1
|
Albrecht E, Pelz K, Gress A, Trung HN, Kalinina OV, Kacprowski T, Baumbach J, List M, Tsoy O. DIGGER 2.0: digging into the functional impact of differential splicing on human and mouse disorders. Nucleic Acids Res 2025:gkaf384. [PMID: 40337913 DOI: 10.1093/nar/gkaf384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Revised: 04/11/2025] [Accepted: 04/25/2025] [Indexed: 05/09/2025] Open
Abstract
Changes in alternative splicing between groups or conditions contribute to protein-protein interaction rewiring, a consequence often neglected in data analysis. The web server and database DIGGER overcomes this limitation by augmenting a protein-protein interaction network with domain-domain interactions and splicing information. Here, we present DIGGER 2.0, which now features both experimental and newly added predicted domain-domain interactions. In addition to the human interactome, DIGGER 2.0 adds support for mouse as an important model organism. Additionally, we integrated the splicing analysis tool NEASE, which allows users to perform online splicing- and interactome-informed enrichment analysis on RNA-seq data. In two application cases (multiple sclerosis and mice models of cardiac diseases), we show the utility of DIGGER 2.0 for deeper exploration and functional interpretation of changes in alternative splicing in human and mouse disorders. DIGGER 2.0 is available at https://exbio.wzw.tum.de/digger/.
Collapse
Affiliation(s)
- Elias Albrecht
- Data Science in Systems Biology, TUM School of Life Sciences, Technical University of Munich, Maximus-von-Imhof Forum 3, 85354 Freising, Germany
- Institute for Computational Systems Biology, University of Hamburg, Albert-Einstein-Ring 8-10, 22761 Hamburg, Germany
| | - Konstantin Pelz
- Data Science in Systems Biology, TUM School of Life Sciences, Technical University of Munich, Maximus-von-Imhof Forum 3, 85354 Freising, Germany
| | - Alexander Gress
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), Campus E8.1, 66123 Saarbrücken, Germany
- Graduate School of Computer Science, Saarland University, Campus E1.3, 66123 Saarbrücken, Germany
| | - Hieu Nguyen Trung
- Data Science in Systems Biology, TUM School of Life Sciences, Technical University of Munich, Maximus-von-Imhof Forum 3, 85354 Freising, Germany
| | - Olga V Kalinina
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), Campus E8.1, 66123 Saarbrücken, Germany
- Drug Bioinformatics, Medical Faculty, Saarland University, Gebäude 15, 66421 Homburg, Germany
- Center for Bioinformatics, Saarland University, Campus E2.1, 66123 Saarbrücken, Germany
| | - Tim Kacprowski
- Division Data Science in Biomedicine, Peter L. Reichertz Institute for Medical Informatics of Technische Universität Braunschweig and Hannover Medical School, Rebenring 56 Lower Saxony, 38106 Braunschweig, Germany
- Braunschweig Integrated Centre of Systems Biology (BRICS), Technische Universität Braunschweig, Rebenring 56 Lower Saxony, 38106 Braunschweig, Germany
| | - Jan Baumbach
- Institute for Computational Systems Biology, University of Hamburg, Albert-Einstein-Ring 8-10, 22761 Hamburg, Germany
- Institute of Mathematics and Computer Science, University of Southern Denmark, Campusvej 55, 5230 Odense, Denmark
| | - Markus List
- Data Science in Systems Biology, TUM School of Life Sciences, Technical University of Munich, Maximus-von-Imhof Forum 3, 85354 Freising, Germany
- Munich Data Science Institute (MDSI), Technical University of Munich, Walther-von-Dyck-Straße 10, 85748 Garching, Germany
| | - Olga Tsoy
- Institute for Computational Systems Biology, University of Hamburg, Albert-Einstein-Ring 8-10, 22761 Hamburg, Germany
| |
Collapse
|
2
|
Yang L, Shao Q, Su J, Liu Y, Chen L, Ndzie Noah ML, Li N, Coorssen JR, Zhan X. What does one-dimensional gel electrophoresis-based western blotting data really mean in the reality of proteoforms? Talanta 2025; 295:128266. [PMID: 40347635 DOI: 10.1016/j.talanta.2025.128266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Revised: 04/27/2025] [Accepted: 05/02/2025] [Indexed: 05/14/2025]
Abstract
One-dimensional gel electrophoresis-based western blotting (1DE-WB) is a routine technique for protein analysis. However, multiple immunoreactive bands are often found for a single protein target in 1DE-WB, yet only the band corresponding to the theoretical molecular weight of the canonical protein is considered relevant and additional immunoreactive bands are dismissed as false positives. However, what additional bands actually contain has not been established. Proteoforms are the final structural and functional entities of a gene product. Here, multiple immunopositive bands in the 1DE-WB analysis of growth hormone and prolactin in human pituitary tissue samples were identified by MS, 2DE-WB, or 2DE-WB combined with MS, and immunopositive bands were found to contain multiple proteoforms; a range of published 1DE-WB data indicate this is likely a common phenomenon. Thus, multiple 1DE-WB positive bands are likely to represent different proteoforms rather than non-specific reactions and MS, 2DE-WB, or 2DE-WB combined with MS are effective approaches to identify proteoforms and thus more thoroughly understand 1DE-WB data.
Collapse
Affiliation(s)
- Lamei Yang
- Shandong Provincial Key Laboratory of Precision Oncology, Shandong Cancer HospItal and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, 440 Jiyan Road, Jinan, Shandong, 250117, PR China
| | - Qianwen Shao
- Shandong Provincial Key Laboratory of Precision Oncology, Shandong Cancer HospItal and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, 440 Jiyan Road, Jinan, Shandong, 250117, PR China; Shandong Provincial Key Medical and Health Laboratory of Ovarian Cancer Multiomics, & Jinan Key Laboratory of Cancer Multiomics, Medical Science and Technology Innovation Center, Shandong First Medical University, 6699 Qingdao Road, Jinan, Shandong, 250117, PR China
| | - Juwen Su
- Shandong Provincial Key Laboratory of Precision Oncology, Shandong Cancer HospItal and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, 440 Jiyan Road, Jinan, Shandong, 250117, PR China
| | - Yingchao Liu
- Department of Neurosurgery, Shandong Provincial Hospital, Shandong First Medical University, 324 Jinwu Weiqi Road, Jinan, Shandong, 250021, PR China
| | - Liang Chen
- Department of Gynecological Oncology, Shandong Cancer HospItal and Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, 440 Jiyan Road, Jinan, 250117, PR China
| | - Marie Louise Ndzie Noah
- Shandong Provincial Key Laboratory of Precision Oncology, Shandong Cancer HospItal and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, 440 Jiyan Road, Jinan, Shandong, 250117, PR China
| | - Na Li
- Shandong Provincial Key Laboratory of Precision Oncology, Shandong Cancer HospItal and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, 440 Jiyan Road, Jinan, Shandong, 250117, PR China
| | - Jens R Coorssen
- Departments of Health Sciences and Biological Sciences, Faculties of Applied Health Sciences and Mathematics & Science, Brock University, St. Catharines, ON, L2S 3A1, Canada; Institute for Globally Distributed Open Research and Education (IGDORE), St. Catharines, ON, Canada
| | - Xianquan Zhan
- Shandong Provincial Key Laboratory of Precision Oncology, Shandong Cancer HospItal and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, 440 Jiyan Road, Jinan, Shandong, 250117, PR China; Shandong Provincial Key Medical and Health Laboratory of Ovarian Cancer Multiomics, & Jinan Key Laboratory of Cancer Multiomics, Medical Science and Technology Innovation Center, Shandong First Medical University, 6699 Qingdao Road, Jinan, Shandong, 250117, PR China.
| |
Collapse
|
3
|
Wang Q, Wang Q, Zhu G, Sun L. Capillary Electrophoresis-Mass Spectrometry for Top-Down Proteomics. ANNUAL REVIEW OF ANALYTICAL CHEMISTRY (PALO ALTO, CALIF.) 2025; 18:125-147. [PMID: 39847747 PMCID: PMC12081194 DOI: 10.1146/annurev-anchem-071124-092242] [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] [Indexed: 01/25/2025]
Abstract
Mass spectrometry (MS)-based top-down proteomics (TDP) characterizes proteoforms in cells, tissues, and biological fluids (e.g., human plasma) to better our understanding of protein function and to discover new protein biomarkers for disease diagnosis and therapeutic development. Separations of proteoforms with high peak capacity are needed due to the high complexity of biological samples. Capillary electrophoresis (CE)-MS has been recognized as a powerful analytical tool for protein analysis since the 1980s owing to its high separation efficiency and sensitivity of CE-MS for proteoforms. Here, we review benefits of CE-MS for advancing TDP, challenges and solutions of the method, and the main research areas in which CE-MS-based TDP can make significant contributions. We provide a brief perspective of CE-MS-based TDP moving forward.
Collapse
Affiliation(s)
- Qianjie Wang
- Department of Chemistry, Michigan State University, East Lansing, Michigan, USA;
| | - Qianyi Wang
- Department of Chemistry, Michigan State University, East Lansing, Michigan, USA;
| | - Guijie Zhu
- Department of Chemistry, Michigan State University, East Lansing, Michigan, USA;
| | - Liangliang Sun
- Department of Chemistry, Michigan State University, East Lansing, Michigan, USA;
| |
Collapse
|
4
|
Yong C, Liang Y, Wang M, Jin W, Fan X, Wang Z, Cao K, Wu T, Li Q, Chang C. Alternative splicing: A key regulator in T cell response and cancer immunotherapy. Pharmacol Res 2025; 215:107713. [PMID: 40147681 DOI: 10.1016/j.phrs.2025.107713] [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: 12/27/2024] [Revised: 03/03/2025] [Accepted: 03/24/2025] [Indexed: 03/29/2025]
Abstract
Alternative splicing (AS), a key post-transcriptional regulatory mechanism, is frequently dysregulated in cancer, driving both tumor progression and immune modulation. Aberrant AS influences antigen presentation, T cell activation, immune checkpoint regulation, and cytokine signaling, contributing to immune evasion but also presenting unique therapeutic vulnerabilities. Targeting AS has emerged as a promising strategy in cancer immunotherapy. Splicing-derived neoantigens have been identified as potent inducers of CD8⁺ T cell responses, offering potential for personalized treatment. AS modulators such as PRMT5 inhibitor GSK3326595 enhance immunotherapy efficacy by upregulating MHC class II expression and promoting T cell infiltration, while RBM39 inhibitor indisulam induces tumor-specific neoantigens. Furthermore, combining AS-targeting drugs with immune checkpoint inhibitors (ICIs) has demonstrated synergistic effects, improved response rates and overcoming resistance in preclinical models. Despite these advances, challenges remain in optimizing drug specificity and minimizing toxicity. Future efforts should focus on refining AS-targeting therapies, identifying predictive biomarkers, and integrating these approaches into clinical applications. This review highlights the therapeutic potential of AS modulation in cancer immunotherapy and its implications for advancing precision oncology.
Collapse
Affiliation(s)
- Caiyu Yong
- School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, PR China; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, PR China
| | - Yexin Liang
- School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, PR China; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, PR China
| | - Minmin Wang
- School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, PR China; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, PR China
| | - Weiwei Jin
- School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, PR China; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, PR China
| | - Xuefei Fan
- School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, PR China; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, PR China
| | - Zhengwen Wang
- School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, PR China; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, PR China
| | - Kui Cao
- School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, PR China; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, PR China
| | - Tong Wu
- School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, PR China; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, PR China
| | - Qian Li
- School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, PR China; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, PR China
| | - Cunjie Chang
- School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, PR China; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, PR China.
| |
Collapse
|
5
|
Lee EJ, Park S, Jeong KS. Sirt2 deficiency aggravates intramuscular adipose tissue infiltration and impairs myogenesis with aging in male mice. Biogerontology 2025; 26:93. [PMID: 40257511 DOI: 10.1007/s10522-025-10238-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Accepted: 04/08/2025] [Indexed: 04/22/2025]
Abstract
Sarcopenia, closely associated with other diseases such as diabetes, metabolic syndrome, and osteoporosis, significantly impacts aging populations. It is characterized by muscle atrophy, increased intramuscular adipose tissue, impaired myogenesis, chronic low-grade inflammation, and reduced muscle function. The mechanisms behind aging muscle remain incompletely understood. This study aims to elucidate the role of Sirt2 in the aging process of skeletal muscles and enhance our understanding of the underlying mechanisms. Sirt2 expression was reduced in aging muscle of male mice by 40%, compared to young muscle. Aged male Sirt2 knockout mice exhibit increased intramuscular adipose tissue infiltration by 8.5-fold changes. Furthermore, the deletion of Sirt2 exacerbated myogenesis impairment in aged muscle by decreasing the expression of Pax7 (50%) and NogoA (80%), compared to age- and sex- matched counterparts, emphasizing the role of Sirt2 in pathology of aging muscle. Additionally, long-term Sirt2 deletion affected other Sirtuin subfamily members, with decreased expressions of Sirt1 (65%), Sirt4 (94%), and Sirt5 (71%), and increased expressions of Sirt6 (4.6-fold) and Sirt7 (2.8-fold) in old male Sirt2 knockout mice, while there was no difference of these gene expression in young male mice. This study underscores the critical need for a deeper investigation into Sirt2, promising new insights that could lead to targeted therapies for sarcopenia, ultimately improving the quality of life in the elderly.
