL97 S13A (serine to alanine substitution at residue 13) retains kinase activity but the mutant protein accumulated at lower levels than WT UL97. Finally, we show both laboratory (AD169) and clinical (TB40/E) strains of HCMV encoding UL97 S13A replicated with WT kinetics in fibroblasts but showed decreased UL97 accumulation. Taken together, we conclude that 14-3-3 proteins interact with and stabilize UL97 during HCMV infection.
Collapse
Affiliation(s)
- Satoko Iwahori
- Department of Virology and Parasitology, Fujita Health University School of Medicine, 1-98 Dengakugakubo, Kutsukake-cho, Toyoake, Aichi 470-1192, Japan; Institute for Molecular Virology and McArdle Laboratory for Cancer Research, University of Wisconsin-Madison, Madison, WI, USA
| | - Angie C Umaña
- Institute for Molecular Virology and McArdle Laboratory for Cancer Research, University of Wisconsin-Madison, Madison, WI, USA
| | - Robert F Kalejta
- Institute for Molecular Virology and McArdle Laboratory for Cancer Research, University of Wisconsin-Madison, Madison, WI, USA.
| | - Takayuki Murata
- Department of Virology and Parasitology, Fujita Health University School of Medicine, 1-98 Dengakugakubo, Kutsukake-cho, Toyoake, Aichi 470-1192, Japan.
| |
Collapse
|
25
|
Identification of Transferrin Receptor 1 (TfR1) Overexpressed in Lung Cancer Cells, and Internalization of Magnetic Au-CoFe2O4 Core-Shell Nanoparticles Functionalized with Its Ligand in a Cellular Model of Small Cell Lung Cancer (SCLC). Pharmaceutics 2022; 14:pharmaceutics14081715. [PMID: 36015341 PMCID: PMC9413248 DOI: 10.3390/pharmaceutics14081715] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 07/27/2022] [Accepted: 08/02/2022] [Indexed: 11/26/2022] Open
Abstract
Lung cancer is, currently, one of the main malignancies causing deaths worldwide. To date, early prognostic and diagnostic markers for small cell lung cancer (SCLC) have not been systematically and clearly identified, so most patients receive standard treatment. In the present study, we combine quantitative proteomics studies and the use of magnetic core-shell nanoparticles (mCSNP’s), first to identify a marker for lung cancer, and second to functionalize the nanoparticles and their possible application for early and timely diagnosis of this and other types of cancer. In the present study, we used label-free mass spectrometry in combination with an ion-mobility approach to identify 220 proteins with increased abundance in small cell lung cancer (SCLC) cell lines. Our attention was focused on cell receptors for their potential application as mCSNP’s targets; in this work, we report the overexpression of Transferrin Receptor (TfR1) protein, also known as Cluster of Differentiation 71 (CD71) up to a 30-fold increase with respect to the control cell. The kinetics of endocytosis, evaluated by a flow cytometry methodology based on fluorescence quantification, demonstrated that receptors were properly activated with the transferrin supported on the magnetic core-shell nanoparticles. Our results are important in obtaining essential information for monitoring the disease and/or choosing better treatments, and this finding will pave the way for future synthesis of nanoparticles including chemotherapeutic drugs for lung cancer treatments.
Collapse
|
26
|
Zhang Y, Luo B, Liu MC, OuYang RH, Fan XM, Jiang N, Yang FJ, Wang LJ, Zhou BY. Analysis of immune response in BALB/c mice immunized with recombinant plasmids pMZ-X3-Ts14-3-3.3 and pMZ-X3-sp-Ts14-3-3.3 of Taenia solium. Acta Trop 2022; 232:106517. [PMID: 35595093 DOI: 10.1016/j.actatropica.2022.106517] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 04/29/2022] [Accepted: 05/13/2022] [Indexed: 11/01/2022]
Abstract
There is a lack of vaccine against human cysticercosis, thus making a huge population at the risk of infection. In this study, we chose a novel potential antigen molecule Taenia solium 14-3-3.3 (Ts14-3-3.3) and optimized it as sp-Ts14-3-3.3 (sp is immunoglobulin H chain V-region precursor, partial) in order to construct recombinant plasmids pMZ-X3-Ts14-3-3.3 and pMZ-X3-sp-Ts14-3-3.3. BALB/c mice were divided into four groups for immunization: pMZ-X3-Ts14-3-3.3, pMZ-X3-sp-Ts14-3-3.3, pMZ-X3 plasmid control group and PBS control group. Compared with two control groups, the proliferation level of splenic lymphocytes increased significantly in pMZ-X3-Ts14-3-3.3 and pMZ-X3-sp-Ts14-3-3.3 groups and reached the maximum in week 6. And the same case arose as cytokines associated with Th1 response, IFN-γ, and IL-2 while those with Th2 response, IL-4, IL-10 went up and reached the maximum in week 4. The levels of serum specific IgG, IgG1 and IgG2a rose and reached the maximum in week 6, 4 and 6, respectively. Meanwhile, the proportion of CD4+/CD8+ splenic T lymphocytes increased and reached the peak in week 6. The results indicated that the recombinant plasmids pMZ-X3-Ts14-3-3.3 and pMZ-X3-sp-Ts14-3-3.3 can induce specific cellular and humoral immune responses in BALB/c mice with immunization. Notably, the recombinant plasmid pMZ-X3-sp-Ts14-3-3.3 has a better immune effect, which proves that Ts14-3-3.3 enjoys a higher possibility as a potential antigen molecule to T. solium vaccine.
Collapse
|
27
|
Gao Z, Fan T, Chen L, Yang M, Wai Wong VK, Chen D, Liu Z, Zhou Y, Wu W, Qiu Z, Zhang C, Li Y, Jiang Y. Design, synthesis and antitumor evaluation of novel 1H-indole-2-carboxylic acid derivatives targeting 14-3-3η protein. Eur J Med Chem 2022; 238:114402. [DOI: 10.1016/j.ejmech.2022.114402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 04/06/2022] [Accepted: 04/17/2022] [Indexed: 11/04/2022]
|
28
|
Mugabo Y, Zhao C, Tan JJ, Ghosh A, Campbell SA, Fadzeyeva E, Paré F, Pan SS, Galipeau M, Ast J, Broichhagen J, Hodson DJ, Mulvihill EE, Petropoulos S, Lim GE. 14-3-3ζ constrains insulin secretion by regulating mitochondrial function in pancreatic β-cells. JCI Insight 2022; 7:156378. [PMID: 35298439 PMCID: PMC9089799 DOI: 10.1172/jci.insight.156378] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Accepted: 03/11/2022] [Indexed: 11/17/2022] Open
Abstract
While critical for neurotransmitter synthesis, 14-3-3 proteins are often assumed to have redundant functions due to their ubiquitous expression, but despite this assumption, various 14-3-3 isoforms have been implicated in regulating metabolism. We previously reported contributions of 14-3-3ζ in β cell function, but these studies were performed in tumor-derived MIN6 cells and systemic KO mice. To further characterize the regulatory roles of 14-3-3ζ in β cell function, we generated β cell–specific 14-3-3ζ–KO mice. Although no effects on β cell mass were detected, potentiated glucose-stimulated insulin secretion (GSIS), mitochondrial function, and ATP synthesis were observed. Deletion of 14-3-3ζ also altered the β cell transcriptome, as genes associated with mitochondrial respiration and oxidative phosphorylation were upregulated. Acute 14-3-3 protein inhibition in mouse and human islets recapitulated the enhancements in GSIS and mitochondrial function, suggesting that 14-3-3ζ is the critical isoform in β cells. In dysfunctional db/db islets and human islets from type 2 diabetic donors, expression of Ywhaz/YWHAZ, the gene encoding 14-3-3ζ, was inversely associated with insulin secretion, and pan–14-3-3 protein inhibition led to enhanced GSIS and mitochondrial function. Taken together, this study demonstrates important regulatory functions of 14-3-3ζ in the regulation of β cell function and provides a deeper understanding of how insulin secretion is controlled in β cells.
