1
|
Sun R, Yan Q, Deng W, Chen P, Wu D, Zheng P. N-glycosylation-modifications-driven conformational dynamics attenuate substrate inhibition of d-lactonohydrolase. Bioorg Chem 2025; 155:108122. [PMID: 39787912 DOI: 10.1016/j.bioorg.2025.108122] [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: 11/17/2024] [Revised: 12/25/2024] [Accepted: 01/01/2025] [Indexed: 01/12/2025]
Abstract
Achieving enzyme catalysis at high substrate concentrations is a substantial challenge in industrial biocatalysis, and the role of glycosylation in post-translational modifications that modulate enzyme substrate inhibition remains poorly understood. This study provides insights into the role of N-glycosylation in substrate inhibition by comparing the catalytic properties of d-lactonohydrolase (d-Lac) derived from Fusarium moniliforme expressed in prokaryotic and eukaryotic hosts. Experimental evidence indicates that recombinant d-Lac expressed in Pichia pastoris (PpLac-WT) exhibits higher hydrolysis rates at a substrate concentration of 400 g/L, with reduced substrate inhibition and enhanced stability compared to the recombinant d-Lac expressed in Escherichia coli (EcLac-WT). Mutant PpLac-M1 achieves a conversion rate of 40 % at a substrate concentration of 400 g/L, with a space-time yield of d-pantoic acid reaching 91.1 g/L/h. Proteomics analysis reveals that residues N29 and N278, located approximately 10-20 Å from the active site undergo N-glycosylation in PpLac-WT. Using microsecond-scale molecular dynamics simulations and Markov state models, we elucidate the effects of glycosylation on the conformational flexibility of two key loops at the entrance of the binding pocket. Specifically, the loops in PpLac-WT can transition between open and closed states, whereas those in EcLac-WT tend to remain open. In high substrate concentration conditions, the open state causes congestion, leading to substrate inhibition. Shortest-path map analysis confirms that substrate entry is dynamically controlled by residue N29 on the loops surrounding the active site. Our findings enhance the understanding of the effects of glycosylation on enzyme conformational dynamics and provide insights into mitigating inhibition at high substrate concentrations.
Collapse
Affiliation(s)
- Ruobin Sun
- School of Biotechnology and Key Laboratory of Industrial Biotechnology of Education, School of Biotechnology, Jiangnan University, Wuxi 214122 China
| | - Qipeng Yan
- Hunan Provincial Key Laboratory of Animal Models and Molecular Medicine, State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, School of Biomedical Sciences, Hunan University, Changsha 410082 China; Greater Bay Area Institute for Innovation, Hunan University, Guangzhou 511300 Guangdong Province, China
| | - Wenhao Deng
- Key Laboratory of Material Chemistry for Energy Conversion and Storage, Ministry of Education, School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology, Wuhan 430074, China; Department of Physics and Astronomy, University College London, London WC1E 6BT, United Kingdom
| | - Pengcheng Chen
- School of Biotechnology and Key Laboratory of Industrial Biotechnology of Education, School of Biotechnology, Jiangnan University, Wuxi 214122 China
| | - Dan Wu
- School of Biotechnology and Key Laboratory of Industrial Biotechnology of Education, School of Biotechnology, Jiangnan University, Wuxi 214122 China
| | - Pu Zheng
- School of Biotechnology and Key Laboratory of Industrial Biotechnology of Education, School of Biotechnology, Jiangnan University, Wuxi 214122 China.
| |
Collapse
|
2
|
Cheng Z, Cheng Z, Zhang Y, Zhang S. "Intrinsic disorder-protein modification-LLPS-tumor" regulatory axis: From regulatory mechanisms to precision medicine. Biochim Biophys Acta Rev Cancer 2025; 1880:189242. [PMID: 39672280 DOI: 10.1016/j.bbcan.2024.189242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Revised: 12/07/2024] [Accepted: 12/08/2024] [Indexed: 12/15/2024]
Abstract
Liquid-Liquid Phase Separation (LLPS) is an important mechanism for the formation of functional droplets. Protein modification is an important pathway to regulate LLPS, in which series of modifying groups realize dynamic regulation by changing the charge and spatial resistance of the modified proteins. Meanwhile, uncontrolled protein modifications associated with LLPS dysregulation are highly correlated with tumorigenesis and development, suggesting the existence of a potential regulatory axis between the three. In this review, we pioneered "protein modification-LLPS-tumor" regulatory axis and summarized protein modifications that regulate LLPS in cancer cells (including phosphorylation, acetylation, methylation, ubiquitination, SUMOylation, lactate, ADP-ribosylation, O-glycosylation, and acylation) and their associated modification mechanisms. Finally, we outline advances in precision medicine based on this regulatory axis. The aim of this review is to expand the understanding of protein modifications regulating LLPS under normal or abnormal cellular conditions and to provide possible ideas for precision therapy.
Collapse
Affiliation(s)
- Zekun Cheng
- Xiangya School of Medicine, Central South University, Changsha, Hunan 410013, PR China
| | - Zehao Cheng
- Xiangya School of Medicine, Central South University, Changsha, Hunan 410013, PR China
| | - Yikai Zhang
- Xiangya School of Medicine, Central South University, Changsha, Hunan 410013, PR China
| | - Shubing Zhang
- Department of Cell Biology, School of Life Sciences, Central South University, Changsha, Hunan 410013, PR China.
| |
Collapse
|
3
|
Chen J, Ye H. Expanding horizons: genetic code expansion technology in the study of PTM functions. Bioorg Med Chem 2025; 118:118049. [PMID: 39729921 DOI: 10.1016/j.bmc.2024.118049] [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: 10/01/2024] [Revised: 12/14/2024] [Accepted: 12/18/2024] [Indexed: 12/29/2024]
Abstract
Recent advancements in Genetic Code Expansion (GCE) have significantly enhanced our understanding of post-translational modifications (PTMs), which are critical for protein regulation. GCE facilitates the precise incorporation of unnatural amino acids (UAAs) at specific sites within proteins of interest (POIs), making it a powerful tool for modulating PTMs in vivo. This review summarizes the various UAAs utilized to directly incorporate PTMs into proteins through GCE, with a focus on their applications in both histone and non-histone PTMs research. We also discuss the challenges associated with incorporating certain PTMs into target proteins via GCE and provide an overview of the latest strategies developed to overcome these hurdles.
Collapse
Affiliation(s)
- Jingzhuo Chen
- Jiangsu Provincial Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Tongjiaxiang No. 24, Nanjing 210009, Jiangsu, China
| | - Hui Ye
- Jiangsu Provincial Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Tongjiaxiang No. 24, Nanjing 210009, Jiangsu, China.
| |
Collapse
|
4
|
Mihut A, O'Neill JS, Partch CL, Crosby P. PERspectives on circadian cell biology. Philos Trans R Soc Lond B Biol Sci 2025; 380:20230483. [PMID: 39842483 DOI: 10.1098/rstb.2023.0483] [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: 03/14/2024] [Revised: 05/18/2024] [Accepted: 05/24/2024] [Indexed: 01/24/2025] Open
Abstract
Daily rhythms in the activities of PERIOD proteins are critical to the temporal regulation of mammalian physiology. While the molecular partners and genetic circuits that allow PERIOD to effect auto-repression and regulate transcriptional programmes are increasingly well understood, comprehension of the time-resolved mechanisms that allow PERIOD to conduct this daily dance is incomplete. Here, we consider the character and controversies of this central mammalian clock protein with a focus on its intrinsically disordered nature.This article is part of the Theo Murphy meeting issue 'Circadian rhythms in infection and immunity'.
Collapse
Affiliation(s)
- Andrei Mihut
- MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge CB2 0QH, UK
| | - John S O'Neill
- MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge CB2 0QH, UK
| | - Carrie L Partch
- Department of Chemistry and Biochemistry, University of California, 1156 High Street, Santa Cruz, CA 95064, USA
| | - Priya Crosby
- Institute of Cell Biology, School of Biological Sciences, University of Edinburgh, Edinburgh EH9 3BF, UK
| |
Collapse
|
5
|
Abid MSR, Naldrett MJ, Alvarez S, Eichhorn CD, Andrews MT, Checco JW. Rapid Microwave Fixation of the Brain Reveals Seasonal Changes in the Phosphoproteome of Hibernating Thirteen-Lined Ground Squirrels. ACS Chem Neurosci 2025. [PMID: 39840768 DOI: 10.1021/acschemneuro.4c00635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2025] Open
Abstract
Hibernating mammals such as the thirteen-lined ground squirrel (Ictidomys tridecemlineatus) experience significant reductions in oxidative metabolism and body temperature when entering a state known as torpor. Animals entering or exiting torpor do not experience permanent loss of brain function or other injuries, and the processes that enable such neuroprotection are not well understood. To gain insight into changes in protein function that occur in the dramatically different physiological states of hibernation, we performed quantitative phosphoproteomics experiments on thirteen-lined ground squirrels that are summer-active, winter-torpid, and spring-active. An important aspect of our approach was the use of focused microwave irradiation of the brain to sacrifice the animals and rapidly inactivate phosphatases and kinases to preserve the native phosphoproteome. Overall, our results showed pronounced changes in phosphorylated proteins for the transitions into and out of torpor, including proteins involved in gene expression, DNA maintenance and repair, cellular plasticity, and human disease. In contrast, the transition between the active states showed minimal changes. This study offers valuable insight into the global changes in brain phosphorylation in hibernating mammals, the results of which may be relevant to future therapeutic strategies for brain injury.
Collapse
Affiliation(s)
- Md Shadman Ridwan Abid
- Department of Chemistry, University of Nebraska-Lincoln, Lincoln, Nebraska 68588, United States
| | - Michael J Naldrett
- The Nebraska Center for Biotechnology, University of Nebraska-Lincoln, Lincoln, Nebraska 68588, United States
| | - Sophie Alvarez
- The Nebraska Center for Biotechnology, University of Nebraska-Lincoln, Lincoln, Nebraska 68588, United States
| | - Catherine D Eichhorn
- Department of Chemistry, University of Nebraska-Lincoln, Lincoln, Nebraska 68588, United States
- The Nebraska Center for Integrated Biomolecular Communication (NCIBC), University of Nebraska-Lincoln, Lincoln, Nebraska 68588, United States
| | - Matthew T Andrews
- School of Natural Resources, University of Nebraska-Lincoln, Lincoln, Nebraska 68583, United States
| | - James W Checco
- Department of Chemistry, University of Nebraska-Lincoln, Lincoln, Nebraska 68588, United States
- The Nebraska Center for Integrated Biomolecular Communication (NCIBC), University of Nebraska-Lincoln, Lincoln, Nebraska 68588, United States
| |
Collapse
|
6
|
Little MJ, Mason JM, Mehrban N. Evolution of branched peptides as novel biomaterials. J Mater Chem B 2025. [PMID: 39835399 PMCID: PMC11747965 DOI: 10.1039/d4tb01897d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Accepted: 01/11/2025] [Indexed: 01/22/2025]
Abstract
Branched peptide-based materials draw inspiration from dendritic structures to emulate the complex architecture of native tissues, aiming to enhance the performance of biomaterials in medical applications. These innovative materials benefit from several key features: they exhibit slower degradation rates, greater stiffness, and the ability to self-assemble. These properties are crucial for maintaining the structural integrity and functionality of the materials over time. By integrating bioactive peptides and natural polymers within their branched frameworks, these materials offer modularity and tunability and can accommodate a range of mechanical properties, degradation rates, and biological functions making them suitable for biomedical applications, including drug delivery systems, wound healing scaffolds, and tissue engineering constructs. In drug delivery, these materials can be engineered to release therapeutic agents in a controlled manner, enhancing the efficacy and safety of treatments. In wound healing, they provide a supportive environment which promotes rapid and efficient tissue repair. The combination of biomimetic design and functional adaptability makes branched peptide-based materials a promising candidate for the development of next-generation biomaterials, paving the way for significant advancements in healthcare.
Collapse
Affiliation(s)
| | - Jody M Mason
- University of Bath, Claverton Down, Bath, BA2 7AY, UK.
| | - Nazia Mehrban
- University of Bath, Claverton Down, Bath, BA2 7AY, UK.
| |
Collapse
|
7
|
Jo CS, Zhao H, Hwang JS. Acetylation-enhanced Sp1 transcriptional activity suppresses Mlph expression. Sci Rep 2025; 15:2338. [PMID: 39824975 PMCID: PMC11748735 DOI: 10.1038/s41598-025-86282-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Accepted: 01/09/2025] [Indexed: 01/20/2025] Open
Abstract
Melanosome transport is regulated by major proteins, including Rab27a, Melanophilin (Mlph), and Myosin Va (Myo-Va), that form a tripartite complex. Mutation of these proteins causes melanosome aggregation around the nucleus. Among these proteins, Mlph is a linker between Rab27a and Myo-Va. There are some studies about the regulation of Mlph transcriptional expression. However, its regulation by post-translational modifications remains unclear. In this study, inhibition of HDACs by SAHA and TSA disrupted melanosome transport, causing melanosome aggregation. Specifically, we identified a novel mechanism in which HDAC5 regulates Mlph expression via Sp1. Knockdown of HDAC5 increased the acetylation of Sp1 and the binding to the Mlph promoter, thereby modulating its expression. This study highlights the crucial role of HDAC5 in melanosome transport through its interaction with Sp1. These findings suggest that HDAC5-mediated deacetylation is pivotal in the post-translational modification of melanosome transport, providing insights into the molecular mechanisms underlying this process.
Collapse
Affiliation(s)
- Chan Song Jo
- Department of Genetics and Biotechnology, Graduate School of Biotechnology, College of Life Sciences, Kyung Hee University, Yongin, Korea
| | - Hairu Zhao
- Department of Genetics and Biotechnology, Graduate School of Biotechnology, College of Life Sciences, Kyung Hee University, Yongin, Korea
| | - Jae Sung Hwang
- Department of Genetics and Biotechnology, Graduate School of Biotechnology, College of Life Sciences, Kyung Hee University, Yongin, Korea.
| |
Collapse
|
8
|
Xiao B, Zhang S, Ainiwaer M, Liu H, Ning L, Hong Y, Sun Y, Ji Y. Deep learning-based assessment of missense variants in the COG4 gene presented with bilateral congenital cataract. BMJ Open Ophthalmol 2025; 10:e001906. [PMID: 39809522 DOI: 10.1136/bmjophth-2024-001906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Accepted: 12/11/2024] [Indexed: 01/16/2025] Open
Abstract
OBJECTIVE We compared the protein structure and pathogenicity of clinically relevant variants of the COG4 gene with AlphaFold2 (AF2), Alpha Missense (AM), and ThermoMPNN for the first time. METHODS AND ANALYSIS The sequences of clinically relevant Cog4 missense variants (one novel identified p.Y714F and three pre-existing p.G512R, p.R729W and p.L769R from Uniprot Q9H9E3) were imported into AF2 for protein structural prediction, and the pathogenicity was estimated using AM and ThermoMPNN. Different pathogenicity metrics were aggregated with principal component analysis (PCA) and further analysed at three levels (amino acid position, substitution and post-translation) based on all possible Cog4 missense variants (n=14 915). RESULTS Localised protein structural impact including change of conformation and amino acid polarity, breakage of hydrogen bond and salt-bridge, and formation of alpha-helix were identified among clinically relevant Cog4 variants. The global structural comparison with multidimensional scaling demonstrated variants with similar protein structures (AF2) tended to exhibit similar clinical and biological phenotypes. The Cog4 p.Y714F variant exhibited greater protein structural similarity to mutated Cog4 found in Saul‒Wilson syndrome (p.G512R) and shared similar clinical phenotype (congenital cataract and psychomotor retardation). PCA of included pathogenic metrics demonstrated p.Y714F occurred at a critical position in Cog4 amino acid sequence with disrupted post-translational phosphorylation. CONCLUSION Deep learning algorithms, including AF2, AM and ThermoMPNN, can be useful for evaluating variant of uncertain significance (VUS) by structural and pathogenicity prediction. Despite classified as VUS (American College of Medical Genetics and Genomics criteria: PM1, PP4), the pathogenicity in this Cog4 variant cannot be ruled out and warrants further investigation.