Collapse
Affiliation(s)
- Eun-Joo Lee
- Department of Pathology, College of Veterinary Medicine, Kyungpook National University, Daegu, 41566, Republic of Korea.
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, 02115, USA.
| | - SunYoung Park
- Department of Pathology, College of Veterinary Medicine, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - Kyu-Shik Jeong
- Department of Pathology, College of Veterinary Medicine, Kyungpook National University, Daegu, 41566, Republic of Korea.
- Department of Companion Animal Health, Daegu Haany University, Gyeongsan, 38610, Republic of Korea.
- Stellamed Co., LTD, Daegu, 41504, Republic of Korea.
| |
Collapse
|
6
|
Murali M, Saquing J, Lu S, Gao Z, Watts EF, Jordan B, Wakefield ZP, Fiszbein A, Cooper DR, Castaldi PJ, Korkin D, Sheynkman GM. Biosurfer for systematic tracking of regulatory mechanisms leading to protein isoform diversity. Genome Res 2025; 35:1012-1024. [PMID: 40086882 PMCID: PMC12047184 DOI: 10.1101/gr.279317.124] [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: 03/15/2024] [Accepted: 01/06/2025] [Indexed: 03/16/2025]
Abstract
Long-read RNA-seq has shed light on transcriptomic complexity, but questions remain about the functionality of downstream protein products. We introduce Biosurfer, a computational approach for comparing protein isoforms, while systematically tracking the transcriptional, splicing, and translational variations that underlie differences in the sequences of the protein products. Using Biosurfer, we analyzed the differences in 35,082 pairs of GENCODE annotated protein isoforms, finding a majority (70%) of variable N-termini are due to the alternative transcription start sites, while only 9% arise from 5' UTR alternative splicing (AS). Biosurfer's detailed tracking of nucleotide-to-residue relationships helps reveal an uncommonly tracked source of single amino acid residue changes arising from the codon splits at junctions. For 17% of internal sequence changes, such split codon patterns lead to single residue differences, termed "ragged codons." Of variable C-termini, 72% involve splice- or intron retention-induced reading frameshifts. We systematically characterize an unusual pattern of reading frame changes, in which the first frameshift is closely followed by a distinct second frameshift that restores the original frame, which we term a "snapback" frameshift. We analyze the long-read RNA-seq-predicted proteome of a human cell line and find similar trends as compared to our GENCODE analysis, with the exception of a higher proportion of transcripts predicted to undergo nonsense-mediated decay. Biosurfer's comprehensive characterization of long-read RNA-seq data sets should accelerate insights of the functional role of protein isoforms, providing mechanistic explanation of the origins of the proteomic diversity driven by the AS. Biosurfer is available as a Python package.
Collapse
Affiliation(s)
- Mayank Murali
- Broad Institute of MIT and Harvard University, Cambridge, Massachusetts 02142, USA
| | - Jamie Saquing
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, Virginia 22903, USA
| | - Senbao Lu
- Bioinformatics and Computational Biology Program, Worcester Polytechnic Institute, Worcester, Massachusetts 01609, USA
- Computer Science Department, Worcester Polytechnic Institute, Worcester, Massachusetts 01609, USA
| | - Ziyang Gao
- Bioinformatics and Computational Biology Program, Worcester Polytechnic Institute, Worcester, Massachusetts 01609, USA
- Computer Science Department, Worcester Polytechnic Institute, Worcester, Massachusetts 01609, USA
| | - Emily F Watts
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, Virginia 22903, USA
| | - Ben Jordan
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, Virginia 22903, USA
| | - Zachary Peters Wakefield
- Bioinformatics Program, Boston University, Boston, Massachusetts 02215, USA
- Department of Biology, Boston University, Boston, Massachusetts 02215, USA
| | - Ana Fiszbein
- Bioinformatics Program, Boston University, Boston, Massachusetts 02215, USA
- Department of Biology, Boston University, Boston, Massachusetts 02215, USA
| | - David R Cooper
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, Virginia 22903, USA
| | - Peter J Castaldi
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts 02115, USA
- Division of General Medicine and Primary Care, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts 02115, USA
| | - Dmitry Korkin
- Bioinformatics and Computational Biology Program, Worcester Polytechnic Institute, Worcester, Massachusetts 01609, USA
- Computer Science Department, Worcester Polytechnic Institute, Worcester, Massachusetts 01609, USA
| | - Gloria M Sheynkman
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, Virginia 22903, USA;
- Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, Virginia 22903, USA
- Center for Public Health Genomics, University of Virginia, Charlottesville, Virginia 22903, USA
- UVA Cancer Center, University of Virginia, Charlottesville, Virginia 22903, USA
| |
Collapse
|
7
|
Najar CFBA, Feng R, Dai C, Fair B, Hauck Q, Li J, Cao X, Dey KK, De Jager P, Bennett D, Liu X, Wang G, Li YI. Genetic and functional analysis of unproductive splicing using LeafCutter2. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.04.06.646893. [PMID: 40291686 PMCID: PMC12026817 DOI: 10.1101/2025.04.06.646893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
Alternative splicing commonly generates unproductive mRNA transcripts that harbor premature termination codons 1 , leading to their degradation by nonsense-mediated decay (NMD). These events reduce overall protein expression levels of affected genes, potentially contributing to gene regulation and disease mechanisms. Here, we present LeafCutter2, which enables identification and quantification of unproductive splicing from short-read RNA-seq data. LeafCutter2 requires minimal gene annotations (start and stop codons) to annotate NMD-inducing splicing events, and identifies differential unproductive splicing between groups, providing insights into its contributions to differential gene expression. Moreover, LeafCutter2 enables mapping of unproductive splicing quantitative trait loci (u-sQTLs), which often colocalize with expression QTLs and GWAS loci. Applying LeafCutter2 to RNA-seq data across human and 6 non-human species, we uncovered a broad landscape of unproductive splicing, which varies widely across tissues. Strikingly, we observed a conserved developmental-stage-specific increase in unproductive splicing during testis maturation across all species. In Alzheimer's disease (AD), we analyzed RNA-seq data from the AD Functional Genomics consortium FunGen-xQTL project and identified unproductive splicing events in 18 AD risk genes, including TSPAN14 , PICALM , and CASS4 , likely mediating genetic effects on disease risk. We performed an integrative analysis using gene expression QTLs, protein expression QTLs, and AD GWAS data, showing that unproductive splicing provides unique regulatory insights beyond traditional approaches. Thus, LeafCutter2 represents a powerful tool for understanding the functional impact of alternative splicing on gene expression and disease mechanisms.
Collapse
|
8
|
Jia Q, Sun X, Li H, Guo J, Niu K, Chan KM, Bernards R, Qin W, Jin H. Perturbation of mRNA splicing in liver cancer: insights, opportunities and challenges. Gut 2025; 74:840-852. [PMID: 39658264 DOI: 10.1136/gutjnl-2024-333127] [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: 07/08/2024] [Accepted: 11/08/2024] [Indexed: 12/12/2024]
Abstract
Perturbation of mRNA splicing is commonly observed in human cancers and plays a role in various aspects of cancer hallmarks. Understanding the mechanisms and functions of alternative splicing (AS) not only enables us to explore the complex regulatory network involved in tumour initiation and progression but also reveals potential for RNA-based cancer treatment strategies. This review provides a comprehensive summary of the significance of AS in liver cancer, covering the regulatory mechanisms, cancer-related AS events, abnormal splicing regulators, as well as the interplay between AS and post-transcriptional and post-translational regulations. We present the current bioinformatic approaches and databases to detect and analyse AS in cancer, and discuss the implications and perspectives of AS in the treatment of liver cancer.
Collapse
Affiliation(s)
- Qi Jia
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaoxiao Sun
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Haoyu Li
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jianglong Guo
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Kongyan Niu
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Kui Ming Chan
- Department of Biomedical Sciences, City University of Hong Kong, HKSAR, China
| | - René Bernards
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Division of Molecular Carcinogenesis, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, Noord-Holland, The Netherlands
| | - Wenxin Qin
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Haojie Jin
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
9
|
Lambourne L, Mattioli K, Santoso C, Sheynkman G, Inukai S, Kaundal B, Berenson A, Spirohn-Fitzgerald K, Bhattacharjee A, Rothman E, Shrestha S, Laval F, Carroll BS, Plassmeyer SP, Emenecker RJ, Yang Z, Bisht D, Sewell JA, Li G, Prasad A, Phanor S, Lane R, Moyer DC, Hunt T, Balcha D, Gebbia M, Twizere JC, Hao T, Holehouse AS, Frankish A, Riback JA, Salomonis N, Calderwood MA, Hill DE, Sahni N, Vidal M, Bulyk ML, Fuxman Bass JI. Widespread variation in molecular interactions and regulatory properties among transcription factor isoforms. Mol Cell 2025; 85:1445-1466.e13. [PMID: 40147441 PMCID: PMC12121496 DOI: 10.1016/j.molcel.2025.03.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 12/06/2024] [Accepted: 03/05/2025] [Indexed: 03/29/2025]
Abstract
Most human transcription factor (TF) genes encode multiple protein isoforms differing in DNA-binding domains, effector domains, or other protein regions. The global extent to which this results in functional differences between isoforms remains unknown. Here, we systematically compared 693 isoforms of 246 TF genes, assessing DNA binding, protein binding, transcriptional activation, subcellular localization, and condensate formation. Relative to reference isoforms, two-thirds of alternative TF isoforms exhibit differences in one or more molecular activities, which often could not be predicted from sequence. We observed two primary categories of alternative TF isoforms: "rewirers" and "negative regulators," both of which were associated with differentiation and cancer. Our results support a model wherein the relative expression levels of, and interactions involving, TF isoforms add an understudied layer of complexity to gene regulatory networks, demonstrating the importance of isoform-aware characterization of TF functions and providing a rich resource for further studies.
