1
|
Deshpande P, Chimata AV, Singh A. Exploring the role of N-acetyltransferases in diseases: a focus on N-acetyltransferase 9 in neurodegeneration. Neural Regen Res 2025; 20:2862-2871. [PMID: 39435604 PMCID: PMC11826463 DOI: 10.4103/nrr.nrr-d-24-00779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 09/04/2024] [Accepted: 09/23/2024] [Indexed: 10/23/2024] Open
Abstract
Acetyltransferases, required to transfer an acetyl group on protein are highly conserved proteins that play a crucial role in development and disease. Protein acetylation is a common post-translational modification pivotal to basic cellular processes. Close to 80%-90% of proteins are acetylated during translation, which is an irreversible process that affects protein structure, function, life, and localization. In this review, we have discussed the various N-acetyltransferases present in humans, their function, and how they might play a role in diseases. Furthermore, we have focused on N-acetyltransferase 9 and its role in microtubule stability. We have shed light on how N-acetyltransferase 9 and acetylation of proteins can potentially play a role in neurodegenerative diseases. We have specifically discussed the N-acetyltransferase 9-acetylation independent function and regulation of c-Jun N-terminal kinase signaling and microtubule stability during development and neurodegeneration.
Collapse
Affiliation(s)
| | | | - Amit Singh
- Department of Biology, University of Dayton, Dayton, OH, USA
- Premedical Program, University of Dayton, Dayton, OH, USA
- Center for Tissue Regeneration and Engineering at Dayton (TREND), University of Dayton, Dayton, OH, USA
- Integrative Science and Engineering Center, University of Dayton, Dayton, OH, USA
- Center for Genomic Advocacy (TCGA), Indiana State University, Terre Haute, IN, USA
| |
Collapse
|
2
|
Hutson HK, Qin G, Cai C, Nestorova GG. Comparative proteomic profiling of glioblastoma and healthy brain cell-derived extracellular vesicles reveals enrichment of cancer-associated proteins. J Proteomics 2025; 316:105418. [PMID: 40058457 DOI: 10.1016/j.jprot.2025.105418] [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: 09/25/2024] [Revised: 01/30/2025] [Accepted: 03/03/2025] [Indexed: 03/15/2025]
Abstract
Extracellular vesicles (EVs)-mediated cellular communication plays a role in cancer development and progression. This study focuses on identifying glioblastoma-specific EV protein markers through a comparative mass spectrometry bottom-up proteomic analysis of the LN-229 cell line and human neurons, astrocytes, and endothelial brain cells (HEBCs) using timsTOF Pro 2 instrument. The statistically significant upregulated proteins with fold change greater than 2 in the glioblastoma-derived EVs were clustered based on physical and functional interactions using the STRING database and analyzed using Gene Ontology enrichment. LN229-derived EVs contained an average of 2635 proteins, while human astrocytes, neurons, and HEBC encapsulated 2647, 716, and 2285 proteins, respectively. NanoParticle Tracking Analysis indicated that glioblastoma-derived EVs exhibited greater size variability compared to EVs from healthy cells. Statistical analysis identified 25 statistically significant proteins with increased levels in LN229 EVs relative to at least two healthy cell lines suggesting their potential as glioblastoma markers. Functional clustering using the STRING database and GO analysis indicated involvement in epigenetic regulation, metastasis, angiogenesis, and protein folding. Post-translational modification analysis identified a subset of 17 proteins unique to the cancer-derived EVs involved in chromatin regulation, extracellular matrix remodeling, and basement membrane organization pathways, highlighting their role in tumor progression.
Collapse
Affiliation(s)
- Hope K Hutson
- Molecular Science and Nanotechnology, Louisiana Tech University, United States
| | - Guoting Qin
- College of Optometry, University of Houston, United States
| | - Chengzhi Cai
- Department of Chemistry, University of Houston, United States
| | | |
Collapse
|
3
|
Nagai R, Milam OL, Niwa T, Howell WJ, Best JA, Yoshida H, Freeburg CD, Koomen JM, Fujii K. Ribosomal expansion segment contributes to translation fidelity via N-terminal processing of ribosomal proteins. Nucleic Acids Res 2025; 53:gkaf448. [PMID: 40433980 DOI: 10.1093/nar/gkaf448] [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: 11/30/2024] [Revised: 04/30/2025] [Accepted: 05/14/2025] [Indexed: 05/29/2025] Open
Abstract
Eukaryotic ribosomes exhibit higher mRNA translation fidelity than prokaryotic ribosomes, partly due to eukaryote-specific ribosomal RNA (rRNA) insertions. Among these, expansion segment 27L (ES27L) on the 60S subunit enhances fidelity by anchoring methionine aminopeptidase (MetAP) at the nascent protein exit tunnel, accelerating co-translational N-terminal initiator methionine (iMet) processing. However, the mechanisms by which iMet processing influences translation fidelity remain unknown. Using yeast in vitro translation (IVT) systems, we found that inhibiting co-translational iMet processing does not impact ribosome decoding of ongoing peptide synthesis. Instead, our novel method to monitor iMet processing in vivo revealed that ribosomes purified from strains lacking MetAP ribosomal association (ES27L Δb1-4) or major yeast MetAP (Δmap1) increase iMet retention on ribosomal proteins (RPs). Given the densely packed structure of ribosomes, iMet retention on RPs may distort ribosomal structure and impair its function. Indeed, reconstituted IVT systems containing iMet-retaining ribosome subunits from ES27L Δb1-4 strain, combined with translation factors from wild-type strains, elucidated that iMet retention on the 40S ribosomal subunit causes translation errors. Our study demonstrated the critical role of ES27L in adjusting ribosome association of universally conserved MetAP enzyme to fine-tune iMet processing of key RPs, thereby ensuring the structural integrity and functional accuracy of eukaryotic ribosomes.
Collapse
Affiliation(s)
- Riku Nagai
- Center for NeuroGenetics, University of Florida, Gainesville, FL 32610, United States
- Department of Molecular Genetics and Microbiology, University of Florida, Gainesville, FL 32610, United States
| | - Olivia L Milam
- Center for NeuroGenetics, University of Florida, Gainesville, FL 32610, United States
- Department of Molecular Genetics and Microbiology, University of Florida, Gainesville, FL 32610, United States
| | - Tatsuya Niwa
- Cell Biology Center, Institute of Integrated Research, Institute of Science Tokyo, Yokohama, Kanagawa 226-8503, Japan
| | - William J Howell
- Center for NeuroGenetics, University of Florida, Gainesville, FL 32610, United States
- Department of Molecular Genetics and Microbiology, University of Florida, Gainesville, FL 32610, United States
| | - Jacob A Best
- Center for NeuroGenetics, University of Florida, Gainesville, FL 32610, United States
- Department of Molecular Genetics and Microbiology, University of Florida, Gainesville, FL 32610, United States
| | - Hideji Yoshida
- Department of Physics, Osaka Medical and Pharmaceutical University, Takatsuki, Osaka 569-8686, Japan
| | - Carver D Freeburg
- Center for NeuroGenetics, University of Florida, Gainesville, FL 32610, United States
- Department of Molecular Genetics and Microbiology, University of Florida, Gainesville, FL 32610, United States
| | - John M Koomen
- Department of Molecular Oncology, Moffitt Cancer Center, Tampa, FL 33612, United States
| | - Kotaro Fujii
- Center for NeuroGenetics, University of Florida, Gainesville, FL 32610, United States
- Department of Molecular Genetics and Microbiology, University of Florida, Gainesville, FL 32610, United States
| |
Collapse
|
4
|
Weber D, Ferrario PG, Bub A. Exercise intensity determines circulating levels of Lac-Phe and other exerkines: a randomized crossover trial. Metabolomics 2025; 21:63. [PMID: 40335829 PMCID: PMC12058925 DOI: 10.1007/s11306-025-02260-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Accepted: 04/18/2025] [Indexed: 05/09/2025]
Abstract
INTRODUCTION Exercise metabolomics research has revealed significant exercise-induced metabolic changes and identified several exerkines as mediators of physiological adaptations to exercise. However, the effect of exercise intensity on metabolic changes and circulating exerkine levels remains to be examined. OBJECTIVES This study compared the metabolic responses to moderate-intensity and vigorous-intensity aerobic exercise. METHODS A two-period crossover trial was conducted under controlled conditions at the Max Rubner-Institute in Karlsruhe, Germany. Seventeen young, healthy, and physically active men performed 30 min moderate-intensity (50% VO2peak) and vigorous-intensity (75% VO2peak) aerobic exercise using two bicycle ergometer protocols in a randomized sequence. Blood samples obtained immediately before exercise and at four time points after exercise were analyzed in an untargeted metabolomics approach, and separate linear mixed models were applied to over 1000 metabolites. RESULTS Vigorous-intensity exercise induced a greater metabolic response than moderate-intensity exercise. Several intensity-dependent metabolites were identified, primarily involved in amino acid metabolism and energy conversion pathways, including N-lactoyl-amino acids, TCA cycle intermediates, N-acetylated amino acids, and acylcholines. The exerkines N-lactoyl-phenylalanine, lactate, and succinate were among the most intensity-dependent metabolites. N-acetylated amino acids and acylcholines were systematically altered by exercise intensity, indicating potential physiological functions. CONCLUSION Exercise intensity significantly affects exercise-induced metabolic alterations and changes in exerkine levels. Our results expand the knowledge about exerkine dynamics and emphasize the role of exercise intensity in promoting physiological adaptations to exercise. The trial was registered on October 5, 2017, at the German Clinical Trials Register under the Registration Number DRKS00009743 (Universal Trial Number of WHO: U1111-1200-2530).
Collapse
Affiliation(s)
- Dirk Weber
- Institute of Sports and Sports Science, Karlsruhe Institute of Technology (KIT), Engler-Bunte-Ring 15, 76131, Karlsruhe, Germany.
| | - Paola G Ferrario
- Department of Physiology and Biochemistry of Nutrition, Max Rubner-Institute, Karlsruhe, Germany
| | - Achim Bub
- Institute of Sports and Sports Science, Karlsruhe Institute of Technology (KIT), Engler-Bunte-Ring 15, 76131, Karlsruhe, Germany
- Department of Physiology and Biochemistry of Nutrition, Max Rubner-Institute, Karlsruhe, Germany
| |
Collapse
|
5
|
Luo T, Li C, Zhou L, Sun H, Yang MM. Protein Acetylation in Age-Related Macular Degeneration: Mechanisms, Roles, and Therapeutic Perspectives. Invest Ophthalmol Vis Sci 2025; 66:30. [PMID: 40402519 DOI: 10.1167/iovs.66.5.30] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/23/2025] Open
Abstract
Age-related macular degeneration (AMD) is a top cause of severe vision loss and blindness in older adults globally. This multifactorial disease arises from genetic, environmental, and age-related factors. Protein acetylation modification plays a key role in AMD progression through both epigenetic and non-epigenetic pathways. This review comprehensively discusses the multidimensional impacts of protein acetylation in AMD, particularly its dynamic regulation of angiogenesis, oxidative stress, inflammatory responses, and cellular senescence. Recent evidence shows that histone acetylation modification inhibits choroidal neovascularization (CNV) formation by regulating vascular endothelial growth factor (VEGF) and hypoxia-inducible factor (HIF-1α) expression, while upregulating the complement inhibitor clusterin to maintain Bruch's membrane integrity. Additionally, the NAD+-dependent deacetylase SIRT1 modulates the deacetylation of transcription factors such as PGC-1α, NF-κB, and FOXO3, enhancing mitochondrial antioxidant function and suppressing inflammatory cascades to disrupt the vicious cycle of oxidative stress and chronic inflammation. In terms of cellular senescence, histone hypoacetylation and hyperacetylation of non-histone proteins (e.g., p53, E2F1) jointly cause retinal pigment epithelial (RPE) cell-cycle arrest and autophagy imbalance, accelerating AMD progression. Genetic evidence further reveals subtype-specific expression changes and epigenetic regulatory mechanisms of histone deacetylases (HDACs), such as HDAC11 and HDAC1/3, in AMD. This article explores the clinical significance of these findings and proposes a novel combined therapeutic strategy. It involves synergistically targeting acetylation homeostasis with HDAC inhibitors (e.g., TSA, AN7) and SIRT1 activators while inhibiting abnormal angiogenesis, repairing metabolic disorders, and restoring autophagy function. This dual-targeting approach may overcome current anti-VEGF therapy limitations and open new precision management avenues for AMD.
Collapse
Affiliation(s)
- Tianyi Luo
- The Second Clinical Medical College of Jinan University, Department of Ophthalmology, Shenzhen People's Hospital, Shenzhen, China
| | - Cunzi Li
- The Second Clinical Medical College of Jinan University, Department of Ophthalmology, Shenzhen People's Hospital, Shenzhen, China
| | - Lan Zhou
- Department of Ophthalmology, Shenzhen People's Hospital (The First Affiliated Hospital, Southern University of Science and Technology; The Second Clinical Medical College, Jinan University), Shenzhen, China
- Post-doctoral Scientific Research Station of Basic Medicine, Jinan University, Guangzhou, China
| | - Hongyan Sun
- Department of Ophthalmology, Shenzhen People's Hospital (The First Affiliated Hospital, Southern University of Science and Technology; The Second Clinical Medical College, Jinan University), Shenzhen, China
| | - Ming Ming Yang
- Department of Ophthalmology, Shenzhen People's Hospital (The First Affiliated Hospital, Southern University of Science and Technology; The Second Clinical Medical College, Jinan University), Shenzhen, China
| |
Collapse
|
6
|
Cui S, Guo C, Yan L, He Y, Wu L. Research on enhancing enzymatic degradation of anti-digestive peptides containing D-amino acids through N-terminal acetylation. Bioorg Chem 2025; 158:108337. [PMID: 40054398 DOI: 10.1016/j.bioorg.2025.108337] [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: 12/30/2024] [Revised: 02/28/2025] [Accepted: 02/28/2025] [Indexed: 03/19/2025]
Abstract
The incorporation of D-amino acids can influence the structure and enzymatic stability of proteins or peptides, especially when they are introduced at enzyme cleavage sites, significantly inhibiting the enzymatic hydrolysis of peptides. The abnormal accumulation of these peptides has been linked to age-related conditions, including cataracts and Alzheimer's disease. N-terminal acetylation, an essential post-translational modification, significantly enhances the physicochemical properties of peptides and plays an essential role in regulating their performance within biological systems. This research examined the impact of N-terminal acetylation on the enzymatic hydrolysis of peptides incorporating D-amino acids. Enzymatic activity assessments showed that N-terminal acetylation greatly promoted the enzymatic breakdown of these peptides by Proteinase K (PROK), with the substrate decay rate constant for the acetylated peptide Ac-6-w increasing by 17.5 times. This enhancement was specific to serine-type proteases, which exhibited a comparable cleavage pattern. Molecular docking further demonstrated that N-terminal acetylation improved interactions within the catalytic triad of serine proteases, leading to faster enzymatic degradation. The results provide novel insights into the impact of N-terminal acetylation on the enzymatic behavior of peptides incorporating D-amino acids, and they propose a potential approach for targeting these peptides to preserve normal physiological functions.