Collapse
Affiliation(s)
- Yves Mugabo
- Department of Medicine, Université de Montréal, Montreal, Canada
| | - Cheng Zhao
- Division of Obstetrics and Gynecology, Department of Clinical Science, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
| | - Ju Jing Tan
- Immunopathology Axis, Centre de Recherche du Centre hospitalier de l'Université de Montréal, Montreal, Canada
| | - Anindya Ghosh
- Department of Medicine, Université de Montréal, Montreal, Canada
| | - Scott A Campbell
- Cardiometabolic Axis, Centre de Recherche du Centre hospitalier de l'Université de Montréal, Montreal, Canada
| | - Evgenia Fadzeyeva
- Energy Substrate Laboratory, University of Ottawa Heart Institute, Ottawa, Canada
| | - Frédéric Paré
- Cardiometabolic Axis, Centre de recherche du CHUM (CRCHUM), Montreal, Canada
| | - Siew Siew Pan
- Department of Medicine, Université de Montréal, Montreal, Canada
| | - Maria Galipeau
- Department of Medicine, Université de Montréal, Montreal, Canada
| | - Julia Ast
- Institute of Metabolism and Systems Research (IMSR), Centre of Membrane Pro, University of Birmingham, Birmingham, United Kingdom
| | - Johannes Broichhagen
- Department of Chemical Biology, Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Berlin, Germany
| | - David J Hodson
- Institute of Metabolism and Systems Research (IMSR), Centre of Membrane Pro, University of Birmingham, Birmingham, United Kingdom
| | - Erin E Mulvihill
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Canada
| | | | - Gareth E Lim
- Department of Medicine, Université de Montréal, Montreal, Canada
| |
Collapse
|
29
|
Kozeleková A, Náplavová A, Brom T, Gašparik N, Šimek J, Houser J, Hritz J. Phosphorylated and Phosphomimicking Variants May Differ—A Case Study of 14-3-3 Protein. Front Chem 2022; 10:835733. [PMID: 35321476 PMCID: PMC8935074 DOI: 10.3389/fchem.2022.835733] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 02/14/2022] [Indexed: 12/12/2022] Open
Abstract
Protein phosphorylation is a critical mechanism that biology uses to govern cellular processes. To study the impact of phosphorylation on protein properties, a fully and specifically phosphorylated sample is required although not always achievable. Commonly, this issue is overcome by installing phosphomimicking mutations at the desired site of phosphorylation. 14-3-3 proteins are regulatory protein hubs that interact with hundreds of phosphorylated proteins and modulate their structure and activity. 14-3-3 protein function relies on its dimeric nature, which is controlled by Ser58 phosphorylation. However, incomplete Ser58 phosphorylation has obstructed the detailed study of its effect so far. In the present study, we describe the full and specific phosphorylation of 14-3-3ζ protein at Ser58 and we compare its characteristics with phosphomimicking mutants that have been used in the past (S58E/D). Our results show that in case of the 14-3-3 proteins, phosphomimicking mutations are not a sufficient replacement for phosphorylation. At physiological concentrations of 14-3-3ζ protein, the dimer-monomer equilibrium of phosphorylated protein is much more shifted towards monomers than that of the phosphomimicking mutants. The oligomeric state also influences protein properties such as thermodynamic stability and hydrophobicity. Moreover, phosphorylation changes the localization of 14-3-3ζ in HeLa and U251 human cancer cells. In summary, our study highlights that phosphomimicking mutations may not faithfully represent the effects of phosphorylation on the protein structure and function and that their use should be justified by comparing to the genuinely phosphorylated counterpart.
Collapse
Affiliation(s)
- Aneta Kozeleková
- Central European Institute of Technology, Masaryk University, Brno, Czechia
- National Centre for Biomolecular Research, Faculty of Science, Masaryk University, Brno, Czechia
| | | | - Tomáš Brom
- National Centre for Biomolecular Research, Faculty of Science, Masaryk University, Brno, Czechia
| | - Norbert Gašparik
- Central European Institute of Technology, Masaryk University, Brno, Czechia
- National Centre for Biomolecular Research, Faculty of Science, Masaryk University, Brno, Czechia
| | - Jan Šimek
- Central European Institute of Technology, Masaryk University, Brno, Czechia
| | - Josef Houser
- Central European Institute of Technology, Masaryk University, Brno, Czechia
- National Centre for Biomolecular Research, Faculty of Science, Masaryk University, Brno, Czechia
| | - Jozef Hritz
- Central European Institute of Technology, Masaryk University, Brno, Czechia
- Department of Chemistry, Faculty of Science, Masaryk University, Brno, Czechia
- *Correspondence: Jozef Hritz,
| |
Collapse
|
30
|
Greaves D, Calle Y. Epithelial Mesenchymal Transition (EMT) and Associated Invasive Adhesions in Solid and Haematological Tumours. Cells 2022; 11:649. [PMID: 35203300 PMCID: PMC8869945 DOI: 10.3390/cells11040649] [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: 01/13/2022] [Revised: 02/07/2022] [Accepted: 02/08/2022] [Indexed: 12/13/2022] Open
Abstract
In solid tumours, cancer cells that undergo epithelial mesenchymal transition (EMT) express characteristic gene expression signatures that promote invasive migration as well as the development of stemness, immunosuppression and drug/radiotherapy resistance, contributing to the formation of currently untreatable metastatic tumours. The cancer traits associated with EMT can be controlled by the signalling nodes at characteristic adhesion sites (focal contacts, invadopodia and microtentacles) where the regulation of cell migration, cell cycle progression and pro-survival signalling converge. In haematological tumours, ample evidence accumulated during the last decade indicates that the development of an EMT-like phenotype is indicative of poor disease prognosis. However, this EMT phenotype has not been directly linked to the assembly of specific forms of adhesions. In the current review we discuss the role of EMT in haematological malignancies and examine its possible link with the progression towards more invasive and aggressive forms of these tumours. We also review the known types of adhesions formed by haematological malignancies and speculate on their possible connection with the EMT phenotype. We postulate that understanding the architecture and regulation of EMT-related adhesions will lead to the discovery of new therapeutic interventions to overcome disease progression and resistance to therapies.
Collapse
Affiliation(s)
| | - Yolanda Calle
- School of Life Sciences and Health, University of Roehampton, London SW15 4JD, UK;
| |
Collapse
|
31
|
Proteomic Research on the Antitumor Properties of Medicinal Mushrooms. Molecules 2021; 26:molecules26216708. [PMID: 34771120 PMCID: PMC8588050 DOI: 10.3390/molecules26216708] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 10/27/2021] [Accepted: 11/02/2021] [Indexed: 12/22/2022] Open
Abstract
Medicinal mushrooms are increasingly being recognized as an important therapeutic modality in complementary oncology. Until now, more than 800 mushroom species have been known to possess significant pharmacological properties, of which antitumor and immunomodulatory properties have been the most researched. Besides a number of medicinal mushroom preparations being used as dietary supplements and nutraceuticals, several isolates from mushrooms have been used as official antitumor drugs in clinical settings for several decades. Various proteomic approaches allow for the identification of a large number of differentially regulated proteins serendipitously, thereby providing an important platform for a discovery of new potential therapeutic targets and approaches as well as biomarkers of malignant disease. This review is focused on the current state of proteomic research into antitumor mechanisms of some of the most researched medicinal mushroom species, including Phellinus linteus, Ganoderma lucidum, Auricularia auricula, Agrocybe aegerita, Grifola frondosa, and Lentinus edodes, as whole body extracts or various isolates, as well as of complex extract mixtures.
Collapse
|
32
|
Gęgotek A, Atalay S, Skrzydlewska E. UV induced changes in proteome of rats plasma are reversed by dermally applied cannabidiol. Sci Rep 2021; 11:20666. [PMID: 34667212 PMCID: PMC8526570 DOI: 10.1038/s41598-021-00134-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 10/07/2021] [Indexed: 11/20/2022] Open
Abstract
UV radiation is known to induce a multiple changes in the metabolism of skin-building cells, what can affect the functioning not only neighboring cells, but also, following signal transduction releasing into the blood vessels, the entire body. Therefore, the aim of this study was to analyze the proteomic disturbances occurred in plasma of chronically UVA/UVB irradiated rats and define the effect on these changes of skin topically applied cannabidiol (CBD). Obtained results showed significant changes in the expression of numerous anti-inflammatory and signaling proteins including: NFκB inhibitor, 14-3-3 protein, protein kinase C, keratin, and protein S100 after UV irradiation and CBD treatment. Moreover, the effects of UVA and UVB were manifested by increased level of lipid peroxidation products-protein adducts formation. CBD partially prevented all of these changes, but in a various degree depending on the UV radiation type. Moreover, topical treatment with CBD resulted in the penetration of CBD into the blood and, as a consequence, in direct modifications to the plasma protein structure by creating CBD adducts with molecules, such as proline-rich protein 30, transcription factor 19, or N-acetylglucosamine-6-sulfatase, what significantly changed the activity of these proteins. In conclusion, it may be suggested that CBD applied topically may be an effective compound against systemic UV-induced oxidative stress, but its effectiveness requires careful analysis of CBD's effects on other tissues of the living organism.