Collapse
Affiliation(s)
- Luke Lambourne
- Center for Cancer Systems Biology (CCSB), Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA; Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Kaia Mattioli
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA.
| | - Clarissa Santoso
- Department of Biology, Boston University, Boston, MA 02215, USA; Bioinformatics Program, Boston University, Boston, MA 02215, USA
| | - Gloria Sheynkman
- Center for Cancer Systems Biology (CCSB), Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA; Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Sachi Inukai
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Babita Kaundal
- Department of Epigenetics and Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Anna Berenson
- Molecular Biology, Cell Biology & Biochemistry Program, Boston University, Boston, MA 02215, USA
| | - Kerstin Spirohn-Fitzgerald
- Center for Cancer Systems Biology (CCSB), Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA; Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Anukana Bhattacharjee
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA; Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Elisabeth Rothman
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | | | - Florent Laval
- Center for Cancer Systems Biology (CCSB), Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA; Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; TERRA Teaching and Research Centre, University of Liège, Gembloux 5030, Belgium; Laboratory of Viral Interactomes, GIGA Institute, University of Liège, Liège 4000, Belgium
| | - Brent S Carroll
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Stephen P Plassmeyer
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, MO, USA; Center for Biomolecular Condensates, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Ryan J Emenecker
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, MO, USA; Center for Biomolecular Condensates, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Zhipeng Yang
- Center for Cancer Systems Biology (CCSB), Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA; Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Deepa Bisht
- Department of Epigenetics and Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jared A Sewell
- Department of Biology, Boston University, Boston, MA 02215, USA
| | - Guangyuan Li
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA; Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Anisa Prasad
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA; Harvard College, Cambridge, MA 02138, USA
| | - Sabrina Phanor
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Ryan Lane
- Department of Biology, Boston University, Boston, MA 02215, USA
| | - Devlin C Moyer
- Bioinformatics Program, Boston University, Boston, MA 02215, USA
| | - Toby Hunt
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Hinxton, Cambridge CD10 1SD, UK
| | - Dawit Balcha
- Center for Cancer Systems Biology (CCSB), Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA; Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Marinella Gebbia
- Center for Cancer Systems Biology (CCSB), Dana-Farber Cancer Institute, Boston, MA 02215, USA; The Donnelly Centre, University of Toronto, Toronto, ON M5S 3E1, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 3E1, Canada; Lunenfeld-Tanenbaum Research Institute (LTRI), Sinai Health System, Toronto, ON M5G 1X5, Canada
| | - Jean-Claude Twizere
- Center for Cancer Systems Biology (CCSB), Dana-Farber Cancer Institute, Boston, MA 02215, USA; TERRA Teaching and Research Centre, University of Liège, Gembloux 5030, Belgium; Laboratory of Viral Interactomes, GIGA Institute, University of Liège, Liège 4000, Belgium
| | - Tong Hao
- Center for Cancer Systems Biology (CCSB), Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA; Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Alex S Holehouse
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, MO, USA; Center for Biomolecular Condensates, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Adam Frankish
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Hinxton, Cambridge CD10 1SD, UK
| | - Josh A Riback
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Nathan Salomonis
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA; Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Michael A Calderwood
- Center for Cancer Systems Biology (CCSB), Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA; Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - David E Hill
- Center for Cancer Systems Biology (CCSB), Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA; Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Nidhi Sahni
- Department of Epigenetics and Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| | - Marc Vidal
- Center for Cancer Systems Biology (CCSB), Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA; Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA.
| | - Martha L Bulyk
- Center for Cancer Systems Biology (CCSB), Dana-Farber Cancer Institute, Boston, MA 02215, USA; Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA; Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA.
| | - Juan I Fuxman Bass
- Center for Cancer Systems Biology (CCSB), Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Biology, Boston University, Boston, MA 02215, USA; Bioinformatics Program, Boston University, Boston, MA 02215, USA; Molecular Biology, Cell Biology & Biochemistry Program, Boston University, Boston, MA 02215, USA.
| |
Collapse
|
10
|
Aguzzoli Heberle B, Brandon JA, Page ML, Nations KA, Dikobe KI, White BJ, Gordon LA, Fox GA, Wadsworth ME, Doyle PH, Williams BA, Fox EJ, Shantaraman A, Ryten M, Goodwin S, Ghiban E, Wappel R, Mavruk-Eskipehlivan S, Miller JB, Seyfried NT, Nelson PT, Fryer JD, Ebbert MTW. Mapping medically relevant RNA isoform diversity in the aged human frontal cortex with deep long-read RNA-seq. Nat Biotechnol 2025; 43:635-646. [PMID: 38778214 PMCID: PMC11863200 DOI: 10.1038/s41587-024-02245-9] [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: 08/06/2023] [Accepted: 04/15/2024] [Indexed: 05/25/2024]
Abstract
Determining whether the RNA isoforms from medically relevant genes have distinct functions could facilitate direct targeting of RNA isoforms for disease treatment. Here, as a step toward this goal for neurological diseases, we sequenced 12 postmortem, aged human frontal cortices (6 Alzheimer disease cases and 6 controls; 50% female) using one Oxford Nanopore PromethION flow cell per sample. We identified 1,917 medically relevant genes expressing multiple isoforms in the frontal cortex where 1,018 had multiple isoforms with different protein-coding sequences. Of these 1,018 genes, 57 are implicated in brain-related diseases including major depression, schizophrenia, Parkinson's disease and Alzheimer disease. Our study also uncovered 53 new RNA isoforms in medically relevant genes, including several where the new isoform was one of the most highly expressed for that gene. We also reported on five mitochondrially encoded, spliced RNA isoforms. We found 99 differentially expressed RNA isoforms between cases with Alzheimer disease and controls.
Collapse
Affiliation(s)
- Bernardo Aguzzoli Heberle
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, USA
- Department of Neuroscience, College of Medicine, University of Kentucky, Lexington, KY, USA
| | - J Anthony Brandon
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, USA
| | - Madeline L Page
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, USA
| | - Kayla A Nations
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, USA
| | - Ketsile I Dikobe
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, USA
| | - Brendan J White
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, USA
| | - Lacey A Gordon
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, USA
| | - Grant A Fox
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, USA
- Department of Neuroscience, College of Medicine, University of Kentucky, Lexington, KY, USA
| | - Mark E Wadsworth
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, USA
| | - Patricia H Doyle
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, USA
- Department of Neuroscience, College of Medicine, University of Kentucky, Lexington, KY, USA
| | - Brittney A Williams
- Department of Pharmacology and Nutritional Sciences, College of Medicine, University of Kentucky, Lexington, KY, USA
| | - Edward J Fox
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, USA
| | | | - Mina Ryten
- UK Dementia Research Institute at The University of Cambridge, Cambridge, UK
- Department of Clinical Neurosciences, School of Clinical Medicine, University of Cambridge, Cambridge, UK
- Department of Genetics and Genomic Medicine, Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Sara Goodwin
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
| | - Elena Ghiban
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
| | - Robert Wappel
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
| | | | - Justin B Miller
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, USA
- Division of Biomedical Informatics, Internal Medicine, College of Medicine, University of Kentucky, Lexington, KY, USA
- Department of Pathology and Laboratory Medicine, University of Kentucky, Lexington, KY, USA
- Microbiology, Immunology and Molecular Genetics, College of Medicine, University of Kentucky, Lexington, KY, USA
| | - Nicholas T Seyfried
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, USA
| | - Peter T Nelson
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, USA
| | - John D Fryer
- Department of Neuroscience, Mayo Clinic, Scottsdale, AZ, USA
| | - Mark T W Ebbert
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, USA.
- Department of Neuroscience, College of Medicine, University of Kentucky, Lexington, KY, USA.
- Division of Biomedical Informatics, Internal Medicine, College of Medicine, University of Kentucky, Lexington, KY, USA.
| |
Collapse
|
11
|
Kim S, Woo Y, Um D, Chun I, Noh SJ, Ji HA, Jung N, Goo BS, Yoo JY, Mun DJ, Nghi TD, Nhung TTM, Han SH, Lee SB, Lee W, Yun J, So KH, Kim DK, Jang H, Suh Y, Rah JC, Baek ST, Yoon KJ, Kim MS, Kim TK, Park SK. Perturbed cell fate decision by schizophrenia-associated AS3MT d2d3 isoform during corticogenesis. SCIENCE ADVANCES 2025; 11:eadp8271. [PMID: 40153497 PMCID: PMC11952104 DOI: 10.1126/sciadv.adp8271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/13/2024] [Accepted: 02/24/2025] [Indexed: 03/30/2025]
Abstract
The neurodevelopmental theory of schizophrenia emphasizes early brain development in its etiology. Genome-wide association studies have linked schizophrenia to genetic variations of AS3MT (arsenite methyltransferase) gene, particularly the increased expression of AS3MTd2d3 isoform. To investigate the biological basis of this association with schizophrenia pathophysiology, we established a transgenic mouse model (AS3MTd2d3-Tg) ectopically expressing AS3MTd2d3 at the cortical neural stem cells. AS3MTd2d3-Tg mice exhibited enlarged ventricles and deficits in sensorimotor gating and sociability. Single-cell and single-nucleus RNA sequencing analyses of AS3MTd2d3-Tg brains revealed cell fate imbalances and altered excitatory neuron composition. AS3MTd2d3 localized to centrosome, disrupting mitotic spindle orientation and differentiation in developing neocortex and organoids, in part through NPM1 (Nucleophosmin 1). The structural analysis identified that hydrophobic residues exposed in AS3MTd2d3 are critical for its pathogenic function. Therefore, our findings may help to explain the early pathological features of schizophrenia.
Collapse
Affiliation(s)
- Seunghyun Kim
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 37673, Republic of Korea
| | - Youngsik Woo
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 37673, Republic of Korea
| | - Dahun Um
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 37673, Republic of Korea
| | - Inseop Chun
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 37673, Republic of Korea
| | - Su-Jin Noh
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 37673, Republic of Korea
| | - Hyeon Ah Ji
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 37673, Republic of Korea
| | - Namyoung Jung
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 37673, Republic of Korea
| | - Bon Seong Goo
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 37673, Republic of Korea
| | - Jin Yeong Yoo
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 37673, Republic of Korea
| | - Dong Jin Mun
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 37673, Republic of Korea
| | - Tran Diem Nghi
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 37673, Republic of Korea
| | - Truong Thi My Nhung
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 37673, Republic of Korea
| | - Seung Hyeon Han
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 37673, Republic of Korea
| | - Su Been Lee
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 37673, Republic of Korea
| | - Wonhyeok Lee
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 37673, Republic of Korea
| | - Jonghyeok Yun
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 37673, Republic of Korea
| | - Ki Hurn So
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 37673, Republic of Korea
| | - Dae-Kyum Kim
- Division of Thoracic and Upper Gastrointestinal Surgery, Department of Surgery, Faculty of Medicine and Health Sciences, McGill University, Montreal, Quebec H3G 1A4, Canada
- Cancer Research Program, Research Institute of McGill University Health Centre, Montreal, Quebec H4A 3J1, Canada
| | - Hyunsoo Jang
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejon 34141, Republic of Korea
| | - Yeongjun Suh
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 37673, Republic of Korea
| | - Jong-Cheol Rah
- Korea Brain Research Institute, Daegu 41062, Republic of Korea
| | - Seung Tae Baek
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 37673, Republic of Korea
- Institute for Convergence Research and Education in Advanced Technology, Yonsei University, Seoul 03772, Republic of Korea
| | - Ki-Jun Yoon
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejon 34141, Republic of Korea
| | - Min-Sung Kim
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 37673, Republic of Korea
- Institute for Convergence Research and Education in Advanced Technology, Yonsei University, Seoul 03772, Republic of Korea
| | - Tae-Kyung Kim
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 37673, Republic of Korea
- Institute for Convergence Research and Education in Advanced Technology, Yonsei University, Seoul 03772, Republic of Korea
| | - Sang Ki Park
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 37673, Republic of Korea
- Institute for Convergence Research and Education in Advanced Technology, Yonsei University, Seoul 03772, Republic of Korea
| |
Collapse
|
12
|
Auxillos J, Stigliani A, Vaagensø C, Garland W, Niazi A, Valen E, Jensen T, Sandelin A. True length of diverse capped RNA sequencing (TLDR-seq): 5'-3'-end sequencing of capped RNAs regardless of 3'-end status. Nucleic Acids Res 2025; 53:gkaf240. [PMID: 40183637 PMCID: PMC11969664 DOI: 10.1093/nar/gkaf240] [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: 10/22/2024] [Revised: 02/20/2025] [Accepted: 03/14/2025] [Indexed: 04/05/2025] Open
Abstract
Analysis of transcript function is greatly aided by knowledge of the full-length RNA sequence. New long-read sequencing enabled by Oxford Nanopore and PacBio devices have the potential to provide full-length transcript information; however, standard methods still lack the ability to capture true RNA 5' ends and select for polyadenylated (pA+) transcripts only. Here, we present a method that, by utilizing cap trapping and 3'-end adapter ligation, sequences transcripts between their exact 5' and 3' ends regardless of polyadenylation status and without the need for ribosomal RNA depletion, with the ability to characterize polyadenylation length of RNAs, if any. The method shows high reproducibility, can faithfully detect 5' ends, 3' ends and splice junctions, and produces gene-expression estimates that are highly correlated to those of short-read sequencing techniques. We also demonstrate that the method can detect and sequence full-length nonadenylated (pA-) RNAs, including long noncoding RNAs, promoter upstream transcripts, and enhancer RNAs, and present cases where pA+ and pA- RNAs show preferences for different but closely located transcription start sites. Our method is therefore useful for the characterization of diverse capped RNA species and analysis of relationships between transcription initiation, termination, and RNA processing.