Collapse
Affiliation(s)
- Shuaishuai Cui
- School of Chemical Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Cunxin Guo
- School of Chemical Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Liang Yan
- School of Chemical Sciences, University of Chinese Academy of Sciences, Beijing 100049, China; Sinopep-Allsino Biopharmaceutical Co., Ltd., Jiangsu 222000, China..
| | - Yujian He
- School of Chemical Sciences, University of Chinese Academy of Sciences, Beijing 100049, China; School of Future Technology, University of Chinese Academy of Sciences, Beijing 100049, China.
| | - Li Wu
- School of Chemical Sciences, University of Chinese Academy of Sciences, Beijing 100049, China; State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China.
| |
Collapse
|
7
|
Yoshinaga D, Craven I, Feng R, Prondzynski M, Shani K, Tharani Y, Mayourian J, Joseph M, Walker D, Bortolin RH, Carreon CK, Boss B, Upton S, Parker KK, Pu WT, Bezzerides VJ. Dysregulation of N-terminal acetylation causes cardiac arrhythmia and cardiomyopathy. Nat Commun 2025; 16:3604. [PMID: 40234403 PMCID: PMC12000442 DOI: 10.1038/s41467-025-58539-2] [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: 08/28/2024] [Accepted: 03/21/2025] [Indexed: 04/17/2025] Open
Abstract
N-terminal acetyltransferases including NAA10 catalyze N-terminal acetylation, an evolutionarily conserved co- and post-translational modification. However, little is known about the role of N-terminal acetylation in cardiac homeostasis. To gain insight into cardiac-dependent NAA10 function, we studied a previously unidentified NAA10 variant p.(Arg4Ser) segregating with QT-prolongation, cardiomyopathy, and developmental delay in a large kindred. Here, we show that the NAA10R4S variant reduced enzymatic activity, decreased NAA10-NAA15 complex formation, and destabilized the enzymatic complex N-terminal acetyltransferase A. In NAA10R4S/Y-induced pluripotent stem-cell-derived cardiomyocytes (iPSC-CMs), dysregulation of the late sodium and slow delayed rectifier potassium currents caused severe repolarization abnormalities, consistent with clinical QT prolongation. Engineered heart tissues generated from NAA10R4S/Y-iPSC-CMs had significantly decreased contractile force and sarcomeric disorganization, consistent with the pedigree's cardiomyopathic phenotype. Proteomic studies revealed dysregulation of metabolic pathways and cardiac structural proteins. We identified small molecule and genetic therapies that normalized the phenotype of NAA10R4S/Y-iPSC-CMs. Our study defines the roles of N-terminal acetylation in cardiac regulation and delineates mechanisms underlying QT prolongation, arrhythmia, and cardiomyopathy caused by NAA10 dysfunction.
Collapse
Affiliation(s)
- Daisuke Yoshinaga
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Isabel Craven
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Rui Feng
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Maksymilian Prondzynski
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Kevin Shani
- Disease Biophysics Group, Wyss Institute for Biologically Inspired Engineering, Harvard John A. Paulson School of Engineering and Applied Sciences, Allston, MA, USA
| | - Yashasvi Tharani
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Joshua Mayourian
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Milosh Joseph
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - David Walker
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Raul H Bortolin
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | | | - Bridget Boss
- Department of Pediatric Cardiology, Dartmouth Hitchcock Medical Center, Manchester, NH, USA
| | - Sheila Upton
- Department of Medical Genetics, Dartmouth Hitchcock Medical Center, Manchester, NH, USA
| | - Kevin Kit Parker
- Disease Biophysics Group, Wyss Institute for Biologically Inspired Engineering, Harvard John A. Paulson School of Engineering and Applied Sciences, Allston, MA, USA
| | - William T Pu
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Vassilios J Bezzerides
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA.
- Department of Pediatric Cardiology, Dartmouth Hitchcock Medical Center, Manchester, NH, USA.
| |
Collapse
|
8
|
Wang M, Li Y, Wang J, Oh SH, Cao Y, Chen R. Integrating short-read and long-read single-cell RNA sequencing for comprehensive transcriptome profiling in mouse retina. Genome Res 2025; 35:740-754. [PMID: 40050124 PMCID: PMC12047235 DOI: 10.1101/gr.279167.124] [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: 02/20/2024] [Accepted: 11/21/2024] [Indexed: 03/28/2025]
Abstract
The vast majority of protein-coding genes in the human genome produce multiple mRNA isoforms through alternative splicing, significantly enhancing the complexity of the transcriptome and proteome. To establish an efficient method for characterizing transcript isoforms within tissue samples, we conducted a systematic comparison between single-cell long-read and conventional short-read RNA sequencing techniques. The transcriptome of approximately 30,000 mouse retina cells was profiled using 1.54 billion Illumina short reads and 1.40 billion Oxford Nanopore Technologies long reads. Consequently, we identify 44,325 transcript isoforms, with a notable 38% previously uncharacterized and 17% expressed exclusively in distinct cellular subclasses. We observe that long-read sequencing not only matches the gene expression and cell-type annotation performance of short-read sequencing but also excel in the precise identification of transcript isoforms. While transcript isoforms are often shared across various cell types, their relative abundance shows considerable cell type-specific variation. The data generated from our study significantly enhance the existing repertoire of transcript isoforms, thereby establishing a resource for future research into the mechanisms and implications of alternative splicing within retinal biology and its links to related diseases.
Collapse
Affiliation(s)
- Meng Wang
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Yumei Li
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas 77030, USA
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Jun Wang
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Soo Hwan Oh
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Yexuan Cao
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Rui Chen
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas 77030, USA;
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas 77030, USA
| |
Collapse
|
9
|
Crawford CEW, Burslem GM. Acetylation: a new target for protein degradation in cancer. Trends Cancer 2025; 11:403-420. [PMID: 40055119 PMCID: PMC11981854 DOI: 10.1016/j.trecan.2025.01.013] [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: 10/08/2024] [Revised: 01/15/2025] [Accepted: 01/24/2025] [Indexed: 04/11/2025]
Abstract
Acetylation is an increasing area of focus for cancer research as it is closely related to a variety of cellular processes through modulation of histone and non-histone proteins. However, broadly targeting acetylation threatens to yield nonselective toxic effects owing to the vital role of acetylation in cellular function. There is thus a pressing need to elucidate and characterize the specific cancer-relevant roles of acetylation for future therapeutic design. Acetylation-mediated protein homeostasis is an example of selective acetylation that affects a myriad of proteins as well as their correlated functions. We review recent examples of acetylation-mediated protein homeostasis that have emerged as key contributors to tumorigenesis, tumor proliferation, metastasis, and/or drug resistance, and we discuss their implications for future exploration of this intriguing phenomenon.
Collapse
Affiliation(s)
- Callie E W Crawford
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, PA 19104, USA
| | - George M Burslem
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, PA 19104, USA; Department of Cancer Biology and Epigenetics Institute, Perelman School of Medicine, University of Pennsylvania, PA 19104, USA.
| |
Collapse
|
10
|
Zhang T, Jin Q, Ji J. Antimicrobial Peptides and Their Mimetics: Promising Candidates of Next-Generation Therapeutic Agents Combating Multidrug-Resistant Bacteria. Adv Biol (Weinh) 2025; 9:e2400461. [PMID: 39913150 DOI: 10.1002/adbi.202400461] [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: 08/03/2024] [Revised: 01/05/2025] [Indexed: 02/07/2025]
Abstract
The increasing morbidity and mortality caused by multidrug-resistant bacteria alerts human beings to the fact that conventional antibiotics are no longer reliable and effective alternatives are imperatively needed. Owing to wide range of sources, diverse structures, and unique mode of action, antimicrobial peptides have been highly anticipated and extensively studied in recent years. Besides, the integration of artificial intelligence helps researchers gain access to the vast unexplored chemical space, which opens more opportunities for the optimization and design of novel structures. Moreover, Due to advances in chemistry and synthetic biology, researchers have also begun to focus on the potential of chemical mimetics of antimicrobial peptides. In this review, a comprehensive discussion about natural and synthesized antimicrobial peptides as well as their chemical mimetics is made, so as to provide a comprehensive summary of this field and inspire follow-up research.
Collapse
Affiliation(s)
- Tianyi Zhang
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310058, P. R. China
| | - Qiao Jin
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310058, P. R. China
| | - Jian Ji
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310058, P. R. China
- State Key Laboratory of Transvascular Implantation Devices, The Second Affiliated Hospital, Zhejiang University School of Medicine, 88 Jiefang Rd, Hangzhou, 310009, P. R. China
| |
Collapse
|
11
|
Sakato-Antoku M, Patel N, Inaba M, Rao Q, Yang J, Patel-King RS, Inaba K, Balsbaugh JL, King SM. Phyloproteomics reveals conserved patterns of axonemal dynein methylation across the motile ciliated eukaryotes. Mol Biol Cell 2025; 36:ar49. [PMID: 39969973 PMCID: PMC12005109 DOI: 10.1091/mbc.e25-02-0055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2025] [Accepted: 02/14/2025] [Indexed: 02/20/2025] Open
Abstract
Axonemal dynein assembly occurs in the cytoplasm and numerous cytosolic factors are specifically required for this process. Recently, one factor (DNAAF3/PF22) was identified as a methyltransferase. Examination of Chlamydomonas dyneins found they are methylated at substoichiometric levels on multiple sites, including Lys and Arg residues in several of the nucleotide-binding domains and on the microtubule-binding region. Given the highly conserved nature of axonemal dyneins, one key question is whether methylation happens only in dyneins from the chlorophyte algae, or whether these modifications occur more broadly throughout the motile ciliated eukaryotes. Here we take a phyloproteomic approach and examine dynein methylation in a wide range of eukaryotic organisms bearing motile cilia. We find unambiguous evidence for methylation of axonemal dyneins in alveolates, chlorophytes, trypanosomes, and a broad range of metazoans. Intriguingly, we were unable to identify a single instance of methylation on Drosophila melanogaster sperm dyneins even though dipterans express a Dnaaf3 orthologue, or in spermatozoids of the fern Ceratopteris, which assembles inner arms but lacks both outer arm dyneins and DNAAF3. Thus, methylation of axonemal dyneins has been broadly conserved in most eukaryotic groups and has the potential to variably modify the function of these motors.
Collapse
Affiliation(s)
- Miho Sakato-Antoku
- Department of Molecular Biology and Biophysics, University of Connecticut Health Center, Farmington, CT 06030-3305
| | - Nikisha Patel
- Department of Biology, Trinity College, Hartford, CT 06106
| | - Mayu Inaba
- Department of Cell Biology, University of Connecticut Health Center, Farmington, CT 06030
| | - Qinhui Rao
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT 06511
| | - Jun Yang
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT 06511
| | - Ramila S. Patel-King
- Department of Molecular Biology and Biophysics, University of Connecticut Health Center, Farmington, CT 06030-3305
| | - Kazuo Inaba
- Shimoda Marine Research Center, University of Tsukuba, Shizuoka 415-0025, Japan
| | - Jeremy L. Balsbaugh
- Proteomics and Metabolomics Facility, Center for Open Research Resources and Equipment, University of Connecticut, Storrs, CT 06269
| | - Stephen M. King
- Department of Molecular Biology and Biophysics, University of Connecticut Health Center, Farmington, CT 06030-3305
| |
Collapse
|
12
|
Kuang YL, Locatelli CA, Qin Y, Zhang Y, Theusch E, Muñoz-Howell A, Sanchez G, Lu M, Nguyen MA, Yalamanchili T, Wang X, Nalula G, Mattis AN, Oni-Orisan A, Iribarren C, Krauss RM, Mulvihill EE, Medina MW. MIR192 Upregulates GLP-1 Receptor and Improves Statin-Induced Impairment of Insulin Secretion. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.18.643960. [PMID: 40166140 PMCID: PMC11956930 DOI: 10.1101/2025.03.18.643960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Statins are a commonly prescribed cholesterol lowering drug class that can increase the risk of new-onset diabetes (NOD). To investigate the molecular mechanisms underlying this effect, we generated human induced pluripotent stem cells (iPSCs) from individuals identified from electronic health records of Kaiser Permanente of Northern California who were susceptible to developing NOD after statin initiation or controls who maintained stable fasting glucose on statin treatment. RNA-seq analysis of iPSCs incubated with atorvastatin, simvastatin or mock buffer for 24 hours identified the long non-coding RNA MIR194-2HG as a top candidate gene. Statin-induced increases in its expression were observed in NOD resistant controls, while statin-induced reductions occurred in NOD susceptible cases. MIR194-2HG encompasses two microRNA genes: MIR192 and MIR194-2. The mature microRNA miR-192-5p, derived from the 5' arm of MIR192, was predicted to bind the 3'UTR of the glucagon like peptide 1 (GLP-1) receptor (GLP1R) transcript. Transfection of a rat insulinoma cell line INS-1 with a miR-192-5p mimic increased Glp1r transcript (1.41-fold) and protein (1.51-fold) levels compared to a scrambled control. Using a luciferase reporter containing the human GLP1R 3'UTR, miR-192-5p overexpression similarly increased luciferase signal (1.44-fold). The miR-192-5p mimic enhanced glucose stimulated insulin secretion (GSIS) in response to GLP1R agonists (1.64-1.81-fold) and rescued simvastatin-induced GSIS impairment in INS-1 cells. Wildtype mice treated with miR-192 AAV8 had improved glucose sensitivity. Islets isolated from these mice exhibited enhanced GLP-1 potentiated GSIS during perifusion ex vivo. These effects were absent in the DIRKO (Glp1r/Gipr double knockout) mouse islets, consistent with the idea that miR-192 promotes GLP-1 mediated GSIS through GLP1R. These findings implicate MIR192 in statin-induced impairment of GSIS by modulating GLP1R, potentially contributing to the susceptibility to NOD in statin users.
Collapse
Affiliation(s)
- Yu-Lin Kuang
- Department of Pediatrics, University of California San Francisco, Oakland, CA, USA
| | - Cassandra A.A. Locatelli
- Department of Biochemistry, Microbiology and Immunology, The University of Ottawa, Ottawa, Ontario, Canada
- University of Ottawa Heart Institute, Ottawa, Ontario, Canada
| | - Yuanyuan Qin
- Department of Pediatrics, University of California San Francisco, Oakland, CA, USA
| | - Yuqing Zhang
- Department of Pediatrics, University of California San Francisco, Oakland, CA, USA
| | - Elizabeth Theusch
- Department of Pediatrics, University of California San Francisco, Oakland, CA, USA
| | - Antonio Muñoz-Howell
- Department of Pediatrics, University of California San Francisco, Oakland, CA, USA
| | - Gabriela Sanchez
- Kaiser Permanente Division of Research, 2000 Broadway, Oakland, CA, USA
| | - Meng Lu
- Kaiser Permanente Division of Research, 2000 Broadway, Oakland, CA, USA
| | - My-Anh Nguyen
- University of Ottawa Heart Institute, Ottawa, Ontario, Canada
| | - Tanvi Yalamanchili
- Department of Pediatrics, University of California San Francisco, Oakland, CA, USA
| | - Xuanwen Wang
- Department of Pediatrics, University of California San Francisco, Oakland, CA, USA
| | - Gilbert Nalula
- Department of Pediatrics, University of California San Francisco, Oakland, CA, USA
| | - Aras N. Mattis
- Department of Pathology, University of California San Francisco, San Francisco, CA, USA
- Board Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA, USA
- The Liver Center, University of California San Francisco, San Francisco, CA, USA
| | - Akinyemi Oni-Orisan
- Department of Clinical Pharmacy, University of California San Francisco, San Francisco, CA, USA
- Institute for Human Genetics, University of California San Francisco, San Francisco, CA, USA
| | - Carlos Iribarren
- Kaiser Permanente Division of Research, 2000 Broadway, Oakland, CA, USA
| | - Ronald M. Krauss
- Department of Pediatrics, University of California San Francisco, Oakland, CA, USA
- Department of Medicine, University of California San Francisco, Oakland, CA, USA
| | - Erin E. Mulvihill
- Department of Biochemistry, Microbiology and Immunology, The University of Ottawa, Ottawa, Ontario, Canada
- University of Ottawa Heart Institute, Ottawa, Ontario, Canada
| | - Marisa W. Medina
- Department of Pediatrics, University of California San Francisco, Oakland, CA, USA
- The Liver Center, University of California San Francisco, San Francisco, CA, USA
- Institute for Human Genetics, University of California San Francisco, San Francisco, CA, USA
| |
Collapse
|
13
|
Larios-Serrato V, Vázquez-Manjarrez N, Resendis-Antonio O, Rios-Sarabia N, Meza B, Fiehn O, Torres J. Analyses of Saliva Metabolome Reveal Patterns of Metabolites That Differentiate SARS-CoV-2 Infection and COVID-19 Disease Severity. Metabolites 2025; 15:192. [PMID: 40137156 PMCID: PMC11944064 DOI: 10.3390/metabo15030192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2025] [Revised: 02/27/2025] [Accepted: 03/04/2025] [Indexed: 03/27/2025] Open
Abstract
BACKGROUND The metabolome of COVID-19 patients has been studied sparsely, with most research focusing on a limited number of plasma metabolites or small cohorts. This is the first study to test saliva metabolites in COVID-19 patients in a comprehensive way, revealing patterns significantly linked to disease and severity, highlighting saliva's potential as a non-invasive tool for pathogenesis or diagnostic studies. METHODS We included 30 asymptomatic subjects with no prior COVID-19 infection or vaccination, 102 patients with mild SARS-CoV-2 infection, and 61 hospitalized patients with confirmed SARS-CoV-2 status. Saliva samples were analyzed using hydrophilic interaction liquid chromatography-mass spectrometry (HILIC-MS/MS) in positive and negative ionization modes. RESULTS Significant differences in metabolites were identified in COVID-19 patients, with distinct patterns associated with disease severity. Dipeptides such as Val-Glu and Met-Gln were highly elevated in moderate cases, suggesting specific protease activity related to SARS-CoV-2. Acetylated amino acids like N-acetylserine and N-acetylhistidine increased in severe cases. Bacterial metabolites, including muramic acid and indole-3-carboxaldehyde, were higher in mild-moderate cases, indicating that oral microbiota differs according to disease severity. In severe cases, polyamines and organ-damage-related metabolites, such as N-acetylspermine and 3-methylcytidine, were significantly increased. Interestingly, most metabolites that were reduced in moderate cases were elevated in severe cases. CONCLUSIONS Saliva metabolomics offers insightful information that is potentially useful in studying COVID-19 severity and for diagnosis.