Collapse
Affiliation(s)
- Binghe Xiao
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai, China
- Key laboratory of Myopia and Related Eye Diseases, NHC, Shanghai, China
- Key laboratory of Myopia and Related Eye Diseases, Chinese Academy of Medical Sciences, Shanghai, China
| | - Shaohua Zhang
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai, China
- Key laboratory of Myopia and Related Eye Diseases, NHC, Shanghai, China
- Key laboratory of Myopia and Related Eye Diseases, Chinese Academy of Medical Sciences, Shanghai, China
| | - Maierdanjiang Ainiwaer
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai, China
- Key laboratory of Myopia and Related Eye Diseases, NHC, Shanghai, China
- Key laboratory of Myopia and Related Eye Diseases, Chinese Academy of Medical Sciences, Shanghai, China
| | - Houyi Liu
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai, China
- Key laboratory of Myopia and Related Eye Diseases, NHC, Shanghai, China
- Key laboratory of Myopia and Related Eye Diseases, Chinese Academy of Medical Sciences, Shanghai, China
| | - Li Ning
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai, China
- Key laboratory of Myopia and Related Eye Diseases, NHC, Shanghai, China
- Key laboratory of Myopia and Related Eye Diseases, Chinese Academy of Medical Sciences, Shanghai, China
| | - Yingying Hong
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai, China
- Key laboratory of Myopia and Related Eye Diseases, NHC, Shanghai, China
- Key laboratory of Myopia and Related Eye Diseases, Chinese Academy of Medical Sciences, Shanghai, China
| | - Yang Sun
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai, China
- Key laboratory of Myopia and Related Eye Diseases, NHC, Shanghai, China
- Key laboratory of Myopia and Related Eye Diseases, Chinese Academy of Medical Sciences, Shanghai, China
| | - Yinghong Ji
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai, China
- Key laboratory of Myopia and Related Eye Diseases, NHC, Shanghai, China
- Key laboratory of Myopia and Related Eye Diseases, Chinese Academy of Medical Sciences, Shanghai, China
| |
Collapse
|
9
|
Han Z, Fu J, Gong A, Ren W. Bacterial indole-3-propionic acid inhibits macrophage IL-1β production through targeting methionine metabolism. SCIENCE CHINA. LIFE SCIENCES 2025:10.1007/s11427-024-2789-1. [PMID: 39825207 DOI: 10.1007/s11427-024-2789-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Accepted: 11/20/2024] [Indexed: 01/20/2025]
Abstract
The gut microbiota plays key roles in host health by shaping the host immune responses through their metabolites, like indole derivatives from tryptophan. However, the direct role of these indole derivatives in macrophage fate decision and the underlying mechanism remains unknown. Here, we found that bacterial indole-3-propionic acid (IPA) downregulates interleukin-1beta (IL-1β) production in M1 macrophages through inhibition of nuclear factor-kappa B (NF-κB) signaling. Mechanistically, IPA binds specifically with methionine adenosyl-transferase 2A (MAT2A) to promote S-adenosylmethionine (SAM) synthesis, which facilitates the DNA methylation of ubiquitin-specific peptidase 16 (USP16, a deubiquitinase), and in turn promotes Toll-like receptor 4 (TLR4) ubiquitination and NF-κB inhibition. Furthermore, IPA administration attenuates sepsis in mouse models induced by lipopolysaccharides (LPS), showcasing its potential as a microbial-derived adjunct in alleviating inflammation. Collectively, our findings reveal a newly found microbial metabolite-immune system regulatory pathway mediated by IPA.
Collapse
Affiliation(s)
- Ziyi Han
- State Key Laboratory of Livestock and Poultry Breeding, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
- Animal Disease-resistant Nutrition, Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, 625014, China
| | - Jian Fu
- State Key Laboratory of Livestock and Poultry Breeding, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Aiyan Gong
- China Institute of Veterinary Drug Control, Beijing, 100081, China
| | - Wenkai Ren
- State Key Laboratory of Livestock and Poultry Breeding, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China.
| |
Collapse
|
10
|
De la Fuente IM, Cortes JM, Malaina I, Pérez-Yarza G, Martinez L, López JI, Fedetz M, Carrasco-Pujante J. The main sources of molecular organization in the cell. Atlas of self-organized and self-regulated dynamic biostructures. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2025:S0079-6107(25)00003-3. [PMID: 39805422 DOI: 10.1016/j.pbiomolbio.2025.01.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/19/2024] [Accepted: 01/10/2025] [Indexed: 01/16/2025]
Abstract
One of the most important goals of contemporary biology is to understand the principles of the molecular order underlying the complex dynamic architecture of cells. Here, we present an overview of the main driving forces involved in the cellular molecular complexity and in the emergent functional dynamic structures, spanning from the most basic molecular organization levels to the complex emergent integrative systemic behaviors. First, we address the molecular information processing which is essential in many complex fundamental mechanisms such as the epigenetic memory, alternative splicing, regulation of transcriptional system, and the adequate self-regulatory adaptation to the extracellular environment. Next, we approach the biochemical self-organization, which is central to understand the emergency of metabolic rhythms, circadian oscillations, and spatial traveling waves. Such a complex behavior is also fundamental to understand the temporal compartmentalization of the cellular metabolism and the dynamic regulation of many physiological activities. Numerous examples of biochemical self-organization are considered here, which show that practically all the main physiological processes in the cell exhibit this type of dynamic molecular organization. Finally, we focus on the biochemical self-assembly which, at a primary level of organization, is a basic but important mechanism for the order in the cell allowing biomolecules in a disorganized state to form complex aggregates necessary for a plethora of essential structures and physiological functions. In total, more than 500 references have been compiled in this review. Due to these main sources of order, systemic functional structures emerge in the cell, driving the metabolic functionality towards the biological complexity.
Collapse
Affiliation(s)
- Ildefonso M De la Fuente
- Department of Mathematics, Faculty of Science and Technology, University of the Basque Country, UPV/EHU, Leioa 48940, Spain.
| | - Jesus M Cortes
- Department of Cell Biology and Histology, Faculty of Medicine and Nursing, University of the Basque Country, UPV/EHU, Leioa 48940, Spain; Biobizkaia Health Research Institute, Barakaldo 48903, Spain; IKERBASQUE: The Basque Foundation for Science, Bilbao
| | - Iker Malaina
- Department of Mathematics, Faculty of Science and Technology, University of the Basque Country, UPV/EHU, Leioa 48940, Spain
| | - Gorka Pérez-Yarza
- Department of Cell Biology and Histology, Faculty of Medicine and Nursing, University of the Basque Country, UPV/EHU, Leioa 48940, Spain
| | - Luis Martinez
- Department of Mathematics, Faculty of Science and Technology, University of the Basque Country, UPV/EHU, Leioa 48940, Spain
| | - José I López
- Biobizkaia Health Research Institute, Barakaldo 48903, Spain
| | - Maria Fedetz
- Department of Cell Biology and Immunology, Institute of Parasitology and Biomedicine "López-Neyra", CSIC, Granada 18016, Spain
| | - Jose Carrasco-Pujante
- Department of Cell Biology and Histology, Faculty of Medicine and Nursing, University of the Basque Country, UPV/EHU, Leioa 48940, Spain
| |
Collapse
|
11
|
Shao C, Tang S, Yu S, Liu C, Zhang Y, Wan T, He Z, Yuan Q, Wu S, Zhang H, Wan N, Zhan M, Tan RX, Hao H, Ye H, Wang N. Genetic code expansion reveals site-specific lactylation in living cells reshapes protein functions. Nat Commun 2025; 16:227. [PMID: 39779673 PMCID: PMC11711764 DOI: 10.1038/s41467-024-55165-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 12/04/2024] [Indexed: 01/11/2025] Open
Abstract
Protein lactylation is an emerging field. To advance the exploration of its biological functions, here we develop a comprehensive workflow that integrates proteomics to identify lactylated sites, genetic code expansion (GCE) for the expression of site-specifically lactylated proteins in living cells, and an integrated functional analysis (IFA) platform to evaluate their biological effects. Using a combined wet-and-dry-lab proteomics strategy, we identify a conserved lactylation at ALDOA-K147, which we hypothesize plays a significant biological role. Expression of this site-specifically lactylated ALDOA in mammalian cells reveals that this modification not only inhibits enzymatic activity but also induces gain-of-function effects. These effects reshaped ALDOA functionality by enhancing protein stability, promoting nuclear translocation, regulating adhesion-related gene expression, altering cell morphology and modulating ALDOA-interacting proteins. Our findings highlight the utility of the GCE-based workflow in establishing causal relationships between specific lactylation events and both target-specific and cell-wide changes, advancing our understanding of protein lactylation's functional impact.
Collapse
Affiliation(s)
- Chang Shao
- Jiangsu Provincial Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Tongjiaxiang No. 24, Nanjing, Jiangsu, China
| | - Shuo Tang
- School of Pharmacy, Nanjing University of Chinese Medicine, Xianlindadao No. 138, Nanjing, Jiangsu, China
| | - Siqin Yu
- Jiangsu Provincial Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Tongjiaxiang No. 24, Nanjing, Jiangsu, China
| | - Chenguang Liu
- Jiangsu Provincial Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Tongjiaxiang No. 24, Nanjing, Jiangsu, China
| | - Yueyang Zhang
- Jiangsu Provincial Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Tongjiaxiang No. 24, Nanjing, Jiangsu, China
| | - Tianyan Wan
- School of Pharmacy, Nanjing University of Chinese Medicine, Xianlindadao No. 138, Nanjing, Jiangsu, China
| | - Zimeng He
- Jiangsu Provincial Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Tongjiaxiang No. 24, Nanjing, Jiangsu, China
| | - Qi Yuan
- Jiangsu Provincial Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Tongjiaxiang No. 24, Nanjing, Jiangsu, China
| | - Shihan Wu
- School of Pharmacy, Nanjing University of Chinese Medicine, Xianlindadao No. 138, Nanjing, Jiangsu, China
| | - Hanqing Zhang
- Jiangsu Provincial Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Tongjiaxiang No. 24, Nanjing, Jiangsu, China
| | - Ning Wan
- Jiangsu Provincial Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Tongjiaxiang No. 24, Nanjing, Jiangsu, China
| | - Mengru Zhan
- School of Pharmacy, Nanjing University of Chinese Medicine, Xianlindadao No. 138, Nanjing, Jiangsu, China
| | - Ren Xiang Tan
- School of Pharmacy, Nanjing University of Chinese Medicine, Xianlindadao No. 138, Nanjing, Jiangsu, China
- State Key Laboratory of Pharmaceutical Biotechnology, Institute of Functional Biomolecules, School of Life Sciences, Nanjing University, Nanjing, Jiangsu, China
| | - Haiping Hao
- Jiangsu Provincial Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Tongjiaxiang No. 24, Nanjing, Jiangsu, China
| | - Hui Ye
- Jiangsu Provincial Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Tongjiaxiang No. 24, Nanjing, Jiangsu, China.
| | - Nanxi Wang
- School of Pharmacy, Nanjing University of Chinese Medicine, Xianlindadao No. 138, Nanjing, Jiangsu, China.
| |
Collapse
|
12
|
Ling L, Xu G, Fang M, Chen J, Gong M, Wang T, Ju R, Nie S. A combined proteomic and metabolomic analysis of the early aborted embryonic tissues with maternal COVID-19 infection. J Proteomics 2025; 313:105383. [PMID: 39793701 DOI: 10.1016/j.jprot.2025.105383] [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: 04/21/2024] [Revised: 12/09/2024] [Accepted: 01/06/2025] [Indexed: 01/13/2025]
Abstract
COVID-19 still spreads worldwide, and repeated infections are hard to avoid. Maternal infection during pregnancy is associated with adverse maternal and neonatal outcomes. Our study used a multi-omics profiling method to explore the proteome and metabolome alteration in early embryonic development after COVID-19 infection. A total of 30 chorionic tissues after artificial abortion (15 infection and 15 no-infection samples) were collected, and the UHPLC-MS/MS and LC-MS/MS were applied in the present study. As a result, 311 significantly differentially expressed proteins were identified. The function annotations revealed that the thermogenesis pathway is the most significantly enriched signaling pathway; PRKAG2, IGF1R, and RPS6KB2 were identified as the hub proteins. There were 359 metabolites significantly altered after infection. The functional annotations revealed that amino acid metabolism was significantly affected, especially beta-alanine metabolism, glutamate metabolism, and histidine metabolism pathways. The metabolites in ovarian steroidogenesis showed a down-regulating trend in the infection group. Finally, we combined the results of proteins and metabolomics analysis. The biosynthesis of the cofactors pathway was identified as significantly enriched in both proteomics and metabolomics datasets. Our findings provide a network of protein regulation and metabolite perturbation during early embryonic development with COVID-19 infection. Our findings can provide valuable insights for further exploration of the complex mechanism of COVID-19-associated pregnancy complications and outcomes. SIGNIFICANCE: COVID-19 has developed into the most prominent and deadliest pandemic respiratory disease in the world, and repeated infections are complicated to avoid. COVID-19 infection during pregnancy increases the risk of adverse maternal and neonatal outcomes, such as preterm birth and stillbirth. However, previous studies mainly focused on its effect on pregnant women, such as the clinical characteristics and gestation outcomes. There is no relevant report about the effects of virus infection on embryos in early pregnancy. The effects of COVID-19 infection changes of the proteins and metabolites during early embryonic development are undefined. Our findings provide an association between protein regulation, metabolite perturbation, and COVID-19 infection, which can provide valuable insights for further exploration of the complex mechanism COVID-19 COVID-19-associated pregnancy complications and adverse pregnancy outcomes.
Collapse
Affiliation(s)
- Ling Ling
- Department of Gynecology and Obstetrics, Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing 211100, Jiangsu, China
| | - Guiqin Xu
- Department of Gynecology and Obstetrics, Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing 211100, Jiangsu, China
| | - Miao Fang
- Department of Gynecology and Obstetrics, Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing 211100, Jiangsu, China
| | - Jianquan Chen
- Central Laboratory, Translational Medicine Research Center, The Affiliated Jiangning Hospital with Nanjing Medical University, Nanjing, Jiangsu, China
| | - Ming Gong
- Department of Gynecology and Obstetrics, Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing 211100, Jiangsu, China
| | - TianMing Wang
- Central Laboratory, Translational Medicine Research Center, The Affiliated Jiangning Hospital with Nanjing Medical University, Nanjing, Jiangsu, China.
| | - Rong Ju
- Department of Gynecology and Obstetrics, Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing 211100, Jiangsu, China.
| | - Sipei Nie
- Department of Gynecology and Obstetrics, Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing 211100, Jiangsu, China.
| |
Collapse
|
13
|
Wang RN, Li L, Zhou J, Ran J. Multifaceted roles of UFMylation in health and disease. Acta Pharmacol Sin 2025:10.1038/s41401-024-01456-9. [PMID: 39775503 DOI: 10.1038/s41401-024-01456-9] [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: 09/05/2024] [Accepted: 12/09/2024] [Indexed: 01/11/2025] Open
Abstract
Ubiquitin fold modifier 1 (UFM1) is a newly identified post-translational modifier that is involved in the UFMylation process. Similar to ubiquitination, UFMylation enables the conjugation of UFM1 to specific target proteins, thus altering their stability, activity, or localization. UFM1 chains have the potential to undergo cleavage from their associated proteins via UFM1-specific proteases, thus highlighting a reversible feature of UFMylation. This modification is conserved across nearly all eukaryotic organisms, and is associated with diverse biological activities such as hematopoiesis and the endoplasmic reticulum stress response. The disruption of UFMylation results in embryonic lethality in mice and is associated with various human diseases, thus underscoring its essential role in embryonic development, tissue morphogenesis, and organismal homeostasis. In this review, we aim to provide an in-depth overview of the UFMylation system, its importance in disease processes, and its potential as a novel target for therapeutic intervention.
Collapse
Affiliation(s)
- Ru-Na Wang
- Center for Cell Structure and Function, Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, Jinan, 250014, China
| | - Lin Li
- Center for Cell Structure and Function, Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, Jinan, 250014, China
| | - Jun Zhou
- Center for Cell Structure and Function, Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, Jinan, 250014, China
- Department of Genetics and Cell Biology, College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, Haihe Laboratory of Cell Ecosystem, Nankai University, Tianjin, 300071, China
| | - Jie Ran
- Center for Cell Structure and Function, Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, Jinan, 250014, China.
| |
Collapse
|
14
|
Tian H, Tao Z, Zhang W, Chen Y, Su T, Wang X, Yang H, Cai H, Liu S, Zhang Y, Zhang Y. Comparative Proteomics and N-Glycoproteomics Reveal the Effects of Different Plasma Protein Enrichment Technologies. J Proteome Res 2025; 24:134-143. [PMID: 39668702 DOI: 10.1021/acs.jproteome.4c00545] [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] [Indexed: 12/14/2024]
Abstract
Human plasma proteomic and glycoproteomic analyses have emerged as an alternate avenue to identify disease biomarkers and therapeutic approaches. However, the vast number of high-abundance proteins in plasma can cause mass spectrometry (MS) suppression, which makes it challenging to detect low-abundance proteins (LAP). Currently, immunoaffinity-based depletion methods and strategies involving nanomaterial protein coronas have been developed to remove high-abundance proteins (HAP) and enhance the depth of plasma protein identification. Despite these advancements, there is a lack of systematic comparison and evaluation of the qualitative and quantitative effects of different strategies on the human plasma proteome and glycoproteome. In this study, we evaluated the performance of four depletion methods including combinatorial peptide ligand libraries (CPLL), Top 2, Top 14, and the nanomaterial protein corona formed by magnetic nanoparticles (MN) in both plasma proteomics and N-glycoproteomics. Compared to the CPLL, Top 2, and Top 14 strategies, the MN approach significantly increased the number of identified peptides and proteins. However, it demonstrated a relatively lower efficacy in identifying intact N-glycopeptides and N-glycoproteins. In contrast, the immunoaffinity-based depletion methods are better suited to glycoproteomics due to higher identification numbers. We believe that this work provides valuable insights and options for various research objectives, as well as clinical applications.