Collapse
Affiliation(s)
- Jamie Auxillos
- Section for Computational and RNA Biology, Department of Biology, University of Copenhagen, DK2200 Copenhagen, Denmark
- Biotech Research and Innovation Centre, University of Copenhagen, DK2200 Copenhagen, Denmark
| | - Arnaud Stigliani
- Section for Computational and RNA Biology, Department of Biology, University of Copenhagen, DK2200 Copenhagen, Denmark
- Biotech Research and Innovation Centre, University of Copenhagen, DK2200 Copenhagen, Denmark
| | - Christian Skov Vaagensø
- Section for Computational and RNA Biology, Department of Biology, University of Copenhagen, DK2200 Copenhagen, Denmark
- Biotech Research and Innovation Centre, University of Copenhagen, DK2200 Copenhagen, Denmark
| | - William Garland
- Department of Molecular Biology and Genetics, Aarhus University, DK8000 Aarhus, Denmark
| | - Adnan Muhammed Niazi
- Computational Biology Unit, Department of Informatics, University of Bergen, N-5008 Bergen, Norway
| | - Eivind Valen
- Computational Biology Unit, Department of Informatics, University of Bergen, N-5008 Bergen, Norway
- Department of Biosciences, University of Oslo, N-0371 Oslo, Norway
| | - Torben Heick Jensen
- Department of Molecular Biology and Genetics, Aarhus University, DK8000 Aarhus, Denmark
| | - Albin Sandelin
- Section for Computational and RNA Biology, Department of Biology, University of Copenhagen, DK2200 Copenhagen, Denmark
- Biotech Research and Innovation Centre, University of Copenhagen, DK2200 Copenhagen, Denmark
| |
Collapse
|
13
|
Even-Ros D, Huertas-Romero J, Marín-Menguiano M, Nusspaumer G, Borge M, Irimia M, Zurita F, González-Reyes A. Drosophila ovarian stem cell niche ageing involves coordinated changes in transcription and alternative splicing. Nat Commun 2025; 16:2596. [PMID: 40091053 PMCID: PMC11911433 DOI: 10.1038/s41467-025-57901-8] [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: 05/17/2024] [Accepted: 03/06/2025] [Indexed: 03/19/2025] Open
Abstract
Gene expression (GE) and alternative splicing (AS) contribute to the formation of new interaction networks with potentially significant cellular functions. Here, we investigate ageing in the Drosophila female germline stem cell (GSC) niche and describe functional changes in both GE and AS. The GSC niche comprises three types of support cells, whose ageing transcriptomes reveal differential GE and AS variations related to cell adhesion, cytoskeleton and neural signalling. Because each population show distinctive GE and AS changes, niche cell types possess unique ageing signatures. Depending on the cell population, groups of genes display changes in both GE and AS, revealing a coordinated regulation of transcription and splicing during niche ageing. One such gene is Fasciclin 2, a neural adhesion molecule that we find is essential for niche functioning. Furthermore, genes involved in AS undergo changes in GE and/or AS themselves, providing a mechanistic explanation for the coordination of these two processes during niche ageing. This is the case of the splicing factor Smu1, described here as a key element necessary for ovarian niche homeostasis.
Collapse
Affiliation(s)
- Dilamm Even-Ros
- Centro Andaluz de Biología del Desarrollo (CABD), CSIC-Junta de Andalucía-UPO, Carretera de Utrera km 1, 41013, Seville, Spain
| | - Judit Huertas-Romero
- Centro Andaluz de Biología del Desarrollo (CABD), CSIC-Junta de Andalucía-UPO, Carretera de Utrera km 1, 41013, Seville, Spain
| | - Miriam Marín-Menguiano
- Centro Andaluz de Biología del Desarrollo (CABD), CSIC-Junta de Andalucía-UPO, Carretera de Utrera km 1, 41013, Seville, Spain
| | - Gretel Nusspaumer
- Centro Andaluz de Biología del Desarrollo (CABD), CSIC-Junta de Andalucía-UPO, Carretera de Utrera km 1, 41013, Seville, Spain
| | - Miguel Borge
- Centre for Genomic Regulation, Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Manuel Irimia
- Centre for Genomic Regulation, Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
- ICREA, Barcelona, Spain
| | - Federico Zurita
- Departamento de Genética e Instituto de Biotecnología, Universidad de Granada, Centro de Investigación Biomédica, 18071, Granada, Spain.
| | - Acaimo González-Reyes
- Centro Andaluz de Biología del Desarrollo (CABD), CSIC-Junta de Andalucía-UPO, Carretera de Utrera km 1, 41013, Seville, Spain.
| |
Collapse
|
14
|
Chi Z, Gupta V, Query C. U2-2 snRNA Mutations Alter the Transcriptome. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.12.642709. [PMID: 40161847 PMCID: PMC11952416 DOI: 10.1101/2025.03.12.642709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Intron removal from pre-mRNA is catalyzed by the spliceosome, which comprises 5 snRNPs containing small nuclear RNAs (snRNAs). U2 snRNA makes critical RNA-RNA and RNA-protein contacts throughout the splicing cycle. Mutations in U2 snRNA, particularly at position C28, have been linked to cancers. To study gene expression changes mediated by mutated U2 snRNAs, U2-2 C28 mutants, U2-2 knockout (KO), and U2-2 overexpression (OE) cell lines were constructed followed by RNA sequencing. We observed significant changes in splicing and over 4,000 differentially expressed genes enriched in pathways like RNA processing and non-coding RNAs upon knocking out U2-2 snRNA. Splicing patterns were more influenced by U2-2 dosage than mutations alone. Therefore, the mutant exhibits a compound phenotype, resulting from reduced U2-2 levels (and thus mostly phenocopying the KO) and additional mutant-specific splicing changes. HIGHLIGHTS U2-2 snRNA BSL mutants alter splicing and the transcriptomeU2-2 KO phenocopies most altered splice events in the mutantsBoth U2-2 levels and mutations alter splicingMany altered splice events lead to NMD.
Collapse
|
15
|
Luo S, Peng H, Shi Y, Cai J, Zhang S, Shao N, Li J. Integration of proteomics profiling data to facilitate discovery of cancer neoantigens: a survey. Brief Bioinform 2025; 26:bbaf087. [PMID: 40052441 PMCID: PMC11886573 DOI: 10.1093/bib/bbaf087] [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: 10/17/2024] [Revised: 12/29/2024] [Accepted: 02/19/2025] [Indexed: 03/10/2025] Open
Abstract
Cancer neoantigens are peptides that originate from alterations in the genome, transcriptome, or proteome. These peptides can elicit cancer-specific T-cell recognition, making them potential candidates for cancer vaccines. The rapid advancement of proteomics technology holds tremendous potential for identifying these neoantigens. Here, we provided an up-to-date survey about database-based search methods and de novo peptide sequencing approaches in proteomics, and we also compared these methods to recommend reliable analytical tools for neoantigen identification. Unlike previous surveys on mass spectrometry-based neoantigen discovery, this survey summarizes the key advancements in de novo peptide sequencing approaches that utilize artificial intelligence. From a comparative study on a dataset of the HepG2 cell line and nine mixed hepatocellular carcinoma proteomics samples, we demonstrated the potential of proteomics for the identification of cancer neoantigens and conducted comparisons of the existing methods to illustrate their limits. Understanding these limits, we suggested a novel workflow for neoantigen discovery as perspectives.
Collapse
Affiliation(s)
- Shifu Luo
- Faculty of Computer Science and Control Engineering, Shenzhen University of Advanced Technology, Shenzhen, 518107, Guangdong, China
- Faculty of Health Sciences, University of Macau, Taipa, Macao SAR 999078, China
| | - Hui Peng
- Faculty of Computer Science and Control Engineering, Shenzhen University of Advanced Technology, Shenzhen, 518107, Guangdong, China
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore
| | - Ying Shi
- Faculty of Computer Science and Control Engineering, Shenzhen University of Advanced Technology, Shenzhen, 518107, Guangdong, China
- School of Computer and Information Technology, Shanxi University, Taiyuan, 030006, Shanxi, China
| | - Jiaxin Cai
- Faculty of Computer Science and Control Engineering, Shenzhen University of Advanced Technology, Shenzhen, 518107, Guangdong, China
| | - Songming Zhang
- Faculty of Computer Science and Control Engineering, Shenzhen University of Advanced Technology, Shenzhen, 518107, Guangdong, China
| | - Ningyi Shao
- Faculty of Health Sciences, University of Macau, Taipa, Macao SAR 999078, China
| | - Jinyan Li
- Faculty of Computer Science and Control Engineering, Shenzhen University of Advanced Technology, Shenzhen, 518107, Guangdong, China
| |
Collapse
|
16
|
Wu H, Zhou Y, Wang X, Tang C, Yang F, Xu K, Ren T. Systematic exploration of prognostic alternative splicing events related to tumor immune microenvironment of Clear Cell Renal Cell Carcinoma. Cancer Biomark 2025; 42:18758592251317402. [PMID: 40171812 DOI: 10.1177/18758592251317402] [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: 04/04/2025]
Abstract
BackgroundPathologically, clear cell renal cell carcinoma (ccRCC) is the most common type of renal carcinoma, with high heterogeneity and poor prognosis. There is increasing evidence that alternative splicing (AS) is involved in tumor evolution and tumor immune microenvironment (TIME). However, studies on the exploration of AS events and TIME in ccRCC are still few but needed.MethodsThe transcriptional data and clinicopathological information of patients with ccRCC in The Cancer Genome Atlas (TCGA) database were extracted completely. Patients were grouped according to the ESTIMATE algorithm and differentially expressed AS events (DEASs) were identified. The relationship between AS events and features of TIME were investigated by functional enrichment analysis and unsupervised consensus analysis. Finally, hub splicing factors (SFs) was identified by the regulatory network of survival-related AS events and intersection SFs, and its biological function was further verified in vitro.ResultsIn total, the data of 515 patients with ccRCC were extracted and analyzed. Patients with low immune-score presented longer overall survival (OS) than high immune-score. 861 AS events were identified as DEASs, and they were enriched in immune-related pathways. 3 AS-based clusters were identified and found to have different prognoses and unique immune features. Finally, MBNL1 was identified as a hub SF, and it was shown to inhibit proliferation and metastasis, promote apoptosis, and block cells in G2/M phase in 786O and A498 cells. Mechanistically, MBNL1 regulates QKI expression through AS.ConclusionThe prognostic risk model constructed base on immune-related AS events has good predictive ability for ccRCC. The hub SF MBNL1 identied in the present study could inhibit the progression of ccRCC. This effect is likely due to the regulation of QKI expression through AS.
Collapse
Affiliation(s)
- Hongwei Wu
- Clinical Medical College, Chengdu Medical College, Chengdu, China
- Department of Oncology, The First Affiliated Hospital of Chengdu Medical College, Chengdu, China
| | - Yuchuan Zhou
- Clinical Medical College, Chengdu Medical College, Chengdu, China
- Department of Oncology, The First Affiliated Hospital of Chengdu Medical College, Chengdu, China
- Department of Oncology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Xi Wang
- Department of Ultrasound, the General Hospital of Western Theater Command, Chengdu, China
| | - Chunhan Tang
- Clinical Medical College, Chengdu Medical College, Chengdu, China
- Department of Oncology, The First Affiliated Hospital of Chengdu Medical College, Chengdu, China
| | - Fang Yang
- Clinical Medical College, Chengdu Medical College, Chengdu, China
- Department of Oncology, The First Affiliated Hospital of Chengdu Medical College, Chengdu, China
| | - Ke Xu
- Clinical Medical College, Chengdu Medical College, Chengdu, China
- Department of Oncology, The First Affiliated Hospital of Chengdu Medical College, Chengdu, China
| | - Tao Ren
- Clinical Medical College, Chengdu Medical College, Chengdu, China
- Department of Oncology, The First Affiliated Hospital of Chengdu Medical College, Chengdu, China
| |
Collapse
|
17
|
van Oostrum M, Schuman EM. Understanding the molecular diversity of synapses. Nat Rev Neurosci 2025; 26:65-81. [PMID: 39638892 DOI: 10.1038/s41583-024-00888-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/08/2024] [Indexed: 12/07/2024]
Abstract
Synapses are composed of thousands of proteins, providing the potential for extensive molecular diversity to shape synapse type-specific functional specializations. In this Review, we explore the landscape of synaptic diversity and describe the mechanisms that expand the molecular complexity of synapses, from the genotype to the regulation of gene expression to the production of specific proteoforms and the formation of localized protein complexes. We emphasize the importance of examining every molecular layer and adopting a systems perspective to understand how these interconnected mechanisms shape the diverse functional and structural properties of synapses. We explore current frameworks for classifying synapses and methodologies for investigating different synapse types at varying scales, from synapse-type-specific proteomics to advanced imaging techniques with single-synapse resolution. We highlight the potential of synapse-type-specific approaches for integrating molecular data with cellular functions, circuit organization and organismal phenotypes to enable a more holistic exploration of neuronal phenomena across different scales.