Collapse
Affiliation(s)
- Violeta Larios-Serrato
- Departamento de Bioquímica, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City 11340, Mexico;
| | - Natalia Vázquez-Manjarrez
- Unidad de Metabolómica, Departamento Fisiologia de la Nutricion, Instituto de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City 14080, Mexico;
| | - Osbaldo Resendis-Antonio
- Human Systems Biology Laboratory, Instituto Nacional de Medicina Genómica, Centro de Ciencias de la Complejidad & Coordinación de la Investigación Científica-Red de Apoyo a la Investigación, Universidad Nacional Autonoma de Mexico, Mexico City 04510, Mexico;
| | - Nora Rios-Sarabia
- Unidad de Investigación Médica en Enfermedades Infecciosas, UMAE Pediatría, Centro Médico Nacional SXXI, Instituto Mexicano del Seguro Social, Mexico City 06920, Mexico;
| | - Beatriz Meza
- Departamento Académico de Ciencia Animal y Conservación del Hábitat, Departamento Académico de Ingenierías en Pesquerías, Universidad Autónoma de Baja California Sur, La Paz 023080, Mexico;
- Centro de Investigaciones Biológicas del Noroeste SC, La Paz 23205, Mexico
| | - Oliver Fiehn
- NIH West Coast Metabolomics Center, UC Davis Genome Center, Davis, CA 95616, USA;
| | - Javier Torres
- Unidad de Investigación Médica en Enfermedades Infecciosas, UMAE Pediatría, Centro Médico Nacional SXXI, Instituto Mexicano del Seguro Social, Mexico City 06920, Mexico;
| |
Collapse
|
14
|
Cates K, Hung V, Barna M. Ribosome-associated proteins: unwRAPping ribosome heterogeneity in the twenty-first century. Philos Trans R Soc Lond B Biol Sci 2025; 380:20230378. [PMID: 40045784 PMCID: PMC11883435 DOI: 10.1098/rstb.2023.0378] [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: 08/27/2024] [Revised: 12/07/2024] [Accepted: 01/06/2025] [Indexed: 03/09/2025] Open
Abstract
The definition of the ribosome as the monolithic machinery in cells that synthesizes all proteins in the cell has persisted for the better part of a century. Yet, research has increasingly revealed that ribosomes are dynamic, multimodal complexes capable of fine-tuning gene expression. This translation regulation may be achieved by ribosome-associated proteins (RAPs), which play key roles as modular trans-acting factors that are dynamic across different cellular contexts and can mediate the recruitment of specific transcripts or the modification of RNA or ribosomal proteins. As a result, RAPs have the potential to rapidly regulate translation within specific subcellular regions, across different cell or tissue types, in response to signalling, or in disease states. In this article, we probe the definition of the eukaryotic ribosome and review the major layers of additional proteins that expand the definition of ribosomes in the twenty-first century. We pose RAPs as key modulators that impart ribosome function in cellular processes, development and disease.This article is part of the discussion meeting issue 'Ribosome diversity and its impact on protein synthesis, development and disease'.
Collapse
Affiliation(s)
- Kitra Cates
- Department of Genetics, Stanford University School of Medicine, Stanford, CA94305, USA
| | - Victoria Hung
- Department of Genetics, Stanford University School of Medicine, Stanford, CA94305, USA
| | - Maria Barna
- Department of Genetics, Stanford University School of Medicine, Stanford, CA94305, USA
| |
Collapse
|
15
|
Ji Y, Gao Y, Li X, Hu H, Zhang Y, Shi Y. Discovery of proteolytically stable monocyte locomotion inhibitory factor peptide through systematic structural optimization. Eur J Med Chem 2025; 285:117237. [PMID: 39808971 DOI: 10.1016/j.ejmech.2025.117237] [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: 11/08/2024] [Revised: 12/23/2024] [Accepted: 01/01/2025] [Indexed: 01/16/2025]
Abstract
The identification of novel molecular candidates capable of treating osteoarthritis (OA) has significant clinical implications. Monocyte locomotion inhibitory factor peptide (MLIF) is a pentapeptide derived from Entamoeba histolytica. It has been found possesses selective anti-inflammatory effects both in vitro and in vivo. Nonetheless, like many peptide therapeutics, MLIF has relatively poor proteolytic stability and short half-life in vivo, hindering its effective clinical applicability. To overcome these limitations, structural optimizations are needed to enhance the stability of MLIF while preserving or even enhancing its anti-inflammatory activities. Herein, a series of MLIF derivatives were designed and synthesized based on diverse structural modifications including N-terminal modifications, d-amino acid replacement, N-methylation, sulfhydryl modification, cyclization, and splicing strategy. Among all the MLIF derivatives, MLIF 30 with replacing l-methionine (Met) with D-Met and linking the polyethylene glycol (PEG) to cysteine (Cys) of MLIF displayed enhanced in vitro anti-inflammatory activities. Further in vivo experiment demonstrated MLIF 30 could reduce cartilage inflammation and attenuate cartilage damage more effectively in the collagenase induced osteoarthritis (CIOA) mice due to its improved serum stability compared to the linear MLIF. These findings laid foundation for the development of potent and stable anti-inflammatory peptide therapeutics and pushed the frontier of MLIF for clinical OA treatment.
Collapse
Affiliation(s)
- Yajing Ji
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China; School of Medicine, Shanghai University, Shanghai, 200444, China
| | - Yuan Gao
- School of Medicine, Shanghai University, Shanghai, 200444, China
| | - Xiang Li
- School of Pharmacy, Naval Medical University, Shanghai, 200433, China
| | - Honggang Hu
- School of Medicine, Shanghai University, Shanghai, 200444, China; Shanghai Integration and Innovation Center of Marine Medical Engineering, China; Shanghai Engineering Research Center of Organ Repair, China
| | - Yuefan Zhang
- School of Medicine, Shanghai University, Shanghai, 200444, China.
| | - Yejiao Shi
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China; Shanghai Integration and Innovation Center of Marine Medical Engineering, China.
| |
Collapse
|
16
|
Roston TM, Bezzerides VJ, Roberts JD, Abrams DJ. Management of ultrarare inherited arrhythmia syndromes. Heart Rhythm 2025; 22:832-843. [PMID: 39154872 DOI: 10.1016/j.hrthm.2024.08.023] [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: 05/01/2024] [Revised: 08/06/2024] [Accepted: 08/08/2024] [Indexed: 08/20/2024]
Abstract
Ultrarare inherited arrhythmia syndromes are increasingly diagnosed as a result of increased awareness as well as increased availability and reduced cost of genetic testing. Yet by definition, their rarity and heterogeneous expression make development of evidence-based management strategies more challenging, typically employing strategies garnered from similar genetic cardiac disorders. For the most part, reliance on anecdotal experiences, expert opinion, and small retrospective cohort studies is the only means to diagnose and to treat these patients. Here we review the management of specific ultrarare inherited arrhythmic syndromes together with the genetic and molecular basis, which will become increasingly important with the development of targeted therapies to correct the biologic basis of these disorders.
Collapse
Affiliation(s)
- Thomas M Roston
- Division of Cardiology and Centre for Cardiovascular Innovation, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Vassilios J Bezzerides
- Center for Cardiovascular Genetics, Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Jason D Roberts
- Population Health Research Institute, McMaster University, and Hamilton Health Sciences, Hamilton, Ontario, Canada
| | - Dominic J Abrams
- Center for Cardiovascular Genetics, Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
17
|
Popovic M, Isermann L, Geißen S, Senft K, Georgomanolis T, Baldus S, Frezza C, Trifunovic A. Tissue-specific adaptations to cytochrome c oxidase deficiency shape physiological outcomes. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167567. [PMID: 39613003 DOI: 10.1016/j.bbadis.2024.167567] [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: 07/31/2024] [Revised: 11/05/2024] [Accepted: 11/05/2024] [Indexed: 12/01/2024]
Abstract
It becomes increasingly clear that the tissue specificity of mitochondrial diseases might in part rely on their ability to compensate for mitochondrial defects, contributing to the heterogeneous nature of mitochondrial diseases. Here, we investigated tissue-specific responses to cytochrome c oxidase (CIV or COX) deficiency using a mouse model with heart and skeletal muscle-specific depletion of the COX assembly factor COX10. At three weeks of age, both tissues exhibit pronounced CIV depletion but respond differently to oxidative phosphorylation (OXPHOS) impairment. Heart-specific COX10 depletion caused severe dilated cardiomyopathy, while skeletal muscle experiences less damage. Cardiac CIV deficiency triggered extensive metabolic remodelling and stress response activation, potentially worsening cardiomyopathy, whereas skeletal muscle showed no stress response or significant metabolic changes. Our findings highlight distinct tissue capacities for managing CIV deficiency, explaining how identical primary defects can lead to different phenotypic outcomes and contribute to the heterogeneous progression of mitochondrial diseases.
Collapse
Affiliation(s)
- Milica Popovic
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, 50931 Cologne, Germany; Institute for Mitochondrial Diseases and Aging, Faculty of Medicine and University Hospital Cologne, University of Cologne, D-50931 Cologne, Germany
| | - Lea Isermann
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, 50931 Cologne, Germany; Institute for Mitochondrial Diseases and Aging, Faculty of Medicine and University Hospital Cologne, University of Cologne, D-50931 Cologne, Germany
| | - Simon Geißen
- Department for Experimental Cardiology, Faculty of Medicine, University of Cologne, 50937, Germany; Clinic III for Internal Medicine, University Hospital Cologne, 50937, Germany; Center for Molecular Medicine (CMMC), University of Cologne, 50931 Cologne, Germany
| | - Katharina Senft
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, 50931 Cologne, Germany; Institute for Mitochondrial Diseases and Aging, Faculty of Medicine and University Hospital Cologne, University of Cologne, D-50931 Cologne, Germany
| | - Theodoros Georgomanolis
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, 50931 Cologne, Germany; Institute for Metabolomics in Ageing, Faculty of Medicine and University Hospital Cologne, University of Cologne, D-50931 Cologne, Germany
| | - Stephan Baldus
- Department for Experimental Cardiology, Faculty of Medicine, University of Cologne, 50937, Germany; Clinic III for Internal Medicine, University Hospital Cologne, 50937, Germany; Center for Molecular Medicine (CMMC), University of Cologne, 50931 Cologne, Germany
| | - Christian Frezza
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, 50931 Cologne, Germany; Institute for Metabolomics in Ageing, Faculty of Medicine and University Hospital Cologne, University of Cologne, D-50931 Cologne, Germany; Institute of Genetics, Faculty of Mathematics and Natural Sciences, University of Cologne, D-50931 Cologne, Germany
| | - Aleksandra Trifunovic
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, 50931 Cologne, Germany; Institute for Mitochondrial Diseases and Aging, Faculty of Medicine and University Hospital Cologne, University of Cologne, D-50931 Cologne, Germany; Center for Molecular Medicine (CMMC), University of Cologne, 50931 Cologne, Germany.
| |
Collapse
|
18
|
Flores JA, O'Neill SE, Jarodsky JM, Reichow SL. Calcium induced N-terminal gating and pore collapse in connexin-46/50 gap junctions. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.12.637955. [PMID: 39990482 PMCID: PMC11844560 DOI: 10.1101/2025.02.12.637955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/25/2025]
Abstract
Gap junctions facilitate electrical and metabolic coupling essential for tissue function. Under ischemic conditions ( e.g., heart attack or stroke), elevated intracellular calcium (Ca 2+ ) levels uncouple these cell-to-cell communication pathways to protect healthy cells from cytotoxic signals. Using single-particle cryo-EM, we elucidate details of the Ca 2+ -induced gating mechanism of native connexin-46/50 (Cx46/50) gap junctions. The resolved structures reveal Ca 2+ binding sites within the channel pore that alter the chemical environment of the permeation pathway and induce diverse occluded and gated states through N-terminal domain remodeling. Moreover, subunit rearrangements lead to pore collapse, enabling steric blockade by the N-terminal domains, reminiscent of the "iris model" of gating proposed over four decades ago. These findings unify and expand key elements of previous gating models, providing mechanistic insights into how Ca 2+ signaling regulates gap junction uncoupling and broader implications for understanding cell stress responses and tissue protection.
Collapse
|
19
|
Makwana R, Patel R, O'Neill R, Marchi E, Lyon GJ. The Cardiovascular Manifestations and Management Recommendations for Ogden Syndrome. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.02.11.25321331. [PMID: 40236393 PMCID: PMC11996587 DOI: 10.1101/2025.02.11.25321331] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
The NatA complex is composed of the NAA10, NAA15, and HYPK subunits. It is primarily responsible for N-terminal acetylation, a critical post-translational modification in eukaryotes. Pathogenic variants within NAA10 cause Ogden Syndrome (OS), which is characterized by varying degrees of intellectual disability, hypotonia, developmental delay, and cardiac abnormalities. Although the cardiac manifestations of the disease have been described extensively in case reports, there has not been a study focusing on the cardiac manifestations and their recommended clinical cardiac management. In this study, we describe the cardiac manifestations of OS in a cohort of 85 probands. We found increased incidence of structural and electrophysiologic abnormalities, with particularly high prevalence of QT interval prolongation. Sub-analysis showed that male probands and those with variants within the NAA15-binding domain had more severe phenotypes than females or those with variants outside of the NAA15-binding domain. Our results suggest that an OS diagnosis should be accompanied by full cardiac workup with emphasis on echocardiogram for structural defects and EKG/Holter monitoring for electrophysiologic abnormalities. Additionally, we strongly recommend that the use of QT-prolonging drugs be followed up with routine electrophysiological monitoring or consultation with a pediatric cardiologist. We hope this study guides clinicians and caregivers treating patients with OS and moves the field toward a standardized diagnostic workup for patients with this condition.
Collapse
|
20
|
Pollitt SL, Levy AD, Anderson MC, Blanpied TA. Large Donor CRISPR for Whole-Coding Sequence Replacement of Cell Adhesion Molecule LRRTM2. J Neurosci 2025; 45:e1461242024. [PMID: 39824639 PMCID: PMC11823385 DOI: 10.1523/jneurosci.1461-24.2024] [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: 08/01/2024] [Revised: 11/01/2024] [Accepted: 12/02/2024] [Indexed: 01/20/2025] Open
Abstract
The cell adhesion molecule leucine-rich repeat transmembrane neuronal protein 2 (LRRTM2) is crucial for synapse development and function. However, our understanding of its endogenous trafficking has been limited due to difficulties in manipulating its coding sequence (CDS) using standard genome editing techniques. Instead, we replaced the entire LRRTM2 CDS by adapting a two-guide CRISPR knock-in method, enabling complete control of LRRTM2. In primary rat hippocampal cultures dissociated from embryos of both sexes, N-terminally tagged, endogenous LRRTM2 was found in 80% of synapses, and synaptic LRRTM2 content correlated with PSD-95 and AMPAR levels. LRRTM2 was also enriched with AMPARs outside synapses, demonstrating the sensitivity of this method to detect relevant new biology. Finally, we leveraged total genomic control to increase the synaptic levels of LRRTM2 via simultaneous mutation of its C-terminal domain, which did not correspondingly increase AMPAR enrichment. The coding region of thousands of genes span lengths suitable for whole-CDS replacement, suggesting this simple approach will enable straightforward structure-function analysis in neurons.