Collapse
Affiliation(s)
- Huohuan Tian
- Department of Respiratory & Critical Care Medicine, Institutes for Systems Genetics, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Ze Tao
- Department of Respiratory & Critical Care Medicine, Institutes for Systems Genetics, West China Hospital, Sichuan University, Chengdu 610041, China
- Transplant Center and NHC Key Lab of Transplant Engineering and Immunology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Wanli Zhang
- Core Facility of West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yuzhe Chen
- Transplant Center and NHC Key Lab of Transplant Engineering and Immunology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Tao Su
- Department of Respiratory & Critical Care Medicine, Institutes for Systems Genetics, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xinyuan Wang
- Department of Respiratory & Critical Care Medicine, Institutes for Systems Genetics, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Hao Yang
- Department of Respiratory & Critical Care Medicine, Institutes for Systems Genetics, West China Hospital, Sichuan University, Chengdu 610041, China
- Transplant Center and NHC Key Lab of Transplant Engineering and Immunology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Hao Cai
- Department of Thoracic Surgery and Institute of Thoracic Oncology, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital of Sichuan University, Chengdu 610097, China
| | - Shuyun Liu
- Core Facility of West China Hospital, Sichuan University, Chengdu 610041, China
- Department of General Surgery, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yi Zhang
- Division of Pancreatic Surgery, Department of General Surgery, West China Hospital of Sichuan University, Chengdu 610041, China
- Department of General Surgery, Chengdu ShangJinNanFu Hospital, Chengdu 610000, China
| | - Yong Zhang
- Department of Respiratory & Critical Care Medicine, Institutes for Systems Genetics, West China Hospital, Sichuan University, Chengdu 610041, China
- Transplant Center and NHC Key Lab of Transplant Engineering and Immunology, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
15
|
Wang G, Chen L, Lian J, Gong L, Tian F, Wang Y, Lin X, Liu Y. Proteomic Insights into the Regulatory Role of CobQ Deacetylase in Aeromonas hydrophila. J Proteome Res 2025; 24:333-343. [PMID: 39659247 DOI: 10.1021/acs.jproteome.4c00847] [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] [Indexed: 12/12/2024]
Abstract
Post-translational modifications are crucial in regulating biological functions across both prokaryotes and eukaryotes. In Aeromonas hydrophila, CobQ, a recently identified novel deacetylase, plays a significant role in lysine deacetylation, influencing bacterial metabolism and stress responses. The present study utilized quantitative proteomics to investigate the impact of cobQ deletion on the global protein expression profile in A. hydrophila. Through data-independent acquisition mass spectrometry, we identified 233 upregulated and 41 downregulated proteins in the cobQ deletion mutant (ΔahcobQ) strain compared to the wild-type (WT) strain. Key differentially expressed proteins were involved in oxidative phosphorylation, bacterial secretion, and ribosomal function. Additionally, phenotypic assays demonstrated that the ΔahcobQ strain exhibited an increased resistance to oxidative phosphorylation inhibitors, suggesting a pivotal role for AhCobQ in energy metabolism. Outer membrane proteins and efflux pumps also showed altered expression, indicating potential implications for membrane permeability and antibiotic resistance. These results suggested that AhCobQ plays a vital regulatory role in maintaining metabolic homeostasis and responding to environmental stress, highlighting its potential as a target for therapeutic interventions against A. hydrophila infections.
Collapse
Affiliation(s)
- Guibin Wang
- Fujian Provincial Key Laboratory of Agroecological Processing and Safety Monitoring, College of Life Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, China
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 102206, China
| | - Linxin Chen
- College of JunCao Science and Ecology, College of Life Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, China
- Fujian Provincial Key Laboratory of Agroecological Processing and Safety Monitoring, College of Life Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, China
- Key Laboratory of Marine Biotechnology of Fujian Province, Institute of Oceanology, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Juanqi Lian
- College of JunCao Science and Ecology, College of Life Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, China
- Fujian Provincial Key Laboratory of Agroecological Processing and Safety Monitoring, College of Life Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, China
- Key Laboratory of Marine Biotechnology of Fujian Province, Institute of Oceanology, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Lanqing Gong
- College of JunCao Science and Ecology, College of Life Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, China
- Fujian Provincial Key Laboratory of Agroecological Processing and Safety Monitoring, College of Life Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, China
- Key Laboratory of Marine Biotechnology of Fujian Province, Institute of Oceanology, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Feng Tian
- College of JunCao Science and Ecology, College of Life Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, China
- Fujian Provincial Key Laboratory of Agroecological Processing and Safety Monitoring, College of Life Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, China
- National Engineering Research Center of JUNCAO Technology, Fujian Agriculture and Forestry University, Fuzhou 350002, China
- Key Laboratory of Marine Biotechnology of Fujian Province, Institute of Oceanology, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Yuqian Wang
- Fujian Provincial Key Laboratory of Agroecological Processing and Safety Monitoring, College of Life Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, China
- Agricultural College, Anhui Science and Technology University, Chuzhou 233100, China
| | - Xiangmin Lin
- Fujian Provincial Key Laboratory of Agroecological Processing and Safety Monitoring, College of Life Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, China
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 102206, China
- Key Laboratory of Marine Biotechnology of Fujian Province, Institute of Oceanology, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Yanling Liu
- College of JunCao Science and Ecology, College of Life Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, China
- Fujian Provincial Key Laboratory of Agroecological Processing and Safety Monitoring, College of Life Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, China
- National Engineering Research Center of JUNCAO Technology, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| |
Collapse
|
16
|
Miao C, Huang Y, Zhang C, Wang X, Wang B, Zhou X, Song Y, Wu P, Chen ZS, Feng Y. Post-translational modifications in drug resistance. Drug Resist Updat 2025; 78:101173. [PMID: 39612546 DOI: 10.1016/j.drup.2024.101173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 10/24/2024] [Accepted: 11/16/2024] [Indexed: 12/01/2024]
Abstract
Resistance to antitumor drugs, antimicrobial drugs, and antiviral drugs severely limits treatment effectiveness and cure rate of diseases. Protein post-translational modifications (PTMs) represented by glycosylation, ubiquitination, SUMOylation, acetylation, phosphorylation, palmitoylation, and lactylation are closely related to drug resistance. PTMs are typically achieved by adding sugar chains (glycosylation), small proteins (ubiquitination), lipids (palmitoylation), or functional groups (lactylation) to amino acid residues. These covalent additions are usually the results of signaling cascades and could be reversible, with the triggering mechanisms depending on the type of modifications. PTMs are involved in antitumor drug resistance, not only as inducers of drug resistance but also as targets for reversing drug resistance. Bacteria exhibit multiple PTMs-mediated antimicrobial drug resistance. PTMs allow viral proteins and host cell proteins to form complex interaction networks, inducing complex antiviral drug resistance. This review summarizes the important roles of PTMs in drug resistance, providing new ideas for exploring drug resistance mechanisms, developing new drug targets, and guiding treatment plans.
Collapse
Affiliation(s)
- Chenggui Miao
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 999077, Hong Kong; Center for Xin'an Medicine and Modernization of Traditional Chinese Medicine of IHM, Anhui University of Chinese Medicine, Hefei 230012, China; Department of Pharmacology, School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei 230012, China
| | - Yurong Huang
- Department of Respiratory Medicine, Center of Infectious Diseases and Pathogen Biology, State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, The First Hospital, Jilin University, Changchun 130021, China
| | - Cheng Zhang
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 999077, Hong Kong
| | - Xiao Wang
- Department of Clinical Nursing, School of Nursing, Anhui University of Chinese Medicine, Hefei, China
| | - Bing Wang
- Department of Pharmacology, School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei 230012, China
| | - Xinyue Zhou
- Department of Pharmacology, School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei 230012, China
| | - Yingqiu Song
- Department of Pharmacology, School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei 230012, China
| | - Peng Wu
- Department of Anatomy, School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei 230012, China
| | - Zhe-Sheng Chen
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 999077, Hong Kong; Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY 11439, USA.
| | - Yibin Feng
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 999077, Hong Kong.
| |
Collapse
|
17
|
Muneer G, Chen C, Chen Y. Advancements in Global Phosphoproteomics Profiling: Overcoming Challenges in Sensitivity and Quantification. Proteomics 2025; 25:e202400087. [PMID: 39696887 PMCID: PMC11735659 DOI: 10.1002/pmic.202400087] [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: 07/24/2024] [Revised: 11/29/2024] [Accepted: 11/29/2024] [Indexed: 12/20/2024]
Abstract
Protein phosphorylation introduces post-genomic diversity to proteins, which plays a crucial role in various cellular activities. Elucidation of system-wide signaling cascades requires high-performance tools for precise identification and quantification of dynamics of site-specific phosphorylation events. Recent advances in phosphoproteomic technologies have enabled the comprehensive mapping of the dynamic phosphoproteomic landscape, which has opened new avenues for exploring cell type-specific functional networks underlying cellular functions and clinical phenotypes. Here, we provide an overview of the basics and challenges of phosphoproteomics, as well as the technological evolution and current state-of-the-art global and quantitative phosphoproteomics methodologies. With a specific focus on highly sensitive platforms, we summarize recent trends and innovations in miniaturized sample preparation strategies for micro-to-nanoscale and single-cell profiling, data-independent acquisition mass spectrometry (DIA-MS) for enhanced coverage, and quantitative phosphoproteomic pipelines for deep mapping of cell and disease biology. Each aspect of phosphoproteomic analysis presents unique challenges and opportunities for improvement and innovation. We specifically highlight evolving phosphoproteomic technologies that enable deep profiling from low-input samples. Finally, we discuss the persistent challenges in phosphoproteomic technologies, including the feasibility of nanoscale and single-cell phosphoproteomics, as well as future outlooks for biomedical applications.
Collapse
Affiliation(s)
- Gul Muneer
- Institute of ChemistryAcademia SinicaTaipeiTaiwan
| | | | - Yu‐Ju Chen
- Institute of ChemistryAcademia SinicaTaipeiTaiwan
- Department of ChemistryNational Taiwan UniversityTaipeiTaiwan
| |
Collapse
|
18
|
Kamran A, Hussain MD, Farooq T, Li F, Khan M, Li X, Yang S, Xie X. Deciphering intricate plant-virus interactions: Potyvirids orchestrate protein posttranslational modifications to regulate pathogenicity. Microbiol Res 2025; 290:127940. [PMID: 39536513 DOI: 10.1016/j.micres.2024.127940] [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: 07/26/2024] [Revised: 10/08/2024] [Accepted: 10/23/2024] [Indexed: 11/16/2024]
Abstract
In a molecular-arm-race between viruses and their hosts, viruses have evolved to harness their host's post-translational modifications (PTMs) machinery to gain a competitive edge. These modifications are the most reliable target of plant viruses to overcome the host defence for successful infection. Relatively fewer PTMs i.e., phosphorylation, O-GlcNAcylation, Ubiquitination, and SUMOylation have been studied regulating the potyvirus-plant interaction. Therefore, it is worth drawing attention towards the importance and potential of this undermined but key strategy of potyvirids (members of family Potyviridae) to abduct their host defence line, suggesting to review in detail the existing knowledge of these PTMs and highlight the unexplored modifications that might have played their part in establishing successful infection. The current review provides an understanding of how PTMs execute viral replication and infection dynamics during plant-potyvirid interactions. We highlighted that PTMs linked to CP, NIa-pro, NIb, and VPg are important to specify their host, virulence, overcoming host innate immunity, and most importantly disarming the host of RNA silencing tool of nailing any intruder. The limitations and potential improvements in studying undermined PTMs, including acetylation, glycosylation, methylation, and neddylation, as well as challenges and future perspectives of this inevitable process are mechanistically deciphered in the course of plant-virus interactions. This communication opens new avenues for investigating the fundamental mechanisms of virus infection and the development of new antiviral strategies for sustainable disease managements.
Collapse
Affiliation(s)
- Ali Kamran
- Key Laboratory of Agricultural Microbiology, College of Agriculture, Guizhou University, Guiyang 550025, China
| | - Muhammad Dilshad Hussain
- Key Laboratory of Agricultural Microbiology, College of Agriculture, Guizhou University, Guiyang 550025, China
| | - Tahir Farooq
- Guangdong Provincial Key Laboratory of High Technology for Plant Protection, Plant Protection Research Institute, Guangdong Academy of Agricultural Sciences, Guangzhou 510640, China
| | - Fangfang Li
- State Key Laboratory for Biology of Plant Diseases and Insect Pests, Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Beijing 100089, China
| | - Mehran Khan
- Key Laboratory of Agricultural Microbiology, College of Agriculture, Guizhou University, Guiyang 550025, China
| | - Xiangyang Li
- State Key Laboratory of Green Pesticide; Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Center for R&D of Fine Chemicals of Guizhou University, Guiyang 550025, China
| | - Sanwei Yang
- Key Laboratory of Agricultural Microbiology, College of Agriculture, Guizhou University, Guiyang 550025, China.
| | - Xin Xie
- Key Laboratory of Agricultural Microbiology, College of Agriculture, Guizhou University, Guiyang 550025, China.
| |
Collapse
|
19
|
Cao Y, Yu T, Zhu Z, Zhang Y, Sun S, Li N, Gu C, Yang Y. Exploring the landscape of post-translational modification in drug discovery. Pharmacol Ther 2025; 265:108749. [PMID: 39557344 DOI: 10.1016/j.pharmthera.2024.108749] [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: 07/21/2024] [Revised: 09/11/2024] [Accepted: 11/04/2024] [Indexed: 11/20/2024]
Abstract
Post-translational modifications (PTMs) play a crucial role in regulating protein function, and their dysregulation is frequently associated with various diseases. The emergence of epigenetic drugs targeting factors such as histone deacetylases (HDACs) and histone methyltransferase enhancers of zeste homolog 2 (EZH2) has led to a significant shift towards precision medicine, offering new possibilities to overcome the limitations of traditional therapeutics. In this review, we aim to systematically explore how small molecules modulate PTMs. We discuss the direct targeting of enzymes involved in PTM pathways, the modulation of substrate proteins, and the disruption of protein-enzyme interactions that govern PTM processes. Additionally, we delve into the emerging strategy of employing multifunctional molecules to precisely regulate the modification levels of proteins of interest (POIs). Furthermore, we examine the specific characteristics of these molecules, evaluating their therapeutic benefits and potential drawbacks. The goal of this review is to provide a comprehensive understanding of PTM-targeting strategies and their potential for personalized medicine, offering a forward-looking perspective on the evolution of precision therapeutics.
Collapse
Affiliation(s)
- Yuhao Cao
- Nanjing Hospital of Chinese Medicine affiliated to Nanjing University of Chinese Medicine, Nanjing 210022, China; School of Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Tianyi Yu
- School of Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Ziang Zhu
- School of Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Yuanjiao Zhang
- School of Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Shanliang Sun
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| | - Nianguang Li
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| | - Chunyan Gu
- Nanjing Hospital of Chinese Medicine affiliated to Nanjing University of Chinese Medicine, Nanjing 210022, China; School of Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| | - Ye Yang
- School of Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| |
Collapse
|
20
|
Xie L, Song D, Ouyang Z, Ning Y, Liu X, Li L, Xia W, Yang Y. USP27 promotes glycolysis and hepatocellular carcinoma progression by stabilizing PFKFB3 through deubiquitination. Cell Signal 2024; 127:111585. [PMID: 39746496 DOI: 10.1016/j.cellsig.2024.111585] [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: 11/10/2024] [Revised: 12/23/2024] [Accepted: 12/30/2024] [Indexed: 01/04/2025]
Abstract
Hepatocellular carcinoma (HCC) is associated with a dismal prognosis, primarily due to its high rates of metastasis and recurrence. Metabolic reprogramming, specifically enhanced glycolysis, is a prominent feature of cancer progression. This study identifies ubiquitin-specific peptidase 27 X-linked (USP27) as a significant regulator of glycolysis in HCC. We demonstrate that USP27 stabilizes PFKFB3, a key glycolytic enzyme, through deubiquitination, thereby increasing glycolytic activity and facilitating tumor progression. Furthermore, we reveal that CTCF, a well-known transcription factor, directly binds to the USP27 promoter and upregulates its expression, thereby establishing a connection between transcriptional regulation and metabolic reprogramming in HCC. Knockdown of USP27 or CTCF in HCC cells considerably decreased glycolysis and proliferation, while overexpression had the opposite effect. In vivo studies confirmed that USP27 knockdown suppresses HCC growth and metastasis. Our findings establish the CTCF/USP27/PFKFB3 axis as a novel mechanism driving HCC progression through glycolysis, indicating that targeting this pathway could offer new therapeutic opportunities. These results provide valuable insights into the molecular mechanisms underlying HCC and emphasize the potential of targeting USP27-mediated metabolic pathways as a strategy for cancer treatment.