Collapse
Affiliation(s)
- Marc van Oostrum
- Max Planck Institute for Brain Research, Frankfurt am Main, Germany
- Biozentrum, University of Basel, Basel, Switzerland
| | - Erin M Schuman
- Max Planck Institute for Brain Research, Frankfurt am Main, Germany.
| |
Collapse
|
18
|
Laine E, Freiberger MI. Toward a comprehensive profiling of alternative splicing proteoform structures, interactions and functions. Curr Opin Struct Biol 2025; 90:102979. [PMID: 39778413 PMCID: PMC7617313 DOI: 10.1016/j.sbi.2024.102979] [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: 09/15/2024] [Revised: 11/26/2024] [Accepted: 12/18/2024] [Indexed: 01/11/2025]
Abstract
The mRNA splicing machinery has been estimated to generate 100,000 known protein-coding transcripts for 20,000 human genes (Ensembl, Sept. 2024). However, this set is expanding with the massive and rapidly growing data coming from high-throughput technologies, particularly single-cell and long-read sequencing. Yet, the implications of splicing complexity at the protein level remain largely uncharted. In this review, we describe the current advances toward systematically assessing the contribution of alternative splicing to proteome function diversification. We discuss the potential and challenges of using artificial intelligence-based techniques in identifying alternative splicing proteoforms and characterising their structures, interactions, and functions.
Collapse
Affiliation(s)
- Elodie Laine
- Sorbonne Université, CNRS, IBPS, Laboratory of Computational and Quantitative Biology (LCQB), UMR 7238, 75005 Paris, France; Institut universitaire de France (IUF), France.
| | - Maria Inés Freiberger
- Sorbonne Université, CNRS, IBPS, Laboratory of Computational and Quantitative Biology (LCQB), UMR 7238, 75005 Paris, France
| |
Collapse
|
19
|
Meißgeier T, Kappelmann‐Fenzl M, Staebler S, Ahari AJ, Mertes C, Gagneur J, Linck‐Paulus L, Bosserhoff AK. Splicing control by PHF5A is crucial for melanoma cell survival. Cell Prolif 2025; 58:e13741. [PMID: 39212334 PMCID: PMC11839196 DOI: 10.1111/cpr.13741] [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: 03/08/2024] [Revised: 08/02/2024] [Accepted: 08/19/2024] [Indexed: 09/04/2024] Open
Abstract
Abnormalities in alternative splicing are a hallmark of cancer formation. In this study, we investigated the role of the splicing factor PHD finger protein 5A (PHF5A) in melanoma. Malignant melanoma is the deadliest form of skin cancer, and patients with a high PHF5A expression show poor overall survival. Our data revealed that an siRNA-mediated downregulation of PHF5A in different melanoma cell lines leads to massive splicing defects of different tumour-relevant genes. The loss of PHF5A results in an increased rate of apoptosis by triggering Fas- and unfolded protein response (UPR)-mediated apoptosis pathways in melanoma cells. These findings are tumour-specific because we did not observe this regulation in fibroblasts. Our study identifies a crucial role of PHF5A as driver for melanoma malignancy and the described underlying splicing network provides an interesting basis for the development of new therapeutic targets for this aggressive form of skin cancer.
Collapse
Affiliation(s)
- Tina Meißgeier
- Institute of BiochemistryFriedrich‐Alexander‐University Erlangen‐Nürnberg (FAU)ErlangenGermany
| | - Melanie Kappelmann‐Fenzl
- Institute of BiochemistryFriedrich‐Alexander‐University Erlangen‐Nürnberg (FAU)ErlangenGermany
- Faculty of Computer ScienceDeggendorf Institute of TechnologyDeggendorfGermany
| | - Sebastian Staebler
- Institute of BiochemistryFriedrich‐Alexander‐University Erlangen‐Nürnberg (FAU)ErlangenGermany
| | - Ata Jadid Ahari
- School of Computation, Information and TechnologyTechnical University of MunichGarchingGermany
| | - Christian Mertes
- School of Computation, Information and TechnologyTechnical University of MunichGarchingGermany
| | - Julien Gagneur
- School of Computation, Information and TechnologyTechnical University of MunichGarchingGermany
| | - Lisa Linck‐Paulus
- Institute of BiochemistryFriedrich‐Alexander‐University Erlangen‐Nürnberg (FAU)ErlangenGermany
| | - Anja Katrin Bosserhoff
- Institute of BiochemistryFriedrich‐Alexander‐University Erlangen‐Nürnberg (FAU)ErlangenGermany
| |
Collapse
|
20
|
Pauper M, Hentschel A, Tiburcy M, Beltran S, Ruck T, Schara-Schmidt U, Roos A. Proteomic Profiling Towards a Better Understanding of Genetic Based Muscular Diseases: The Current Picture and a Look to the Future. Biomolecules 2025; 15:130. [PMID: 39858524 PMCID: PMC11763865 DOI: 10.3390/biom15010130] [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: 11/10/2024] [Revised: 12/25/2024] [Accepted: 01/07/2025] [Indexed: 01/27/2025] Open
Abstract
Proteomics accelerates diagnosis and research of muscular diseases by enabling the robust analysis of proteins relevant for the manifestation of neuromuscular diseases in the following aspects: (i) evaluation of the effect of genetic variants on the corresponding protein, (ii) prediction of the underlying genetic defect based on the proteomic signature of muscle biopsies, (iii) analysis of pathophysiologies underlying different entities of muscular diseases, key for the definition of new intervention concepts, and (iv) patient stratification according to biochemical fingerprints as well as (v) monitoring the success of therapeutic interventions. This review presents-also through exemplary case studies-the various advantages of mass proteomics in the investigation of genetic muscle diseases, discusses technical limitations, and provides an outlook on possible future application concepts. Hence, proteomics is an excellent large-scale analytical tool for the diagnostic workup of (hereditary) muscle diseases and warrants systematic profiling of underlying pathophysiological processes. The steady development may allow to overcome existing limitations including a quenched dynamic range and quantification of different protein isoforms. Future directions may include targeted proteomics in diagnostic settings using not only muscle biopsies but also liquid biopsies to address the need for minimally invasive procedures.
Collapse
Affiliation(s)
- Marc Pauper
- Centro Nacional de Análisis Genómico (CNAG), Baldiri Reixac 4, 08028 Barcelona, Spain; (M.P.); (S.B.)
- Departament de Genètica, Microbiologia i Estadística, Facultat de Biologia, Universitat de Barcelona (UB), 08028 Barcelona, Spain
| | - Andreas Hentschel
- Leibniz-Institut für Analytische Wissenschaften-ISAS-e.V., 44227 Dortmund, Germany;
| | - Malte Tiburcy
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Georg August University, 37075 Göttingen, Germany;
- ZHK (German Centre for Cardiovascular Research), Partner Site Göttingen, 37075 Göttingen, Germany
| | - Sergi Beltran
- Centro Nacional de Análisis Genómico (CNAG), Baldiri Reixac 4, 08028 Barcelona, Spain; (M.P.); (S.B.)
- Departament de Genètica, Microbiologia i Estadística, Facultat de Biologia, Universitat de Barcelona (UB), 08028 Barcelona, Spain
- Universitat Pompeu Fabra (UPF), 08002 Barcelona, Spain
| | - Tobias Ruck
- Department of Neurology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University, 40225 Düsseldorf, Germany;
- Department of Neurology, BG-University Hospital Bergmannsheil, Ruhr University Bochum, 44789 Bochum, Germany
- Heimer Institute for Muscle Research, BG-University Hospital Bergmannsheil, 44789 Bochum, Germany
| | - Ulrike Schara-Schmidt
- Department of Pediatric Neurology, Centre for Neuromuscular Disorders, University Duisburg-Essen, 45147 Essen, Germany;
| | - Andreas Roos
- Department of Neurology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University, 40225 Düsseldorf, Germany;
- Department of Pediatric Neurology, Centre for Neuromuscular Disorders, University Duisburg-Essen, 45147 Essen, Germany;
- Brain and Mind Research Institute, Children’s Hospital of Eastern Ontario Research Institute, Ottawa, ON K1H 8L1, Canada
| |
Collapse
|
21
|
Kerr C, Schneider OL, Tichy S, Huge BJ, Champion MM. Capillary Isoelectric Focusing of Proteins and Peptides Using an In-Line cIEF-ESI Interface with Improved MS Characteristics. Anal Chem 2025; 97:649-657. [PMID: 39742429 PMCID: PMC11740900 DOI: 10.1021/acs.analchem.4c05010] [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: 09/17/2024] [Revised: 12/11/2024] [Accepted: 12/17/2024] [Indexed: 01/03/2025]
Abstract
Intact protein analysis using mass spectrometry (MS) is an important technique to characterize and provide a comprehensive overview of protein complexity. It is also the basis of "top-down" approaches in proteomics to describe the proteoforms of single protein's post-translational modifications (PTMs). MS-based analysis of intact proteins benefits from high-resolution separations prior to electrospray ionization. Capillary isoelectric focusing (cIEF) is a high-resolution separation for proteins and peptides which is capable of separating proteoforms. MS detection coupled to cIEF can separate, detect, and characterize proteoforms at the molecular level. However, cIEF with MS detection is a compromised process. The concentration of ampholytes required for cIEF is mutually exclusive with mass spectrometer contamination. We have improved an online cIEF-ESI-MS interface to reduce (desalt) amino acid ampholytes in-line after cIEF and prior to electrospray ionization. In proof of principle experiments, >90% increase in area under the curve of the electropherograms was observed with the interface compared to without the interface. Protein standards including proteoforms of cytochrome C, myoglobin, and α-casein were separated and resolved with high reproducibility. The interface did not compromise the linearity of the cIEF pH gradient separations, achieving a high linearity with a R2 of 0.99. In addition, a tryptic digest of BSA demonstrates baseline resolution of peptides with as little as 0.02 pI unit difference and a full width at half-maximum average of 7.1 s.
Collapse
Affiliation(s)
- Caitlin
M. Kerr
- Department
of Chemistry and Biochemistry, University
of Notre Dame, Notre
Dame, Indiana 46556, United States
- Berthiaume
Institute for Precision Health, University
of Notre Dame, Notre Dame, Indiana 46556, United States
| | - Olivia L. Schneider
- Department
of Chemistry and Biochemistry, University
of Notre Dame, Notre
Dame, Indiana 46556, United States
| | - Shane Tichy
- Agilent
Technologies, 5301 Stevens Creek Blvd., Santa Clara, California 95015, United States
- Center
for Bioanalytic Metrology, University of
Notre Dame, Notre Dame, Indiana 46556, United States
| | - Bonnie Jaskowski Huge
- Department
of Chemistry and Biochemistry, University
of Notre Dame, Notre
Dame, Indiana 46556, United States
| | - Matthew M. Champion
- Department
of Chemistry and Biochemistry, University
of Notre Dame, Notre
Dame, Indiana 46556, United States
- Berthiaume
Institute for Precision Health, University
of Notre Dame, Notre Dame, Indiana 46556, United States
- Center
for Bioanalytic Metrology, University of
Notre Dame, Notre Dame, Indiana 46556, United States
| |
Collapse
|
22
|
Zhang Y, Liu K, Xu Z, Li B, Wu X, Fan R, Yao X, Wu H, Duan C, Gong Y, Chen K, Zeng J, Li L, Xu H. OncoSplicing 3.0: an updated database for identifying RBPs regulating alternative splicing events in cancers. Nucleic Acids Res 2025; 53:D1460-D1466. [PMID: 39558172 PMCID: PMC11701682 DOI: 10.1093/nar/gkae1098] [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: 08/31/2024] [Revised: 10/06/2024] [Accepted: 11/04/2024] [Indexed: 11/20/2024] Open
Abstract
Alternative splicing (AS) is a crucial mechanism to regulate gene expression and protein complexity. RNA-binding proteins (RBPs) play an important role in regulating abnormal alternative splicing in cancers. However, few resources are available to identify specific RBPs responsible for regulating individual AS event. We have developed the OncoSplicing database for integrative analysis of clinically relevant alternative splicing events in TCGA cancers. Here, we further updated the OncoSplicing database by performing correlation analysis between the splicing and mRNA expression data from the TCGA cancers or GTEx tissues, mapping known RNA-binding motifs and eCLIP-seq peaks to all AS events, conducting splicing analysis for RNA-seq data from RBP perturbation experiments in the ENCODE project, and integrating exon and intron sequences for each AS event. With this updated database, users can easily identify potential RBPs responsible for the queried AS event and obtain sequences to design AS-specific primers and minigene constructs for experiment validation. Overall, compared to the previous version, the substantially updated OncoSplicing database (www.oncosplicing.com) offers a more valuable resource for users to identify RBPs responsible for regulating alternative splicing events in cancers.