Collapse
Affiliation(s)
- Agnieszka Gęgotek
- Department of Analytical Chemistry, Medical University of Bialystok, Mickiewicza 2D, 15-222, Bialystok, Poland
| | - Sinemyiz Atalay
- Department of Analytical Chemistry, Medical University of Bialystok, Mickiewicza 2D, 15-222, Bialystok, Poland
| | - Elżbieta Skrzydlewska
- Department of Analytical Chemistry, Medical University of Bialystok, Mickiewicza 2D, 15-222, Bialystok, Poland.
| |
Collapse
|
33
|
Soini L, Redhead M, Westwood M, Leysen S, Davis J, Ottmann C. Identification of molecular glues of the SLP76/14-3-3 protein-protein interaction. RSC Med Chem 2021; 12:1555-1564. [PMID: 34667951 PMCID: PMC8459327 DOI: 10.1039/d1md00172h] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Accepted: 07/01/2021] [Indexed: 12/12/2022] Open
Abstract
The stabilisation of protein-protein interactions (PPIs) through molecular glues is a novel and promising approach in drug discovery. In stark contrast to research in protein-protein inhibition the field of stabilisation remains underdeveloped with comparatively few examples of small-molecule stabilisers of PPIs reported to date. At the same time identifying molecular glues has received recent sustained interest, especially in the fields of targeted protein degradation and 14-3-3 PPIs. The hub-protein 14-3-3 has a broad interactome with more than 500 known protein partners which presents a great opportunity for therapeutic intervention. In this study we have developed an HTRF assay suitable for HTS of the 14-3-3/SLP76 PPI and have completed a proof of concept screen against a chemically diverse library of 20 K molecules. The adaptor protein SLP76 has been reported to interact with 14-3-3 proteins downstream of the TCR playing an important role in mediating its own proteasomal degradation. We believe that stabilisation of this PPI could be exploited to potentiate degradation of SLP76 and therefore inhibit TCR signalling. This would represent an interesting alternative to other approaches in the field of targeted protein degradation. Here we disclose 16 novel stabilisers of the 14-3-3/SLP76 PPI across multiple different chemotypes. Based on the early results presented here we would recommend this approach to find molecular glues with broad applicability in the field of 14-3-3 PPIs.
Collapse
Affiliation(s)
- Lorenzo Soini
- Laboratory of Chemical Biology, Department of Biomedical Engineering and Institute for Complex Molecular Systems, Eindhoven University of Technology Eindhoven The Netherlands .,Department of Chemistry, UCB Celltech Slough UK
| | - Martin Redhead
- Exscientia Ltd, Schrodinger Building, Oxford Science Park Oxford OX44GE UK
| | - Marta Westwood
- Structural Biology, Discovery, Charles River, Chesterford Research Park UK
| | - Seppe Leysen
- Department of Structural Biology and Biophysics, UCB Celltech Slough UK
| | | | - Christian Ottmann
- Laboratory of Chemical Biology, Department of Biomedical Engineering and Institute for Complex Molecular Systems, Eindhoven University of Technology Eindhoven The Netherlands
| |
Collapse
|
34
|
Tivon B, Gabizon R, Somsen BA, Cossar PJ, Ottmann C, London N. Covalent flexible peptide docking in Rosetta. Chem Sci 2021; 12:10836-10847. [PMID: 34476063 PMCID: PMC8372624 DOI: 10.1039/d1sc02322e] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Accepted: 07/09/2021] [Indexed: 11/21/2022] Open
Abstract
Electrophilic peptides that form an irreversible covalent bond with their target have great potential for binding targets that have been previously considered undruggable. However, the discovery of such peptides remains a challenge. Here, we present Rosetta CovPepDock, a computational pipeline for peptide docking that incorporates covalent binding between the peptide and a receptor cysteine. We applied CovPepDock retrospectively to a dataset of 115 disulfide-bound peptides and a dataset of 54 electrophilic peptides. It produced a top-five scoring, near-native model, in 89% and 100% of the cases when docking from the native conformation, and 20% and 90% when docking from an extended peptide conformation, respectively. In addition, we developed a protocol for designing electrophilic peptide binders based on known non-covalent binders or protein-protein interfaces. We identified 7154 peptide candidates in the PDB for application of this protocol. As a proof-of-concept we validated the protocol on the non-covalent complex of 14-3-3σ and YAP1 phosphopeptide. The protocol identified seven highly potent and selective irreversible peptide binders. The predicted binding mode of one of the peptides was validated using X-ray crystallography. This case-study demonstrates the utility and impact of CovPepDock. It suggests that many new electrophilic peptide binders can be rapidly discovered, with significant potential as therapeutic molecules and chemical probes.
Collapse
Affiliation(s)
- Barr Tivon
- Department of Chemical and Structural Biology, The Weizmann Institute of Science Rehovot 7610001 Israel
| | - Ronen Gabizon
- Department of Chemical and Structural Biology, The Weizmann Institute of Science Rehovot 7610001 Israel
| | - Bente A Somsen
- Laboratory of Chemical Biology, Department of Biomedical Engineering, Institute for Complex Molecular Systems, Eindhoven University of Technology P.O. Box 513 5600MB Eindhoven The Netherlands
| | - Peter J Cossar
- Laboratory of Chemical Biology, Department of Biomedical Engineering, Institute for Complex Molecular Systems, Eindhoven University of Technology P.O. Box 513 5600MB Eindhoven The Netherlands
| | - Christian Ottmann
- Laboratory of Chemical Biology, Department of Biomedical Engineering, Institute for Complex Molecular Systems, Eindhoven University of Technology P.O. Box 513 5600MB Eindhoven The Netherlands
| | - Nir London
- Department of Chemical and Structural Biology, The Weizmann Institute of Science Rehovot 7610001 Israel
| |
Collapse
|
35
|
Soini L, Leysen S, Crabbe T, Davis J, Ottmann C. The identification and structural analysis of potential 14-3-3 interaction sites on the bone regulator protein Schnurri-3. Acta Crystallogr F Struct Biol Commun 2021; 77:254-261. [PMID: 34341191 PMCID: PMC8329713 DOI: 10.1107/s2053230x21006658] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Accepted: 06/25/2021] [Indexed: 11/10/2022] Open
Abstract
14-3-3 proteins regulate many intracellular processes and their ability to bind in subtly different fashions to their numerous partner proteins provides attractive drug-targeting points for a range of diseases. Schnurri-3 is a suppressor of mouse bone formation and a candidate target for novel osteoporosis therapeutics, and thus it is of interest to determine whether it interacts with 14-3-3. In this work, potential 14-3-3 interaction sites on mammalian Schnurri-3 were identified by an in silico analysis of its protein sequence. Using fluorescence polarization, isothermal titration calorimetry and X-ray crystallography, it is shown that synthetic peptides containing either phosphorylated Thr869 or Ser542 can indeed interact with 14-3-3, with the latter capable of forming an interprotein disulfide bond with 14-3-3σ: a hitherto unreported phenomenon.