Collapse
Affiliation(s)
- Longhui Xie
- Department of Hepatobiliary Pancreatic Spleen Surgery, The Central Hospital of Yongzhou, Yongzhou 425000, PR China
| | - Dekun Song
- Department of Hepatobiliary Surgery, Binzhou People's Hospital, Binzhou 256600, PR China
| | - Zhengsheng Ouyang
- Department of clinical medicine, YongZhou Vocational Technical College, Yongzhou 425000, PR China; Department of Hepatobiliary Pancreatic Spleen Surgery, The Central Hospital of Yongzhou, Yongzhou 425000, PR China
| | - Yinkuan Ning
- Department of Interventional Vascular Surgery, The Central Hospital of Shaoyang, Shaoyang 422000, PR China
| | - Xintao Liu
- Department of Hepatobiliary Pancreatic Spleen Surgery, The Central Hospital of Yongzhou, Yongzhou 425000, PR China
| | - Lai Li
- Department of Hepatobiliary Pancreatic Spleen Surgery, The Central Hospital of Yongzhou, Yongzhou 425000, PR China
| | - Wangning Xia
- Department of Hepatobiliary Pancreatic Spleen Surgery, The Central Hospital of Yongzhou, Yongzhou 425000, PR China
| | - Yang Yang
- Department of Oncology, The Central Hospital of Shaoyang, Shaoyang 422000, PR China.
| |
Collapse
|
21
|
Kim S, Kim E, Park M, Kim SH, Kim BG, Na S, Sadongo VW, Wijesinghe WCB, Eom YG, Yoon G, Jeong H, Hwang E, Lee C, Myung K, Kim CU, Choi JM, Min SK, Kwon TH, Min D. Hidden route of protein damage through oxygen-confined photooxidation. Nat Commun 2024; 15:10873. [PMID: 39738007 DOI: 10.1038/s41467-024-55168-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Accepted: 12/04/2024] [Indexed: 01/01/2025] Open
Abstract
Oxidative modifications can disrupt protein folds and functions, and are strongly associated with human aging and diseases. Conventional oxidation pathways typically involve the free diffusion of reactive oxygen species (ROS), which primarily attack the protein surface. Yet, it remains unclear whether and how internal protein folds capable of trapping oxygen (O2) contribute to oxidative damage. Here, we report a hidden pathway of protein damage, which we refer to as O2-confined photooxidation. In this process, O2 is captured in protein cavities and subsequently converted into multiple ROS, primarily mediated by tryptophan residues under blue light irradiation. The generated ROS then attack the protein interior through constrained diffusion, causing protein damage. The effects of this photooxidative reaction appear to be extensive, impacting a wide range of cellular proteins, as supported by whole-cell proteomic analysis. This photooxidative mechanism may represent a latent oxidation pathway in human tissues directly exposed to visible light, such as skin and eyes.
Collapse
Affiliation(s)
- Seoyoon Kim
- Department of Chemistry, Ulsan National Institute of Science and Technology, Ulsan, 44919, Republic of Korea
| | - Eojin Kim
- Department of Chemistry, Ulsan National Institute of Science and Technology, Ulsan, 44919, Republic of Korea
| | - Mingyu Park
- Department of Chemistry, Ulsan National Institute of Science and Technology, Ulsan, 44919, Republic of Korea
| | - Seong Ho Kim
- Department of Chemistry, Ulsan National Institute of Science and Technology, Ulsan, 44919, Republic of Korea
| | - Byung-Gyu Kim
- Center for Genomic Integrity, Institute for Basic Science, Ulsan, 44919, Republic of Korea
| | - Seungjin Na
- Digital Omics Research Center, Korea Basic Science Institute, Cheongju, 28119, Republic of Korea
| | - Victor W Sadongo
- Department of Chemistry, Ulsan National Institute of Science and Technology, Ulsan, 44919, Republic of Korea
| | - W C Bhashini Wijesinghe
- Department of Chemistry, Ulsan National Institute of Science and Technology, Ulsan, 44919, Republic of Korea
| | - Yu-Gon Eom
- Department of Chemistry, Pusan National University, Busan, 46241, Republic of Korea
| | - Gwangsu Yoon
- Department of Chemistry, Ulsan National Institute of Science and Technology, Ulsan, 44919, Republic of Korea
| | - Hannah Jeong
- Department of Physics, Ulsan National Institute of Science and Technology, Ulsan, 44919, Republic of Korea
| | - Eunhye Hwang
- Department of Chemistry, Ulsan National Institute of Science and Technology, Ulsan, 44919, Republic of Korea
| | - Chaiheon Lee
- Department of Chemistry, Ulsan National Institute of Science and Technology, Ulsan, 44919, Republic of Korea
| | - Kyungjae Myung
- Center for Genomic Integrity, Institute for Basic Science, Ulsan, 44919, Republic of Korea
- Department of Biomedical Engineering, Ulsan National Institute of Science and Technology, Ulsan, 44919, Republic of Korea
| | - Chae Un Kim
- Department of Physics, Ulsan National Institute of Science and Technology, Ulsan, 44919, Republic of Korea
| | - Jeong-Mo Choi
- Department of Chemistry, Pusan National University, Busan, 46241, Republic of Korea
- Chemistry Institute for Functional Materials, Pusan National University, Busan, 46241, Republic of Korea
| | - Seung Kyu Min
- Department of Chemistry, Ulsan National Institute of Science and Technology, Ulsan, 44919, Republic of Korea.
| | - Tae-Hyuk Kwon
- Department of Chemistry, Ulsan National Institute of Science and Technology, Ulsan, 44919, Republic of Korea.
- X-Dynamic Research Center, Ulsan National Institute of Science and Technology, Ulsan, 44919, Republic of Korea.
| | - Duyoung Min
- Department of Chemistry, Ulsan National Institute of Science and Technology, Ulsan, 44919, Republic of Korea.
- X-Dynamic Research Center, Ulsan National Institute of Science and Technology, Ulsan, 44919, Republic of Korea.
| |
Collapse
|
22
|
Chen L, Hu X, Wang G, Yu F, Dai Z, Jian X, Li Y, Xiang W, Meng Z. E3 ubiquitin ligase TRIM2 identified as a novel suppressor of CYP11B2 and aldosterone production. Cell Mol Life Sci 2024; 82:27. [PMID: 39725733 DOI: 10.1007/s00018-024-05545-0] [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: 09/19/2024] [Revised: 12/07/2024] [Accepted: 12/09/2024] [Indexed: 12/28/2024]
Abstract
Aldosterone-producing adenoma (APA) is a leading cause of primary aldosteronism (PA), a condition marked by excessive aldosterone secretion. CYP11B2, the aldosterone synthase, plays a critical role in aldosterone biosynthesis and the development of APA. Despite its significance, encoding regulatory mechanisms governing CYP11B2, particularly its degradation, remain poorly understood. In this study, we sought to uncover novel regulators of CYP11B2 stability by conducting a siRNA screen targeting E3 ubiquitin ligases. Our results identified TRIM2 as a key negative regulator of CYP11B2, where its overexpression led to a significant reduction in CYP11B2 protein levels and a concomitant decrease in aldosterone production in adrenal tumor cells. Mechanistically, we demonstrated that TRIM2 interacts with CYP11B2 via its RBCC domain, promoting K29/48-linked polyubiquitination and destabilization of CYP11B2. Further results revealed that TRIM2 is downregulated in APA tissues, showing differential expression between the zona glomerulosa (ZG) and zona fasciculata (ZF) of normal adrenal tissue. These findings highlight TRIM2 as a novel modulator of aldosterone synthesis through CYP11B2 degradation, offering a potential therapeutic target for APA.
Collapse
Affiliation(s)
- Liang Chen
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Urological Diseases, Wuhan, China
- Hubei Clinical Research Center for Laparoscopic/Endoscopic Urologic Surgery, Wuhan, China
- Institute of Urology, Wuhan University, Wuhan, China
- Hubei Medical Quality Control Center for Laparoscopic/Endoscopic Urologic Surgery, Wuhan, China
- Wuhan Clinical Research Center for Urogenital Tumors, Wuhan, China
| | - Xuan Hu
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Gang Wang
- Department of Biological Repositories, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Fang Yu
- Department of Pathology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Zhe Dai
- Department of Endocrinology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Xiaobin Jian
- Department of Pathology, The Second Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Yong Li
- Department of Urology, Jinning District People's Hospital, Kunming, China.
| | - Wan Xiang
- Department of Biological Repositories, Zhongnan Hospital of Wuhan University, Wuhan, China.
| | - Zhe Meng
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, China.
- Hubei Key Laboratory of Urological Diseases, Wuhan, China.
- Hubei Clinical Research Center for Laparoscopic/Endoscopic Urologic Surgery, Wuhan, China.
- Institute of Urology, Wuhan University, Wuhan, China.
- Hubei Medical Quality Control Center for Laparoscopic/Endoscopic Urologic Surgery, Wuhan, China.
- Wuhan Clinical Research Center for Urogenital Tumors, Wuhan, China.
- Department of Urology, Daye Hospital of Traditional Chinese Medicine, Huangshi, China.
| |
Collapse
|
23
|
Xu K, Fu H, Chen Q, Sun R, Li R, Zhao X, Zhou J, Wang X. Engineering thermostability of industrial enzymes for enhanced application performance. Int J Biol Macromol 2024; 291:139067. [PMID: 39730046 DOI: 10.1016/j.ijbiomac.2024.139067] [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: 11/14/2024] [Revised: 12/17/2024] [Accepted: 12/19/2024] [Indexed: 12/29/2024]
Abstract
Thermostability is a key factor for the industrial application of enzymes. This review categorizes enzymes by their applications and discusses the importance of engineering thermostability for practical use. It summarizes fundamental theories and recent advancements in enzyme thermostability modification, including directed evolution, semi-rational design, and rational design. Directed evolution uses high-throughput screening to generate random mutations, while semi-rational design combines hotspot identification with screening. Rational design focuses on key residues to enhance stability by improving rigidity, foldability, and reducing aggregation. The review also covers rational strategies like engineering folding energy, surface charge, machine learning methods, and consensus design, along with tools that support these approaches. Practical examples are critically assessed to highlight the benefits and limitations of these strategies. Finally, the challenges and potential contributions of artificial intelligence in enzyme thermostability engineering are discussed.
Collapse
Affiliation(s)
- Kangjie Xu
- Engineering Research Center of Ministry of Education on Food Synthetic Biotechnology and School of Biotechnology, Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu 214122, China; Science Center for Future Foods, Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu 214122, China
| | - Haoran Fu
- Engineering Research Center of Ministry of Education on Food Synthetic Biotechnology and School of Biotechnology, Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu 214122, China; Science Center for Future Foods, Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu 214122, China
| | - Qiming Chen
- Engineering Research Center of Ministry of Education on Food Synthetic Biotechnology and School of Biotechnology, Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu 214122, China; Science Center for Future Foods, Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu 214122, China
| | - Ruoxi Sun
- Engineering Research Center of Ministry of Education on Food Synthetic Biotechnology and School of Biotechnology, Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu 214122, China; Science Center for Future Foods, Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu 214122, China
| | - Ruosong Li
- Engineering Research Center of Ministry of Education on Food Synthetic Biotechnology and School of Biotechnology, Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu 214122, China; Science Center for Future Foods, Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu 214122, China
| | - Xinyi Zhao
- Engineering Research Center of Ministry of Education on Food Synthetic Biotechnology and School of Biotechnology, Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu 214122, China; Science Center for Future Foods, Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu 214122, China
| | - Jingwen Zhou
- Engineering Research Center of Ministry of Education on Food Synthetic Biotechnology and School of Biotechnology, Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu 214122, China; Science Center for Future Foods, Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu 214122, China; Jiangsu Province Engineering Research Center of Food Synthetic Biotechnology, Jiangnan University, Wuxi 214122, China; School of Biotechnology, Jiangnan University, 1800 Lihu Road, Wuxi 214122, China.
| | - Xinglong Wang
- Engineering Research Center of Ministry of Education on Food Synthetic Biotechnology and School of Biotechnology, Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu 214122, China; Science Center for Future Foods, Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu 214122, China.
| |
Collapse
|
24
|
Oliveira JC, Negreiro JM, Nunes FM, Barbosa FG, Mafezoli J, Mattos MC, Fernandes MCR, Pessoa C, Furtado CLM, Zanatta G, Oliveira MCF. In Silico Study of the Anti-MYC Potential of Lanostane-Type Triterpenes. ACS OMEGA 2024; 9:50844-50858. [PMID: 39741863 PMCID: PMC11683602 DOI: 10.1021/acsomega.4c10201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/09/2024] [Revised: 11/30/2024] [Accepted: 12/05/2024] [Indexed: 01/03/2025]
Abstract
One of the most investigated molecular targets for anticancer therapy is the proto-oncogene MYC, which is amplified and thus overexpressed in many types of cancer. Due to its structural characteristics, developing inhibitors for the target has proven to be challenging. In this study, the anti-MYC potential of lanostane-type triterpenes was investigated for the first time, using computational approaches that involved ensemble docking, prediction of structural properties and pharmacokinetic parameters, molecular dynamics (MD), and binding energy calculation using the molecular mechanics-generalized born surface area (MM-GBSA) method. The analysis of physicochemical properties, druglikeness, and pharmacokinetic parameters showed that ligands ganoderic acid E (I), ganoderlactone D (II), ganoderic acid Y (III), ganoderic acid Df (IV), lucidenic acid F (V), ganoderic acid XL4 (VI), mariesiic acid A (VII), and phellinol E (VIII) presented properties within the filter used. These eight ligands, in general, could interact with the molecular target favorably, with interaction energy values between -8.3 and -8.6 kcal mol-1. In MD, the results of RMSD, RMSF, radius of gyration, and hydrogen bonds of the complexes revealed that ligands I, IV, VI, and VII interacted satisfactorily with the protein during the simulations and assisted in its conformational and energetic stabilization. The binding energy calculation using the MM-GBSA method showed better results for the MYC-VII and MYC-I complexes (-44.98 and -41.96 kcal mol-1, respectively). These results support the hypothesis that such molecules can interact with MYC for a considerable period, which would be an essential condition for them to exert their inhibitory activity effectively.
Collapse
Affiliation(s)
- José
A. C. Oliveira
- Department
of Organic and Inorganic Chemistry, Science Center, Federal University of Ceará, Fortaleza, CE 60455-760, Brazil
| | - Jonatas M. Negreiro
- Department
of Organic and Inorganic Chemistry, Science Center, Federal University of Ceará, Fortaleza, CE 60455-760, Brazil
| | - Fátima M. Nunes
- Department
of Organic and Inorganic Chemistry, Science Center, Federal University of Ceará, Fortaleza, CE 60455-760, Brazil
| | - Francisco G. Barbosa
- Department
of Organic and Inorganic Chemistry, Science Center, Federal University of Ceará, Fortaleza, CE 60455-760, Brazil
| | - Jair Mafezoli
- Department
of Organic and Inorganic Chemistry, Science Center, Federal University of Ceará, Fortaleza, CE 60455-760, Brazil
| | - Marcos C. Mattos
- Department
of Organic and Inorganic Chemistry, Science Center, Federal University of Ceará, Fortaleza, CE 60455-760, Brazil
| | - Maria C. R. Fernandes
- Drug Research
and Development Center, Federal University
of Ceará, Rua Coronel Nunes de Melo, 1000, Fortaleza, CE 60430-275, Brazil
| | - Claudia Pessoa
- Drug Research
and Development Center, Federal University
of Ceará, Rua Coronel Nunes de Melo, 1000, Fortaleza, CE 60430-275, Brazil
| | - Cristiana L. M. Furtado
- Drug Research
and Development Center, Federal University
of Ceará, Rua Coronel Nunes de Melo, 1000, Fortaleza, CE 60430-275, Brazil
- Graduate
Program in Medical Sciences, University
of Fortaleza, Rua Francisco
Segundo da Costa, 23-57, Fortaleza, CE 60811-650, Brazil
| | - Geancarlo Zanatta
- Department
of Biophysics, Bioscience Institute, Federal
University of Rio Grande do Sul, Av. Bento Gonçalves, 9500, Building 43422, Laboratory
204, Porto Alegre, RS 91501-970, Brazil
| | - Maria C. F. Oliveira
- Department
of Organic and Inorganic Chemistry, Science Center, Federal University of Ceará, Fortaleza, CE 60455-760, Brazil
| |
Collapse
|
25
|
Pan Q, Zhang Z, Xiong Y, Bao Y, Chen T, Xu P, Liu Z, Ma H, Yu Y, Zhou Z, Wei W. Mapping functional elements of the DNA damage response through base editor screens. Cell Rep 2024; 43:115047. [PMID: 39661519 DOI: 10.1016/j.celrep.2024.115047] [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: 01/28/2024] [Revised: 09/05/2024] [Accepted: 11/19/2024] [Indexed: 12/13/2024] Open
Abstract
Maintaining genomic stability is vital for cellular equilibrium. In this study, we combined CRISPR-mediated base editing with pooled screening technologies to identify numerous mutations in lysine residues and protein-coding genes. The loss of these lysine residues and genes resulted in either sensitivity or resistance to DNA-damaging agents. Among the identified variants, we characterized both loss-of-function and gain-of-function mutations in response to DNA damage. Notably, we discovered that the K494 mutation of C17orf53 disrupts its interaction with RPA proteins, leading to increased sensitivity to cisplatin. Additionally, our analysis identified STK35 as a previously unrecognized gene involved in DNA damage response (DDR) pathways, suggesting that it may play a critical role in DNA repair. We believe that this resource will offer valuable insights into the broader functions of DNA damage response genes and accelerate research on variants relevant to cancer therapy.