Collapse
Affiliation(s)
- Yangjun Zhang
- Cancer Precision Diagnosis and Treatment and Translational Medicine Hubei Engineering Research Center, Zhongnan Hospital, Wuhan University, Wuhan 430000, China
- Department of Biological Repositories, Zhongnan Hospital, Wuhan University, Wuhan 430000, China
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan 430000, China
- Department of Urology, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230000, China
| | - Kai Liu
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan 430000, China
| | - Zhenzhen Xu
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan 430000, China
| | - Bo Li
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan 430000, China
| | - Xiaoliang Wu
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China
| | - Ruixin Fan
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan 430000, China
| | - Xiangyang Yao
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan 430000, China
| | - Huahui Wu
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan 430000, China
| | - Chen Duan
- Cancer Precision Diagnosis and Treatment and Translational Medicine Hubei Engineering Research Center, Zhongnan Hospital, Wuhan University, Wuhan 430000, China
| | - Yan Gong
- Cancer Precision Diagnosis and Treatment and Translational Medicine Hubei Engineering Research Center, Zhongnan Hospital, Wuhan University, Wuhan 430000, China
| | - Ke Chen
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China
| | - Jin Zeng
- Department of Urology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330000, China
| | - Lu Li
- Department of Radiation Oncology, Sichuan Cancer Hospital & Institute, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610041, China
| | - Hua Xu
- Cancer Precision Diagnosis and Treatment and Translational Medicine Hubei Engineering Research Center, Zhongnan Hospital, Wuhan University, Wuhan 430000, China
- Department of Biological Repositories, Zhongnan Hospital, Wuhan University, Wuhan 430000, China
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan 430000, China
- Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan 430000, China
| |
Collapse
|
23
|
Saei AA, Sun L, Mahmoudi M. The role of protein corona in advancing plasma proteomics. Proteomics 2025; 25:e2400028. [PMID: 39221533 PMCID: PMC11735278 DOI: 10.1002/pmic.202400028] [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: 07/10/2024] [Revised: 08/19/2024] [Accepted: 08/21/2024] [Indexed: 09/04/2024]
Abstract
The protein corona, a layer of biomolecules forming around nanoparticles in biological environments, critically influences nanoparticle interactions with biosystems, affecting pharmacokinetics and biological outcomes. Initially, the protein corona presented challenges for nanomedicine and nanotoxicology, such as nutrient depletion in cell cultures and masking of nanoparticle-targeting species. However, recent advancements have highlighted its potential in environmental toxicity, proteomics, and immunology. This viewpoint focuses on leveraging the protein corona to enhance the depth of plasma proteome analysis, addressing challenges posed by the high dynamic range of protein concentrations in plasma. The protein corona simplifies sample preparation, enriches low-abundance proteins, and improves proteome coverage. Innovations include using diverse nanoparticles and spiking small molecules to increase the number of quantified proteins. Reproducibility issues across core facilities necessitate standardized protocols. Moreover, top-down proteomics enables proteoform-specific measurements, providing deeper insights into protein corona composition. Future research should aim at improving top-down proteomics techniques and integrating protein corona studies and proteomics for personalized medicine and advanced diagnostics.
Collapse
Affiliation(s)
- Amir Ata Saei
- Center for Translational Microbiome ResearchDepartment of Microbiology, Tumor and Cell BiologyKarolinska InstitutetStockholmSweden
| | - Liangliang Sun
- Department of ChemistryMichigan State UniversityEast LansingMichiganUSA
| | - Morteza Mahmoudi
- Department of Radiology and Precision Health ProgramMichigan State UniversityEast LansingMichiganUSA
| |
Collapse
|
24
|
Shimpi AA, Naegle KM. Linguistic networks uncover grammatical constraints of protein sentences comprised of domain-based words. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.04.626803. [PMID: 39677636 PMCID: PMC11643033 DOI: 10.1101/2024.12.04.626803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
Evolution has developed a set of principles that determine feasible domain combinations analogous to grammar within natural languages. Treating domains as words and proteins as sentences, made up of words, we apply a linguistic approach to represent the human proteome as an n-gram network. Combining this with network theory and application, we explore the functional language and rules of the human proteome. Additionally, we explored subnetwork languages by focusing on reversible post-translational modifications (PTMs) systems that follow a reader-writer-eraser paradigm. We find that PTM systems appear to sample grammar rules near the onset of the system expansion, but then convergently evolve towards similar grammar rules, which stabilize during the post-metazoan switch. For example, reader and writer domains are typically tightly connected through shared n-grams, but eraser domains are almost always loosely or completely disconnected from readers and writers. Additionally, after grammar fixation, domains with verb-like properties, such as writers and erasers, never appear - consistent with the idea of natural grammar that leads to clarity and limits futile enzymatic cycles. Then, given how some cancer fusion genes represent the possibility for the emergence of novel language, we investigate how cancer fusion genes alter the human proteome n-gram network. We find most cancer fusion genes follow existing grammar rules. Collectively, these results suggest that n-gram based analysis of proteomes is a complement to the more direct protein-protein interaction networks. N-grams can capture abstract functional connections in a more fully described manner, limited only by the definition of domains within the proteome and not by the combinatorial challenge of capturing all protein interaction connections.
Collapse
Affiliation(s)
- Adrian A. Shimpi
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, 22903
- Department of Genome Sciences, University of Virginia, Charlottesville, VA, 22903
| | - Kristen M. Naegle
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, 22903
- Department of Genome Sciences, University of Virginia, Charlottesville, VA, 22903
| |
Collapse
|
25
|
Sun Y, Pang Y, Wu X, Zhu R, Wang L, Tian M, He X, Liu D, Yang X. Landscape of alternative splicing and polyadenylation during growth and development of muscles in pigs. Commun Biol 2024; 7:1607. [PMID: 39627472 PMCID: PMC11614907 DOI: 10.1038/s42003-024-07332-w] [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: 04/24/2024] [Accepted: 11/28/2024] [Indexed: 12/06/2024] Open
Abstract
Alternative polyadenylation (APA) is emerging as a post-transcriptional regulatory mechanism, similar as that of alternative splicing (AS), and plays a prominent role in regulating gene expression and increasing the complexity of the transcriptome and proteome. We use polyadenylation selected long-read isoform sequencing to obtain full-length transcript sequences in porcine muscles at five developmental stages. We identify numerous novel transcripts unannotated in the existing pig genome, including transcripts mapping to known and unknown gene loci, and widespread transcript diversity in porcine muscles. The top 100 most isoformic genes are mainly enriched in Gene Ontology terms related to muscle growth and development. It is revealed that intron retention/exon inclusion and the usage of distal polyadenylation site (PAS) are associated with ageing through analyzing changes of AS and PAS during muscle development. We also identify developmental changes in major transcripts and major PASs. Furthermore, genes/transcripts important for muscle development are identified. The results confirm the importance of AS and APA in pig muscles, substantially increasing transcriptional diversity and showing an important mechanism underlying gene regulation in muscles.
Collapse
Affiliation(s)
- Yuanlu Sun
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, 150030, China
| | - Yu Pang
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, 150030, China
| | - Xiaoxu Wu
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, 150030, China
| | - Rongru Zhu
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, 150030, China
| | - Liang Wang
- Institute of Animal Husbandry, Heilongjiang Academy of Agricultural Sciences, Harbin, 150086, China
| | - Ming Tian
- Institute of Animal Husbandry, Heilongjiang Academy of Agricultural Sciences, Harbin, 150086, China
| | - Xinmiao He
- Institute of Animal Husbandry, Heilongjiang Academy of Agricultural Sciences, Harbin, 150086, China
| | - Di Liu
- Institute of Animal Husbandry, Heilongjiang Academy of Agricultural Sciences, Harbin, 150086, China.
| | - Xiuqin Yang
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, 150030, China.
| |
Collapse
|
26
|
Belchikov N, Hsu J, Li XJ, Jarroux J, Hu W, Joglekar A, Tilgner HU. Understanding isoform expression by pairing long-read sequencing with single-cell and spatial transcriptomics. Genome Res 2024; 34:1735-1746. [PMID: 39567235 DOI: 10.1101/gr.279640.124] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2024]
Abstract
RNA isoform diversity, produced via alternative splicing, and alternative usage of transcription start and poly(A) sites, results in varied transcripts being derived from the same gene. Distinct isoforms can play important biological roles, including by changing the sequences or expression levels of protein products. The first single-cell approaches to RNA sequencing-and later, spatial approaches-which are now widely used for the identification of differentially expressed genes, rely on short reads and offer the ability to transcriptomically compare different cell types but are limited in their ability to measure differential isoform expression. More recently, long-read sequencing methods have been combined with single-cell and spatial technologies in order to characterize isoform expression. In this review, we provide an overview of the emergence of single-cell and spatial long-read sequencing and discuss the challenges associated with the implementation of these technologies and interpretation of these data. We discuss the opportunities they offer for understanding the relationships between the distinct variable elements of transcript molecules and highlight some of the ways in which they have been used to characterize isoforms' roles in development and pathology. Single-nucleus long-read sequencing, a special case of the single-cell approach, is also discussed. We attempt to cover both the limitations of these technologies and their significant potential for expanding our still-limited understanding of the biological roles of RNA isoforms.
Collapse
Affiliation(s)
- Natan Belchikov
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, New York 10065, USA
- Center for Neurogenetics, Weill Cornell Medicine, New York, New York 10021, USA
- Physiology, Biophysics, and Systems Biology Program, Weill Cornell Medicine, New York, New York 10065, USA
| | - Justine Hsu
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, New York 10065, USA
- Center for Neurogenetics, Weill Cornell Medicine, New York, New York 10021, USA
| | - Xiang Jennie Li
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, New York 10065, USA
- Center for Neurogenetics, Weill Cornell Medicine, New York, New York 10021, USA
- Computational Biology Master's Program, Weill Cornell Medicine, New York, New York 10065, USA
| | - Julien Jarroux
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, New York 10065, USA
- Center for Neurogenetics, Weill Cornell Medicine, New York, New York 10021, USA
| | - Wen Hu
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, New York 10065, USA
- Center for Neurogenetics, Weill Cornell Medicine, New York, New York 10021, USA
| | - Anoushka Joglekar
- New York Genome Center, New York, New York 10013, USA
- Department of Biomedical Informatics, Columbia University, New York, New York 10032, USA
| | - Hagen U Tilgner
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, New York 10065, USA;
- Center for Neurogenetics, Weill Cornell Medicine, New York, New York 10021, USA
| |
Collapse
|
27
|
Qin G, Narsinh K, Wei Q, Roach JC, Joshi A, Goetz SL, Moxon ST, Brush MH, Xu C, Yao Y, Glen AK, Morris ED, Ralevski A, Roper R, Belhu B, Zhang Y, Shmulevich I, Hadlock J, Glusman G. Generating Biomedical Knowledge Graphs from Knowledge Bases, Registries, and Multiomic Data. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.14.623648. [PMID: 39605475 PMCID: PMC11601480 DOI: 10.1101/2024.11.14.623648] [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: 11/29/2024]
Abstract
As large clinical and multiomics datasets and knowledge resources accumulate, they need to be transformed into computable and actionable information to support automated reasoning. These datasets range from laboratory experiment results to electronic health records (EHRs). Barriers to accessibility and sharing of such datasets include diversity of content, size and privacy. Effective transformation of data into information requires harmonization of stakeholder goals, implementation, enforcement of standards regarding quality and completeness, and availability of resources for maintenance and updates. Systems such as the Biomedical Data Translator leverage knowledge graphs (KGs), structured and machine learning readable knowledge representation, to encode knowledge extracted through inference. We focus here on the transformation of data from multiomics datasets and EHRs into compact knowledge, represented in a KG data structure. We demonstrate this data transformation in the context of the Translator ecosystem, including clinical trials, drug approvals, cancer, wellness, and EHR data. These transformations preserve individual privacy. We provide access to the five resulting KGs through the Translator framework. We show examples of biomedical research questions supported by our KGs, and discuss issues arising from extracting biomedical knowledge from multiomics data.