Collapse
Affiliation(s)
- Stephanie L Pollitt
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland 21201-1509
- University of Maryland-Medicine Institute for Neuroscience Discovery (UM-MIND), University of Maryland School of Medicine, Baltimore, Maryland 21201-1509
| | - Aaron D Levy
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland 21201-1509
- University of Maryland-Medicine Institute for Neuroscience Discovery (UM-MIND), University of Maryland School of Medicine, Baltimore, Maryland 21201-1509
| | - Michael C Anderson
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland 21201-1509
- University of Maryland-Medicine Institute for Neuroscience Discovery (UM-MIND), University of Maryland School of Medicine, Baltimore, Maryland 21201-1509
- Program in Neuroscience, University of Maryland School of Medicine, Baltimore, Maryland 21201-1509
| | - Thomas A Blanpied
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland 21201-1509
- University of Maryland-Medicine Institute for Neuroscience Discovery (UM-MIND), University of Maryland School of Medicine, Baltimore, Maryland 21201-1509
- Program in Neuroscience, University of Maryland School of Medicine, Baltimore, Maryland 21201-1509
| |
Collapse
|
21
|
Bergendorf A, Park JH, Ball BK, Brubaker DK. Mouse-to-human modeling of microglia single-nuclei transcriptomics identifies immune signaling pathways and potential therapeutic candidates associated with Alzheimer's disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.07.637100. [PMID: 39975195 PMCID: PMC11839086 DOI: 10.1101/2025.02.07.637100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disease characterized by memory loss and behavior change. Studies have found that dysregulation of microglial cells is pivotal to AD pathology. These mechanisms have been studied in mouse models to uncover potential therapeutic biomarkers. Despite these findings, there are limitations to the translatable biological information from mice to humans due to differences in physiology, timeline of disease, and the heterogeneity of humans. To address the inter-species discrepancies, we developed a novel implementation of the Translatable Components Regression (TransComp-R) framework, which integrated microglia single-nuclei mouse and human transcriptomics data to identify biological pathways in mice predictive of human AD. We compared model variations with sparse and traditional principal component analysis. We found that both dimensionality reduction techniques encoded similar AD disease biology on mouse principal components with limited differences in technical performance. Several mouse sparse principal components explained high amounts of variance in humans and significantly differentiated human AD from control microglial cells. Additionally, we identified FDA-approved medications that induced gene expression profiles correlated with projections of healthy human microglia on mouse principal components. Such medications included cabergoline, selumetinib, and palbociclib. This computational framework may support uncovering cross-species disease insights and candidate pharmacological solutions from single-cell datasets.
Collapse
Affiliation(s)
- Alexander Bergendorf
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, USA
- Center for Global Health & Diseases, Department of Pathology, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Jee Hyun Park
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, USA
| | - Brendan K. Ball
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, USA
| | - Douglas K. Brubaker
- Center for Global Health & Diseases, Department of Pathology, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
- The Blood, Heart, Lung, and Immunology Research Center, Case Western Reserve University, University Hospitals of Cleveland, Cleveland, OH 44106, USA
| |
Collapse
|
22
|
Huggler KS, Mellado Fritz CA, Flickinger KM, Chang GR, McGuire MF, Cantor JR. Hexokinase detachment from mitochondria drives the Warburg effect to support compartmentalized ATP production. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.07.637120. [PMID: 39975027 PMCID: PMC11839068 DOI: 10.1101/2025.02.07.637120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Hexokinase (HK) catalyzes the synthesis of glucose-6-phosphate, marking the first committed step of glucose metabolism. Most cancer cells express two homologous isoforms (HK1 and HK2) that can each bind to the outer mitochondrial membrane (OMM). CRISPR screens across hundreds of cancer cell lines indicate that both are dispensable for cell growth in traditional culture media. By contrast, HK2 deletion impairs cell growth in Human Plasma-Like Medium (HPLM). Here, we find that HK2 is required to maintain sufficient cytosolic (OMM-detached) HK activity under conditions that enhance HK1 binding to the OMM. Notably, OMM-detached rather than OMM-docked HK promotes "aerobic glycolysis" (Warburg effect), an enigmatic phenotype displayed by most proliferating cells. We show that several proposed theories for this phenotype cannot explain the HK2 dependence and instead find that HK2 deletion severely impairs glycolytic ATP production with little impact on total ATP yield for cells in HPLM. Our results reveal a basis for conditional HK2 essentiality and suggest that demand for compartmentalized ATP synthesis underlies the Warburg effect.
Collapse
|
23
|
Tsui WHA, Ding SC, Jiang P, Lo YMD. Artificial intelligence and machine learning in cell-free-DNA-based diagnostics. Genome Res 2025; 35:1-19. [PMID: 39843210 PMCID: PMC11789496 DOI: 10.1101/gr.278413.123] [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] [Indexed: 01/24/2025]
Abstract
The discovery of circulating fetal and tumor cell-free DNA (cfDNA) molecules in plasma has opened up tremendous opportunities in noninvasive diagnostics such as the detection of fetal chromosomal aneuploidies and cancers and in posttransplantation monitoring. The advent of high-throughput sequencing technologies makes it possible to scrutinize the characteristics of cfDNA molecules, opening up the fields of cfDNA genetics, epigenetics, transcriptomics, and fragmentomics, providing a plethora of biomarkers. Machine learning (ML) and/or artificial intelligence (AI) technologies that are known for their ability to integrate high-dimensional features have recently been applied to the field of liquid biopsy. In this review, we highlight various AI and ML approaches in cfDNA-based diagnostics. We first introduce the biology of cell-free DNA and basic concepts of ML and AI technologies. We then discuss selected examples of ML- or AI-based applications in noninvasive prenatal testing and cancer liquid biopsy. These applications include the deduction of fetal DNA fraction, plasma DNA tissue mapping, and cancer detection and localization. Finally, we offer perspectives on the future direction of using ML and AI technologies to leverage cfDNA fragmentation patterns in terms of methylomic and transcriptional investigations.
Collapse
Affiliation(s)
- W H Adrian Tsui
- Center for Novostics, Hong Kong Science Park, Pak Shek Kok, New Territories, Hong Kong SAR, China
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China
- Department of Chemical Pathology, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China
| | - Spencer C Ding
- Center for Novostics, Hong Kong Science Park, Pak Shek Kok, New Territories, Hong Kong SAR, China
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China
- Department of Chemical Pathology, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China
| | - Peiyong Jiang
- Center for Novostics, Hong Kong Science Park, Pak Shek Kok, New Territories, Hong Kong SAR, China
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China
- Department of Chemical Pathology, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China
- State Key Laboratory of Translational Oncology, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China
| | - Y M Dennis Lo
- Center for Novostics, Hong Kong Science Park, Pak Shek Kok, New Territories, Hong Kong SAR, China;
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China
- Department of Chemical Pathology, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China
- State Key Laboratory of Translational Oncology, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China
| |
Collapse
|
24
|
Doyle TM, Janes K, Xiao WH, Kolar GR, Luecke HF, Gratton MA, Tosh DK, Jacobson KA, Bennett GJ, Salvemini D. Mitochondrial A 3 Adenosine Receptor as a Mechanism for the Protective Effects of A 3AR Agonists on Chemotherapy-Induced Neuropathic Pain. J Neurosci 2025; 45:e1268242024. [PMID: 39653498 PMCID: PMC11735668 DOI: 10.1523/jneurosci.1268-24.2024] [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: 07/02/2024] [Revised: 10/11/2024] [Accepted: 10/29/2024] [Indexed: 01/18/2025] Open
Abstract
Alterations in mitochondrial function are the linchpin in numerous disease states including in the development of chemotherapy-induced neuropathic pain (CIPN), a major dose-limiting toxicity of widely used chemotherapeutic cytotoxins. In CIPN, mitochondrial dysfunction is characterized by deficits in mitochondrial bioenergetics (e.g., decreased ATP production) that are thought to drive the degeneration of the peripheral nerve sensory axon terminal sensory arbors in the skin (the intraepidermal nerve fibers; IENFs) and induce abnormal spontaneous discharge in peripheral nerve sensory axons. Preserving mitochondrial function is anticipated to prevent CIPN. We have now discovered that the G-protein-coupled receptor, A3 adenosine receptor subtype (A3AR), is expressed on the mitochondrial outer membrane. Ex vivo application of a highly selective A3AR agonist, MRS5980, to saphenous nerve microfilaments harvested from male oxaliplatin-treated rats reversed the loss in ATP production underscoring mitoprotective effects resulting from A3AR activation on mitochondria. Moreover, in vivo administration of A3AR agonists to rats during oxaliplatin treatment was associated with reduced IENF loss and a lower incidence of spontaneous discharge in peripheral afferent axons. These effects are accompanied by improved mitochondrial ATP production in primary afferent sensory axons and overall inhibition of the development of neuropathic pain. These data identify for the first time mitochondrial A3AR and indicate that activation of A3AR protects mitochondrial function in primary afferent sensory axons against chemotherapy-induced neurotoxicity. Repurposing A3AR agonists that are already in clinical trials as anticancer agents as adjunct to chemotherapeutics will address a major unmet medical need for which there are no FDA-approved drugs.
Collapse
Affiliation(s)
- Timothy M Doyle
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, Saint Louis, Missouri 63104
- Institute for Translational Neuroscience, Saint Louis University, Saint Louis, Missouri 63104
| | - Kali Janes
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, Saint Louis, Missouri 63104
| | - Wen Hua Xiao
- Department of Anesthesiology, University of California San Diego, San Diego, California 92103
| | - Grant R Kolar
- Institute for Translational Neuroscience, Saint Louis University, Saint Louis, Missouri 63104
- Department of Pathology, Saint Louis University School of Medicine, Saint Louis, Missouri 63104
| | - Hans F Luecke
- Chemical Proteomics, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland 20892
| | - Michael Anne Gratton
- Center for Sensory Neuroscience, Boys Town National Research Hospital, Boys Town, Nebraska 68010
| | - Dilip K Tosh
- Molecular Recognition Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland 20892
| | - Kenneth A Jacobson
- Molecular Recognition Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland 20892
| | - Gary J Bennett
- Department of Anesthesiology, University of California San Diego, San Diego, California 92103
| | - Daniela Salvemini
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, Saint Louis, Missouri 63104
- Institute for Translational Neuroscience, Saint Louis University, Saint Louis, Missouri 63104
| |
Collapse
|
25
|
De los Santos-Jiménez J, Campos-Sandoval JA, Rosales T, Ko B, Alonso FJ, Márquez J, DeBerardinis RJ, Matés JM. Glutaminase-2 Expression Induces Metabolic Changes and Regulates Pyruvate Dehydrogenase Activity in Glioblastoma Cells. Int J Mol Sci 2025; 26:427. [PMID: 39796278 PMCID: PMC11721818 DOI: 10.3390/ijms26010427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 12/29/2024] [Accepted: 01/03/2025] [Indexed: 01/13/2025] Open
Abstract
Glutaminase controls the first step in glutaminolysis, impacting bioenergetics, biosynthesis and oxidative stress. Two isoenzymes exist in humans, GLS and GLS2. GLS is considered prooncogenic and overexpressed in many tumours, while GLS2 may act as prooncogenic or as a tumour suppressor. Glioblastoma cells usually lack GLS2 while they express high GLS. We investigated how GLS2 expression modifies the metabolism of glioblastoma cells, looking for changes that may explain GLS2's potential tumour suppressive role. We developed LN-229 glioblastoma cells stably expressing GLS2 and performed isotope tracing using U-13C-glutamine and metabolomic quantification to analyze metabolic changes. Treatment with GLS inhibitor CB-839 was also included to concomitantly inhibit endogenous GLS. GLS2 overexpression resulted in extensive metabolic changes, altering the TCA cycle by upregulating part of the cycle but blocking the synthesis of the 6-carbon intermediates from acetyl-CoA. Expression of GLS2 caused downregulation of PDH activity through phosphorylation of S293 of PDHA1. GLS2 also altered nucleotide levels and induced the accumulation of methylated metabolites and S-adenosyl methionine. These changes suggest that GLS2 may be a key regulator linking glutamine and glucose metabolism, also impacting nucleotides and epigenetics. Future research should ascertain the mechanisms involved and the generalizability of these findings in cancer or physiological conditions.
Collapse
Affiliation(s)
- Juan De los Santos-Jiménez
- Departamento de Biología Molecular y Bioquímica, Universidad de Málaga, 29071 Málaga, Spain; (J.A.C.-S.); (F.J.A.); (J.M.)
- Instituto de Investigación Biomédica de Málaga (IBIMA-Plataforma BIONAND), Universidad de Málaga, 29071 Málaga, Spain
| | - José A. Campos-Sandoval
- Departamento de Biología Molecular y Bioquímica, Universidad de Málaga, 29071 Málaga, Spain; (J.A.C.-S.); (F.J.A.); (J.M.)
- Instituto de Investigación Biomédica de Málaga (IBIMA-Plataforma BIONAND), Universidad de Málaga, 29071 Málaga, Spain
| | - Tracy Rosales
- Children’s Medical Center Research Institute, University of Texas Southwestern Medical Center (UTSMC), Dallas, TX 75390, USA; (T.R.); (B.K.); (R.J.D.)
| | - Bookyung Ko
- Children’s Medical Center Research Institute, University of Texas Southwestern Medical Center (UTSMC), Dallas, TX 75390, USA; (T.R.); (B.K.); (R.J.D.)
| | - Francisco J. Alonso
- Departamento de Biología Molecular y Bioquímica, Universidad de Málaga, 29071 Málaga, Spain; (J.A.C.-S.); (F.J.A.); (J.M.)
- Instituto de Investigación Biomédica de Málaga (IBIMA-Plataforma BIONAND), Universidad de Málaga, 29071 Málaga, Spain
| | - Javier Márquez
- Departamento de Biología Molecular y Bioquímica, Universidad de Málaga, 29071 Málaga, Spain; (J.A.C.-S.); (F.J.A.); (J.M.)
- Instituto de Investigación Biomédica de Málaga (IBIMA-Plataforma BIONAND), Universidad de Málaga, 29071 Málaga, Spain
| | - Ralph J. DeBerardinis
- Children’s Medical Center Research Institute, University of Texas Southwestern Medical Center (UTSMC), Dallas, TX 75390, USA; (T.R.); (B.K.); (R.J.D.)
- McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center (UTSMC), Dallas, TX 75390, USA
- Department of Pediatrics, University of Texas Southwestern Medical Center (UTSMC), Dallas, TX 75390, USA
| | - José M. Matés
- Departamento de Biología Molecular y Bioquímica, Universidad de Málaga, 29071 Málaga, Spain; (J.A.C.-S.); (F.J.A.); (J.M.)
- Instituto de Investigación Biomédica de Málaga (IBIMA-Plataforma BIONAND), Universidad de Málaga, 29071 Málaga, Spain
| |
Collapse
|
26
|
Harris TJ, Trader DJ. Exploration of degrons and their ability to mediate targeted protein degradation. RSC Med Chem 2025:d4md00787e. [PMID: 39867589 PMCID: PMC11758578 DOI: 10.1039/d4md00787e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 12/12/2024] [Indexed: 01/28/2025] Open
Abstract
Degrons are short amino acid sequences that can facilitate the degradation of protein substrates. They can be classified as either ubiquitin-dependent or -independent based on their interactions with the ubiquitin proteasome system (UPS). These amino acid sequences are often found in exposed regions of proteins serving as either a tethering point for an interaction with an E3 ligase or initiating signaling for the direct degradation of the protein. Recent advancements in the protein degradation field have shown the therapeutic potential of both classes of degrons through leveraging their degradative effects to engage specific protein targets. This review explores what targeted protein degradation applications degrons can be used in and how they have inspired new degrader technology to target a wide variety of protein substrates.