Collapse
Affiliation(s)
- Qian Pan
- Biomedical Pioneering Innovation Center, Beijing Advanced Innovation Center for Genomics, Peking-Tsinghua Center for Life Sciences, Peking University Genome Editing Research Center, State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking University, Beijing 100871, China
| | - Zhixuan Zhang
- Biomedical Pioneering Innovation Center, Beijing Advanced Innovation Center for Genomics, Peking-Tsinghua Center for Life Sciences, Peking University Genome Editing Research Center, State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking University, Beijing 100871, China
| | - Yangfang Xiong
- Biomedical Pioneering Innovation Center, Beijing Advanced Innovation Center for Genomics, Peking-Tsinghua Center for Life Sciences, Peking University Genome Editing Research Center, State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking University, Beijing 100871, China
| | - Ying Bao
- Biomedical Pioneering Innovation Center, Beijing Advanced Innovation Center for Genomics, Peking-Tsinghua Center for Life Sciences, Peking University Genome Editing Research Center, State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking University, Beijing 100871, China; Changping Laboratory, Beijing 102206, China
| | - Tianxin Chen
- Biomedical Pioneering Innovation Center, Beijing Advanced Innovation Center for Genomics, Peking-Tsinghua Center for Life Sciences, Peking University Genome Editing Research Center, State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking University, Beijing 100871, China
| | - Ping Xu
- Biomedical Pioneering Innovation Center, Beijing Advanced Innovation Center for Genomics, Peking-Tsinghua Center for Life Sciences, Peking University Genome Editing Research Center, State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking University, Beijing 100871, China
| | - Zhiheng Liu
- Biomedical Pioneering Innovation Center, Beijing Advanced Innovation Center for Genomics, Peking-Tsinghua Center for Life Sciences, Peking University Genome Editing Research Center, State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking University, Beijing 100871, China
| | - Huazheng Ma
- Biomedical Pioneering Innovation Center, Beijing Advanced Innovation Center for Genomics, Peking-Tsinghua Center for Life Sciences, Peking University Genome Editing Research Center, State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking University, Beijing 100871, China
| | - Ying Yu
- Biomedical Pioneering Innovation Center, Beijing Advanced Innovation Center for Genomics, Peking-Tsinghua Center for Life Sciences, Peking University Genome Editing Research Center, State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking University, Beijing 100871, China
| | - Zhuo Zhou
- Biomedical Pioneering Innovation Center, Beijing Advanced Innovation Center for Genomics, Peking-Tsinghua Center for Life Sciences, Peking University Genome Editing Research Center, State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking University, Beijing 100871, China; State Key Laboratory of Common Mechanism Research for Major Diseases, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou 215123, Jiangsu, China
| | - Wensheng Wei
- Biomedical Pioneering Innovation Center, Beijing Advanced Innovation Center for Genomics, Peking-Tsinghua Center for Life Sciences, Peking University Genome Editing Research Center, State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking University, Beijing 100871, China; Changping Laboratory, Beijing 102206, China.
| |
Collapse
|
26
|
Sun Y, Gu X, Qu C, Jin N, Qin T, Jin L, Huang J. OsPUB75-OsHDA716 mediates deactivation and degradation of OsbZIP46 to negatively regulate drought tolerance in rice. PLANT PHYSIOLOGY 2024; 197:kiae545. [PMID: 39405437 DOI: 10.1093/plphys/kiae545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Accepted: 08/20/2024] [Indexed: 12/24/2024]
Abstract
Histone deacetylases (HDACs) play crucial roles in plant stress responses via modification of histone as well as nonhistone proteins; however, how HDAC-mediated deacetylation of nonhistone substrates affects protein functions remains elusive. Here, we report that the reduced potassium dependency3/histone deacetylase1-type histone deacetylase OsHDA716 and plant U-box E3 ubiquitin ligase OsPUB75 form a complex to regulate rice drought response via deactivation and degradation of basic leucine zipper (bZIP) transcription factor OsbZIP46 in rice (Oryza sativa). OsHDA716 decreases abscisic acid (ABA)-induced drought tolerance, and mechanistic investigations showed that OsHDA716 interacts with and deacetylates OsbZIP46, a key regulator in ABA signaling and drought response, thus inhibiting its transcriptional activity. Furthermore, OsHDA716 recruits OsPUB75 to facilitate ubiquitination and degradation of deacetylated OsbZIP46. Therefore, the OsPUB75-OsHDA716 complex exerts double restrictions on the transcriptional activity and protein stability of OsbZIP46, leading to repression of downstream drought-responsive gene expression and consequently resulting in reduced drought tolerance. Conversely, OsbZIP46 acts as an upstream repressor to repress OsHDA716 expression, and therefore OsHDA716 and OsbZIP46 form an antagonistic pair to reciprocally inhibit each other. Genetic evidence showed that OsHDA716 works with OsbZIP46 in a common pathway to antagonistically regulate rice drought response, revealing that plants can fine-tune stress responses by the complex interplay between chromatin regulators and transcription factors. Our findings unveil an acetylation-dependent regulatory mechanism governing protein functions and shed light on the precise coordination of activity and stability of key transcription factors through a combination of different posttranslational modifications.
Collapse
Affiliation(s)
- Ying Sun
- Key Laboratory of Biorheological Science and Technology of Ministry of Education, Bioengineering College, Chongqing University, Chongqing 400044, China
| | - Xinyue Gu
- Key Laboratory of Biorheological Science and Technology of Ministry of Education, Bioengineering College, Chongqing University, Chongqing 400044, China
| | - Chengfeng Qu
- Key Laboratory of Biorheological Science and Technology of Ministry of Education, Bioengineering College, Chongqing University, Chongqing 400044, China
| | - Ning Jin
- Key Laboratory of Biorheological Science and Technology of Ministry of Education, Bioengineering College, Chongqing University, Chongqing 400044, China
| | - Tian Qin
- Key Laboratory of Biorheological Science and Technology of Ministry of Education, Bioengineering College, Chongqing University, Chongqing 400044, China
| | - Liang Jin
- Key Laboratory of Biorheological Science and Technology of Ministry of Education, Bioengineering College, Chongqing University, Chongqing 400044, China
| | - Junli Huang
- Key Laboratory of Biorheological Science and Technology of Ministry of Education, Bioengineering College, Chongqing University, Chongqing 400044, China
| |
Collapse
|
27
|
Sun X, LaVoie M, Lefebvre PA, Gallaher SD, Glaesener AG, Strenkert D, Mehta R, Merchant SS, Silflow CD. Identification of a gene controlling levels of the copper response regulator 1 transcription factor in Chlamydomonas reinhardtii. THE PLANT CELL 2024; 37:koae300. [PMID: 39777451 PMCID: PMC11708838 DOI: 10.1093/plcell/koae300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Accepted: 11/08/2024] [Indexed: 01/11/2025]
Abstract
Oxygen prevents hydrogen production in Chlamydomonas (Chlamydomonas reinhardtii), in part by inhibiting the transcription of hydrogenase genes. We developed a screen for mutants showing constitutive accumulation of iron hydrogenase 1 (HYDA1) transcripts in normoxia. A reporter gene required for ciliary motility placed under the control of the HYDA1 promoter conferred motility only in hypoxia. By selecting for mutants able to swim even in normoxia, we obtained strains that constitutively express the reporter gene. One identified mutant was affected in a gene encoding an F-box protein 3 (FBXO3) that participates in ubiquitylation and proteasomal degradation pathways in other eukaryotes. Transcriptome profiles revealed that the mutation, termed cehc1-1 (constitutive expression of hydrogenases and copper-responsive genes), triggers the upregulation of genes known to be targets of copper response regulator 1 (CRR1), a transcription factor involved in the nutritional copper signaling pathway and in the hypoxia response pathway. CRR1 was required for upregulating the HYDA1 reporter gene expression in response to hypoxia and for the constitutive expression of the reporter gene in cehc1-1 mutant cells. The CRR1 protein, normally degraded in Cu-supplemented cells, was stabilized in cehc1-1 cells, supporting the conclusion that CEHC1 facilitates CRR1 degradation. Our results describe a previously unknown pathway for CRR1 inhibition and possibly other pathways leading to complex metabolic changes.
Collapse
Affiliation(s)
- Xiaoqing Sun
- Department of Plant and Microbial Biology, University of Minnesota, St. Paul, MN 55108, USA
| | - Matthew LaVoie
- Department of Plant and Microbial Biology, University of Minnesota, St. Paul, MN 55108, USA
| | - Paul A Lefebvre
- Department of Plant and Microbial Biology, University of Minnesota, St. Paul, MN 55108, USA
| | - Sean D Gallaher
- Quantitative Biosciences Institute, University of California, Berkeley, CA 94720, USA
| | - Anne G Glaesener
- Quantitative Biosciences Institute, University of California, Berkeley, CA 94720, USA
| | - Daniela Strenkert
- Quantitative Biosciences Institute, University of California, Berkeley, CA 94720, USA
| | - Radhika Mehta
- Quantitative Biosciences Institute, University of California, Berkeley, CA 94720, USA
| | - Sabeeha S Merchant
- Quantitative Biosciences Institute, University of California, Berkeley, CA 94720, USA
| | - Carolyn D Silflow
- Department of Plant and Microbial Biology, University of Minnesota, St. Paul, MN 55108, USA
| |
Collapse
|
28
|
Wang J, Miao J, Zhu P. Insights into the complexities of Citrullination: From immune regulation to autoimmune disease. Autoimmun Rev 2024; 24:103734. [PMID: 39719187 DOI: 10.1016/j.autrev.2024.103734] [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: 11/07/2024] [Revised: 12/12/2024] [Accepted: 12/19/2024] [Indexed: 12/26/2024]
Abstract
Citrullination, a post-translational modification that changes arginine to citrulline in proteins, is vital for immune response modulation and cell signaling. Catalyzed by peptidyl arginine deiminases (PADs), citrullination is linked to various diseases, particularly autoimmune disorders like rheumatoid arthritis (RA). Citrullinated proteins can trigger the production of anti-citrullinated protein antibodies (ACPAs), included in RA classification criteria. The immune response to citrullination involves both innate and adaptive immunity, affecting monocytes/macrophages, neutrophils, dendritic cells, natural killer cells, B cells, and T cells. Citrullination contributes to disease development in RA and other conditions such as multiple sclerosis, sepsis, and cancer. Therapeutic strategies targeting citrullination and its effects are being explored, including B cell depletion therapies, T cell-directed approaches, PAD inhibitors, and citrullinated peptide-based vaccines. Understanding the interplay between citrullination and the immune system may lead to novel diagnostic tools and targeted therapies for autoimmune diseases and beyond.
Collapse
Affiliation(s)
- Jiawei Wang
- Department of Clinical Immunology of Xijing Hospital and Department of Cell Biology of National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi'an, 710032, Shaanxi, China
| | - Jinlin Miao
- Department of Clinical Immunology of Xijing Hospital and Department of Cell Biology of National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi'an, 710032, Shaanxi, China.
| | - Ping Zhu
- Department of Clinical Immunology of Xijing Hospital and Department of Cell Biology of National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi'an, 710032, Shaanxi, China.
| |
Collapse
|
29
|
Yamamoto T, Furukawa A, Zhou Y, Kono N, Kitajima S, Ohguchi H, Kawano Y, Ito S, Araki N, Ohtsuki S, Masuda T. Increased CSN5 expression enhances the sensitivity to lenalidomide in multiple myeloma cells. iScience 2024; 27:111399. [PMID: 39687025 PMCID: PMC11647120 DOI: 10.1016/j.isci.2024.111399] [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: 03/27/2024] [Revised: 08/21/2024] [Accepted: 11/12/2024] [Indexed: 12/18/2024] Open
Abstract
Lenalidomide (LEN) is commonly used as an effective therapeutic agent for multiple myeloma (MM). However, in some patients, primary resistance to LEN is observed, the mechanisms of which remain poorly understood. In this study, we combined a LEN sensitivity assay with proteomics data from 15 MM cell lines to identify protein expression profiles associated with primary LEN resistance. Our findings revealed that CSN5 expression is lower in LEN-resistant cell lines than in LEN-sensitive lines. Moreover, we established that CSN5 is degraded via the cullin-RING ubiquitin ligase (CRL)-mediated ubiquitin-proteasome pathway through ubiquitination at lysine 194. Our data suggest that reduced CSN5 expression leads to abnormalities in the ubiquitination cycle of CRL4A, resulting in the inhibition of LEN-mediated degradation of IKZF1 and IKZF3. These findings delineate an additional mechanism of LEN resistance in MM cells and may contribute to the development of alternative therapeutic strategies to overcome LEN resistance.
Collapse
Affiliation(s)
- Takumi Yamamoto
- Department of Pharmaceutical Microbiology, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto 862-0973, Japan
| | - Arisu Furukawa
- Department of Pharmaceutical Microbiology, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto 862-0973, Japan
| | - Yue Zhou
- Department of Cancer Cell Biology, Faculty of Pharmaceutical Sciences, University of Toyama, Toyama 930-0194, Japan
| | - Nobuaki Kono
- Institute for Advanced Biosciences, Keio University, Yamagata 997-0017, Japan
| | - Shojiro Kitajima
- Institute for Advanced Biosciences, Keio University, Yamagata 997-0017, Japan
| | - Hiroto Ohguchi
- Division of Disease Epigenetics, Institute of Resource Development and Analysis, Kumamoto University, Kumamoto 860-0811, Japan
| | - Yawara Kawano
- Department of Hematology, Rheumatology, and Infectious Disease, Faculty of Life Sciences, Kumamoto University, Kumamoto 860-8556, Japan
| | - Shingo Ito
- Department of Pharmaceutical Microbiology, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto 862-0973, Japan
- Department of Pharmaceutical Microbiology, Faculty of Life Sciences, Kumamoto University, Kumamoto 862-0973, Japan
| | - Norie Araki
- Department of Tumor Genetics and Biology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto 860-8556, Japan
| | - Sumio Ohtsuki
- Department of Pharmaceutical Microbiology, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto 862-0973, Japan
- Department of Pharmaceutical Microbiology, Faculty of Life Sciences, Kumamoto University, Kumamoto 862-0973, Japan
| | - Takeshi Masuda
- Institute for Advanced Biosciences, Keio University, Yamagata 997-0017, Japan
| |
Collapse
|
30
|
An D, Kim J, Moon B, Kim H, Nguyen H, Park S, Lee JE, Kim JA, Kim J. PRMT1-mediated methylation regulates MLL2 stability and gene expression. Nucleic Acids Res 2024:gkae1227. [PMID: 39698834 DOI: 10.1093/nar/gkae1227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 11/01/2024] [Accepted: 11/28/2024] [Indexed: 12/20/2024] Open
Abstract
The interplay between multiple transcription factors precisely regulates eukaryotic transcription. Here, we report that the protein methyltransferases, MLL2/KMT2B and PRMT1, interact directly and act collectively to regulate gene expression. PRMT1 binds to the N-terminal region of MLL2, considered an intrinsically disordered region, and methylates multiple arginine residues within its RGG/RG motifs. Notably, overexpression of PRMT1 decreased poly-ubiquitylation of MLL2, whereas mutations on methylation sites in MLL2 increased MLL2 poly-ubiquitylation, suggesting that PRMT1-mediated methylation stabilizes MLL2. MLL2 and PRMT1 cooperatively stimulated the expression of a chromosomal reporter gene in a PRMT1-mediated, MLL2-methylation-dependent manner. RNA-seq analysis found that MLL2 and PRMT1 jointly regulate the expression of genes involved in cell membrane and extracellular matrix functions, and depletion of either resulted in impaired cell migration and invasion. Our study provides evidence that PRMT1-mediated MLL2 methylation regulates MLL2 protein stability and the expression of their target genes.
Collapse
Affiliation(s)
- Dongju An
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon 34141, South Korea
| | - Jihyun Kim
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon 34141, South Korea
| | - Byul Moon
- Aging Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, South Korea
| | - Hyoungmin Kim
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon 34141, South Korea
| | - Hoa Nguyen
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon 34141, South Korea
| | - Sunghu Park
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon 34141, South Korea
| | - J Eugene Lee
- Division of Biomedical Metrology, Korea Research Institute of Standards and Science, Daejeon 34113, South Korea
| | - Jung-Ae Kim
- Aging Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, South Korea
- Disease Target Structure Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, South Korea
- Department of Bioscience, University of Science and Technology, Daejeon 34113, South Korea
| | - Jaehoon Kim
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon 34141, South Korea
| |
Collapse
|
31
|
Tian K, Yao Z, Pan D. Leveraging single-cell and multi-omics approaches to identify MTOR-centered deubiquitination signatures in esophageal cancer therapy. Front Immunol 2024; 15:1490623. [PMID: 39742278 PMCID: PMC11685190 DOI: 10.3389/fimmu.2024.1490623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Accepted: 11/28/2024] [Indexed: 01/03/2025] Open
Abstract
Background Esophageal squamous cell carcinoma (ESCC) remains a significant challenge in oncology due to its aggressive nature and heterogeneity. As one of the deadliest malignancies, ESCC research lags behind other cancer types. The balance between ubiquitination and deubiquitination processes plays a crucial role in cellular functions, with its disruption linked to various diseases, including cancer. Methods Our study utilized diverse analytical approaches, encompassing Cox regression models, single-cell RNA sequencing, intercellular communication analysis, and Gene Ontology enrichment. We also conducted mutation profiling and explored potential immunotherapeutic agents. Furthermore, in vitro cellular experiments and in vivo mouse models were performed to validate findings. These methodologies aimed to establish deubiquitination-related gene signatures (DRGS) for predicting ESCC patient outcomes and response to immunotherapy. Results By integrating datasets from TCGA-ESCC and GSE53624, we developed a DRGS model based on 14 deubiquitination-related genes (DUBGs). This signature effectively forecasts ESCC prognosis, drug responsiveness, and immune cell infiltration patterns. It also influences the mutational landscape of patients. Those classified as high-risk exhibited reduced survival rates, increased genetic alterations, and more complex cellular interactions, potentially explaining their poor outcomes. Notably, in vitro and in vivo experiments identified MTOR, a key component of the signature, as a promising therapeutic target for ESCC treatment. Conclusion Our research highlights the significance of 14 DUBGs in ESCC progression. The risk score derived from this gene set enables clinical stratification of patients into distinct prognostic groups. Moreover, MTOR emerges as a potential target for personalized ESCC therapy, offering new avenues for treatment strategies.