Collapse
Affiliation(s)
- Guangrong Qin
- Institute for Systems Biology, 401 Terry Ave N, Seattle, WA 98109, USA
| | - Kamileh Narsinh
- Institute for Systems Biology, 401 Terry Ave N, Seattle, WA 98109, USA
| | - Qi Wei
- Institute for Systems Biology, 401 Terry Ave N, Seattle, WA 98109, USA
| | - Jared C. Roach
- Institute for Systems Biology, 401 Terry Ave N, Seattle, WA 98109, USA
| | - Arpita Joshi
- The Scripps Research Institute, 10550 N Torrey Pines Rd, La Jolla, CA 92037, USA
| | - Skye L. Goetz
- Institute for Systems Biology, 401 Terry Ave N, Seattle, WA 98109, USA
| | - Sierra T. Moxon
- Lawrence Berkeley National Laboratory, 1 Cyclotron Road, Berkeley, CA 94720, USA
| | - Matthew H. Brush
- UNC Chapel Hill, Department of Genetics, 120 Mason Farm Rd, Chapel Hill, NC 27599, USA
| | - Colleen Xu
- The Scripps Research Institute, 10550 N Torrey Pines Rd, La Jolla, CA 92037, USA
| | - Yao Yao
- Oregon State University, 1500 SW Jefferson Way, Corvallis, OR 97331
| | - Amy K. Glen
- Oregon State University, 1500 SW Jefferson Way, Corvallis, OR 97331
| | - Evan D. Morris
- Renaissance Computing Institute, 100 Europa Dr, Ste 540, Chapel Hill, NC 27517, USA
| | | | - Ryan Roper
- Institute for Systems Biology, 401 Terry Ave N, Seattle, WA 98109, USA
| | - Basazin Belhu
- Institute for Systems Biology, 401 Terry Ave N, Seattle, WA 98109, USA
| | - Yue Zhang
- Institute for Systems Biology, 401 Terry Ave N, Seattle, WA 98109, USA
| | - Ilya Shmulevich
- Institute for Systems Biology, 401 Terry Ave N, Seattle, WA 98109, USA
| | - Jennifer Hadlock
- Institute for Systems Biology, 401 Terry Ave N, Seattle, WA 98109, USA
| | - Gwênlyn Glusman
- Institute for Systems Biology, 401 Terry Ave N, Seattle, WA 98109, USA
| |
Collapse
|
28
|
Zhao Y, Tan J, Fang L, Jiang L. Harnessing meta-omics to unveil and mitigate methane emissions in ruminants: Integrative approaches and future directions. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 951:175732. [PMID: 39182764 DOI: 10.1016/j.scitotenv.2024.175732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 08/17/2024] [Accepted: 08/21/2024] [Indexed: 08/27/2024]
Abstract
Methane emissions from enteric fermentation present a dual challenge globally: they not only contribute significantly to atmospheric greenhouse gases but also represent a considerable energy loss for ruminant animals. Utilizing high-throughput omics technologies to analyze rumen microbiome samples (meta-omics, i.e., metagenomics, metatranscriptomics, metaproteomics, metabolomics) holds vast potential for uncovering the intricate interplay between diet, microbiota, and methane emissions in these animals. The primary obstacle is the effective integration of diverse meta-omic approaches and their broader application across different ruminant species. Genetic variability significantly impacts methane production in ruminants, suggesting that genomic selection could be a viable strategy to reduce emissions. While substantial research has been conducted on the microbiological aspects of methane production, there remains a critical need to delineate the specific genetic interactions between the host and its microbiome. Advancements in meta-omics technologies are poised to shed light on these interactions, enhancing our understanding of the genetic factors that govern methane output. This review explores the potential of meta-omics to accelerate genetic advancements that could lead to reduced methane emissions in ruminants. By employing a systems biology approach, the integration of various omics technologies allows for the identification of key genomic regions and genetic markers linked to methane production. These markers can then be leveraged in selective breeding programs to cultivate traits associated with lower emissions. Moreover, the review addresses current challenges in applying genomic selection for this purpose and discusses how omics technologies can overcome these obstacles. The systematic integration and analysis of diverse biological data provide deeper insights into the genetic underpinnings and overall biology of methane production traits in ruminants. Ultimately, this comprehensive approach not only aids in reducing the environmental impact of agriculture but also contributes to the sustainability and efficiency of livestock management.
Collapse
Affiliation(s)
- Yuchao Zhao
- Beijing Key Laboratory of Dairy Cow Nutrition, College of Animal Science and Technology, Beijing University of Agriculture, Beijing 102206, China
| | - Jian Tan
- Beijing Key Laboratory of Dairy Cow Nutrition, College of Animal Science and Technology, Beijing University of Agriculture, Beijing 102206, China
| | - Luoyun Fang
- Beijing Key Laboratory of Dairy Cow Nutrition, College of Animal Science and Technology, Beijing University of Agriculture, Beijing 102206, China
| | - Linshu Jiang
- Beijing Key Laboratory of Dairy Cow Nutrition, College of Animal Science and Technology, Beijing University of Agriculture, Beijing 102206, China.
| |
Collapse
|
29
|
Fernando MB, Fan Y, Zhang Y, Tokolyi A, Murphy AN, Kammourh S, Michael Deans P, Ghorbani S, Onatzevitch R, Pero A, Padilla C, Williams S, Flaherty EK, Prytkova IA, Cao L, Knowles DA, Fang G, Slesinger PA, Brennand KJ. Phenotypic complexities of rare heterozygous neurexin-1 deletions. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.10.28.564543. [PMID: 37961635 PMCID: PMC10634884 DOI: 10.1101/2023.10.28.564543] [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] [Indexed: 11/15/2023]
Abstract
Given the large number of genes significantly associated with risk for neuropsychiatric disorders, a critical unanswered question is the extent to which diverse mutations --sometimes impacting the same gene-- will require tailored therapeutic strategies. Here we consider this in the context of rare neuropsychiatric disorder-associated copy number variants (2p16.3) resulting in heterozygous deletions in NRXN1, a pre-synaptic cell adhesion protein that serves as a critical synaptic organizer in the brain. Complex patterns of NRXN1 alternative splicing are fundamental to establishing diverse neurocircuitry, vary between the cell types of the brain, and are differentially impacted by unique (non-recurrent) deletions. We contrast the cell-type-specific impact of patient-specific mutations in NRXN1 using human induced pluripotent stem cells, finding that perturbations in NRXN1 splicing result in divergent cell-type-specific synaptic outcomes. Via distinct loss-of-function (LOF) and gain-of-function (GOF) mechanisms, NRXN1 +/- deletions cause decreased synaptic activity in glutamatergic neurons, yet increased synaptic activity in GABAergic neurons. Reciprocal isogenic manipulations causally demonstrate that aberrant splicing drives these changes in synaptic activity. For NRXN1 deletions, and perhaps more broadly, precision medicine will require stratifying patients based on whether their gene mutations act through LOF or GOF mechanisms, in order to achieve individualized restoration of NRXN1 isoform repertoires by increasing wildtype, or ablating mutant isoforms. Given the increasing number of mutations predicted to engender both LOF and GOF mechanisms in brain disorders, our findings add nuance to future considerations of precision medicine.
Collapse
Affiliation(s)
- Michael B. Fernando
- Graduate School of Biomedical Science, Icahn School of Medicine at Mount Sinai, New York, NY 10029
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029
- Friedman Brain Institute, Black Family Stem Cell Institute, Pamela Sklar Division of Psychiatric Genomics, Icahn School of Medicine at Mount Sinai, New York, NY 10029
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, 06520
| | - Yu Fan
- Department of Genetics and Genomics, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Yanchun Zhang
- Department of Genetics and Genomics, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | | | - Aleta N. Murphy
- Graduate School of Biomedical Science, Icahn School of Medicine at Mount Sinai, New York, NY 10029
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029
- Friedman Brain Institute, Black Family Stem Cell Institute, Pamela Sklar Division of Psychiatric Genomics, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Sarah Kammourh
- Department of Genetics and Genomics, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | | | - Sadaf Ghorbani
- Haukeland University Hospital, Bergen, Norway
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, 06520
| | - Ryan Onatzevitch
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Adriana Pero
- Department of Genetics and Genomics, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Christopher Padilla
- Department of Genetics and Genomics, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Sarah Williams
- Graduate School of Biomedical Science, Icahn School of Medicine at Mount Sinai, New York, NY 10029
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029
- Friedman Brain Institute, Black Family Stem Cell Institute, Pamela Sklar Division of Psychiatric Genomics, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Erin K. Flaherty
- Graduate School of Biomedical Science, Icahn School of Medicine at Mount Sinai, New York, NY 10029
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029
- Friedman Brain Institute, Black Family Stem Cell Institute, Pamela Sklar Division of Psychiatric Genomics, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Iya A. Prytkova
- Graduate School of Biomedical Science, Icahn School of Medicine at Mount Sinai, New York, NY 10029
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029
- Friedman Brain Institute, Black Family Stem Cell Institute, Pamela Sklar Division of Psychiatric Genomics, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Lei Cao
- Department of Genetics and Genomics, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - David A. Knowles
- New York Genome Center, New York, NY, 10013
- Departments of Computer Science, Systems Biology, and Data Science Institute, Columbia University, New York, NY, USA, 10027
| | - Gang Fang
- Department of Genetics and Genomics, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Paul A. Slesinger
- Graduate School of Biomedical Science, Icahn School of Medicine at Mount Sinai, New York, NY 10029
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Kristen J. Brennand
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029
- Department of Genetics and Genomics, Icahn School of Medicine at Mount Sinai, New York, NY 10029
- Friedman Brain Institute, Black Family Stem Cell Institute, Pamela Sklar Division of Psychiatric Genomics, Icahn School of Medicine at Mount Sinai, New York, NY 10029
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, 06520
| |
Collapse
|
30
|
Kjer-Hansen P, Phan TG, Weatheritt RJ. Protein isoform-centric therapeutics: expanding targets and increasing specificity. Nat Rev Drug Discov 2024; 23:759-779. [PMID: 39232238 DOI: 10.1038/s41573-024-01025-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/31/2024] [Indexed: 09/06/2024]
Abstract
Most protein-coding genes produce multiple protein isoforms; however, these isoforms are commonly neglected in drug discovery. The expression of protein isoforms can be specific to a disease, tissue and/or developmental stage, and this specific expression can be harnessed to achieve greater drug specificity than pan-targeting of all gene products and to enable improved treatments for diseases caused by aberrant protein isoform production. In recent years, several protein isoform-centric therapeutics have been developed. Here, we collate these studies and clinical trials to highlight three distinct but overlapping modes of action for protein isoform-centric drugs: isoform switching, isoform introduction or depletion, and modulation of isoform activity. In addition, we discuss how protein isoforms can be used clinically as targets for cell type-specific drug delivery and immunotherapy, diagnostic biomarkers and sources of cancer neoantigens. Collectively, we emphasize the value of a focus on isoforms as a route to discovering drugs with greater specificity and fewer adverse effects. This approach could enable the targeting of proteins for which pan-inhibition of all isoforms is toxic and poorly tolerated.