Collapse
Affiliation(s)
- Timothy J Harris
- Department of Pharmaceutical Sciences, University of California Irvine California 92617 USA
| | - Darci J Trader
- Department of Pharmaceutical Sciences, University of California Irvine California 92617 USA
- Department of Chemistry, University of California Irvine California 92617 USA
| |
Collapse
|
27
|
Herrnreiter CJ, Murray MG, Luck M, Ganesa C, Kuprys PV, Li X, Choudhry MA. Bacterial dysbiosis and decrease in SCFA correlate with intestinal inflammation following alcohol intoxication and burn injury. EGASTROENTEROLOGY 2025; 3:e100145. [PMID: 40098807 PMCID: PMC11911813 DOI: 10.1136/egastro-2024-100145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Accepted: 02/21/2025] [Indexed: 03/19/2025]
Abstract
Background Patients intoxicated at the time of burn experience increased rates of sepsis and death compared with that observed in similarly sized burns alone. We sought to characterise changes in the intestinal microbiome and short-chain fatty acids (SCFAs) following alcohol intoxication and burn injury and to determine whether these changes are associated with intestinal inflammation. Methods 10-12-week-old C57BL/6 male and female mice were subjected to ethanol intoxication and a 12.5% total body surface area scald burn injury. The following day, mice were euthanised and faecal contents from the caecum and small intestine (SI) were harvested for 16S sequencing for microbial analysis and caecum contents underwent high-performance liquid chromatography mass spectroscopy to assess SCFAs. Results The intestinal microbiome of ethanol burn (EB) mice exhibited decreased alpha diversity and distinct beta diversity compared with sham vehicle (SV). EB faeces were marked by increased Proteobacteria and many pathobionts. EB caecum faeces exhibited a significant decrease in butyrate and a downward trend in acetate and total SCFAs. SCFA changes correlated with microbial changes particularly in the SI. Treatment of murine duodenal cell clone-K (MODE-K) cells with faecal slurries led to upregulation of interleukin-6 (IL-6) from EB faeces compared with SV faeces which correlated with levels of Enterobacteriaceae. However, supplementation of butyrate reduced faecal slurry-induced MODE-K cells IL-6 release. Conclusion Together, these findings suggest that alcohol and burn injury induce bacterial dysbiosis and a decrease in SCFAs, which together can promote intestinal inflammation and barrier disruption, predisposing to postinjury pathology.
Collapse
Affiliation(s)
- Caroline J Herrnreiter
- Burn and Shock Trauma Research Institute, Loyola University Chicago Health Sciences Division, Maywood, Illinois, USA
- Alcohol Research Program, Loyola University Chicago Health Sciences Division, Maywood, Illinois, USA
- Biochemistry and Molecular Biology Program, Loyola University Chicago Graduate School, Maywood, Illinois, USA
- Department of Surgery, Loyola University Chicago Health Sciences Division, Maywood, Illinois, USA
| | - Mary Grace Murray
- Burn and Shock Trauma Research Institute, Loyola University Chicago Health Sciences Division, Maywood, Illinois, USA
- Alcohol Research Program, Loyola University Chicago Health Sciences Division, Maywood, Illinois, USA
- Biochemistry and Molecular Biology Program, Loyola University Chicago Graduate School, Maywood, Illinois, USA
- Department of Surgery, Loyola University Chicago Health Sciences Division, Maywood, Illinois, USA
| | - Marisa Luck
- Burn and Shock Trauma Research Institute, Loyola University Chicago Health Sciences Division, Maywood, Illinois, USA
- Alcohol Research Program, Loyola University Chicago Health Sciences Division, Maywood, Illinois, USA
- Department of Surgery, Loyola University Chicago Health Sciences Division, Maywood, Illinois, USA
- Department of Microbiology and Immunology, Loyola University Chicago Health Sciences Division, Maywood, Illinois, USA
| | - Chirag Ganesa
- Burn and Shock Trauma Research Institute, Loyola University Chicago Health Sciences Division, Maywood, Illinois, USA
| | - Paulius V Kuprys
- Burn and Shock Trauma Research Institute, Loyola University Chicago Health Sciences Division, Maywood, Illinois, USA
- Alcohol Research Program, Loyola University Chicago Health Sciences Division, Maywood, Illinois, USA
- Department of Surgery, Loyola University Chicago Health Sciences Division, Maywood, Illinois, USA
- Department of Microbiology and Immunology, Loyola University Chicago Health Sciences Division, Maywood, Illinois, USA
| | - Xiaoling Li
- Burn and Shock Trauma Research Institute, Loyola University Chicago Health Sciences Division, Maywood, Illinois, USA
- Alcohol Research Program, Loyola University Chicago Health Sciences Division, Maywood, Illinois, USA
- Department of Surgery, Loyola University Chicago Health Sciences Division, Maywood, Illinois, USA
| | - Mashkoor A Choudhry
- Burn and Shock Trauma Research Institute, Loyola University Chicago Health Sciences Division, Maywood, Illinois, USA
- Alcohol Research Program, Loyola University Chicago Health Sciences Division, Maywood, Illinois, USA
- Department of Surgery, Loyola University Chicago Health Sciences Division, Maywood, Illinois, USA
- Department of Microbiology and Immunology, Loyola University Chicago Health Sciences Division, Maywood, Illinois, USA
| |
Collapse
|
28
|
Gessner A, König J, Wenisch P, Heinrich MR, Stopfer P, Fromm MF, Müller F. New Biomarkers for Renal Transporter-Mediated Drug-Drug Interactions: Metabolomic Effects of Cimetidine, Probenecid, Verapamil, and Rifampin in Humans. Clin Pharmacol Ther 2025; 117:130-142. [PMID: 39148267 PMCID: PMC11652812 DOI: 10.1002/cpt.3414] [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: 05/14/2024] [Accepted: 07/19/2024] [Indexed: 08/17/2024]
Abstract
The inhibition of renal transport proteins organic cation transporter 2 (OCT2), multidrug and toxin extrusion proteins (MATE1, MATE2-K), and organic anion transporters (OAT1, OAT3) causes clinically relevant drug-drug interactions (DDI). Endogenous biomarkers could be used to improve risk prediction of such renal DDIs. While a number of biomarkers for renal DDIs have been described so far, multiple criteria for valid biomarkers have frequently not been investigated, for example, specificity, metabolism, or food effects. Therefore, there is a need for novel biomarkers of renal DDIs. Here, we investigated the global metabolomic effects following the administration of two classical inhibitors of renal transport proteins [cimetidine (OCT2/MATEs), probenecid (OATs)] in human plasma and urine of healthy volunteers. Additionally, we investigated metabolomic effects of two inhibitors of other transporters [verapamil (P-glycoprotein), rifampin (organic anion transporting polypeptides)] as controls. This analysis shows that both cimetidine and probenecid affect compounds involved in caffeine metabolism, carnitines, and sulfates. Hierarchical cluster analysis of the effects of all four inhibitors on endogenous compounds identified multiple promising new sensitive and specific biomarker candidates for OCT2/MATE- or OAT-mediated DDIs. For OCT2/MATEs, 5-amino valeric acid betaine (median log2-fold change of estimated renal elimination: -3.62) presented itself as a promising candidate. For OATs, estimated renal elimination of 7-methyluric acid and cinnamoylglycine (median log2-fold changes -3.10 and -1.92, respectively) was both sensitive and specific. This study provides comprehensive information on metabolomic effects of transport protein inhibition in humans and identifies putative new sensitive and specific biomarkers for renal transporter-mediated DDIs.
Collapse
Affiliation(s)
- Arne Gessner
- Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich‐Alexander‐Universität Erlangen‐NürnbergErlangenGermany
- FAU NeW – Research Center New Bioactive CompoundsFriedrich‐Alexander‐Universität Erlangen‐NürnbergErlangenGermany
| | - Jörg König
- Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich‐Alexander‐Universität Erlangen‐NürnbergErlangenGermany
- FAU NeW – Research Center New Bioactive CompoundsFriedrich‐Alexander‐Universität Erlangen‐NürnbergErlangenGermany
| | - Pia Wenisch
- Department of Chemistry and Pharmacy, Pharmaceutical ChemistryFriedrich‐Alexander‐Universität Erlangen‐NürnbergErlangenGermany
| | - Markus R. Heinrich
- FAU NeW – Research Center New Bioactive CompoundsFriedrich‐Alexander‐Universität Erlangen‐NürnbergErlangenGermany
- Department of Chemistry and Pharmacy, Pharmaceutical ChemistryFriedrich‐Alexander‐Universität Erlangen‐NürnbergErlangenGermany
| | - Peter Stopfer
- Boehringer Ingelheim Pharma GmbH & Co. KGBiberach an der RissGermany
| | - Martin F. Fromm
- Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich‐Alexander‐Universität Erlangen‐NürnbergErlangenGermany
- FAU NeW – Research Center New Bioactive CompoundsFriedrich‐Alexander‐Universität Erlangen‐NürnbergErlangenGermany
| | - Fabian Müller
- Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich‐Alexander‐Universität Erlangen‐NürnbergErlangenGermany
- Boehringer Ingelheim Pharma GmbH & Co. KGBiberach an der RissGermany
| |
Collapse
|
29
|
Lihon MV, Tuchscherer NA, Tao WA. Isolation and Identification of Brain Tissue Extracellular Vesicles for Translational Proteomics. Methods Mol Biol 2025; 2884:225-239. [PMID: 39716007 DOI: 10.1007/978-1-0716-4298-6_15] [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: 12/25/2024]
Abstract
Extracellular vesicles (EVs) are small membrane-bound structures that play important roles in intercellular communication and the transfer of biomolecules between cells. EVs have become a topic of interest for research in translational proteomics for disease biomarker discovery due to their ability to reflect changes in the cellular proteome, including diseases affecting the brain. Utilizing the proteome analysis of EVs to its fullest potential requires proper isolation and purity. In this chapter, we describe a detailed method for the isolation and identification of brain tissue EVs for translational proteomics using our in-house chemical affinity magnetic bead-based (non-antibody) method, the EVtrap. We also discuss various methods for quantification, characterization, and functional analysis of isolated brain tissue EVs, including western blotting, and proteomic profiling of post-translational modifications (PTMs) involved in neurodegenerative diseases, such as protein N-terminal acetylation. This protocol provides a valuable resource for studies conducted on brain tissue EVs and their potential as biomarkers and therapeutic targets for neurological diseases.
Collapse
Affiliation(s)
- Michelle V Lihon
- Department of Biochemistry, Purdue University, West Lafayette, IN, USA
| | | | - W Andy Tao
- Department of Biochemistry, Purdue University, West Lafayette, IN, USA.
- Department of Chemistry, Purdue University, West Lafayette, IN, USA.
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN, USA.
- Purdue Institute for Cancer Research, Purdue University, West Lafayette, IN, USA.
- Tymora Analytical Operations, West Lafayette, IN, USA.
| |
Collapse
|
30
|
Good KV, Kalani L, Vincent JB, Ausió J. Multifaceted roles of MeCP2 in cellular regulation and phase separation: implications for neurodevelopmental disorders, depression, and oxidative stress. Biochem Cell Biol 2025; 103:1-12. [PMID: 39761540 DOI: 10.1139/bcb-2024-0237] [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: 01/28/2025] Open
Abstract
Methyl CpG binding protein 2 (MeCP2) is a chromatin-associated protein that remains enigmatic despite more than 30 years of research, primarily due to the ever-growing list of its molecular functions, and, consequently, its related pathologies. Loss of function MECP2 mutations cause the neurodevelopmental disorder Rett syndrome (RTT); in addition, dysregulation of MeCP2 expression and/ or function are involved in numerous other pathologies, but the mechanisms of MeCP2 regulation are unclear. Advancing technologies and burgeoning mechanistic theories assist our understanding of the complexity of MeCP2 but may inadvertently cloud it if not rigorously tested. Here, rather than focus on RTT, we examine relatively underexplored aspects of MeCP2, such as its dosage homeostasis at the gene and protein levels, its controversial participation in phase separation, and its overlooked role in depression and oxidative stress. All these factors may be essential to understanding the full scope of MeCP2 function in healthy and diseased states, but are relatively infrequently studied and require further criticism. The aim of this review is to discuss the esoteric facets of MeCP2 at the molecular and pathological levels and to consider to what extent they may be necessary for general MeCP2 function.
Collapse
Affiliation(s)
- Katrina V Good
- Department of Biochemistry and Microbiology, University of Victoria, Victoria, BC V8W 2Y2, Canada
- Molecular Neuropsychiatry & Development (MiND) Lab, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON M5T 1R8, Canada
| | - Ladan Kalani
- Department of Biochemistry and Microbiology, University of Victoria, Victoria, BC V8W 2Y2, Canada
| | - John B Vincent
- Molecular Neuropsychiatry & Development (MiND) Lab, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON M5T 1R8, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Juan Ausió
- Department of Biochemistry and Microbiology, University of Victoria, Victoria, BC V8W 2Y2, Canada
| |
Collapse
|
31
|
Lam T, Su A, Gomez Martinez AE, Fomitcheva-Khartchenko A, Herr AE. Single-cell Organelle Extraction with Cellular Indexing. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.23.630180. [PMID: 39763945 PMCID: PMC11703196 DOI: 10.1101/2024.12.23.630180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/12/2025]
Abstract
Bulk methods to fractionate organelles lack the resolution to capture single-cell heterogeneity. While microfluidic approaches attempt to fractionate organelles at the cellular level, they fail to map each organelle back to its cell of origin-crucial for multiomics applications. To address this, we developed VacTrap, a high-throughput microfluidic device for isolating and spatially indexing single nuclei from mammalian cells. VacTrap consists of three aligned layers: (1) a Bis-gel microwells layer with a 'trapdoors' (BAC-gel) base, fabricated atop a through-hole glass slide; (2) a PDMS microwell layer to receive transferred nuclei; and (3) a vacuum manifold. VacTrap operation begins with cell lysis using DDF to release intact nuclei into the Bis-gel microwells, while cytoplasmic proteins are electrophoresed into the Bis-gel. Upon exposure to DTT and vacuum force, the trapdoors open, allowing nuclei to transfer to the PDMS microwells. VacTrap dissolves the trapdoors within 3-5 minutes and achieve synchronized nuclei transfer with 98% efficiency across 80% of trapdoors in a 256-microwell array, surpassing the <1% efficiency of passive transfer without vacuum. Morphology analysis confirmed preservation of organelle integrity throughout VacTrap operation. By enabling spatial indexing of nuclei back to their original cell, VacTrap provides a robust, high-throughput tool for single-cell multiomics applications.
Collapse
Affiliation(s)
- Trinh Lam
- Department of Bioengineering, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Alison Su
- Department of Bioengineering, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Ana E Gomez Martinez
- Department of Bioengineering, University of California, Berkeley, Berkeley, CA 94720, USA
| | | | - Amy E Herr
- Department of Bioengineering, University of California, Berkeley, Berkeley, CA 94720, USA
- Chan Zuckerberg Biohub San Francisco, San Francisco, CA 94158, USA
| |
Collapse
|
32
|
Ha S, Wong VWS, Zhang X, Yu J. Interplay between gut microbiome, host genetic and epigenetic modifications in MASLD and MASLD-related hepatocellular carcinoma. Gut 2024; 74:141-152. [PMID: 38950910 PMCID: PMC11671994 DOI: 10.1136/gutjnl-2024-332398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 06/08/2024] [Indexed: 07/03/2024]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) encompasses a wide spectrum of liver injuries, ranging from hepatic steatosis, metabolic dysfunction-associated steatohepatitis (MASH), fibrosis, cirrhosis to MASLD-associated hepatocellular carcinoma (MASLD-HCC). Recent studies have highlighted the bidirectional impacts between host genetics/epigenetics and the gut microbial community. Host genetics influence the composition of gut microbiome, while the gut microbiota and their derived metabolites can induce host epigenetic modifications to affect the development of MASLD. The exploration of the intricate relationship between the gut microbiome and the genetic/epigenetic makeup of the host is anticipated to yield promising avenues for therapeutic interventions targeting MASLD and its associated conditions. In this review, we summarise the effects of gut microbiome, host genetics and epigenetic alterations in MASLD and MASLD-HCC. We further discuss research findings demonstrating the bidirectional impacts between gut microbiome and host genetics/epigenetics, emphasising the significance of this interconnection in MASLD prevention and treatment.