Collapse
Affiliation(s)
- Lorenzo Soini
- Laboratory of Chemical Biology, Department of Biomedical Engineering and Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, The Netherlands
- Department of Chemistry, UCB Celltech, Slough, United Kingdom
| | - Seppe Leysen
- Department of Structural Biology and Biophysics, UCB Celltech, Slough, United Kingdom
| | - Tom Crabbe
- New Targets, UCB Celltech, Slough, United Kingdom
| | - Jeremy Davis
- Department of Chemistry, UCB Celltech, Slough, United Kingdom
| | - Christian Ottmann
- Laboratory of Chemical Biology, Department of Biomedical Engineering and Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, The Netherlands
| |
Collapse
|
36
|
Garcia BCB, Horie M, Kojima S, Makino A, Tomonaga K. BUD23-TRMT112 interacts with the L protein of Borna disease virus and mediates the chromosomal tethering of viral ribonucleoproteins. Microbiol Immunol 2021; 65:492-504. [PMID: 34324219 DOI: 10.1111/1348-0421.12934] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 07/05/2021] [Accepted: 07/17/2021] [Indexed: 11/28/2022]
Abstract
Persistent intranuclear infection is an uncommon infection strategy among RNA viruses. However, Borna disease virus 1 (BoDV-1), a nonsegmented, negative-strand RNA virus, maintains viral infection in the cell nucleus by forming structured aggregates of viral ribonucleoproteins (vRNPs), and by tethering these vRNPs onto the host chromosomes. To better understand the nuclear infection strategy of BoDV-1, we determined the host protein interactors of the BoDV-1 large (L) protein. By proximity-dependent biotinylation, we identified several nuclear host proteins interacting with BoDV-1 L, one of which is TRMT112, a partner of several RNA methyltransferases (MTase). TRMT112 binds with BoDV-1 L at the RNA-dependent RNA polymerase domain, together with BUD23, an 18S rRNA MTase and 40S ribosomal maturation factor. We then discovered that BUD23-TRMT112 mediates the chromosomal tethering of BoDV-1 vRNPs, and that the MTase activity is necessary in the tethering process. These findings provide us a better understanding on how nuclear host proteins assist the chromosomal tethering of BoDV-1, as well as new prospects of host-viral interactions for intranuclear infection strategy of orthobornaviruses. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Bea Clarise B Garcia
- Laboratory of RNA Viruses, Institute for Frontier Life and Medical Sciences (inFRONT), Kyoto University, Kyoto.,Laboratory of RNA Viruses, Department of Mammalian Regulatory Network, Graduate School of Biostudies, Kyoto University, Kyoto
| | - Masayuki Horie
- Laboratory of RNA Viruses, Institute for Frontier Life and Medical Sciences (inFRONT), Kyoto University, Kyoto.,Hakubi Center for Advanced Research, Kyoto University, Kyoto
| | - Shohei Kojima
- Laboratory of RNA Viruses, Institute for Frontier Life and Medical Sciences (inFRONT), Kyoto University, Kyoto
| | - Akiko Makino
- Laboratory of RNA Viruses, Institute for Frontier Life and Medical Sciences (inFRONT), Kyoto University, Kyoto.,Laboratory of RNA Viruses, Department of Mammalian Regulatory Network, Graduate School of Biostudies, Kyoto University, Kyoto
| | - Keizo Tomonaga
- Laboratory of RNA Viruses, Institute for Frontier Life and Medical Sciences (inFRONT), Kyoto University, Kyoto.,Laboratory of RNA Viruses, Department of Mammalian Regulatory Network, Graduate School of Biostudies, Kyoto University, Kyoto.,Department of Molecular Virology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| |
Collapse
|
37
|
Lim Y, Ku NO. Revealing the Roles of Keratin 8/18-Associated Signaling Proteins Involved in the Development of Hepatocellular Carcinoma. Int J Mol Sci 2021; 22:6401. [PMID: 34203895 PMCID: PMC8232640 DOI: 10.3390/ijms22126401] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 06/08/2021] [Accepted: 06/11/2021] [Indexed: 02/08/2023] Open
Abstract
Although hepatocellular carcinoma (HCC) is developed with various etiologies, protection of hepatocytes seems basically essential to prevent the incidence of HCC. Keratin 8 and keratin 18 (K8/K18) are cytoskeletal intermediate filament proteins that are expressed in hepatocytes. They maintain the cell shape and protect cells under stress conditions. Their protective roles in liver damage have been described in studies of mouse models, and K8/K18 mutation frequency in liver patients. Interestingly, K8/K18 bind to signaling proteins such as transcription factors and protein kinases involved in HCC development. Since K8/K18 are abundant cytoskeletal proteins, K8/K18 binding with the signaling factors can alter the availability of the factors. Herein, we discuss the potential roles of K8/K18 in HCC development.
Collapse
Affiliation(s)
- Younglan Lim
- Interdisciplinary Program of Integrated OMICS for Biomedical Sciences, Yonsei University, Seoul 03722, Korea;
| | - Nam-On Ku
- Interdisciplinary Program of Integrated OMICS for Biomedical Sciences, Yonsei University, Seoul 03722, Korea;
- Department of Bio-Convergence ISED, Underwood International College, Yonsei University, Seoul 03722, Korea
| |
Collapse
|
38
|
Cao Y, Gao A, Li X, Min H, He C, Sun X, Ding WQ, Zhou J. Elevated TAB182 enhances the radioresistance of esophageal squamous cell carcinoma through G2-M checkpoint modulation. Cancer Med 2021; 10:3101-3112. [PMID: 33787085 PMCID: PMC8085956 DOI: 10.1002/cam4.3879] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Revised: 03/01/2021] [Accepted: 03/16/2021] [Indexed: 11/09/2022] Open
Abstract
Background Radiotherapy is one of the main strategies for the treatment of esophageal squamous cell carcinoma (ESCC). However, treatment failure often occurs due to the emergence of radioresistance. In this study, we report a key regulator of radiation sensitivity, termed TAB182 that may become an ideal biomarker and therapeutic target to overcome radioresistance. Materials and Methods By applying qRT‐PCR and immunohistochemical staining, the expression of TAB182 was detected in patient tissues. We next assessed the influence of TAB182 downregulation to radiosensitivity using clonogenic survival assay and γ‐H2A.X foci analysis in TE‐1, TE‐10, and radioresistant TE‐1R cell lines after ionizing radiation. To unveil the mechanism underlying, TAB182 interacting proteins were identified by mass spectrometry following co‐immunoprecipitation. Furthermore, flow cytometry and western blot assay were applied to validate the identified proteins. Results Our results demonstrated that the expression of TAB182 is higher in cancer tissues than normal tissues and elevated expression of TAB182 correlates with poor outcomes of postoperative radiotherapy. Downregulation of TAB182 sensitized cancer cells to ionizing radiation, particularly in radioresistant TE‐1R cells that spontaneously overexpress TAB182. Mechanically, TAB182 interacts with FHL2 to induce G2‐M arrest through wiring the CHK2/CDC25C/CDC2 signaling pathway. Finally, overexpression of shRNA‐resistant TAB182 restored the checkpoint and radioresistance. Conclusion TAB182 potentiates the radioresistance of ESCC cells by modulating the G2‐M checkpoint through its interaction with FHL2. Thus, TAB182 may become an ideal biomarker and therapeutic target of ESCC radiotherapy.
Collapse
Affiliation(s)
- Yuandong Cao
- Department of Radiation Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, P.R. China
| | - Aidi Gao
- Suzhou Cancer Center Core Laboratory, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, Jiangsu, P.R. China
| | - Xiaoqing Li
- Suzhou Cancer Center Core Laboratory, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, Jiangsu, P.R. China
| | - Han Min
- Suzhou Cancer Center Core Laboratory, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, Jiangsu, P.R. China
| | - Chao He
- Suzhou Cancer Center Core Laboratory, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, Jiangsu, P.R. China
| | - Xinchen Sun
- Department of Radiation Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, P.R. China
| | - Wei-Qun Ding
- Department of Pathology, University of Oklahoma Health Science Center, Oklahoma City, OK, USA
| | - Jundong Zhou
- Suzhou Cancer Center Core Laboratory, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, Jiangsu, P.R. China
| |
Collapse
|
39
|
Shao W, Chen W, Zhu X, Zhou X, Jin Y, Zhan C, Liu G, Liu X, Ma D, Qiao Y. Genome-Wide Identification and Characterization of Wheat 14-3-3 Genes Unravels the Role of TaGRF6-A in Salt Stress Tolerance by Binding MYB Transcription Factor. Int J Mol Sci 2021; 22:ijms22041904. [PMID: 33673010 PMCID: PMC7918857 DOI: 10.3390/ijms22041904] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 02/09/2021] [Accepted: 02/09/2021] [Indexed: 11/16/2022] Open
Abstract
14-3-3 proteins are a large multigenic family of general regulatory factors (GRF) ubiquitously found in eukaryotes and play vital roles in the regulation of plant growth, development, and response to stress stimuli. However, so far, no comprehensive investigation has been performed in the hexaploid wheat. In the present study, A total of 17 potential 14-3-3 gene family members were identified from the Chinese Spring whole-genome sequencing database. The phylogenetic comparison with six 14-3-3 families revealed that the majority of wheat 14-3-3 genes might have evolved as an independent branch and grouped into ε and non-ε group using the phylogenetic comparison. Analysis of gene structure and motif indicated that 14-3-3 protein family members have relatively conserved exon/intron arrangement and motif composition. Physical mapping showed that wheat 14-3-3 genes are mainly distributed on chromosomes 2, 3, 4, and 7. Moreover, most 14-3-3 members in wheat exhibited significantly down-regulated expression in response to alkaline stress. VIGS assay and protein-protein interaction analysis further confirmed that TaGRF6-A positively regulated slat stress tolerance by interacting with a MYB transcription factor, TaMYB64. Taken together, our findings provide fundamental information on the involvement of the wheat 14-3-3 family in salt stress and further investigating their molecular mechanism.
Collapse
Affiliation(s)
- Wenna Shao
- Engineering Research Center of Ecology and Agricultural Use of Wetland, Ministry of Education, Hubei Collaborative Innovation Center for Grain Industry, College of Agriculture, Yangtze University, Jingzhou 434000, China; (W.S.); (X.Z.); (Y.J.); (C.Z.); (G.L.); (X.L.)