Collapse
Affiliation(s)
- Kang Tian
- Department of Oncology, The Affiliated Suqian Hospital of Xuzhou Medical University, Suqian, China
| | - Ziang Yao
- Department of Traditional Chinese Medicine, Peking University People’s Hospital, Beijing, China
| | - Da Pan
- Department of Gastroenterology, Wenzhou Central Hospital, Wenzhou, China
| |
Collapse
|
32
|
Binoy A, Nanjan P, Chellamuthu K, Liu H, Zhu S. A click chemistry-based biorthogonal approach for the detection and identification of protein lysine malonylation for osteoarthritis research. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.12.628274. [PMID: 39713453 PMCID: PMC11661220 DOI: 10.1101/2024.12.12.628274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/24/2024]
Abstract
Lysine malonylation is a post-translational modification where a malonyl group, characterized by a negatively charged carboxylate, is covalently attached to the Ɛ-amino side chain of lysine, influencing protein structure and function. Our laboratory identified Mak upregulation in cartilage under aging and obesity, contributing to osteoarthritis (OA). Current antibody-based detection methods face limitations in identifying Mak targets. Here, we introduce an alkyne-functionalized probe, MA-diyne, which metabolically incorporates into proteins, enabling copper(I) ion-catalyzed click reactions to conjugate labeled proteins with azide-based fluorescent dyes or affinity purification tags. In-gel fluorescence confirms MA-diyne incorporation into proteins across various cell types and species, including mouse chondrocytes, adipocytes, Hek293T cells, and C. elegans. Pull-down experiments identified known Mak proteins such as GAPDH and Aldolase. The extent of MA-diyne modification was higher in Sirtuin 5-deficient cells suggesting these modified proteins are Sirtuin 5 substrates. Pulse-chase experiments confirmed the dynamic nature of protein malonylation. Quantitative proteomics identified 1136 proteins corresponding to 8903 peptides with 429 proteins showing 1-fold increase in labeled group. Sirtuin 5 regulated 374 of these proteins. Pull down of newly identified proteins such as β-actin and Stat3 was also done. This study highlights MA-diyne as a powerful chemical tool to investigate the molecular targets and functions of lysine malonylation in OA conditions.
Collapse
Affiliation(s)
- Anupama Binoy
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine (HCOM), Ohio University, Athens, OH, 45701, USA
- Ohio Musculoskeletal and Neurological Institute (OMNI), Heritage College of Osteopathic Medicine (HCOM), Ohio University, Athens, OH, 45701, USA
| | - Pandurangan Nanjan
- Department of Chemistry, Amrita School of Physical Sciences, Amrita Vishwa Vidyapeetham, Coimbatore Campus, Tamilnadu, 641112, India
| | - Kavya Chellamuthu
- Department of Chemistry, Amrita School of Physical Sciences, Amrita Vishwa Vidyapeetham, Coimbatore Campus, Tamilnadu, 641112, India
| | - Huanhuan Liu
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine (HCOM), Ohio University, Athens, OH, 45701, USA
- Ohio Musculoskeletal and Neurological Institute (OMNI), Heritage College of Osteopathic Medicine (HCOM), Ohio University, Athens, OH, 45701, USA
| | - Shouan Zhu
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine (HCOM), Ohio University, Athens, OH, 45701, USA
- Ohio Musculoskeletal and Neurological Institute (OMNI), Heritage College of Osteopathic Medicine (HCOM), Ohio University, Athens, OH, 45701, USA
- Diabetes Institute (DI), Heritage College of Osteopathic Medicine (HCOM), Ohio University, Athens, OH, 45701, USA
| |
Collapse
|
33
|
Gou Q, Yan B, Duan Y, Guo Y, Qian J, Shi J, Hou Y. Ubiquitination of CD47 Regulates Innate Anti-Tumor Immune Response. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024:e2412205. [PMID: 39665172 DOI: 10.1002/advs.202412205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 11/16/2024] [Indexed: 12/13/2024]
Abstract
In addition to adaptive immune checkpoint of PD-1/PD-L1, the innate immune checkpoint SIRPα/CD47 plays an important role in regulation of tumor immune escape. However, the mechanism of CD47 ubiquitination on tumor immune escape remains unclear. Here it is found that TRAF2 bound to the C-terminal of CD47 cytoplasmic fragment and induced its ubiquitination, leading to inhibition of CD47 autophagic degradation by disrupting its binding to LC3, which in turn inhibited macrophage phagocytosis and promoted tumor immune escape. In contrast, loss of TRAF2 facilitated CD47 autophagic degradation and inhibited tumor immune escape. Moreover, autophagy induction promoted CD47 degradation and enhanced the efficacy of CD47 antibody anti-tumor immunotherapy. These findings revealed a novel mechanism of ubiquitination of CD47 on tumor immune escape.
Collapse
Affiliation(s)
- Qian Gou
- School of Life Science, Jiangsu University, Zhenjiang, Jiangsu Province, 212013, People's Republic of China
| | - Bingjun Yan
- School of Life Science, Jiangsu University, Zhenjiang, Jiangsu Province, 212013, People's Republic of China
| | - Yalan Duan
- School of Life Science, Jiangsu University, Zhenjiang, Jiangsu Province, 212013, People's Republic of China
| | - Yilei Guo
- School of Life Science, Jiangsu University, Zhenjiang, Jiangsu Province, 212013, People's Republic of China
| | - Jing Qian
- School of Life Science, Jiangsu University, Zhenjiang, Jiangsu Province, 212013, People's Republic of China
| | - Juanjuan Shi
- School of Life Science, Jiangsu University, Zhenjiang, Jiangsu Province, 212013, People's Republic of China
| | - Yongzhong Hou
- School of Life Science, Jiangsu University, Zhenjiang, Jiangsu Province, 212013, People's Republic of China
| |
Collapse
|
34
|
Soni KK, Gurjar K, Ranjan A, Sinha S, Srivastava M, Verma V. Post-translational modifications control the signal at the crossroads of plant-pathogen interactions. JOURNAL OF EXPERIMENTAL BOTANY 2024; 75:6957-6979. [PMID: 39177255 DOI: 10.1093/jxb/erae358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Accepted: 08/22/2024] [Indexed: 08/24/2024]
Abstract
The co-evolution of plants and pathogens has enabled them to 'outsmart' each other by promoting their own defence responses and suppressing those of the other. While plants are reliant on their sophisticated immune signalling pathways, pathogens make use of effector proteins to achieve the objective. This entails rapid regulation of underlying molecular mechanisms for prompt induction of associated signalling events in both plants as well as pathogens. The past decade has witnessed the emergence of post-translational modification (PTM) of proteins as a key a factor in modulating cellular responses. The ability of PTMs to expand the functional diversity of the proteome and induce rapid changes at the appropriate time enables them to play crucial roles in the regulation of plant-pathogen interactions. Therefore, this review will delve into the intricate interplay of five major PTMs involved in plant defence and pathogen countermeasures. We discuss how plants employ PTMs to fortify their immune networks, and how pathogen effectors utilize/target host modification systems to gain entry into plants and cause disease. We also emphasize the need for identification of novel PTMs and propose the use of PTM pathways as potential targets for genome editing approaches.
Collapse
Affiliation(s)
- Kamlesh Kumar Soni
- Department of Biotechnology, AKS University, Satna, Madhya Pradesh-485001, India
| | - Kishan Gurjar
- Department of Biotechnology, School of Life Sciences, Central University of Rajasthan, Kishangarh, Ajmer, Rajasthan-305817, India
| | - Aastha Ranjan
- Department of Biotechnology, School of Life Sciences, Central University of Rajasthan, Kishangarh, Ajmer, Rajasthan-305817, India
| | - Shashank Sinha
- Department of Biotechnology, School of Life Sciences, Central University of Rajasthan, Kishangarh, Ajmer, Rajasthan-305817, India
| | - Moumita Srivastava
- Plant Biotechnology and Disease Biology, Rajiv Gandhi Centre for Biotechnology, Thycaud Post, Poojappura, Thiruvananthapuram, Kerala-695014, India
| | - Vivek Verma
- Department of Biotechnology, School of Life Sciences, Central University of Rajasthan, Kishangarh, Ajmer, Rajasthan-305817, India
- Plant Biotechnology Department, Gujarat Biotechnology University, Near Gujarat International Finance Tec City, Gandhinagar, Gujarat-382355, India
| |
Collapse
|
35
|
Ma Y, Jia R, Chen S, Ma J, Yin L, Pan X, He Y, Wu T, Zhao Z, Ma L, Wu S, Wang H, Liang G, Huang S, Sun X. Ubiquitin‐Proteasome System in Periodontitis: Mechanisms and Clinical Implications. Cell Prolif 2024. [DOI: 10.1111/cpr.13781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Accepted: 11/09/2024] [Indexed: 01/03/2025] Open
Abstract
ABSTRACTThe progression of periodontitis, a bacteria‐driven inflammatory and bone‐destructive disease, involves myriad cellular and molecular mechanisms. Protein regulation significantly influences the pathogenesis and management of periodontitis. However, research regarding its regulatory role in periodontitis remains relatively limited. The ubiquitin‐proteasome system (UPS), which mainly involves ubiquitination by E3 ubiquitin ligases (E3s) and deubiquitination by deubiquitinating enzymes (DUBs), is the primary intracellular and non‐lysosomal mechanism of protein degradation. Recent studies have provided compelling evidence to support the involvement of UPS in periodontitis progression. Increasing evidence indicated that E3s, such as CUL3, Nedd4‐2, Synoviolin, FBXL19, PDLIM2, TRIMs and TRAFs, modulate inflammatory responses and bone resorption in periodontitis through multiple classical signalling pathways, including NLRP3, GSDMD, NF‐κB, Wnt/β‐catenin and Nrf2. Meanwhile, DUBs, including OTUD1, A20, CYLD, UCH‐L1 and USPs, also broadly modulate periodontitis progression by regulating signalling pathways such as NF‐κB, Wnt/β‐catenin, NLRP3, and BMP2. Therefore, the modulation of E3s and DUBs has proven to be an effective therapy against periodontitis. This review provides a comprehensive overview of the regulatory role of ubiquitinating and deubiquitinating enzymes in periodontitis progression and the underlying mechanisms. Finally, we summarise several chemical and genetic methods that regulate UPS enzymes and pave the way for the development of targeted therapies for periodontitis.
Collapse
Affiliation(s)
- Yilin Ma
- Institute of Stomatology, School and Hospital of Stomatology Wenzhou Medical University Wenzhou China
| | - Ruiwei Jia
- Institute of Stomatology, School and Hospital of Stomatology Wenzhou Medical University Wenzhou China
| | - Shuhong Chen
- Institute of Stomatology, School and Hospital of Stomatology Wenzhou Medical University Wenzhou China
| | - Jun Ma
- Institute of Stomatology, School and Hospital of Stomatology Wenzhou Medical University Wenzhou China
| | - Lei Yin
- Institute of Stomatology, School and Hospital of Stomatology Wenzhou Medical University Wenzhou China
| | - Xingbei Pan
- Institute of Stomatology, School and Hospital of Stomatology Wenzhou Medical University Wenzhou China
| | - Yunuo He
- Institute of Stomatology, School and Hospital of Stomatology Wenzhou Medical University Wenzhou China
| | - Tong Wu
- Institute of Stomatology, School and Hospital of Stomatology Wenzhou Medical University Wenzhou China
| | - Zheyu Zhao
- Institute of Stomatology, School and Hospital of Stomatology Wenzhou Medical University Wenzhou China
| | - Lulu Ma
- Institute of Stomatology, School and Hospital of Stomatology Wenzhou Medical University Wenzhou China
| | - Shengzhuang Wu
- Institute of Stomatology, School and Hospital of Stomatology Hangzhou Medical College Zhejiang Hangzhou China
| | - Huining Wang
- Department of Periodontics, School and Hospital of Stomatology Wenzhou Medical University Wenzhou China
| | - Guang Liang
- Institute of Stomatology, School and Hospital of Stomatology Hangzhou Medical College Zhejiang Hangzhou China
| | - Shengbin Huang
- Institute of Stomatology, School and Hospital of Stomatology Wenzhou Medical University Wenzhou China
- Department of Prosthodontics, School and Hospital of Stomatology Wenzhou Medical University Wenzhou China
| | - Xiaoyu Sun
- Institute of Stomatology, School and Hospital of Stomatology Wenzhou Medical University Wenzhou China
- Department of Periodontics, School and Hospital of Stomatology Wenzhou Medical University Wenzhou China
| |
Collapse
|
36
|
Ren H, Luan Z, Zhang R, Zhang H, Bian C. A novel approach to explore metabolic diseases: Neddylation. Pharmacol Res 2024; 210:107532. [PMID: 39637955 DOI: 10.1016/j.phrs.2024.107532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 11/27/2024] [Accepted: 11/28/2024] [Indexed: 12/07/2024]
Abstract
Protein post translational modification (PTM) is the main regulatory mechanism for eukaryotic cell function, among which ubiquitination is based on the reversible degradation of proteins by the ubiquitin proteasome system to regulate cell homeostasis. The neural precursor cell expressed developmental downregulated gene 8 (NEDD8) is a kind of ubiquitin like protein that shares 80 % homology and 60 % identity with ubiquitin. The PTM process by covalently binding NEDD8 to lysine residues in proteins is called neddylation. The neddylation reaction could be regulated by NEDD8, its precursors, substrates, E1 activating enzymes, E2 binding enzymes, E3 ligases, de-neddylases, and its inhibitors, such as MLN4924. NEDD8 is widely expressed in the whole cell structure of multiple tissues and species, and neddylation related factors are highly expressed in metabolism related adrenal glands, thyroid glands, parathyroid glands, skeletal muscles, myocardium, and adipose tissues, related to metabolic cardiovascular, cerebrovascular and liver diseases, adipogenic and osteogenic differentiation, as well as tumor glycolysis and glucose metabolism resulting from angiogenesis and endothelial disfunction, hepatotoxicity, adipogenesis, osteogenesis, Warburg effect, and insulin function. This review provides researchers with a new approach to explore metabolic diseases via searching and analyzing the histological, cytological, and subcellular localization of neddylation specific molecules in databases, and exploring specific mechanism neddylation mediating metabolic diseases by searching for neddylation related terms with the development of pre-clinical neddylation pharmacological inhibitors.
Collapse
Affiliation(s)
- Huiwen Ren
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, Liaoning, China
| | - Zhilin Luan
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, Liaoning, China
| | - Ruijing Zhang
- Department of Nephrology, Xijing Hospital, the Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Haibo Zhang
- College of Basic Medicine, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, China
| | - Che Bian
- Department of General Medicine, the Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning, China.
| |
Collapse
|
37
|
Li J, Chen Z, Jin M, Gu X, Wang Y, Huang G, Zhao W, Lu C. Histone H4K12 lactylation promotes malignancy progression in triple-negative breast cancer through SLFN5 downregulation. Cell Signal 2024; 124:111468. [PMID: 39395526 DOI: 10.1016/j.cellsig.2024.111468] [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: 07/18/2024] [Revised: 09/15/2024] [Accepted: 10/08/2024] [Indexed: 10/14/2024]
Abstract
Lactylation, a newly identified post-translational modification, is uncertain in its implication in triple-negative breast cancer (TNBC). In this study, we analyzed 60 TNBC samples using immunohistochemical staining and revealed elevated levels of pan-lactylated proteins and specific histone H4K12 lactylation in tumor tissues, correlating with TNBC progression. Lactate exposure in TNBC cell lines significantly induced lysine lactylation at the H4K12 site, leading to alterations in gene profiles and reduced apoptosis. These effects were attenuated by DCA or sodium Oxamate, inhibitors of endogenous lactate production. Gene sequencing showed an increase in Schlafen 5 (SLFN5) expression in TNBC cells treated with Oxamate, contrasting with the effects of lactate exposure. Analysis of TNBC tissues showed a negative correlation between H4K12 lactylation and SLFN5 protein levels. Overexpression of SLFN5 countered the effects of lactate on apoptosis and tumor growth, highlighting its pivotal role in TNBC malignancy. CUT&Tag sequencing indicated that lactylated H4K12 potentially binds to the SLFN5 promoter region. Luciferase reporter assays further verified that lactate-induced suppression of SLFN5 promoter activity is mediated by wild-type H4K12, but not by its R or A mutants, verified by both in vitro and in vivo apoptosis detection in response to lactate and Oxamate stimulation. These results establish that H4K12 lactylation, induced by lactate in TNBC cells, specifically suppresses SLFN5 expression, contributing to TNBC malignancy. Our findings illuminate a critical histone lactylation-dependent carcinogenic pathway in TNBC.