Collapse
Affiliation(s)
- Suki Ha
- 1Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Vincent Wai-Sun Wong
- 1Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Xiang Zhang
- 1Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Jun Yu
- 1Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|
33
|
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
|
34
|
Ree R, Lin SJ, Sti Dahl LO, Huang K, Petree C, Varshney GK, Arnesen T. Naa80 is required for actin N-terminal acetylation and normal hearing in zebrafish. Life Sci Alliance 2024; 7:e202402795. [PMID: 39384430 PMCID: PMC11465159 DOI: 10.26508/lsa.202402795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 09/27/2024] [Accepted: 09/27/2024] [Indexed: 10/11/2024] Open
Abstract
Actin is a critical component of the eukaryotic cytoskeleton. In animals, actins undergo unique N-terminal processing by dedicated enzymes resulting in mature acidic and acetylated forms. The final step, N-terminal acetylation, is catalyzed by NAA80 in humans. N-terminal acetylation of actin is crucial for maintaining normal cytoskeletal dynamics and cell motility in human cell lines. However, the physiological impact of actin N-terminal acetylation remains to be fully understood. We developed a zebrafish naa80 knockout model and demonstrated that Naa80 acetylates both muscle and non-muscle actins in vivo. Assays with purified Naa80 revealed a preference for acetylating actin N-termini. Zebrafish lacking actin N-terminal acetylation exhibited normal development, morphology, and behavior. In contrast, humans with pathogenic actin variants can present with hypotonia and hearing impairment. Whereas zebrafish lacking naa80 showed no obvious muscle defects or abnormalities, we observed abnormal inner ear development, small otoliths, and impaired response to sound. In conclusion, we have established that zebrafish Naa80 N-terminally acetylates actins in vitro and in vivo, and that actin N-terminal acetylation is essential for normal hearing.
Collapse
Affiliation(s)
- Rasmus Ree
- Department of Biomedicine, University of Bergen, Bergen, Norway
| | - Sheng-Jia Lin
- Genes and Human Disease Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | | | - Kevin Huang
- Genes and Human Disease Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Cassidy Petree
- Genes and Human Disease Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Gaurav K Varshney
- Genes and Human Disease Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Thomas Arnesen
- Department of Biomedicine, University of Bergen, Bergen, Norway
- Department of Surgery, Haukeland University Hospital, Bergen, Norway
| |
Collapse
|
35
|
Morales PN, Coons AN, Koopman AJ, Patel S, Chase PB, Parvatiyar MS, Pinto JR. Post-translational modifications of vertebrate striated muscle myosin heavy chains. Cytoskeleton (Hoboken) 2024; 81:832-842. [PMID: 38587113 PMCID: PMC11458826 DOI: 10.1002/cm.21857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 03/06/2024] [Accepted: 03/25/2024] [Indexed: 04/09/2024]
Abstract
Post-translational modifications (PTMs) play a crucial role in regulating the function of many sarcomeric proteins, including myosin. Myosins comprise a family of motor proteins that play fundamental roles in cell motility in general and muscle contraction in particular. A myosin molecule consists of two myosin heavy chains (MyHCs) and two pairs of myosin light chains (MLCs); two MLCs are associated with the neck region of each MyHC's N-terminal head domain, while the two MyHC C-terminal tails form a coiled-coil that polymerizes with other MyHCs to form the thick filament backbone. Myosin undergoes extensive PTMs, and dysregulation of these PTMs may lead to abnormal muscle function and contribute to the development of myopathies and cardiovascular disorders. Recent studies have uncovered the significance of PTMs in regulating MyHC function and showed how these PTMs may provide additional modulation of contractile processes. Here, we discuss MyHC PTMs that have been biochemically and/or functionally studied in mammals' and rodents' striated muscle. We have identified hotspots or specific regions in three isoforms of myosin (MYH2, MYH6, and MYH7) where the prevalence of PTMs is more frequent and could potentially play a significant role in fine-tuning the activity of these proteins.
Collapse
Affiliation(s)
- Paula Nieto Morales
- Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, FL 32306 USA
| | - Arianna N. Coons
- Department of Biological Science, Florida State University, Tallahassee, FL 32306 USA
| | - Amelia J. Koopman
- Department of Biological Science, Florida State University, Tallahassee, FL 32306 USA
| | - Sonu Patel
- Department of Health, Nutrition and Food Sciences, Florida State University, Tallahassee, FL 32306 USA
| | - P. Bryant Chase
- Department of Biological Science, Florida State University, Tallahassee, FL 32306 USA
| | - Michelle S. Parvatiyar
- Department of Health, Nutrition and Food Sciences, Florida State University, Tallahassee, FL 32306 USA
| | - Jose R. Pinto
- Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, FL 32306 USA
| |
Collapse
|
36
|
Tan R, Hoare M, Bellomio P, Broas S, Camacho K, Swovick K, Welle KA, Hryhorenko JR, Ghaemmaghami S. Formylation facilitates the reduction of oxidized initiator methionines. Proc Natl Acad Sci U S A 2024; 121:e2403880121. [PMID: 39499632 PMCID: PMC11572973 DOI: 10.1073/pnas.2403880121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Accepted: 10/02/2024] [Indexed: 11/07/2024] Open
Abstract
Within a cell, protein-bound methionines can be chemically or enzymatically oxidized, and subsequently reduced by methionine sulfoxide reductases (Msrs). Methionine oxidation can result in structural damage or be the basis of functional regulation of enzymes. In addition to participating in redox reactions, methionines play an important role as the initiator residue of translated proteins where they are commonly modified at their α-amine group by formylation or acetylation. Here, we investigated how formylation and acetylation of initiator methionines impact their propensity for oxidation and reduction. We show that in vitro, N-terminal methionine residues are particularly prone to chemical oxidation and that their modification by formylation or acetylation greatly enhances their subsequent enzymatic reduction by MsrA and MsrB. Concordantly, in vivo ablation of methionyl-tRNA formyltransferase (MTF) in Escherichia coli increases the prevalence of oxidized methionines within synthesized proteins. We show that oxidation of formylated initiator methionines is detrimental in part because it obstructs their ensuing deformylation by peptide deformylase (PDF) and hydrolysis by methionyl aminopeptidase (MAP). Thus, by facilitating their reduction, formylation mitigates the misprocessing of oxidized initiator methionines.
Collapse
Affiliation(s)
- Ruiyue Tan
- Department of Biology, University of Rochester, Rochester, NY14627
| | - Margaret Hoare
- Department of Biology, University of Rochester, Rochester, NY14627
| | - Philip Bellomio
- Department of Biology, University of Rochester, Rochester, NY14627
| | - Sarah Broas
- Department of Biology, University of Rochester, Rochester, NY14627
| | | | - Kyle Swovick
- Mass Spectrometry Resource Laboratory, University of Rochester Medical Center, Rochester, NY14627
| | - Kevin A. Welle
- Mass Spectrometry Resource Laboratory, University of Rochester Medical Center, Rochester, NY14627
| | - Jennifer R. Hryhorenko
- Mass Spectrometry Resource Laboratory, University of Rochester Medical Center, Rochester, NY14627
| | - Sina Ghaemmaghami
- Department of Biology, University of Rochester, Rochester, NY14627
- Mass Spectrometry Resource Laboratory, University of Rochester Medical Center, Rochester, NY14627
| |
Collapse
|
37
|
Wen J, Zhang X, Wong CC, Zhang Y, Pan Y, Zhou Y, Cheung AHK, Liu Y, Ji F, Kang X, Liu D, Yu J. Targeting squalene epoxidase restores anti-PD-1 efficacy in metabolic dysfunction-associated steatohepatitis-induced hepatocellular carcinoma. Gut 2024; 73:2023-2036. [PMID: 38744443 PMCID: PMC11671884 DOI: 10.1136/gutjnl-2023-331117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 04/29/2024] [Indexed: 05/16/2024]
Abstract
OBJECTIVE Squalene epoxidase (SQLE) promotes metabolic dysfunction-associated steatohepatitis-associated hepatocellular carcinoma (MASH-HCC), but its role in modulating the tumour immune microenvironment in MASH-HCC remains unclear. DESIGN We established hepatocyte-specific Sqle transgenic (tg) and knockout mice, which were subjected to a choline-deficient high-fat diet plus diethylnitrosamine to induce MASH-HCC. SQLE function was also determined in orthotopic and humanised mice. Immune landscape alterations of MASH-HCC mediated by SQLE were profiled by single-cell RNA sequencing and flow cytometry. RESULTS Hepatocyte-specific Sqle tg mice exhibited a marked increase in MASH-HCC burden compared with wild-type littermates, together with decreased tumour-infiltrating functional IFN-γ+ and Granzyme B+ CD8+ T cells while enriching Arg-1+ myeloid-derived suppressor cells (MDSCs). Conversely, hepatocyte-specific Sqle knockout suppressed tumour growth with increased cytotoxic CD8+ T cells and reduced Arg-1+ MDSCs, inferring that SQLE promotes immunosuppression in MASH-HCC. Mechanistically, SQLE-driven cholesterol accumulation in tumour microenvironment underlies its effect on CD8+ T cells and MDSCs. SQLE and its metabolite, cholesterol, impaired CD8+ T cell activity by inducing mitochondrial dysfunction. Cholesterol depletion in vitro abolished the effect of SQLE-overexpressing MASH-HCC cell supernatant on CD8+ T cell suppression and MDSC activation, whereas cholesterol supplementation had contrasting functions on CD8+ T cells and MDSCs treated with SQLE-knockout supernatant. Targeting SQLE with genetic ablation or pharmacological inhibitor, terbinafine, rescued the efficacy of anti-PD-1 treatment in MASH-HCC models. CONCLUSION SQLE induces an impaired antitumour response in MASH-HCC via attenuating CD8+ T cell function and augmenting immunosuppressive MDSCs. SQLE is a promising target in boosting anti-PD-1 immunotherapy for MASH-HCC.
Collapse
Affiliation(s)
- Jun Wen
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Xiang Zhang
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Chi Chun Wong
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Yating Zhang
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Yasi Pan
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Yunfei Zhou
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Alvin Ho-Kwan Cheung
- Department of Anatomical and Cellular Pathology, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Yali Liu
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Fenfen Ji
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Xing Kang
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Dabin Liu
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Jun Yu
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|
38
|
Graf LG, Moreno-Yruela C, Qin C, Schulze S, Palm GJ, Schmöker O, Wang N, Hocking DM, Jebeli L, Girbardt B, Berndt L, Dörre B, Weis DM, Janetzky M, Albrecht D, Zühlke D, Sievers S, Strugnell RA, Olsen CA, Hofmann K, Lammers M. Distribution and diversity of classical deacylases in bacteria. Nat Commun 2024; 15:9496. [PMID: 39489725 PMCID: PMC11532494 DOI: 10.1038/s41467-024-53903-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 10/25/2024] [Indexed: 11/05/2024] Open
Abstract
Classical Zn2+-dependent deac(et)ylases play fundamental regulatory roles in life and are well characterized in eukaryotes regarding their structures, substrates and physiological roles. In bacteria, however, classical deacylases are less well understood. We construct a Generalized Profile (GP) and identify thousands of uncharacterized classical deacylases in bacteria, which are grouped into five clusters. Systematic structural and functional characterization of representative enzymes from each cluster reveal high functional diversity, including polyamine deacylases and protein deacylases with various acyl-chain type preferences. These data are supported by multiple crystal structures of enzymes from different clusters. Through this extensive analysis, we define the structural requirements of substrate selectivity, and discovered bacterial de-D-/L-lactylases and long-chain deacylases. Importantly, bacterial deacylases are inhibited by archetypal HDAC inhibitors, as supported by co-crystal structures with the inhibitors SAHA and TSA, and setting the ground for drug repurposing strategies to fight bacterial infections. Thus, we provide a systematic structure-function analysis of classical deacylases in bacteria and reveal the basis of substrate specificity, acyl-chain preference and inhibition.
Collapse
Affiliation(s)
- Leonie G Graf
- Department Synthetic and Structural Biochemistry, Institute of Biochemistry, University of Greifswald, Greifswald, Germany
| | - Carlos Moreno-Yruela
- Center for Biopharmaceuticals & Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Institute of Chemical Sciences and Engineering (ISIC), School of Basic Sciences (SB), EPFL, Lausanne, Switzerland
| | - Chuan Qin
- Department Synthetic and Structural Biochemistry, Institute of Biochemistry, University of Greifswald, Greifswald, Germany
| | - Sabrina Schulze
- Department Synthetic and Structural Biochemistry, Institute of Biochemistry, University of Greifswald, Greifswald, Germany
| | - Gottfried J Palm
- Department Synthetic and Structural Biochemistry, Institute of Biochemistry, University of Greifswald, Greifswald, Germany
| | - Ole Schmöker
- Department Synthetic and Structural Biochemistry, Institute of Biochemistry, University of Greifswald, Greifswald, Germany
| | - Nancy Wang
- Peter Doherty Institute for Infection and Immunity, Department of Microbiology and Immunology, The University of Melbourne, Melbourne, VIC, Australia
| | - Dianna M Hocking
- Peter Doherty Institute for Infection and Immunity, Department of Microbiology and Immunology, The University of Melbourne, Melbourne, VIC, Australia
| | - Leila Jebeli
- Peter Doherty Institute for Infection and Immunity, Department of Microbiology and Immunology, The University of Melbourne, Melbourne, VIC, Australia
| | - Britta Girbardt
- Department Synthetic and Structural Biochemistry, Institute of Biochemistry, University of Greifswald, Greifswald, Germany
| | - Leona Berndt
- Department Synthetic and Structural Biochemistry, Institute of Biochemistry, University of Greifswald, Greifswald, Germany
| | - Babett Dörre
- Department Synthetic and Structural Biochemistry, Institute of Biochemistry, University of Greifswald, Greifswald, Germany
| | - Daniel M Weis
- Department Synthetic and Structural Biochemistry, Institute of Biochemistry, University of Greifswald, Greifswald, Germany
| | - Markus Janetzky
- Department Synthetic and Structural Biochemistry, Institute of Biochemistry, University of Greifswald, Greifswald, Germany
| | - Dirk Albrecht
- Department of Microbial Physiology and Molecular Biology, Institute of Microbiology, University of Greifswald, Greifswald, Germany
| | - Daniela Zühlke
- Department of Microbial Physiology and Molecular Biology, Institute of Microbiology, University of Greifswald, Greifswald, Germany
| | - Susanne Sievers
- Department of Microbial Physiology and Molecular Biology, Institute of Microbiology, University of Greifswald, Greifswald, Germany
| | - Richard A Strugnell
- Peter Doherty Institute for Infection and Immunity, Department of Microbiology and Immunology, The University of Melbourne, Melbourne, VIC, Australia
| | - Christian A Olsen
- Center for Biopharmaceuticals & Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Kay Hofmann
- Institute for Genetics, University of Cologne, Cologne, Germany
| | - Michael Lammers
- Department Synthetic and Structural Biochemistry, Institute of Biochemistry, University of Greifswald, Greifswald, Germany.
| |
Collapse
|
39
|
de Vasconcellos Racorti N, Martinelli M, Bustos SO, Salardani M, Camacho MF, Barcick U, Fonseca Lima LR, Jedlicka LDL, Ladeira de Campos CB, Valente RH, Chammas R, Zelanis A. Mannose-6-Phosphate Isomerase Functional Status Shapes a Rearrangement in the Proteome and Degradome of Mannose-Treated Melanoma Cells. J Proteome Res 2024; 23:5177-5192. [PMID: 39420811 PMCID: PMC11536439 DOI: 10.1021/acs.jproteome.4c00705] [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: 08/19/2024] [Revised: 10/01/2024] [Accepted: 10/09/2024] [Indexed: 10/19/2024]
Abstract
Metabolic reprogramming is a ubiquitous feature of transformed cells, comprising one of the hallmarks of cancer and enabling neoplastic cells to adapt to new environments. Accumulated evidence reports on the failure of some neoplastic cells to convert mannose-6-phosphate into fructose-6-phosphate, thereby impairing tumor growth in cells displaying low levels of mannose-6-phosphate isomerase (MPI). Thus, we performed functional analyses and profiled the proteome landscape and the repertoire of substrates of proteases (degradome) of melanoma cell lines with distinct mutational backgrounds submitted to treatment with mannose. Our results suggest a significant rearrangement in the proteome and degradome of melanoma cell lines upon mannose treatment including the activation of catabolic pathways (such as protein turnover) and differences in protein N-terminal acetylation. Even though MPI protein abundance and gene expression status are not prognostic markers, perturbation in the network caused by an exogenous monosaccharide source (i.e., mannose) significantly affected the downstream interconnected biological circuitry. Therefore, as reported in this study, the proteomic/degradomic mapping of mannose downstream effects due to the metabolic rewiring caused by the functional status of the MPI enzyme could lead to the identification of specific molecular players from affected signaling circuits in melanoma.