- Shanghai Key Laboratory of Plant Molecular Sciences, College of Life Sciences, Shanghai Normal University, Shanghai 200234, China;
| | - Wang Chen
- Oil Crops Research Institute, Chinese Academy of Agricultural Sciences, Wuhan 430062, China;
| | - Xiaoguo Zhu
- Shanghai Key Laboratory of Plant Molecular Sciences, College of Life Sciences, Shanghai Normal University, Shanghai 200234, China;
| | - Xiaoyi Zhou
- Engineering Research Center of Ecology and Agricultural Use of Wetland, Ministry of Education, Hubei Collaborative Innovation Center for Grain Industry, College of Agriculture, Yangtze University, Jingzhou 434000, China; (W.S.); (X.Z.); (Y.J.); (C.Z.); (G.L.); (X.L.)
- Shanghai Key Laboratory of Plant Molecular Sciences, College of Life Sciences, Shanghai Normal University, Shanghai 200234, China;
| | - Yingying Jin
- Engineering Research Center of Ecology and Agricultural Use of Wetland, Ministry of Education, Hubei Collaborative Innovation Center for Grain Industry, College of Agriculture, Yangtze University, Jingzhou 434000, China; (W.S.); (X.Z.); (Y.J.); (C.Z.); (G.L.); (X.L.)
| | - Chuang Zhan
- Engineering Research Center of Ecology and Agricultural Use of Wetland, Ministry of Education, Hubei Collaborative Innovation Center for Grain Industry, College of Agriculture, Yangtze University, Jingzhou 434000, China; (W.S.); (X.Z.); (Y.J.); (C.Z.); (G.L.); (X.L.)
| | - Gensen Liu
- Engineering Research Center of Ecology and Agricultural Use of Wetland, Ministry of Education, Hubei Collaborative Innovation Center for Grain Industry, College of Agriculture, Yangtze University, Jingzhou 434000, China; (W.S.); (X.Z.); (Y.J.); (C.Z.); (G.L.); (X.L.)
| | - Xi Liu
- Engineering Research Center of Ecology and Agricultural Use of Wetland, Ministry of Education, Hubei Collaborative Innovation Center for Grain Industry, College of Agriculture, Yangtze University, Jingzhou 434000, China; (W.S.); (X.Z.); (Y.J.); (C.Z.); (G.L.); (X.L.)
| | - Dongfang Ma
- Engineering Research Center of Ecology and Agricultural Use of Wetland, Ministry of Education, Hubei Collaborative Innovation Center for Grain Industry, College of Agriculture, Yangtze University, Jingzhou 434000, China; (W.S.); (X.Z.); (Y.J.); (C.Z.); (G.L.); (X.L.)
- Correspondence: (D.M.); (Y.Q.)
| | - Yongli Qiao
- Shanghai Key Laboratory of Plant Molecular Sciences, College of Life Sciences, Shanghai Normal University, Shanghai 200234, China;
- Correspondence: (D.M.); (Y.Q.)
| |
Collapse
|
40
|
Sanchez DJD, Vasconcelos FR, Teles-Filho ACA, Viana AGA, Martins AMA, Sousa MV, Castro MS, Ricart CA, Fontes W, Bertolini M, Bustamante-Filho IC, Moura AA. Proteomic profile of pre-implantational ovine embryos produced in vivo. Reprod Domest Anim 2021; 56:586-603. [PMID: 33460477 DOI: 10.1111/rda.13897] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Revised: 01/12/2021] [Accepted: 01/14/2021] [Indexed: 12/11/2022]
Abstract
The present study was conducted to decipher the proteome of in vivo-produced pre-implantation ovine embryos. Ten locally adapted Morana Nova ewes received hormonal treatment and were inseminated 12 hr after ovulation. Six days later, 54 embryos (morula and blastocyst developmental state) were recovered from eight ewes and pooled to obtain sufficient protein for proteomic analysis. Extracted embryo proteins were analysed by LC-MS/MS, followed by identification based on four database searches (PEAKS, Proteome Discoverer software, SearchGUI software, PepExplorer). Identified proteins were analysed for gene ontology terms, protein clusters and interactions. Genes associated with the ovine embryo proteome were screened for miRNA targets using data sets of TargetScan (http://www.targetscan.org) and mIRBase (http://www.mirbase.org) servers. There were 667 proteins identified in the ovine embryos. Biological processes of such proteins were mainly related to cellular process and regulation, and molecular functions, to binding and catalytic activity. Analysis of the embryo proteins revealed 49 enriched functional clusters, linked to energy metabolism (TCA cycle, pyruvate and glycolysis metabolism), zona pellucida (ZP), MAPK signalling pathway, tight junction, binding of sperm to ZP, translation, proteasome, cell cycle and calcium/phospholipid binding. Sixteen miRNAs were related to 25 pre-implantation ovine embryo genes, all conserved in human, bovine and ovine species. The interaction network generated by miRNet showed four key miRNAs (hsa-mir-106b-5p; hsa-mir-30-5p; hsa-mir-103a-5p and hsa-mir-106a-5p) with potential interactions with embryo-expressed genes. Functional analysis of the network indicated that miRNAs modulate genes related to cell cycle, regulation of stem cell and embryonic cell differentiation, among others. Retrieved miRNAs also modulate the expression of genes involved in cell signalling pathways, such as MAPK, Wnt, TGF-beta, p53 and Toll-like receptor. The current study describes the first major proteomic profile of 6-day-old ovine embryos produced in vivo, setting a comprehensive foundation for our understanding of embryo physiology in the ovine species.
Collapse
Affiliation(s)
- Deisy J D Sanchez
- Department of Animal Science, Federal University of Ceará, Fortaleza, Brazil
| | - Fabio R Vasconcelos
- Department of Animal Science, Federal University of Ceará, Fortaleza, Brazil
| | | | - Arabela G A Viana
- Department of Animal Science, Federal University of Ceará, Fortaleza, Brazil
| | - Aline M A Martins
- Laboratory of Protein Chemistry and Biochemistry, University of Brasília, Brasília, Brazil
| | - Marcelo V Sousa
- Laboratory of Protein Chemistry and Biochemistry, University of Brasília, Brasília, Brazil
| | - Mariana S Castro
- Laboratory of Protein Chemistry and Biochemistry, University of Brasília, Brasília, Brazil
| | - Carlos A Ricart
- Laboratory of Protein Chemistry and Biochemistry, University of Brasília, Brasília, Brazil
| | - Wagner Fontes
- Laboratory of Protein Chemistry and Biochemistry, University of Brasília, Brasília, Brazil
| | - Marcelo Bertolini
- The School of Veterinay Medicine, Federal University of Rio Grande do Sul, Porto Alegre, Brazil
| | | | - Arlindo A Moura
- Department of Animal Science, Federal University of Ceará, Fortaleza, Brazil
| |
Collapse
|
41
|
Zuo X, Wang S, Xiang W, Yang H, Tahir MM, Zheng S, An N, Han M, Zhao C, Zhang D. Genome-wide identification of the 14-3-3 gene family and its participation in floral transition by interacting with TFL1/FT in apple. BMC Genomics 2021; 22:41. [PMID: 33419402 PMCID: PMC7796649 DOI: 10.1186/s12864-020-07330-2] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Accepted: 12/15/2020] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND Apple (Malus domestica Borkh.) is a popular cultivated fruit crop with high economic value in China. Apple floral transition is an important process but liable to be affected by various environmental factors. The 14-3-3 proteins are involved in regulating diverse biological processes in plants, and some 14-3-3 members play vital roles in flowering. However, little information was available about the 14-3-3 members in apple. RESULTS In the current study, we identified eighteen 14-3-3 gene family members from the apple genome database, designated MdGF14a to MdGF14r. The isoforms possess a conserved core region comprising nine antiparallel α-helices and divergent N and C termini. According to their structural and phylogenetic features, Md14-3-3 proteins could be classified into two major evolutionary branches, the epsilon (ɛ) group and the non-epsilon (non-ɛ) group. Moreover, expression profiles derived from transcriptome data and quantitative real-time reverse transcription PCR analysis showed diverse expression patterns of Md14-3-3 genes in various tissues and in response to different sugars and hormone treatments during the floral transition phase. Four Md14-3-3 isoforms (MdGF14a, MdGF14d, MdGF14i, and MdGF14j) exhibiting prominent transcriptional responses to sugars and hormones were selected for further investigation. Furthermore, yeast two-hybrid and bimolecular fluorescence complementation experiments showed that the four Md14-3-3 proteins interact with key floral integrators, MdTFL1 (TERMINAL FLOWER1) and MdFT (FLOWERING LOCUS T). Subcellular localization of four selected Md14-3-3 proteins demonstrated their localization in both the cytoplasm and nucleus. CONCLUSION We identified the Md14-3-3 s family in apple comprehensively. Certain Md14-3-3 genes are expressed predominantly during the apple floral transition stage, and may participate in the regulation of flowering through association with flower control genes. Our results provide a preliminary framework for further investigation into the roles of Md14-3-3 s in floral transition.