Collapse
Affiliation(s)
- Jingyi Li
- Shanghai Key Laboratory of Molecular Imaging, Jiading District Central Hospital Affiliated Shanghai University of Medicine and Health Sciences, Shanghai, China; Heilongjiang University of Traditional Chinese Medicine, Harbin, Heilongjiang, China
| | - Ziyu Chen
- School of Pharmacy, Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Mingming Jin
- Shanghai Key Laboratory of Molecular Imaging, Jiading District Central Hospital Affiliated Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Xuefeng Gu
- School of Pharmacy, Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Yuhan Wang
- Shanghai Key Laboratory of Molecular Imaging, Jiading District Central Hospital Affiliated Shanghai University of Medicine and Health Sciences, Shanghai, China; Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Gang Huang
- Shanghai Key Laboratory of Molecular Imaging, Jiading District Central Hospital Affiliated Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Weiming Zhao
- Heilongjiang University of Traditional Chinese Medicine, Harbin, Heilongjiang, China
| | - Changlian Lu
- Shanghai Key Laboratory of Molecular Imaging, Jiading District Central Hospital Affiliated Shanghai University of Medicine and Health Sciences, Shanghai, China.
| |
Collapse
|
38
|
Gnanaolivu R, Hart SN. Using AI-predicted protein structures as a reference to predict loss-of-function activity in tumor suppressor breast cancer genes. Comput Struct Biotechnol J 2024; 23:3472-3480. [PMID: 39430403 PMCID: PMC11490748 DOI: 10.1016/j.csbj.2024.10.008] [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: 07/08/2024] [Revised: 10/03/2024] [Accepted: 10/03/2024] [Indexed: 10/22/2024] Open
Abstract
Background The loss-of-function (LOF) classification of most missense variants in tumor suppressor breast cancer genes BRCA1, BRCA2, PALB2, and RAD51C remains unclassified and confounds clinical actionability. Classifying these variants is challenging due to their rarity, leading clinicians to rely on in silico predictive methods. Protein stability changes are associated with function, making stability predictors valuable. Stability predictions upon missense variant perturbations require high-resolution protein structures. However, the availability of these high-resolution structures is lacking. This study explores using generative AI to predict high-resolution protein structures, which can then be analyzed with in silico protein stability prediction methods to assess LOF activity in ordered regions of the protein. This study also determines the appropriate in silico protein stability and dedicated in silico missense prediction methods in dbNSFP v4.7 database to predict LOF activity in ordered regions of these four genes. Functional classifications from homology recombination DNA repair (HDR) assays and variant classifications from the ClinVar database provide a reliable dataset for evaluating the performance of these in silico prediction methods. Results Complex AlphaFold2 structures of the BRCA1-C terminal (BRCT) domain and the DNA-binding (DB) domain of BRCA2, analyzed using protein stability tool FoldX predicts LOF activity from missense variants significantly better than experimentally-derived structures in ordered regions. The BRCT domain achieved an Area Under the Curve (AUC)= 0.861 (95 % CI:0.858-0.863) and AUC= 0.842 (95 % CI:0.840-0.845), while the DB domain achieved an AUC= 0.836 (95 % CI:0.8322-0.841), compared to AUC= 0.847 (95 % CI:0.844-0.850) and AUC= 0.835 (95 % CI:0.832-0.837) from the BRCT domain, and AUC= 0.830 (95 % CI:0.821-0.8320) from the DB domain from experimentally-derived structures. Protein stability does not predict LOF activity from missense variants better than dedicated in silico missense predictors. Overall, we find that AlphaMissense ranks highly, with an average AUC= 0.890 (95 % CI 0.886-0.895) from ordered regions across these four cancer genes, compared to all other in silico missense predictors present in the dbNSFP database. Conclusions The study reveals that generative AI protein predicted structures can outperform experimentally-derived structures in evaluating LOF activity from predicted protein stability in ordered regions of genes BRCA1, BRCA2, PALB2 and RAD51C. The study also highlights the predictive performance of AlphaMissense as the premier in silico missense prediction method to predict LOF activity from missense variants in these four tumor suppressor breast cancer genes. The code for this study can be downloaded for free on GitHub (https://github.com/rohandavidg/CarePred).
Collapse
Affiliation(s)
- Rohan Gnanaolivu
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, United States
| | - Steven N. Hart
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, United States
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, United States
| |
Collapse
|
39
|
Liu Y, Liu X, Pan C. Advances in Factors Affecting ALDH2 Activity and its Mechanisms. Cardiovasc Toxicol 2024; 24:1428-1438. [PMID: 39365551 DOI: 10.1007/s12012-024-09923-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Accepted: 09/15/2024] [Indexed: 10/05/2024]
Abstract
Aldehyde dehydrogenase 2 (ALDH2) is a mitochondrial enzyme primarily involved in the detoxification of alcohol-derived aldehyde and endogenous toxic aldehydes. It exhibits widespread expression across various organs and exerts a broad and significant impact on diverse acute cardiovascular diseases, including acute coronary syndrome, acute aortic dissection, hypoxic pulmonary hypertension, and heart failure. The ALDH2 rs671 variant represents the most prevalent genetic variant in East Asian populations, with carriage rates ranging from 30 to 50% among the Chinese population. Given its widespread presence in the body, the wide range of diseases it affects, and its high rate of variation, it can serve as a crucial tool for the precise prevention and treatment of acute cardiovascular diseases, while offering individualized medication guidance. This review aims to provide a comprehensive overview of the latest advancements in factors affecting ALDH2 activity, encompassing post-transcriptional modifications, modulators of ALDH2, and relevant clinical drugs.
Collapse
Affiliation(s)
- Yun Liu
- Department of Emergency Medicine, Qilu Hospital of Shandong University, Jinan, 250012, China
- Chest Pain Center, Shandong Provincial Clinical Research Center for Emergency and Critical Care Medicine, Institute of Emergency and Critical Care Medicine of Shandong University, Qilu Hospital of Shandong University, Jinan, 250012, China
- Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Shandong Provincial Engineering Laboratory for Emergency and Critical Care Medicine, Qilu Hospital of Shandong University, Jinan, 250012, China
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, Jinan, 250012, China
| | - Xuemei Liu
- Department of Nephrology, The Fifth People's Hospital of Jinan, Jinan, 250022, China
| | - Chang Pan
- Department of Emergency Medicine, Qilu Hospital of Shandong University, Jinan, 250012, China.
- Chest Pain Center, Shandong Provincial Clinical Research Center for Emergency and Critical Care Medicine, Institute of Emergency and Critical Care Medicine of Shandong University, Qilu Hospital of Shandong University, Jinan, 250012, China.
- Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Shandong Provincial Engineering Laboratory for Emergency and Critical Care Medicine, Qilu Hospital of Shandong University, Jinan, 250012, China.
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, Jinan, 250012, China.
| |
Collapse
|
40
|
Sena S, Prakash A, Van Staden J, Kumar V. Epigenetic control of plant regeneration: Unraveling the role of histone methylation. CURRENT PLANT BIOLOGY 2024; 40:100408. [DOI: 10.1016/j.cpb.2024.100408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2025]
|
41
|
Wan QK, Li TT, Liu BB, He B. USP5 promotes tumor progression by stabilizing SLUG in bladder cancer. Oncol Lett 2024; 28:572. [PMID: 39397799 PMCID: PMC11467842 DOI: 10.3892/ol.2024.14705] [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: 04/16/2024] [Accepted: 08/07/2024] [Indexed: 10/15/2024] Open
Abstract
Bladder cancer ranks as the second most prevalent urology malignancy globally. Invasive metastasis is a significant contributor to mortality among patients with bladder cancer, yet the underlying mechanisms remain elusive. Deubiquitinases are pivotal in carcinogenesis, with USP5 implicated in the malignant progression of hepatocellular carcinoma, colorectal cancer and non-small cell lung cancer. The present study assessed the role and mechanism of ubiquitin-specific proteinase 5 (USP5) in the malignant progression of bladder cancer. The association between USP5 expression and bladder cancer prognosis and stage was analyzed using The Cancer Genome Atlas database. Moreover, to elucidate the role of USP5 in bladder cancer, USP5 overexpression and knockdown cell lines were established using T24 cells. Cell viability, proliferation and migration were assessed using Cell Counting Kit-8, Transwell and scratch assays, respectively. Cyclohexanamide was used to evaluate the effect of USP5 expression on Snail family zinc finger 2 (SLUG) stability. Immunoprecipitation and immunofluorescence co-localization were utilized to probe the interaction between USP5 and SLUG. Changes in mRNA and protein levels were assessed using reverse transcription-quantitative PCR and western blotting, respectively. The results revealed that patients with bladder cancer with high USP5 expression had significantly shorter survival (P<0.05) and a higher clinicopathologic stage (P<0.05) than those with low USP5 expression. T24 cells overexpressing USP5 demonstrated significantly increased proliferation (P<0.05), invasion (P<0.05) and expression of epithelial-mesenchymal transition markers (P<0.05); whereas T24 cells with knocked-down USP5 expression revealed significantly reduced proliferation (P<0.05), invasion (P<0.05) and epithelial-mesenchymal transition markers (P<0.05). Immunoprecipitation experiments demonstrated the binding of USP5 to SLUG in bladder cancer cells, with further analysis revealing that USP5 upregulated protein levels of SLUG by inhibiting its ubiquitination. Furthermore, the treatment of bladder cancer cells with Degrasyn, a USP5 inhibitor, was associated with a significant inhibition of the proliferation (P<0.05) and invasion (P<0.05) of T24 cells. In conclusion, the findings of the present study underscore the role of USP5 in promoting the malignant progression of bladder cancer through the stabilization of SLUG. Targeting USP5 holds promise as a strategy for inhibiting bladder cancer progression.
Collapse
Affiliation(s)
- Qiang-Kun Wan
- Department of Radiotherapy, The First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui 233004, P.R. China
| | - Ting-Ting Li
- Department of Radiotherapy, The First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui 233004, P.R. China
| | - Bei-Bei Liu
- Department of Urology, The First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui 233004, P.R. China
| | - Bin He
- Department of Radiotherapy, The First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui 233004, P.R. China
| |
Collapse
|
42
|
Aldrete CA, An C, Call CC, Gao XJ, Vlahos AE. Perspectives on Synthetic Protein Circuits in Mammalian Cells. CURRENT OPINION IN BIOMEDICAL ENGINEERING 2024; 32:100555. [PMID: 39372446 PMCID: PMC11448451 DOI: 10.1016/j.cobme.2024.100555] [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] [Indexed: 10/08/2024]
Abstract
Mammalian synthetic biology aims to engineer cellular behaviors for therapeutic applications, such as enhancing immune cell efficacy against cancers or improving cell transplantation outcomes. Programming complex biological functions necessitates an understanding of molecular mechanisms governing cellular responses to stimuli. Traditionally, synthetic biology has focused on transcriptional circuits, but recent advances have led to the development of synthetic protein circuits, leveraging programmable binding, proteolysis, or phosphorylation to modulate protein interactions and cellular functions. These circuits offer advantages including robust performance, rapid functionality, and compact design, making them suitable for cellular engineering or gene therapies. This review outlines the post-translational toolkit, emphasizing synthetic protein components utilizing proteolysis or phosphorylation to program mammalian cell behaviors. Finally, we focus on key differences between rewiring native signaling pathways and creating orthogonal behaviors, alongside a proposed framework for translating synthetic protein circuits from tool development to pre-clinical applications in biomedicine.
Collapse
Affiliation(s)
- Carlos A. Aldrete
- Department of Chemical Engineering, Stanford University, CA, USA, 94305
| | - Connie An
- Department of Chemical Engineering, Stanford University, CA, USA, 94305
| | - Connor C. Call
- Department of Chemical Engineering, Stanford University, CA, USA, 94305
| | - Xiaojing J. Gao
- Department of Chemical Engineering, Stanford University, CA, USA, 94305
| | | |
Collapse
|
43
|
Howes JM, Harper MT. Application of the Cellular Thermal Shift Assay (CETSA) to validate drug target engagement in platelets. Platelets 2024; 35:2354833. [PMID: 38767506 DOI: 10.1080/09537104.2024.2354833] [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: 03/08/2024] [Accepted: 05/06/2024] [Indexed: 05/22/2024]
Abstract
Small molecule drugs play a major role in the study of human platelets. Effective action of a drug requires it to bind to one or more targets within the platelet (target engagement). However, although in vitro assays with isolated proteins can be used to determine drug affinity to these targets, additional factors affect target engagement and its consequences in an intact platelet, including plasma membrane permeability, intracellular metabolism or compartmentalization, and level of target expression. Mechanistic interpretation of the effect of drugs on platelet activity requires comprehensive investigation of drug binding in the proper cellular context, i.e. in intact platelets. The Cellular Thermal Shift Assay (CETSA) is a valuable method to investigate target engagement within complex cellular environments. The assay is based on the principle that drug binding to a target protein increases that protein's thermal stability. In this technical report, we describe the application of CETSA to platelets. We highlight CETSA as a quick and informative technique for confirming the direct binding of drugs to platelet protein targets, providing a platform for understanding the mechanism of action of drugs in platelets, and which will be a valuable tool for investigating platelet signaling and function.
Collapse
Affiliation(s)
| | - Matthew T Harper
- Department of Pharmacology, University of Cambridge, Cambridge, UK
| |
Collapse
|
44
|
Xian Y, Ye J, Tang Y, Zhang N, Peng C, Huang W, He G. Deubiquitinases as novel therapeutic targets for diseases. MedComm (Beijing) 2024; 5:e70036. [PMID: 39678489 PMCID: PMC11645450 DOI: 10.1002/mco2.70036] [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: 07/10/2024] [Revised: 11/24/2024] [Accepted: 11/26/2024] [Indexed: 12/17/2024] Open
Abstract
Deubiquitinating enzymes (DUBs) regulate substrate ubiquitination by removing ubiquitin or cleaving within ubiquitin chains, thereby maintaining cellular homeostasis. Approximately 100 DUBs in humans counteract E3 ubiquitin ligases, finely balancing ubiquitination and deubiquitination processes to maintain cellular proteostasis and respond to various stimuli and stresses. Given their role in modulating ubiquitination levels of various substrates, DUBs are increasingly linked to human health and disease. Here, we review the DUB family, highlighting their distinctive structural characteristics and chain-type specificities. We show that DUB family members regulate key signaling pathways, such as NF-κB, PI3K/Akt/mTOR, and MAPK, and play crucial roles in tumorigenesis and other diseases (neurodegenerative disorders, cardiovascular diseases, inflammatory disorders, and developmental diseases), making them promising therapeutic targets Our review also discusses the challenges in developing DUB inhibitors and underscores the critical role of the DUBs in cellular signaling and cancer. This comprehensive analysis enhances our understanding of the complex biological functions of the DUBs and underscores their therapeutic potential.
Collapse
Affiliation(s)
- Yali Xian
- Department of Dermatology & VenerologyState Key Laboratory of BiotherapyWest China HospitalSichuan UniversityChengduChina
| | - Jing Ye
- Department of Dermatology & VenerologyState Key Laboratory of BiotherapyWest China HospitalSichuan UniversityChengduChina
| | - Yu Tang
- Department of Dermatology & VenerologyState Key Laboratory of BiotherapyWest China HospitalSichuan UniversityChengduChina
| | - Nan Zhang
- State Key Laboratory of Southwestern Chinese Medicine ResourcesSchool of PharmacyChengdu University of Traditional Chinese MedicineChengduChina
| | - Cheng Peng
- State Key Laboratory of Southwestern Chinese Medicine ResourcesSchool of PharmacyChengdu University of Traditional Chinese MedicineChengduChina
| | - Wei Huang
- State Key Laboratory of Southwestern Chinese Medicine ResourcesSchool of PharmacyChengdu University of Traditional Chinese MedicineChengduChina
| | - Gu He
- Department of Dermatology & VenerologyState Key Laboratory of BiotherapyWest China HospitalSichuan UniversityChengduChina
| |
Collapse
|
45
|
Herman N, Vitenberg T, Hayouka Z, Opatovsky I. Regulation of antimicrobial peptides in Hermetia illucens in response to fungal exposure. Sci Rep 2024; 14:29561. [PMID: 39609510 PMCID: PMC11605112 DOI: 10.1038/s41598-024-80133-7] [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: 07/28/2024] [Accepted: 11/15/2024] [Indexed: 11/30/2024] Open
Abstract
The black soldier fly (Hermetia illucens) is important for antimicrobial peptide (AMP) research due to its exposure to diverse microorganisms. However, the impact of different fungi on AMP abundance in H. illucens remains unexplored. We studied the induction of AMP expression under basal conditions and with three fungi: non-pathogenic Candida tropicalis, Saccharomyces cerevisiae, and pathogenic Beauveria bassiana, using RNA-sequencing and liquid chromatography with tandem mass spectrometry. Under naive conditions, most AMPs belonged to the lysozyme, cecropin, and defensin classes, with defensins most abundant. We demonstrate that dietary supplementation with fungi is sufficient to induce AMP expression in H. illucens. However, exposure to C. tropicalis and B. bassiana also caused downregulation of certain AMPs, suggesting that these fungi may suppress or modulate the host immune response to aid in their survival and colonization. Evidently, S. cerevisiae and B. bassiana trigger similar AMP pathways, whereas C. tropicalis elicits a distinct reaction with upregulation of defensins and cecropins. Lysozymes were upregulated by S. cerevisiae and B. bassiana, but downregulated by C. tropicalis, potentially facilitating fungal survival in the larval gut. Understanding these mechanisms opens possibilities for leveraging AMPs to combat C. tropicalis, which is implicated in human diseases.