Collapse
Affiliation(s)
| | - Matheus Martinelli
- Functional
Proteomics Laboratory, Federal University
of São Paulo − UNIFESP, São José dos Campos, São Paulo 12231-280, Brazil
| | - Silvina Odete Bustos
- Grupo
de Oncologia Experimental, Instituto do
Câncer do Estado de São Paulo − ICESP, São Paulo, São
Paulo 01246-000, Brazil
| | - Murilo Salardani
- Functional
Proteomics Laboratory, Federal University
of São Paulo − UNIFESP, São José dos Campos, São Paulo 12231-280, Brazil
| | - Maurício Frota Camacho
- Functional
Proteomics Laboratory, Federal University
of São Paulo − UNIFESP, São José dos Campos, São Paulo 12231-280, Brazil
| | - Uilla Barcick
- Functional
Proteomics Laboratory, Federal University
of São Paulo − UNIFESP, São José dos Campos, São Paulo 12231-280, Brazil
| | - Luis Roberto Fonseca Lima
- Functional
Proteomics Laboratory, Federal University
of São Paulo − UNIFESP, São José dos Campos, São Paulo 12231-280, Brazil
| | - Letícia Dias Lima Jedlicka
- Functional
Proteomics Laboratory, Federal University
of São Paulo − UNIFESP, São José dos Campos, São Paulo 12231-280, Brazil
- Instituto
de Estudos em Saúde e Biológicas, Universidade Federal do Sul e Sudeste do Pará- Unifesspa, Marabá, Pará 68507-590, Brazil
| | - Claudia Barbosa Ladeira de Campos
- Laboratory
of Biochemistry and Molecular and Cellular Biology of Fungi, Federal University of São Paulo − UNIFESP, São José dos Campos, São Paulo 12231-280, Brazil
| | - Richard Hemmi Valente
- Laboratory
of Toxinology, Center for Research, Innovation, and Surveillance in
COVID-19 and Health Emergencies, FIOCRUZ, Rio de Janeiro 21040-900, Brazil
| | - Roger Chammas
- Grupo
de Oncologia Experimental, Instituto do
Câncer do Estado de São Paulo − ICESP, São Paulo, São
Paulo 01246-000, Brazil
- Faculdade
de Medicina da Universidade
de São Paulo, São
Paulo 01246-903, Brazil
| | - André Zelanis
- Functional
Proteomics Laboratory, Federal University
of São Paulo − UNIFESP, São José dos Campos, São Paulo 12231-280, Brazil
| |
Collapse
|
40
|
Kolář MH, McGrath H, Nepomuceno FC, Černeková M. Three Stages of Nascent Protein Translocation Through the Ribosome Exit Tunnel. WILEY INTERDISCIPLINARY REVIEWS. RNA 2024; 15:e1873. [PMID: 39496527 DOI: 10.1002/wrna.1873] [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: 04/14/2024] [Revised: 08/31/2024] [Accepted: 09/16/2024] [Indexed: 11/06/2024]
Abstract
All proteins in living organisms are produced in ribosomes that facilitate the translation of genetic information into a sequence of amino acid residues. During translation, the ribosome undergoes initiation, elongation, termination, and recycling. In fact, peptide bonds are formed only during the elongation phase, which comprises periodic association of transfer RNAs and multiple auxiliary proteins with the ribosome and the addition of an amino acid to the nascent polypeptide one at a time. The protein spends a considerable amount of time attached to the ribosome. Here, we conceptually divide this portion of the protein lifetime into three stages. We define each stage on the basis of the position of the N-terminus of the nascent polypeptide within the ribosome exit tunnel and the context of the catalytic center. We argue that nascent polypeptides experience a variety of forces that determine how they translocate through the tunnel and interact with the tunnel walls. We review current knowledge about nascent polypeptide translocation and identify several white spots in our understanding of the birth of proteins.
Collapse
Affiliation(s)
- Michal H Kolář
- Department of Physical Chemistry, University of Chemistry and Technology, Prague, Czech Republic
| | - Hugo McGrath
- Department of Physical Chemistry, University of Chemistry and Technology, Prague, Czech Republic
| | - Felipe C Nepomuceno
- Department of Physical Chemistry, University of Chemistry and Technology, Prague, Czech Republic
| | - Michaela Černeková
- Department of Physical Chemistry, University of Chemistry and Technology, Prague, Czech Republic
| |
Collapse
|
41
|
Larsen SK, Bekkelund ÅK, Glomnes N, Arnesen T, Aksnes H. Assessing N-terminal acetylation status of cellular proteins via an antibody specific for acetylated methionine. Biochimie 2024; 226:113-120. [PMID: 39038730 DOI: 10.1016/j.biochi.2024.07.007] [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: 01/11/2024] [Revised: 07/01/2024] [Accepted: 07/16/2024] [Indexed: 07/24/2024]
Abstract
N-terminal acetylation is being recognized as a factor affecting protein lifetime and proteostasis. It is a modification where an acetyl group is added to the N-terminus of proteins, and this occurs in 80 % of the human proteome. N-terminal acetylation is catalyzed by enzymes called N-terminal acetyltransferases (NATs). The various NATs acetylate different N-terminal amino acids, and methionine is a known target for some of the NATs. Currently, the acetylation status of most proteins can only be assessed with a limited number of methods, including mass spectrometry, which although powerful and robust, remains laborious and can only survey a fraction of the proteome. We here present testing of an antibody that was developed to specifically recognize Nt-acetylated methionine-starting proteins. We have used dot blots with synthetic acetylated and non-acetylated peptides in addition to protein analysis of lysates from NAT knockout cell lines to assess the specificity and application of this anti-Nt-acetylated methionine antibody (anti-NtAc-Met). Our results demonstrate that this antibody is indeed NtAc-specific and further show that it has selectivity for some subtypes of methionine-starting N-termini, specifically potential substrates of the NatC, NatE and NatF enzymes. We propose that this antibody may be a powerful tool to identify NAT substrates or to analyse changes in N-terminal acetylation for specific cellular proteins of interest.
Collapse
Affiliation(s)
| | - Åse K Bekkelund
- Department of Biomedicine, University of Bergen, Bergen, Norway
| | - Nina Glomnes
- Department of Biomedicine, University of Bergen, Bergen, Norway; Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Thomas Arnesen
- Department of Biomedicine, University of Bergen, Bergen, Norway; Department of Surgery, Haukeland University Hospital, Bergen, Norway
| | - Henriette Aksnes
- Department of Biomedicine, University of Bergen, Bergen, Norway.
| |
Collapse
|
42
|
Borghys H, Schwab A, Keppler B. Middle-aged dogs with low and high Aβ CSF concentrations show differences in energy and stress related metabolic profiles in CSF. Heliyon 2024; 10:e39104. [PMID: 39498015 PMCID: PMC11532822 DOI: 10.1016/j.heliyon.2024.e39104] [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: 01/19/2024] [Revised: 10/01/2024] [Accepted: 10/07/2024] [Indexed: 11/07/2024] Open
Abstract
Background Amyloid beta (Aβ) accumulation in the brain is one of the earliest findings in Alzheimer's disease (AD). The dog is a natural animal model for amyloid processing and early brain amyloid pathology. The goal of this study is to examine which differences in metabolomic profiles in cerebrospinal fluid (CSF) could be detected in dogs with a difference in CSF Aβ concentrations before amyloid accumulation occurs. Method Metabolic profiling was performed on CSF from 4 to 8 year old dogs with different CSF Aβ concentrations. Results Metabolomic profiling of CSF showed differences in brain energy metabolism. More specifically, increases in N-acetylation of amino acids and amino sugars, creatine and pentose metabolism, and a decrease in tricarboxylic acid (TCA) cycle were seen in dogs with a high CSF Aβ concentration. In addition, signs of elevated oxidative stress, higher methionine, lipid and nucleotide metabolism and increased levels of cysteine, myo-inositol and trimethylamine N-oxide were noted in these animals. Conclusions Differences in energy metabolism and stress mediated metabolic changes are seen in the brain of dogs with different CSF Aβ concentrations, before any amyloid deposition occurs. Similar metabolic changes, as in the high Aβ dogs, have been described in AD in humans and/or transgenic AD mice, some of them in very early phases. General significance The differences observed in metabolomic profiles could help in identifying potential biomarkers for an increased risk of developing amyloid pathology in the brain and open the door to the evaluation of preventive treatments for amyloid pathology in humans.
Collapse
Affiliation(s)
- Herman Borghys
- Janssen Research & Development, a division of Janssen Pharmaceutica N.V., Beerse, Belgium
| | | | | |
Collapse
|
43
|
Harrison BR, Partida-Aguilar M, Marye A, Djukovic D, Kauffman M, Dunbar MD, Mariner BL, McCoy BM, Algavi YM, Muller E, Baum S, Bamberger T, Raftery D, Creevy KE, Dog Aging Project Consortium, Avery A, Borenstein E, Snyder-Mackler N, Promislow DE. Protein catabolites as blood-based biomarkers of aging physiology: Findings from the Dog Aging Project. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.17.618956. [PMID: 39484426 PMCID: PMC11526923 DOI: 10.1101/2024.10.17.618956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Our understanding of age-related physiology and metabolism has grown through the study of systems biology, including transcriptomics, single-cell analysis, proteomics and metabolomics. Studies in lab organisms in controlled environments, while powerful and complex, fall short of capturing the breadth of genetic and environmental variation in nature. Thus, there is now a major effort in geroscience to identify aging biomarkers and to develop aging interventions that might be applied across the diversity of humans and other free-living species. To meet this challenge, the Dog Aging Project (DAP) is designed to identify cross-sectional and longitudinal patterns of aging in complex systems, and how these are shaped by the diversity of genetic and environmental variation among companion dogs. Here we surveyed the plasma metabolome from the first year of sampling of the Precision Cohort of the DAP. By incorporating extensive metadata and whole genome sequencing information, we were able to overcome the limitations inherent in breed-based estimates of genetic and physiological effects, and to probe the physiological and dietary basis of the age-related metabolome. We identified a significant effect of age on approximately 40% of measured metabolites. Among other insights, we discovered a potentially novel biomarker of age in the post-translationally modified amino acids (ptmAAs). The ptmAAs, which can only be generated by protein hydrolysis, covaried both with age and with other biomarkers of amino acid metabolism, and in a way that was robust to diet. Clinical measures of kidney function mediated about half of the higher ptmAA levels in older dogs. This work identifies ptmAAs as robust indicators of age in dogs, and points to kidney function as a physiological mediator of age-associated variation in the plasma metabolome.
Collapse
Affiliation(s)
- Benjamin R. Harrison
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
| | - Maria Partida-Aguilar
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
| | - Abbey Marye
- University of Utah, Department of Microbiology and Immunology, Salt Lake City, UT, USA
| | - Danijel Djukovic
- Center for Studies in Ecology and Demography, University of Washington, Seattle, WA, USA
| | - Mandy Kauffman
- Center for Studies in Ecology and Demography, University of Washington, Seattle, WA, USA
| | - Matthew D. Dunbar
- Center for Studies in Ecology and Demography, University of Washington, Seattle, WA, USA
| | | | - Brianah M. McCoy
- School of Life Sciences, Arizona State University, Tempe, AZ, USA
| | - Yadid M. Algavi
- Department of Clinical Microbiology and Immunology, Tel Aviv University, Tel Aviv, Israel
| | - Efrat Muller
- Blavatnik School of Computer Science, Tel Aviv University, Tel Aviv, Israel
| | - Shiri Baum
- Blavatnik School of Computer Science, Tel Aviv University, Tel Aviv, Israel
| | - Tal Bamberger
- Department of Clinical Microbiology and Immunology, Tel Aviv University, Tel Aviv, Israel
| | - Dan Raftery
- Northwest Metabolomics Research Center, Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA, USA
| | - Kate E. Creevy
- Department of Small Animal Clinical Sciences, Texas A&M University, College Station, TX, USA
| | | | - Anne Avery
- College of Veterinary Medicine and Biomedical Sciences, Colorado State University, CO, USA
| | - Elhanan Borenstein
- Department of Clinical Microbiology and Immunology, Tel Aviv University, Tel Aviv, Israel
- Blavatnik School of Computer Science, Tel Aviv University, Tel Aviv, Israel
- Faculty of Medical & Health Sciences, Tel Aviv University, Tel Aviv, Israel
| | | | - Daniel E. Promislow
- Jean Mayer USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA, USA
| |
Collapse
|
44
|
Totty MS, Juanes RC, Bach SV, Ameur LB, Valentine MR, Simons E, Romac M, Trinh H, Henderson K, Del Rosario I, Tippani M, Miller RA, Kleinman JE, Page SC, Saunders A, Hyde TM, Martinowich K, Hicks SC, Costa VD. Transcriptomic diversity of amygdalar subdivisions across humans and nonhuman primates. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.18.618721. [PMID: 39463931 PMCID: PMC11507838 DOI: 10.1101/2024.10.18.618721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
The amygdaloid complex mediates learning, memory, and emotions. Understanding the cellular and anatomical features that are specialized in the amygdala of primates versus other vertebrates requires a systematic, anatomically-resolved molecular analysis of constituent cell populations. We analyzed five nuclear subdivisions of the primate amygdala with single-nucleus RNA sequencing in macaques, baboons, and humans to examine gene expression profiles for excitatory and inhibitory neurons and confirmed our results with single-molecule FISH analysis. We identified distinct subtypes of FOXP2 + interneurons in the intercalated cell masses and protein-kinase C-δ interneurons in the central nucleus. We also establish that glutamatergic, pyramidal-like neurons are transcriptionally specialized within the basal, lateral, or accessory basal nuclei. Understanding the molecular heterogeneity of anatomically-resolved amygdalar neuron types provides a cellular framework for improving existing models of how amygdalar neural circuits contribute to cognition and mental health in humans by using nonhuman primates as a translational bridge.
Collapse
Affiliation(s)
- Michael S. Totty
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Rita Cervera Juanes
- Department of Translational Neuroscience, Center for Precision Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Svitlana V. Bach
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA
| | - Lamya Ben Ameur
- Vollum Institute, Oregon Health and Science University, Portland, OR, USA
| | - Madeline R. Valentine
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA
| | - Evan Simons
- Division of Neuroscience, Oregon National Primate Research Center, Beaverton, OR, USA
| | - McKenna Romac
- Division of Neuroscience, Oregon National Primate Research Center, Beaverton, OR, USA
- Division of Developmental and Cognitive Neuroscience, Emory National Primate Research Center, Atlanta, GA, USA
| | - Hoa Trinh
- Vollum Institute, Oregon Health and Science University, Portland, OR, USA
| | - Krystal Henderson
- Division of Neuroscience, Oregon National Primate Research Center, Beaverton, OR, USA
| | - Ishbel Del Rosario
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA
| | - Madhavi Tippani
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA
| | - Ryan A. Miller
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA
| | - Joel E. Kleinman
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Stephanie Cerceo Page
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA
| | - Arpiar Saunders
- Vollum Institute, Oregon Health and Science University, Portland, OR, USA
| | - Thomas M. Hyde
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Keri Martinowich
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD, USA
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Johns Hopkins Kavli Neuroscience Discovery Institute, Baltimore, MD, USA
| | - Stephanie C. Hicks
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
- Center for Computational Biology, Johns Hopkins University, Baltimore, MD, USA
- Malone Center for Engineering in Healthcare, Johns Hopkins University, Baltimore, MD, USA
| | - Vincent D. Costa
- Division of Neuroscience, Oregon National Primate Research Center, Beaverton, OR, USA
- Division of Developmental and Cognitive Neuroscience, Emory National Primate Research Center, Atlanta, GA, USA
- Department of Psychiatry and Behavioral Sciences, Emory University, Atlanta, GA 30329, USA
| |
Collapse
|
45
|
Derstine N, Laremore T, Amsalem E. Post-transcriptional regulation of Dufour's gland reproductive signals in bumble bees. BMC Genomics 2024; 25:976. [PMID: 39420273 PMCID: PMC11488150 DOI: 10.1186/s12864-024-10873-3] [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: 04/04/2024] [Accepted: 10/07/2024] [Indexed: 10/19/2024] Open
Abstract
Pheromone communication is a key mechanism by which the reproductive division of labor is maintained within insect communities. Understanding how pheromones evolved to regulate social behavior requires knowledge of the molecular regulation of their production. However, even in cases where pheromones were identified, our understanding of their biosynthesis and molecular regulation remains limited. Bumble bees provide a unique system to explore pheromone biosynthesis since workers produce ester sterility signals in their Dufour's gland that differ from gyne-specific esters and are not produced by queens. These esters are hypothesized to be produced in the exocrine gland where they are stored, and indeed queens, gynes and workers differ significantly in the expression of Dufour's gland genes coding to enzymes involved in the biosynthesis of esters. However, a previous transcriptome analysis revealed no gene expression differences in the Dufour's gland of workers despite differences in both ester production and ovarian activation, suggesting that ester production may be regulated lower down. Proteomics of the Dufour's gland of queens, gynes, and workers recovered over 2400 proteins and broadly matched the previous RNAseq data. However, more than 100 differentially expressed proteins were found between the worker groups, including key enzymes in fatty acid biosynthesis, indicating that the regulation of reproductive signal biosynthesis in workers is done post-transcription. Overall, our data provide evidence that pheromone biosynthesis in the Dufour's gland is caste specific, that gynes and workers are likely using different enzymes to make their respective wax esters, and that the regulation on pheromone production in queens, gynes and workers is likely done at different regulatory levels, with workers signals being subjected to regulation at the protein level.