Collapse
Affiliation(s)
- Xiya Zuo
- College of Horticulture, Northwest A & F University, Yangling, 712100, China
| | - Shixiang Wang
- College of Horticulture, Northwest A & F University, Yangling, 712100, China
| | - Wen Xiang
- College of Horticulture, Northwest A & F University, Yangling, 712100, China
| | - Huiru Yang
- College of Horticulture, Northwest A & F University, Yangling, 712100, China
| | | | - Shangong Zheng
- College of Horticulture, Northwest A & F University, Yangling, 712100, China
| | - Na An
- College of Life Sciences, Northwest A & F University, Yangling, 712100, China
| | - Mingyu Han
- College of Horticulture, Northwest A & F University, Yangling, 712100, China
| | - Caiping Zhao
- College of Horticulture, Northwest A & F University, Yangling, 712100, China
| | - Dong Zhang
- College of Horticulture, Northwest A & F University, Yangling, 712100, China.
| |
Collapse
|
42
|
Abstract
14-3-3 proteins are mostly expressed in the brain and are closely involved in numerous brain functions and various brain disorders. Among the isotypes of the 14-3-3 proteins, 14-3-3γ is mainly expressed in neurons and is highly produced during brain development, which could indicate that it has a significance in neural development. Furthermore, the distinctive levels of temporally and locally regulated 14-3-3γ expression in various brain disorders suggest that it could play a substantial role in brain plasticity of the diseased states. In this review, we introduce the various brain disorders reported to be involved with 14-3-3γ, and summarize the changes of 14-3-3γ expression in each brain disease. We also discuss the potential of 14-3-3γ for treatment and the importance of research on specific 14-3-3 isotypes for an effective therapeutic approach.
Collapse
Affiliation(s)
- Eunsil Cho
- School of Biosystem and Biomedical Science, College of Health Science, Korea University, Seoul 02708, Korea
| | - Jae-Yong Park
- School of Biosystem and Biomedical Science, College of Health Science, Korea University, Seoul 02708, Korea
| |
Collapse
|
43
|
Farhadi P, Yarani R, Dokaneheifard S, Mansouri K. The emerging role of targeting cancer metabolism for cancer therapy. Tumour Biol 2020; 42:1010428320965284. [PMID: 33028168 DOI: 10.1177/1010428320965284] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Glucose, as the main consuming nutrient of the body, faces different destinies in cancer cells. Glycolysis, oxidative phosphorylation, and pentose phosphate pathways produce different glucose-derived metabolites and thus affect cells' bioenergetics differently. Tumor cells' dependency to aerobic glycolysis and other cancer-specific metabolism changes are known as the cancer hallmarks, distinct cancer cells from normal cells. Therefore, these tumor-specific characteristics receive the limelight as targets for cancer therapy. Glutamine, serine, and fatty acid oxidation together with 5-lipoxygenase are main pathways that have attracted lots of attention for cancer therapy. In this review, we not only discuss different tumor metabolism aspects but also discuss the metabolism roles in the promotion of cancer cells at different stages and their difference with normal cells. Besides, we dissect the inhibitors potential in blocking the main metabolic pathways to introduce the effective and non-effective inhibitors in the field.
Collapse
Affiliation(s)
- Pegah Farhadi
- Medical Biology Research Center, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Reza Yarani
- Translational Type 1 Diabetes Research, Department of Clinical Research, Steno Diabetes Center Copenhagen, Gentofte, Denmark
| | - Sadat Dokaneheifard
- Sylvester Comprehensive Cancer Center, Department of Human Genetics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Kamran Mansouri
- Medical Biology Research Center, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran.,Department of Molecular Medicine, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| |
Collapse
|
44
|
Characterization of the expression and prognostic value of 14-3-3 isoforms in breast cancer. Aging (Albany NY) 2020; 12:19597-19617. [PMID: 33052135 PMCID: PMC7732302 DOI: 10.18632/aging.103919] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2020] [Accepted: 07/25/2020] [Indexed: 01/24/2023]
Abstract
The tyrosine 3-monooxygenase/tryptophan 5-monooxygenase activation proteins (14-3-3) participate in the tumorigenesis and progression of numerous malignances, but their precise prognostic values in breast cancer (BrCa) remain unknown. Here, we investigated the expression profiles and prognostic roles of 14-3-3 isoforms by employing multiple online databases. The transcriptional levels of most 14-3-3 isoforms in BrCa tissues were significantly higher than those in normal tissues. High mRNA expression of 14-3-3 beta/sigma/theta/zeta was significantly associated with poor overall survival (OS) in BrCa patients, while high mRNA expression of 14-3-3 epsilon was notably related to favorable OS. High mRNA expression of 14-3-3 beta/gamma/sigma/theta/zeta was significantly associated with poor relapse-free survival (RFS) in BrCa patients. A high mutation rate of 14-3-3 was determined to be associated with poor clinical outcomes. In addition, 14-3-3 expression was correlated with the infiltration of specific immune cells types. Analysis of the breast-specific protein-protein interaction (PPI) network suggested that 14-3-3 proteins were involved in several potential oncogenic mechanisms in BrCa. Finally, we performed experimentally validated their oncogenic roles in BrCa. Overall, our findings systematically elucidate the expression and distinct prognostic value of 14-3-3 isoforms in BrCa, which may provide potential therapeutic targets and prognostic biomarkers for BrCa.
Collapse
|
45
|
Lu H, Zhou Q, He J, Jiang Z, Peng C, Tong R, Shi J. Recent advances in the development of protein-protein interactions modulators: mechanisms and clinical trials. Signal Transduct Target Ther 2020; 5:213. [PMID: 32968059 PMCID: PMC7511340 DOI: 10.1038/s41392-020-00315-3] [Citation(s) in RCA: 461] [Impact Index Per Article: 92.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 07/15/2020] [Accepted: 07/23/2020] [Indexed: 02/05/2023] Open
Abstract
Protein-protein interactions (PPIs) have pivotal roles in life processes. The studies showed that aberrant PPIs are associated with various diseases, including cancer, infectious diseases, and neurodegenerative diseases. Therefore, targeting PPIs is a direction in treating diseases and an essential strategy for the development of new drugs. In the past few decades, the modulation of PPIs has been recognized as one of the most challenging drug discovery tasks. In recent years, some PPIs modulators have entered clinical studies, some of which been approved for marketing, indicating that the modulators targeting PPIs have broad prospects. Here, we summarize the recent advances in PPIs modulators, including small molecules, peptides, and antibodies, hoping to provide some guidance to the design of novel drugs targeting PPIs in the future.
Collapse
Affiliation(s)
- Haiying Lu
- Personalized Drug Therapy Key Laboratory of Sichuan Province, Department of Pharmacy, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, 610072, Chengdu, China
| | - Qiaodan Zhou
- Department of Ultrasonic, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, 610072, Chengdu, China
| | - Jun He
- Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, 610041, Sichuan, China
| | - Zhongliang Jiang
- Miller School of Medicine, University of Miami, Miami, FL, 33136, USA
| | - Cheng Peng
- The Ministry of Education Key Laboratory of Standardization of Chinese Herbal Medicines of Ministry, State Key Laboratory Breeding Base of Systematic Research, Development and Utilization of Chinese Medicine Resources, Pharmacy College, Chengdu University of Traditional Chinese Medicine, 611137, Chengdu, China.
| | - Rongsheng Tong
- Personalized Drug Therapy Key Laboratory of Sichuan Province, Department of Pharmacy, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, 610072, Chengdu, China.
| | - Jianyou Shi
- Personalized Drug Therapy Key Laboratory of Sichuan Province, Department of Pharmacy, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, 610072, Chengdu, China.
| |
Collapse
|
46
|
14-3-3 σ: A potential biomolecule for cancer therapy. Clin Chim Acta 2020; 511:50-58. [PMID: 32950519 DOI: 10.1016/j.cca.2020.09.009] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 09/04/2020] [Accepted: 09/09/2020] [Indexed: 12/22/2022]
Abstract
As more studies have focused on the function of 14-3-3 proteins, their role in tumor progression has gradually improved. In the 14-3-3 protein family, 14-3-3σ is the protein that is most associated with tumor occurrence and development. In some malignancies, 14-3-3σ acts as a tumor suppressor via p53 and tumor suppressor genes. In most tumors, 14-3-3σ overexpression increases resistance to chemotherapy and radiotherapy and mediates the G2-M checkpoint after DNA damage. Although 14-3-3σ overexpression has been closely associated with poorer prognosis in pancreatic, gastric and colorectal cancer, its role in gallbladder and nasopharyngeal cancer remains less clear. As such, the function of 14-3-3σ in specific cancer types needs to be further clarified. It has been hypothesized that a role may be related to its molecular chaperone function combined with various protein ligands. In this review, we examine the role of 14-3-3σ in tumor development and drug resistance. We discuss the potential of targeting 14-3-3σ regulators in cancer therapy and treatment.