Collapse
Affiliation(s)
- Neta Herman
- Laboratory of Insect Nutrition and Metabolism, Department of Nutrition and Natural Products, MIGAL - Galilee Research Centre, 1101202, Kiryat Shmona, Israel.
- Department of Animal Science, Faculty of Sciences and Technology, Tel-Hai Academic College, 1220800, Upper Galilee, Israel.
| | - Tzach Vitenberg
- Laboratory of Insect Nutrition and Metabolism, Department of Nutrition and Natural Products, MIGAL - Galilee Research Centre, 1101202, Kiryat Shmona, Israel
- Department of Animal Science, Faculty of Sciences and Technology, Tel-Hai Academic College, 1220800, Upper Galilee, Israel
| | - Zvi Hayouka
- Institute of Biochemistry, Food Science and Nutrition, The Robert H. Smith Faculty of Agricultural, Food & Environment, The Hebrew University of Jerusalem, 76100, Rehovot, Israel
| | - Itai Opatovsky
- Laboratory of Insect Nutrition and Metabolism, Department of Nutrition and Natural Products, MIGAL - Galilee Research Centre, 1101202, Kiryat Shmona, Israel.
- Department of Animal Science, Faculty of Sciences and Technology, Tel-Hai Academic College, 1220800, Upper Galilee, Israel.
| |
Collapse
|
46
|
Hang Q, Zuo S, Yang Y, Wang Y, Li C, Li W, Guo J, Hou S, Huang H. USP33 is an integrin α6 deubiquitinase and promotes esophageal squamous cell carcinoma cell migration and metastasis. J Cancer Res Clin Oncol 2024; 150:511. [PMID: 39589547 PMCID: PMC11599434 DOI: 10.1007/s00432-024-06041-5] [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: 10/22/2024] [Accepted: 11/16/2024] [Indexed: 11/27/2024]
Abstract
PURPOSE The deubiquitinating enzymes (DUBs) have been linked to cancer initiation and progression. Although ubiquitin-specific protease 33 (USP33) represents a significant factor in regulating various tumor cell behaviors, its specific biological functions and precise mechanisms in esophageal squamous cell carcinoma (ESCC) progression remain unclear. METHODS The expressions of USP33 mRNA in GEO databases, clinical ESCC samples, and USP33 protein were analyzed using bioinformatics, RT-PCR, and immunohistochemistry, respectively. Using Kaplan-Meier survival curves, the log-rank test was used to determine the cumulative survival rate. Western blotting was used to determine indicated protein expression. The cell biological functions were evaluated by cell growth assay, transwell, cell adhesion, and cell spreading assay, respectively. The interaction between USP33 and integrins was detected by immunoprecipitation, and the deubiquitination was performed by deubiquitination assay. The metastatic ability was checked by tail vein injection. RESULTS A significant positive correlation was found between USP33 expression and clinical TNM stage, T classification, and poor prognosis in patients with ESCC. USP33 promoted laminin-dependent adhesion, spreading, and migration of ESCC cells but not their proliferation. Mechanistically, USP33 mediates cell migration through binding, deubiquinating, and stabilizing integrin α6. USP33 knockdown could inhibit ESCC cell migration and metastasis majorly through integrin α6. CONCLUSION This study reveals a novel mechanism of USP33 in promoting laminin-dependent ESCC cell migration and metastasis through integrin α6, suggesting that USP33 may be a promising target for treating ESCC.
Collapse
Affiliation(s)
- Qinglei Hang
- Department of Clinical Medicine, Medical College, Key laboratory of Jiangsu province university for Nucleic Acid & Cell Fate Manipulation, Yangzhou University, Yangzhou, Jiangsu Province, 225009, China.
- Jiangsu Provincial Innovation and Practice Base for Postdoctors, Suining People's Hospital, Affiliated Hospital of Xuzhou Medical University, Suining, Jiangsu Province, 221200, China.
| | - Shiying Zuo
- Department of Clinical Medicine, Medical College, Key laboratory of Jiangsu province university for Nucleic Acid & Cell Fate Manipulation, Yangzhou University, Yangzhou, Jiangsu Province, 225009, China
- Department of Gastroenterology, Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu Province, 225001, China
| | - Yawen Yang
- Department of Clinical Medicine, Medical College, Key laboratory of Jiangsu province university for Nucleic Acid & Cell Fate Manipulation, Yangzhou University, Yangzhou, Jiangsu Province, 225009, China
| | - Yuanzhi Wang
- Department of Clinical Medicine, Medical College, Key laboratory of Jiangsu province university for Nucleic Acid & Cell Fate Manipulation, Yangzhou University, Yangzhou, Jiangsu Province, 225009, China
- Department of Gastroenterology, Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu Province, 225001, China
| | - Caimin Li
- Department of Clinical Medicine, Medical College, Key laboratory of Jiangsu province university for Nucleic Acid & Cell Fate Manipulation, Yangzhou University, Yangzhou, Jiangsu Province, 225009, China
- Department of Gastroenterology, Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu Province, 225001, China
| | - Wenqian Li
- Department of Clinical Medicine, Medical College, Key laboratory of Jiangsu province university for Nucleic Acid & Cell Fate Manipulation, Yangzhou University, Yangzhou, Jiangsu Province, 225009, China
- Department of Gastroenterology, Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu Province, 225001, China
| | - Jingya Guo
- Department of Clinical Medicine, Medical College, Key laboratory of Jiangsu province university for Nucleic Acid & Cell Fate Manipulation, Yangzhou University, Yangzhou, Jiangsu Province, 225009, China
- Department of Gastroenterology, Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu Province, 225001, China
| | - Sicong Hou
- Department of Clinical Medicine, Medical College, Key laboratory of Jiangsu province university for Nucleic Acid & Cell Fate Manipulation, Yangzhou University, Yangzhou, Jiangsu Province, 225009, China.
- Department of Gastroenterology, Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu Province, 225001, China.
| | - Haifeng Huang
- Department of Laboratory Medicine, The First People's Hospital of Yancheng, Yancheng, Jiangsu Province, 224006, China.
- Department of Laboratory Medicine, Yancheng First Hospital, Affiliated Hospital of Nanjing University Medical School, Yancheng, Jiangsu Province, 224006, China.
| |
Collapse
|
47
|
Keller A, Bakhtina A, Bruce JE. Large-Scale Quantitative Cross-Linking and Mass Spectrometry Provides New Insight on Protein Conformational Plasticity within Organelles, Cells, and Tissues. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.14.623288. [PMID: 39651194 PMCID: PMC11623499 DOI: 10.1101/2024.11.14.623288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2024]
Abstract
Many proteins can exist in multiple conformational states in vivo to achieve distinct functional roles. These states include alternative conformations, variable PTMs, and association with interacting protein, nucleotide, and ligand partners. Quantitative chemical cross-linking of live cells, organelles, or tissues together with mass spectrometry provides the relative abundance of cross-link levels formed in two or more compared samples, which depends both on the relative levels of existent protein conformational states in the compared samples as well as the relative likelihood of the cross-link originating from each. Because cross-link conformational state preferences can vary widely, one expects intra-protein cross-link levels from proteins with high conformational plasticity to display divergent quantitation among samples with differing conformational ensembles. Here we use the large volume of quantitative cross-linking data available on the public XLinkDB database to cluster intra-protein cross-links according to their quantitation in many diverse compared samples to provide the first widescale glimpse of cross-links grouped according to the protein conformational state(s) from which they predominantly originate. We further demonstrate how cluster cross-links can be aligned with any protein structure to assess the likelihood that they were derived from it.
Collapse
|
48
|
Khaitin AM, Guzenko VV, Bachurin SS, Demyanenko SV. c-Myc and FOXO3a-The Everlasting Decision Between Neural Regeneration and Degeneration. Int J Mol Sci 2024; 25:12621. [PMID: 39684331 DOI: 10.3390/ijms252312621] [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: 10/26/2024] [Revised: 11/20/2024] [Accepted: 11/22/2024] [Indexed: 12/18/2024] Open
Abstract
The transcription factors c-Myc and FoxO3a play significant roles in neurodegenerative processes, yet their interaction in neurological disorders remains largely unexplored. In contrast, much of the available information about their relationship comes from cancer research. While it is well-established that FoxO3a inhibits c-Myc activity, this interaction represents only a basic understanding of a far more complex dynamic, which includes exceptions under specific conditions and the involvement of additional regulatory factors. Given the critical need to address this gap for the treatment and prevention of neurodegenerative disorders, this review consolidates current knowledge on the joint roles of these two factors in neuropathology. It also highlights their conformational flexibility, post-translational modifications, and outlines potential directions for future research.
Collapse
Affiliation(s)
- Andrey M Khaitin
- Laboratory of Molecular Neuroscience, Academy of Biology and Biotechnology, Southern Federal University, 194/1 Stachky Ave., Rostov-on-Don 344090, Russia
| | - Valeria V Guzenko
- Laboratory of Molecular Neuroscience, Academy of Biology and Biotechnology, Southern Federal University, 194/1 Stachky Ave., Rostov-on-Don 344090, Russia
| | - Stanislav S Bachurin
- Laboratory of Molecular Neuroscience, Academy of Biology and Biotechnology, Southern Federal University, 194/1 Stachky Ave., Rostov-on-Don 344090, Russia
| | - Svetlana V Demyanenko
- Laboratory of Molecular Neuroscience, Academy of Biology and Biotechnology, Southern Federal University, 194/1 Stachky Ave., Rostov-on-Don 344090, Russia
| |
Collapse
|
49
|
Van Scoyk AN, Antelope O, Ayer DE, Peterson RT, Pomicter AD, Owen SC, Deininger MW. Bioluminescence assay of lysine deacylase sirtuin activity. Cell Chem Biol 2024; 31:2002-2014.e4. [PMID: 39515338 DOI: 10.1016/j.chembiol.2024.10.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Revised: 06/08/2024] [Accepted: 10/18/2024] [Indexed: 11/16/2024]
Abstract
Lysine acylation can direct protein function, localization, and interactions. Sirtuins deacylate lysine toward maintaining cellular homeostasis, and their aberrant expression contributes to the pathogenesis of multiple conditions, including cancer. Measuring sirtuins' activity is essential to exploring their potential as therapeutic targets, but accurate quantification is challenging. We developed "SIRTify", a high-sensitivity assay for measuring sirtuin activity in vitro and in vivo. SIRTify is based on a split-version of the NanoLuc luciferase consisting of a truncated, catalytically inactive N-terminal moiety (LgBiT) that complements with a high-affinity C-terminal peptide (p86) to form active luciferase. Acylation of two lysines within p86 disrupts binding to LgBiT and abates luminescence. Deacylation by sirtuins reestablishes p86 and restores binding, generating a luminescence signal proportional to sirtuin activity. Measurements accurately reflect reported sirtuin specificity for lysine-acylations and confirm the effects of sirtuin modulators. SIRTify quantifies lysine deacylation dynamics and may be adaptable to monitoring additional post-translational modifications.
Collapse
Affiliation(s)
| | - Orlando Antelope
- University of Utah, Department of Pharmacology and Toxicology, Salt Lake City
| | - Donald E Ayer
- University of Utah, Department of Oncological Sciences, Salt Lake City
| | - Randall T Peterson
- University of Utah, Department of Pharmacology and Toxicology, Salt Lake City
| | - Anthony D Pomicter
- University of Utah, Division of Hematology Biorepository, Salt Lake City
| | - Shawn C Owen
- University of Utah, Department of Molecular Pharmaceutics, Salt Lake City; University of Utah, Department of Medicinal Chemistry, Department of Biomedical Engineering, Salt Lake City.
| | - Michael W Deininger
- Versiti Blood Research Institute, Milwaukee; Division of Hematology and Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee.
| |
Collapse
|
50
|
Wu T, Sun Y, Wang D, Isaji T, Fukuda T, Suzuki C, Hanamatsu H, Nishikaze T, Tsumoto H, Miura Y, Furukawa JI, Gu J. The acetylglucosaminyltransferase GnT-Ⅲ regulates erythroid differentiation through ERK/MAPK signaling. J Biol Chem 2024; 300:108010. [PMID: 39571652 PMCID: PMC11699732 DOI: 10.1016/j.jbc.2024.108010] [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: 07/17/2024] [Revised: 11/05/2024] [Accepted: 11/07/2024] [Indexed: 12/15/2024] Open
Abstract
Differentiation therapy is an alternative strategy used in treating chronic myelogenous leukemia to induce the differentiation of immature or cancerous cells toward mature cells and inhibit tumor cell proliferation. We aimed to explore N-glycans' roles in erythroid differentiation using the sodium butyrate (NaBu)-induced model of K562 cells (WT/NaBu cells). Here, using lectin blot, flow cytometry, real-time PCR, and mass spectrometry analyses, we demonstrated that the mRNA levels of N-acetylglucosaminyltransferase Ⅲ ((encoded by the MGAT3 gene) and its product (bisected N-glycans) were significantly increased during erythroid differentiation. To address the importance of GnTN-acetylglucosaminyltransferase-Ⅲ in this progress, we established a stable MGAT3 KO K562 cell line using the CRISPR/Cas9 technology. Compared to WT/NaBu cells, MGAT3 KO significantly impeded the progression of erythroid differentiation, as shown in decreased cell color and levels of erythroid markers, glycophorin A (CD235a), and β-globin. Consistently, MGAT3 KO mitigated the inhibitory impact of NaBu on cell proliferation. During induction, MGAT3 KO suppressed the cellular phosphorylated tyrosine and phospho-extracellular signal-regulated kinase (ERK)1/2 levels. Inhibition of the ERK/mitogen-activated protein kinase signaling pathway using U0126 blocked erythroid differentiation while concurrently suppressing the expression levels of MGAT3 and bisected N-glycans. Furthermore, the lack of bisecting GlcNAc modification on c-Kit and transferrin receptor 1 (CD71) suppressed cellular signaling and accelerated the degradation of the CD71 protein, respectively. Our study highlights the critical role of MGAT3 in regulating erythroid differentiation associated with the ERK/mitogen-activated protein kinase signaling pathway, which may shed light on identifying new differentiation therapy in chronic myelogenous leukemia.
Collapse
Affiliation(s)
- Tiangui Wu
- Division of Regulatory Glycobiology, Institute of Molecular Biomembrane and Glycobiology, Tohoku Medical and Pharmaceutical University, Sendai, Miyagi, Japan
| | - Yuhan Sun
- Division of Regulatory Glycobiology, Institute of Molecular Biomembrane and Glycobiology, Tohoku Medical and Pharmaceutical University, Sendai, Miyagi, Japan
| | - Dan Wang
- Division of Regulatory Glycobiology, Institute of Molecular Biomembrane and Glycobiology, Tohoku Medical and Pharmaceutical University, Sendai, Miyagi, Japan
| | - Tomoya Isaji
- Division of Regulatory Glycobiology, Institute of Molecular Biomembrane and Glycobiology, Tohoku Medical and Pharmaceutical University, Sendai, Miyagi, Japan
| | - Tomohiko Fukuda
- Division of Regulatory Glycobiology, Institute of Molecular Biomembrane and Glycobiology, Tohoku Medical and Pharmaceutical University, Sendai, Miyagi, Japan
| | - Chiharu Suzuki
- Division of Glyco-Systems Biology, Institute for Glyco-Core Research, Tokai National Higher Education and Research System, Nagoya, Japan
| | - Hisatoshi Hanamatsu
- Division of Glyco-Systems Biology, Institute for Glyco-Core Research, Tokai National Higher Education and Research System, Nagoya, Japan
| | - Takashi Nishikaze
- Solutions COE, Analytical & Measuring Instruments Division, Shimadzu Corporation, Kyoto, Japan
| | - Hiroki Tsumoto
- Research Team for Mechanism of Aging, Tokyo Metropolitan Institute for Geriatrics and Gerontology, Tokyo, Japan
| | - Yuri Miura
- Research Team for Mechanism of Aging, Tokyo Metropolitan Institute for Geriatrics and Gerontology, Tokyo, Japan
| | - Jun-Ichi Furukawa
- Division of Glyco-Systems Biology, Institute for Glyco-Core Research, Tokai National Higher Education and Research System, Nagoya, Japan; Department of Orthopedic Surgery, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Jianguo Gu
- Division of Regulatory Glycobiology, Institute of Molecular Biomembrane and Glycobiology, Tohoku Medical and Pharmaceutical University, Sendai, Miyagi, Japan.
| |
Collapse
|