Collapse
Affiliation(s)
- Nathan Derstine
- Department of Entomology, Center for Chemical Ecology, Center for Pollinator Research, Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, PA, 16802, USA.
| | - Tatiana Laremore
- Proteomics and Mass Spectrometry Core Facility, Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, PA, 16802, USA
| | - Etya Amsalem
- Department of Entomology, Center for Chemical Ecology, Center for Pollinator Research, Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, PA, 16802, USA
| |
Collapse
|
46
|
Hassanzadeh K, Liu J, Maddila S, Mouradian MM. Posttranslational Modifications of α-Synuclein, Their Therapeutic Potential, and Crosstalk in Health and Neurodegenerative Diseases. Pharmacol Rev 2024; 76:1254-1290. [PMID: 39164116 PMCID: PMC11549938 DOI: 10.1124/pharmrev.123.001111] [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: 10/27/2023] [Revised: 07/28/2024] [Accepted: 08/09/2024] [Indexed: 08/22/2024] Open
Abstract
α-Synuclein (α-Syn) aggregation in Lewy bodies and Lewy neurites has emerged as a key pathogenetic feature in Parkinson's disease, dementia with Lewy bodies, and multiple system atrophy. Various factors, including posttranslational modifications (PTMs), can influence the propensity of α-Syn to misfold and aggregate. PTMs are biochemical modifications of a protein that occur during or after translation and are typically mediated by enzymes. PTMs modulate several characteristics of proteins including their structure, activity, localization, and stability. α-Syn undergoes various posttranslational modifications, including phosphorylation, ubiquitination, SUMOylation, acetylation, glycation, O-GlcNAcylation, nitration, oxidation, polyamination, arginylation, and truncation. Different PTMs of a protein can physically interact with one another or work together to influence a particular physiological or pathological feature in a process known as PTMs crosstalk. The development of detection techniques for the cooccurrence of PTMs in recent years has uncovered previously unappreciated mechanisms of their crosstalk. This has led to the emergence of evidence supporting an association between α-Syn PTMs crosstalk and synucleinopathies. In this review, we provide a comprehensive evaluation of α-Syn PTMs, their impact on misfolding and pathogenicity, the pharmacological means of targeting them, and their potential as biomarkers of disease. We also highlight the importance of the crosstalk between these PTMs in α-Syn function and aggregation. Insight into these PTMS and the complexities of their crosstalk can improve our understanding of the pathogenesis of synucleinopathies and identify novel targets of therapeutic potential. SIGNIFICANCE STATEMENT: α-Synuclein is a key pathogenic protein in Parkinson's disease and other synucleinopathies, making it a leading therapeutic target for disease modification. Multiple posttranslational modifications occur at various sites in α-Synuclein and alter its biophysical and pathological properties, some interacting with one another to add to the complexity of the pathogenicity of this protein. This review details these modifications, their implications in disease, and potential therapeutic opportunities.
Collapse
Affiliation(s)
- Kambiz Hassanzadeh
- Robert Wood Johnson Medical School Institute for Neurological Therapeutics, and Department of Neurology, Rutgers Biomedical and Health Sciences, Piscataway, New Jersey
| | - Jun Liu
- Robert Wood Johnson Medical School Institute for Neurological Therapeutics, and Department of Neurology, Rutgers Biomedical and Health Sciences, Piscataway, New Jersey
| | - Santhosh Maddila
- Robert Wood Johnson Medical School Institute for Neurological Therapeutics, and Department of Neurology, Rutgers Biomedical and Health Sciences, Piscataway, New Jersey
| | - M Maral Mouradian
- Robert Wood Johnson Medical School Institute for Neurological Therapeutics, and Department of Neurology, Rutgers Biomedical and Health Sciences, Piscataway, New Jersey
| |
Collapse
|
47
|
Pérez-Cova M, Bedia C, Checa A, Meister I, Tauler R, Wheelock CE, Jaumot J. Metabolomic and sphingolipidomic profiling of human hepatoma cells exposed to widely used pharmaceuticals. J Pharm Biomed Anal 2024; 249:116378. [PMID: 39074424 DOI: 10.1016/j.jpba.2024.116378] [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: 04/11/2024] [Revised: 06/08/2024] [Accepted: 07/21/2024] [Indexed: 07/31/2024]
Abstract
Pharmaceutical compounds have become one of the main contaminants of emerging concern (CECs) due to their high usage and increased release into the environment. This study aims to assess the effects caused by three widely consumed hepatotoxic pharmaceutical compounds: an antibiotic (amoxicillin), an antiepileptic (carbamazepine), and an antidepressant (trazodone), on human health when indirectly exposed to toxicologically relevant concentrations (30, 15, and 7.5 μM for amoxicillin and carbamazepine, and 4, 2, and 1 μM for trazodone). A combination of semi-targeted metabolomic and targeted sphingolipid analyses was chosen to unravel the metabolic alterations in human hepatic cells exposed to these CECs at three concentrations for 24 h. HepG2 hepatoma cells were encapsulated in sodium alginate spheroids to improve the physiological relevance of this in vitro approach. Statistical analysis was used to identify the most affected metabolites and sphingolipids for each drug exposure. The results revealed small but significant changes in response to carbamazepine and trazodone exposures, affecting sphingolipid, glycerophospholipid precursors, and amino acid metabolism. Under both drug treatments, a decrease in various ceramide species (related to cell signaling) was observed, along with reduced taurine levels (related to the biosynthesis of bile acid conjugates) and carnitine levels (suggesting an impact on energy production). These and other drug-specific changes indicate that cellular functions in liver cells might be altered under low doses of these CECs, potentially affecting the health of other organs.
Collapse
Affiliation(s)
- Miriam Pérez-Cova
- Department of Environmental Chemistry, IDAEA-CSIC, Jordi Girona 18-26, Barcelona E08034, Spain; Department of Chemical Engineering and Analytical Chemistry, University of Barcelona, Diagonal 647, Barcelona, Barcelona E08028, Spain
| | - Carmen Bedia
- Department of Environmental Chemistry, IDAEA-CSIC, Jordi Girona 18-26, Barcelona E08034, Spain
| | - Antonio Checa
- Unit of Integrative Metabolomics, Institute of Environmental Medicine, Karolinska Institute, Stockholm, Sweden
| | - Isabel Meister
- Unit of Integrative Metabolomics, Institute of Environmental Medicine, Karolinska Institute, Stockholm, Sweden; Gunma University Initiative for Advanced Research (GIAR), Gunma University, 3-39-22 Showa-machi, Maebashi, Gunma 371-8511, Japan
| | - Romà Tauler
- Department of Environmental Chemistry, IDAEA-CSIC, Jordi Girona 18-26, Barcelona E08034, Spain
| | - Craig E Wheelock
- Unit of Integrative Metabolomics, Institute of Environmental Medicine, Karolinska Institute, Stockholm, Sweden; Gunma University Initiative for Advanced Research (GIAR), Gunma University, 3-39-22 Showa-machi, Maebashi, Gunma 371-8511, Japan; Department of Respiratory Medicine and Allergy, Karolinska University Hospital, Stockholm 141-86, Sweden
| | - Joaquim Jaumot
- Department of Environmental Chemistry, IDAEA-CSIC, Jordi Girona 18-26, Barcelona E08034, Spain.
| |
Collapse
|
48
|
Frederick MJ, Perez-Bello D, Yadollahi P, Castro P, Frederick A, Frederick A, Osman RA, Essien F, Yebra I, Hamlin A, Ow TJ, Skinner HD, Sandulache VC. Reliable RNA-seq analysis from FFPE specimens as a means to accelerate cancer-related health disparities research. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.10.617597. [PMID: 39416147 PMCID: PMC11482925 DOI: 10.1101/2024.10.10.617597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
Whole transcriptome sequencing (WTS/ RNA-Seq) is a ubiquitous tool for investigating cancer biology. RNA isolated from frozen sources limits possible studies for analysis of associations with phenotypes or clinical variables requiring long-term follow-up. Although good correlations are reported in RNA-Seq data from paired frozen and formalin fixed paraffin embedded (FFPE) samples, uncertainties regarding RNA quality, methods of extraction, and data reliability are hurdles to utilization of archival samples. We compared three different platforms for performing RNA-seq using archival FFPE oropharyngeal squamous carcinoma (OPSCC) specimens stored up to 20 years, as part of an investigation of transcriptional profiles related to health disparities. We developed guidelines to purify DNA and RNA from FFPE tissue and perform downstream RNA-seq and DNA SNP arrays. RNA was extracted from 150 specimens, with an average yield of 401.8 ng/cm 2 of tissue. Most samples yielded sufficient RNA reads >13,000 protein coding genes which could be used to differentiate HPV-associated from HPV-independent OPSCCs. Co-isolated DNA was used to identify patient ancestry. Utilizing the methods described in this study provides a robust, reliable, and standardized means of DNA & RNA extraction from FFPE as well as a means by which to assure the quality of the data generated.
Collapse
|
49
|
Ma Q, Li J, Yu S, Zhou J, Liu Y, Wang X, Ye D, Wu Y, Gong T, Zhang Q, Wang L, Zou J, Li Y. YkuR functions as a protein deacetylase in Streptococcus mutans. Proc Natl Acad Sci U S A 2024; 121:e2407820121. [PMID: 39356671 PMCID: PMC11474102 DOI: 10.1073/pnas.2407820121] [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: 04/18/2024] [Accepted: 08/13/2024] [Indexed: 10/04/2024] Open
Abstract
Protein acetylation is a common and reversible posttranslational modification tightly governed by protein acetyltransferases and deacetylases crucial for various biological processes in both eukaryotes and prokaryotes. Although recent studies have characterized many acetyltransferases in diverse bacterial species, only a few protein deacetylases have been identified in prokaryotes, perhaps in part due to their limited sequence homology. In this study, we identified YkuR, encoded by smu_318, as a unique protein deacetylase in Streptococcus mutans. Through protein acetylome analysis, we demonstrated that the deletion of ykuR significantly upregulated protein acetylation levels, affecting key enzymes in translation processes and metabolic pathways, including starch and sucrose metabolism, glycolysis/gluconeogenesis, and biofilm formation. In particular, YkuR modulated extracellular polysaccharide synthesis and biofilm formation through the direct deacetylation of glucosyltransferases (Gtfs) in the presence of NAD+. Intriguingly, YkuR can be acetylated in a nonenzymatic manner, which then negatively regulated its deacetylase activity, suggesting the presence of a self-regulatory mechanism. Moreover, in vivo studies further demonstrated that the deletion of ykuR attenuated the cariogenicity of S. mutans in the rat caries model, substantiating its involvement in the pathogenesis of dental caries. Therefore, our study revealed a unique regulatory mechanism mediated by YkuR through protein deacetylation that regulates the physiology and pathogenicity of S. mutans.
Collapse
Affiliation(s)
- Qizhao Ma
- Laboratory of Oral Microbiology, State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu610041, China
- Department of Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu610041, China
| | - Jing Li
- Laboratory of Oral Microbiology, State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu610041, China
- Department of Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu610041, China
| | - Shuxing Yu
- Laboratory of Oral Microbiology, State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu610041, China
- Department of Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu610041, China
| | - Jing Zhou
- Laboratory of Oral Microbiology, State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu610041, China
- Department of Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu610041, China
| | - Yaqi Liu
- Laboratory of Oral Microbiology, State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu610041, China
- Department of Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu610041, China
| | - Xinyue Wang
- Laboratory of Oral Microbiology, State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu610041, China
- Department of Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu610041, China
| | - Dingwei Ye
- Laboratory of Oral Microbiology, State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu610041, China
- Department of Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu610041, China
| | - Yumeng Wu
- Laboratory of Oral Microbiology, State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu610041, China
- Department of Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu610041, China
| | - Tao Gong
- Laboratory of Oral Microbiology, State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu610041, China
| | - Qiong Zhang
- Laboratory of Oral Microbiology, State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu610041, China
- Department of Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu610041, China
| | - Lingyun Wang
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT06510
| | - Jing Zou
- Laboratory of Oral Microbiology, State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu610041, China
- Department of Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu610041, China
| | - Yuqing Li
- Laboratory of Oral Microbiology, State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu610041, China
- Laboratory of Archaeological Repository, Center for Archaeological Science, Sichuan University, Chengdu610041, China
| |
Collapse
|
50
|
Hoernstein SNW, Schlosser A, Fiedler K, van Gessel N, Igloi GL, Lang D, Reski R. A snapshot of the Physcomitrella N-terminome reveals N-terminal methylation of organellar proteins. PLANT CELL REPORTS 2024; 43:250. [PMID: 39361041 PMCID: PMC11450134 DOI: 10.1007/s00299-024-03329-1] [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: 06/13/2024] [Accepted: 09/13/2024] [Indexed: 10/05/2024]
Abstract
KEY MESSAGE Analysis of the N-terminome of Physcomitrella reveals N-terminal monomethylation of nuclear-encoded, mitochondria-localized proteins. Post- or co-translational N-terminal modifications of proteins influence their half-life as well as mediating protein sorting to organelles via cleavable N-terminal sequences that are recognized by the respective translocation machinery. Here, we provide an overview on the current modification state of the N-termini of over 4500 proteins from the model moss Physcomitrella (Physcomitrium patens) using a compilation of 24 N-terminomics datasets. Our data reveal distinct proteoforms and modification states and confirm predicted targeting peptide cleavage sites of 1,144 proteins localized to plastids and the thylakoid lumen, to mitochondria, and to the secretory pathway. In addition, we uncover extended N-terminal methylation of mitochondrial proteins. Moreover, we identified PpNTM1 (P. patens alpha N-terminal protein methyltransferase 1) as a candidate for protein methylation in plastids, mitochondria, and the cytosol. These data can now be used to optimize computational targeting predictors, for customized protein fusions and their targeted localization in biotechnology, and offer novel insights into potential dual targeting of proteins.
Collapse
Affiliation(s)
- Sebastian N W Hoernstein
- Plant Biotechnology, Faculty of Biology, University of Freiburg, Schaenzlestr. 1, 79104, Freiburg, Germany
| | - Andreas Schlosser
- Rudolf Virchow Center for Experimental Biomedicine, University of Würzburg, Josef-Schneider-Str. 2, 97080, Würzburg, Germany
| | - Kathrin Fiedler
- Institute of Biology III, University of Freiburg, Schaenzlestr. 1, 79104, Freiburg, Germany
- Lonza, Hochbergerstr. 60A, 4057, Basel, Switzerland
| | - Nico van Gessel
- Plant Biotechnology, Faculty of Biology, University of Freiburg, Schaenzlestr. 1, 79104, Freiburg, Germany
| | - Gabor L Igloi
- Institute of Biology III, University of Freiburg, Schaenzlestr. 1, 79104, Freiburg, Germany
| | - Daniel Lang
- Plant Biotechnology, Faculty of Biology, University of Freiburg, Schaenzlestr. 1, 79104, Freiburg, Germany
- Microbial Genomics and Bioforensics, Bundeswehr Institute of Microbiology, Neuherbergstr. 11, 80937, Munich, Germany
| | - Ralf Reski
- Plant Biotechnology, Faculty of Biology, University of Freiburg, Schaenzlestr. 1, 79104, Freiburg, Germany.
- Signalling Research Centres BIOSS and CIBSS, Schaenzlestr. 18, 79104, Freiburg, Germany.
| |
Collapse
|