Collapse
|
47
|
MicroRNAomic Transcriptomic Analysis Reveal Deregulation of Clustered Cellular Functions in Human Mesenchymal Stem Cells During in Vitro Passaging. Stem Cell Rev Rep 2020; 16:222-238. [PMID: 31848878 DOI: 10.1007/s12015-019-09924-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Clinical trials using human mesenchymal stem/stromal cells (hMSCs) for cell replacement therapy showed varied outcomes, where cells' efficacy has been perceived as the limiting factor. In particular, the quality and number of the expanded cells in vitro. In this study, we aimed to determine molecular signatures of hMSCs derived from the pulp of extracted deciduous teeth (SHED) and Wharton's jelly (WJSCs) that associated with cellular ageing during in vitro passaging. We observed distinct phenotypic changes resembling proliferation reduction, cell enlargement, an increase cell population in G2/M phase, and differentially expressed of tumor suppressor p53 in passage (P) 6 as compared to P3, which indicating in vitro cell senescence. The subsequent molecular analysis showed a set of diverse differentially expressed miRNAs and mRNAs involved in maintaining cell proliferation and stemness properties. Considering the signaling pathway related to G2/M DNA damage regulation is widely recognized as part of anti-proliferation mechanism controlled by p53, we explored possible miRNA-mRNA interaction in this regulatory pathway based on genomic coordinates retrieved from miRanda. Our work reveals the potential reason for SHED underwent proliferation arrest due to the direct impinge on the expression of CKS1 by miRNAs specifically miR-22 and miR-485-5p which lead to down regulation of CDK1 and Cyclin B. It is intended that our study will contribute to the understanding of these miRNA/mRNA driving the biological process and regulating different stages of cell cycle is beneficial in developing effective rejuvenation strategies in order to obtain quality stem cells for transplantation.
Collapse
|
48
|
Torrico B, Antón-Galindo E, Fernàndez-Castillo N, Rojo-Francàs E, Ghorbani S, Pineda-Cirera L, Hervás A, Rueda I, Moreno E, Fullerton JM, Casadó V, Buitelaar JK, Rommelse N, Franke B, Reif A, Chiocchetti AG, Freitag C, Kleppe R, Haavik J, Toma C, Cormand B. Involvement of the 14-3-3 Gene Family in Autism Spectrum Disorder and Schizophrenia: Genetics, Transcriptomics and Functional Analyses. J Clin Med 2020; 9:E1851. [PMID: 32545830 PMCID: PMC7356291 DOI: 10.3390/jcm9061851] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 06/09/2020] [Accepted: 06/10/2020] [Indexed: 12/13/2022] Open
Abstract
The 14-3-3 protein family are molecular chaperones involved in several biological functions and neurological diseases. We previously pinpointed YWHAZ (encoding 14-3-3ζ) as a candidate gene for autism spectrum disorder (ASD) through a whole-exome sequencing study, which identified a frameshift variant within the gene (c.659-660insT, p.L220Ffs*18). Here, we explored the contribution of the seven human 14-3-3 family members in ASD and other psychiatric disorders by investigating the: (i) functional impact of the 14-3-3ζ mutation p.L220Ffs*18 by assessing solubility, target binding and dimerization; (ii) contribution of common risk variants in 14-3-3 genes to ASD and additional psychiatric disorders; (iii) burden of rare variants in ASD and schizophrenia; and iv) 14-3-3 gene expression using ASD and schizophrenia transcriptomic data. We found that the mutant 14-3-3ζ protein had decreased solubility and lost its ability to form heterodimers and bind to its target tyrosine hydroxylase. Gene-based analyses using publicly available datasets revealed that common variants in YWHAE contribute to schizophrenia (p = 6.6 × 10-7), whereas ultra-rare variants were found enriched in ASD across the 14-3-3 genes (p = 0.017) and in schizophrenia for YWHAZ (meta-p = 0.017). Furthermore, expression of 14-3-3 genes was altered in post-mortem brains of ASD and schizophrenia patients. Our study supports a role for the 14-3-3 family in ASD and schizophrenia.
Collapse
Affiliation(s)
- Bàrbara Torrico
- Departament de Genètica, Microbiologia i Estadística, Facultat de Biologia, Universitat de Barcelona, Prevosti Building, floor 2, Av. Diagonal 643, 08028 Barcelona, Spain; (B.T.); (E.A.-G.); (N.F.-C.); (E.R.-F.); (L.P.-C.)
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Institut de Biomedicina de la Universitat de Barcelona (IBUB), 08028 Barcelona, Spain; (E.M.); (V.C.)
- Institut de Recerca Sant Joan de Déu (IR-SJD), 08950 Esplugues de Llobregat, Spain
| | - Ester Antón-Galindo
- Departament de Genètica, Microbiologia i Estadística, Facultat de Biologia, Universitat de Barcelona, Prevosti Building, floor 2, Av. Diagonal 643, 08028 Barcelona, Spain; (B.T.); (E.A.-G.); (N.F.-C.); (E.R.-F.); (L.P.-C.)
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Institut de Biomedicina de la Universitat de Barcelona (IBUB), 08028 Barcelona, Spain; (E.M.); (V.C.)
- Institut de Recerca Sant Joan de Déu (IR-SJD), 08950 Esplugues de Llobregat, Spain
| | - Noèlia Fernàndez-Castillo
- Departament de Genètica, Microbiologia i Estadística, Facultat de Biologia, Universitat de Barcelona, Prevosti Building, floor 2, Av. Diagonal 643, 08028 Barcelona, Spain; (B.T.); (E.A.-G.); (N.F.-C.); (E.R.-F.); (L.P.-C.)
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Institut de Biomedicina de la Universitat de Barcelona (IBUB), 08028 Barcelona, Spain; (E.M.); (V.C.)
- Institut de Recerca Sant Joan de Déu (IR-SJD), 08950 Esplugues de Llobregat, Spain
| | - Eva Rojo-Francàs
- Departament de Genètica, Microbiologia i Estadística, Facultat de Biologia, Universitat de Barcelona, Prevosti Building, floor 2, Av. Diagonal 643, 08028 Barcelona, Spain; (B.T.); (E.A.-G.); (N.F.-C.); (E.R.-F.); (L.P.-C.)
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Institut de Biomedicina de la Universitat de Barcelona (IBUB), 08028 Barcelona, Spain; (E.M.); (V.C.)
- Institut de Recerca Sant Joan de Déu (IR-SJD), 08950 Esplugues de Llobregat, Spain
| | - Sadaf Ghorbani
- Centre for Neuropsychiatric Disorders, Department of Biomedicine, University of Bergen, N5009 Bergen, Norway; (S.G.); (R.K.); (J.H.)
| | - Laura Pineda-Cirera
- Departament de Genètica, Microbiologia i Estadística, Facultat de Biologia, Universitat de Barcelona, Prevosti Building, floor 2, Av. Diagonal 643, 08028 Barcelona, Spain; (B.T.); (E.A.-G.); (N.F.-C.); (E.R.-F.); (L.P.-C.)
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Institut de Biomedicina de la Universitat de Barcelona (IBUB), 08028 Barcelona, Spain; (E.M.); (V.C.)
- Institut de Recerca Sant Joan de Déu (IR-SJD), 08950 Esplugues de Llobregat, Spain
| | - Amaia Hervás
- Child and Adolescent Mental Health Unit, Hospital Universitari Mútua de Terrassa, 08221 Terrassa, Spain; (A.H.); (I.R.)
- IGAIN, Global Institute of Integral Attention to Neurodevelopment, 08007 Barcelona, Spain
| | - Isabel Rueda
- Child and Adolescent Mental Health Unit, Hospital Universitari Mútua de Terrassa, 08221 Terrassa, Spain; (A.H.); (I.R.)
| | - Estefanía Moreno
- Institut de Biomedicina de la Universitat de Barcelona (IBUB), 08028 Barcelona, Spain; (E.M.); (V.C.)
- Department of Biochemistry and Molecular Biomedicine, Faculty of Biology, University of Barcelona, 08028 Barcelona, Spain
| | - Janice M. Fullerton
- Neuroscience Research Australia, Sydney, NSW 2031, Australia;
- School of Medical Sciences, University of New South Wales, Sydney, NSW 2052, Australia
| | - Vicent Casadó
- Institut de Biomedicina de la Universitat de Barcelona (IBUB), 08028 Barcelona, Spain; (E.M.); (V.C.)
- Department of Biochemistry and Molecular Biomedicine, Faculty of Biology, University of Barcelona, 08028 Barcelona, Spain
| | - Jan K. Buitelaar
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, 6525 HR Nijmegen, The Netherlands;
- Karakter Child and Adolescent Psychiatry University Centre, 6525 GC Nijmegen, The Netherlands;
| | - Nanda Rommelse
- Karakter Child and Adolescent Psychiatry University Centre, 6525 GC Nijmegen, The Netherlands;
- Department of Psychiatry, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, 6525 HR Nijmegen, The Netherlands;
| | - Barbara Franke
- Department of Psychiatry, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, 6525 HR Nijmegen, The Netherlands;
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, 6525 HR Nijmegen, The Netherlands
| | - Andreas Reif
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital Frankfurt, 60590 Frankfurt am Main, Germany;
| | - Andreas G. Chiocchetti
- Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, Autism Research and Intervention Center of Excellence Frankfurt, JW Goethe University, 60323 Frankfurt am Main, Germany; (A.G.C.); (C.F.)
| | - Christine Freitag
- Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, Autism Research and Intervention Center of Excellence Frankfurt, JW Goethe University, 60323 Frankfurt am Main, Germany; (A.G.C.); (C.F.)
| | - Rune Kleppe
- Centre for Neuropsychiatric Disorders, Department of Biomedicine, University of Bergen, N5009 Bergen, Norway; (S.G.); (R.K.); (J.H.)
- Division of Psychiatry, Haukeland University Hospital, 5021 Bergen, Norway
| | - Jan Haavik
- Centre for Neuropsychiatric Disorders, Department of Biomedicine, University of Bergen, N5009 Bergen, Norway; (S.G.); (R.K.); (J.H.)
| | - Claudio Toma
- Departament de Genètica, Microbiologia i Estadística, Facultat de Biologia, Universitat de Barcelona, Prevosti Building, floor 2, Av. Diagonal 643, 08028 Barcelona, Spain; (B.T.); (E.A.-G.); (N.F.-C.); (E.R.-F.); (L.P.-C.)
- Neuroscience Research Australia, Sydney, NSW 2031, Australia;
- School of Medical Sciences, University of New South Wales, Sydney, NSW 2052, Australia
- Centro de Biología Molecular “Severo Ochoa”, Universidad Autónoma de Madrid/CSIC, C/Nicolás Cabrera, 1, Campus UAM, 28049 Madrid, Spain
| | - Bru Cormand
- Departament de Genètica, Microbiologia i Estadística, Facultat de Biologia, Universitat de Barcelona, Prevosti Building, floor 2, Av. Diagonal 643, 08028 Barcelona, Spain; (B.T.); (E.A.-G.); (N.F.-C.); (E.R.-F.); (L.P.-C.)
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Institut de Biomedicina de la Universitat de Barcelona (IBUB), 08028 Barcelona, Spain; (E.M.); (V.C.)
- Institut de Recerca Sant Joan de Déu (IR-SJD), 08950 Esplugues de Llobregat, Spain
| |
Collapse
|
49
|
Bosica F, Andrei SA, Neves JF, Brandt P, Gunnarsson A, Landrieu I, Ottmann C, O'Mahony G. Design of Drug-Like Protein-Protein Interaction Stabilizers Guided By Chelation-Controlled Bioactive Conformation Stabilization. Chemistry 2020; 26:7131-7139. [PMID: 32255539 DOI: 10.1002/chem.202001608] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Indexed: 01/29/2023]
Abstract
Protein-protein interactions (PPIs) of 14-3-3 proteins are a model system for studying PPI stabilization. The complex natural product Fusicoccin A stabilizes many 14-3-3 PPIs but is not amenable for use in SAR studies, motivating the search for more drug-like chemical matter. However, drug-like 14-3-3 PPI stabilizers enabling such studies have remained elusive. An X-ray crystal structure of a PPI in complex with an extremely low potency stabilizer uncovered an unexpected non-protein interacting, ligand-chelated Mg2+ leading to the discovery of metal-ion-dependent 14-3-3 PPI stabilization potency. This originates from a novel chelation-controlled bioactive conformation stabilization effect. Metal chelation has been associated with pan-assay interference compounds (PAINS) and frequent hitter behavior, but chelation can evidently also lead to true potency gains and find use as a medicinal chemistry strategy to guide compound optimization. To demonstrate this, we exploited the effect to design the first potent, selective, and drug-like 14-3-3 PPI stabilizers.
Collapse
Affiliation(s)
- Francesco Bosica
- Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Pepparedsleden 1, 43183, Mölndal, Sweden.,Laboratory of Chemical Biology, Department of Biomedical, Engineering and Institute for Complex Molecular Systems (ICMS), Eindhoven University of Technology, Den Dolech 2, 5612, AZ, Eindhoven, The Netherlands
| | - Sebastian A Andrei
- Laboratory of Chemical Biology, Department of Biomedical, Engineering and Institute for Complex Molecular Systems (ICMS), Eindhoven University of Technology, Den Dolech 2, 5612, AZ, Eindhoven, The Netherlands
| | - João Filipe Neves
- ERL9002 Integrative Structural Biology, CNRS, 50 Avenue de Halley, 59658, Villeneuve d'Ascq, Lille, France.,U1167 RID-AGE Risk Factors and Molecular Determinants of Aging-Related Diseases, Inserm, CHU Lille, Institut Pasteur de Lille, Univ. Lille, 1 Rue du Professeur Calmette, 59800, Lille, France
| | - Peter Brandt
- Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Pepparedsleden 1, 43183, Mölndal, Sweden
| | - Anders Gunnarsson
- Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Pepparedsleden 1, 43183, Mölndal, Sweden
| | - Isabelle Landrieu
- ERL9002 Integrative Structural Biology, CNRS, 50 Avenue de Halley, 59658, Villeneuve d'Ascq, Lille, France.,U1167 RID-AGE Risk Factors and Molecular Determinants of Aging-Related Diseases, Inserm, CHU Lille, Institut Pasteur de Lille, Univ. Lille, 1 Rue du Professeur Calmette, 59800, Lille, France
| | - Christian Ottmann
- Laboratory of Chemical Biology, Department of Biomedical, Engineering and Institute for Complex Molecular Systems (ICMS), Eindhoven University of Technology, Den Dolech 2, 5612, AZ, Eindhoven, The Netherlands.,Department of Chemistry, University of Duisburg-Essen, Universitätsstrasse 7, 45117, Essen, Germany
| | - Gavin O'Mahony
- Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Pepparedsleden 1, 43183, Mölndal, Sweden
| |
Collapse
|
50
|
Nathan KG, Lal SK. The Multifarious Role of 14-3-3 Family of Proteins in Viral Replication. Viruses 2020; 12:E436. [PMID: 32294919 PMCID: PMC7232403 DOI: 10.3390/v12040436] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 04/06/2020] [Accepted: 04/07/2020] [Indexed: 02/06/2023] Open
Abstract
The 14-3-3 proteins are a family of ubiquitous and exclusively eukaryotic proteins with an astoundingly significant number of binding partners. Their binding alters the activity, stability, localization, and phosphorylation state of a target protein. The association of 14-3-3 proteins with the regulation of a wide range of general and specific signaling pathways suggests their crucial role in health and disease. Recent studies have linked 14-3-3 to several RNA and DNA viruses that may contribute to the pathogenesis and progression of infections. Therefore, comprehensive knowledge of host-virus interactions is vital for understanding the viral life cycle and developing effective therapeutic strategies. Moreover, pharmaceutical research is already moving towards targeting host proteins in the control of virus pathogenesis. As such, targeting the right host protein to interrupt host-virus interactions could be an effective therapeutic strategy. In this review, we generated a 14-3-3 protein interactions roadmap in viruses, using the freely available Virusmentha network, an online virus-virus or virus-host interaction tool. Furthermore, we summarize the role of the 14-3-3 family in RNA and DNA viruses. The participation of 14-3-3 in viral infections underlines its significance as a key regulator for the expression of host and viral proteins.
Collapse
Affiliation(s)
- Kavitha Ganesan Nathan
- School of Science, Monash University, Bandar Sunway, Selangor Darul Ehsan 47500, Malaysia;
| | - Sunil K. Lal
- School of Science, Monash University, Bandar Sunway, Selangor Darul Ehsan 47500, Malaysia;
- Tropical Medicine & Biology Platform, Monash University, Bandar Sunway, Selangor Darul Ehsan 47500, Malaysia
| |
Collapse
|