Collapse
Affiliation(s)
- Peter Kjer-Hansen
- EMBL Australia, Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia.
- St. Vincent's Healthcare Clinical Campus, School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Darlinghurst, New South Wales, Australia.
| | - Tri Giang Phan
- St. Vincent's Healthcare Clinical Campus, School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Darlinghurst, New South Wales, Australia
- Precision Immunology Program, Garvan Institute of Medical Research, Sydney, New South Wales, Australia
| | - Robert J Weatheritt
- EMBL Australia, Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia.
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, New South Wales, Australia.
| |
Collapse
|
31
|
Xu R, Li S, Chien CJ, Zhong Y, Xiao H, Fang S, Du S. Expression of Smyd1b_tv1 by Alternative Splicing in Cardiac Muscle is Critical for Sarcomere Organization in Cardiomyocytes and Heart Function. Mol Cell Biol 2024; 44:543-561. [PMID: 39320962 PMCID: PMC11583600 DOI: 10.1080/10985549.2024.2402660] [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: 02/07/2024] [Revised: 07/01/2024] [Accepted: 07/05/2024] [Indexed: 09/27/2024] Open
Abstract
Smyd1, a member of the Smyd lysine methyltransferase family, plays an important role in myofibrillogenesis of skeletal and cardiac muscles. Loss of Smyd1b (a Smyd1 ortholog) function in zebrafish results in embryonic death from heart malfunction. smyd1b encodes two isoforms, Smyd1b_tv1 and Smyd1b_tv2, differing by 13 amino acids due to alternative splicing. While smyd1 alternative splicing is evolutionarily conserved, the isoform-specific expression and function of Smyd1b_tv1 and Smyd1b_tv2 remained unknown. Here we analyzed their expression and function in skeletal and cardiac muscles. Our analysis revealed expression of smyd1b_tv1 predominately in cardiac and smyd1b_tv2 in skeletal muscles. Using zebrafish models expressing only one isoform, we demonstrated that Smyd1b_tv1 is essential for cardiomyocyte differentiation and fish viability, whereas Smyd1b_tv2 is dispensable for heart development and fish survival. Cellular and biochemical analyses revealed that Smyd1b_tv1 differs from Smyd1b_tv2 in protein localization and binding with myosin chaperones. While Smyd1b_tv2 diffused in the cytosol of muscle cells, Smyd1b_tv1 was localized to M-lines and essential for sarcomere organization in cardiomyocytes. Co-IP analysis revealed a stronger binding of Smyd1b_tv1 with chaperones and cochaperones compared with Smyd1b_tv2. Collectively, these findings highlight the nonequivalence of Smyd1b isoforms in cardiomyocyte differentiation, emphasizing the critical role of Smyd1b_tv1 in cardiac function.
Collapse
Affiliation(s)
- Rui Xu
- Department of Biochemistry and Molecular Biology, Institute of Marine and Environmental Technology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Siping Li
- Department of Biochemistry and Molecular Biology, Institute of Marine and Environmental Technology, University of Maryland School of Medicine, Baltimore, Maryland, USA
- East China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Shanghai, China
| | - Chien-Ju Chien
- Department of Biochemistry and Molecular Biology, Institute of Marine and Environmental Technology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Yongwang Zhong
- Center for Biomedical Engineering, Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Huanhuan Xiao
- Department of Biochemistry and Molecular Biology, Institute of Marine and Environmental Technology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Shengyun Fang
- Center for Biomedical Engineering, Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Shaojun Du
- Department of Biochemistry and Molecular Biology, Institute of Marine and Environmental Technology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
32
|
Sadeghi S, Ashkarran AA, Wang Q, Zhu G, Mahmoudi M, Sun L. Mass Spectrometry-Based Top-Down Proteomics in Nanomedicine: Proteoform-Specific Measurement of Protein Corona. ACS NANO 2024; 18. [PMID: 39276099 PMCID: PMC11440641 DOI: 10.1021/acsnano.4c04675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 08/30/2024] [Accepted: 09/06/2024] [Indexed: 09/16/2024]
Abstract
Conventional mass spectrometry (MS)-based bottom-up proteomics (BUP) analysis of the protein corona [i.e., an evolving layer of biomolecules, mostly proteins, formed on the surface of nanoparticles (NPs) during their interactions with biomolecular fluids] enabled the nanomedicine community to partly identify the biological identity of NPs. Such an approach, however, fails to pinpoint the specific proteoforms─distinct molecular variants of proteins in the protein corona. The proteoform-level information could potentially advance the prediction of the biological fate and pharmacokinetics of nanomedicines. Recognizing this limitation, this study pioneers a robust and reproducible MS-based top-down proteomics (TDP) technique for characterizing proteoforms in the protein corona. Our TDP approach has successfully identified about 900 proteoforms in the protein corona of polystyrene NPs, ranging from 2 to 70 kDa, revealing proteoforms of 48 protein biomarkers with combinations of post-translational modifications, signal peptide cleavages, and/or truncations─details that BUP could not fully discern. This advancement in MS-based TDP offers a more advanced approach to characterize NP protein coronas, deepening our understanding of NPs' biological identities. We, therefore, propose using both TDP and BUP strategies to obtain more comprehensive information about the protein corona, which, in turn, can further enhance the diagnostic and therapeutic efficacy of nanomedicine technologies.
Collapse
Affiliation(s)
- Seyed
Amirhossein Sadeghi
- Department
of Chemistry, Michigan State University, 578 S Shaw Lane, East Lansing, Michigan 48824, United States
| | - Ali Akbar Ashkarran
- Department
of Radiology and Precision Health Program, Michigan State University, East Lansing, Michigan 48824, United States
| | - Qianyi Wang
- Department
of Chemistry, Michigan State University, 578 S Shaw Lane, East Lansing, Michigan 48824, United States
| | - Guijie Zhu
- Department
of Chemistry, Michigan State University, 578 S Shaw Lane, East Lansing, Michigan 48824, United States
| | - Morteza Mahmoudi
- Department
of Radiology and Precision Health Program, Michigan State University, East Lansing, Michigan 48824, United States
| | - Liangliang Sun
- Department
of Chemistry, Michigan State University, 578 S Shaw Lane, East Lansing, Michigan 48824, United States
| |
Collapse
|
33
|
Kiseleva OI, Arzumanian VA, Kurbatov IY, Poverennaya EV. In silico and in cellulo approaches for functional annotation of human protein splice variants. BIOMEDITSINSKAIA KHIMIIA 2024; 70:315-328. [PMID: 39324196 DOI: 10.18097/pbmc20247005315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/27/2024]
Abstract
The elegance of pre-mRNA splicing mechanisms continues to interest scientists even after over a half century, since the discovery of the fact that coding regions in genes are interrupted by non-coding sequences. The vast majority of human genes have several mRNA variants, coding structurally and functionally different protein isoforms in a tissue-specific manner and with a linkage to specific developmental stages of the organism. Alteration of splicing patterns shifts the balance of functionally distinct proteins in living systems, distorts normal molecular pathways, and may trigger the onset and progression of various pathologies. Over the past two decades, numerous studies have been conducted in various life sciences disciplines to deepen our understanding of splicing mechanisms and the extent of their impact on the functioning of living systems. This review aims to summarize experimental and computational approaches used to elucidate the functions of splice variants of a single gene based on our experience accumulated in the laboratory of interactomics of proteoforms at the Institute of Biomedical Chemistry (IBMC) and best global practices.
Collapse
Affiliation(s)
- O I Kiseleva
- Institute of Biomedical Chemistry, Moscow, Russia
| | | | | | | |
Collapse
|
34
|
Teng L, Sun Y, Chen J, Wang C, Urbach JM, Kobe B, Ye N, Zeng Q. Exon shuffling and alternative splicing of ROCO genes in brown algae enables a diverse repertoire of candidate immune receptors. FRONTIERS IN PLANT SCIENCE 2024; 15:1445022. [PMID: 39246816 PMCID: PMC11378527 DOI: 10.3389/fpls.2024.1445022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Accepted: 08/01/2024] [Indexed: 09/10/2024]
Abstract
The ROCO family is a family of GTPases characterized by a central ROC-COR tandem domain. Interest in the structure and function of ROCO proteins has increased with the identification of their important roles in human disease. Nevertheless, the functions of most ROCO proteins are still unknown. In the present study, we characterized the structure, evolution, and expression of ROCOs in four species of brown algae. Brown algae have a larger number of ROCO proteins than other organisms reported to date. Phylogenetic analyses showed that ROCOs have an ancient origin, likely originated in prokaryotes. ROCOs in brown algae clustered into four groups and showed no strong relationship with red algae or green algae. Brown algal ROCOs retain the ancestral LRR-ROC-COR domain arrangement, which is found in prokaryotes, plants and some basal metazoans. Remarkably, individual LRR motifs in ROCO genes are each encoded by separate exons and exhibit intense exon shuffling and diversifying selection. Furthermore, the tandem LRR exons exhibit alternative splicing to generate multiple transcripts. Both exon shuffling and alternative splicing of LRR repeats may be important mechanisms for generating diverse ligand-binding specificities as immune receptors. Besides their potential immune role, expression analysis shows that many ROCO genes are responsive to other stress conditions, suggesting they could participate in multiple signal pathways, not limited to the immune response. Our results substantially enhance our understanding of the structure and function of this mysterious gene family.
Collapse
Affiliation(s)
- Linhong Teng
- College of Life Sciences, Dezhou University, Dezhou, China
| | - Yuhuan Sun
- College of Life Sciences, Dezhou University, Dezhou, China
| | - Jiayi Chen
- College of Life Sciences, Dezhou University, Dezhou, China
| | - Chenghui Wang
- College of Life Sciences, Dezhou University, Dezhou, China
| | - Jonathan M Urbach
- Ragon Institute of Mass General Brigham, MIT, and Harvard, Cambridge, MA, United States
| | - Bostjan Kobe
- School of Chemistry and Molecular Biosciences, Institute for Molecular Bioscience and Australian Infectious Diseases Research Centre, University of Queensland, Brisbane, QLD, Australia
| | - Naihao Ye
- National Key Laboratory of Mariculture Biobreeding and Sustainable Production, Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao, China
| | | |
Collapse
|
35
|
Verma A, Goel A, Koner N, Gunasekaran G, Radha V. Development and tissue specific expression of RAPGEF1 (C3G) transcripts having exons encoding disordered segments with predicted regulatory function. Mol Biol Rep 2024; 51:907. [PMID: 39141165 DOI: 10.1007/s11033-024-09845-3] [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: 05/18/2024] [Accepted: 08/06/2024] [Indexed: 08/15/2024]
Abstract
BACKGROUND The ubiquitously expressed Guanine nucleotide exchange factor, RAPGEF1 (C3G), is essential for early development of mouse embryos. It functions to regulate gene expression and cytoskeletal reorganization, thereby controlling cell proliferation and differentiation. While multiple transcripts have been predicted, their expression in mouse tissues has not been investigated in detail. METHODS & RESULTS Full length RAPGEF1 isoforms primarily arise due to splicing at two hotspots, one involving exon-3, and the other involving exons 12-14 incorporating amino acids immediately following the Crk binding region of the protein. These isoforms vary in expression across embryonic and adult organs. We detected the presence of unannotated, and unpredicted transcripts with incorporation of cassette exons in various combinations, specifically in the heart, brain, testis and skeletal muscle. Isoform switching was detected as myocytes in culture and mouse embryonic stem cells were differentiated to form myotubes, and embryoid bodies respectively. The cassette exons encode a serine-rich polypeptide chain, which is intrinsically disordered, and undergoes phosphorylation. In silico structural analysis using AlphaFold indicated that the presence of cassette exons alters intra-molecular interactions, important for regulating catalytic activity. LZerD based docking studies predicted that the isoforms with one or more cassette exons differ in interaction with their target GTPase, RAP1A. CONCLUSIONS Our results demonstrate the expression of novel RAPGEF1 isoforms, and predict cassette exon inclusion as an additional means of regulating RAPGEF1 activity in various tissues and during differentiation.
Collapse
Affiliation(s)
- Archana Verma
- CSIR-Centre for Cellular & Molecular Biology, Uppal Road, Habsiguda, Hyderabad, 500 007, India
- Department of Pediatric Hematology and Oncology, University Childrens Hospital, Muenster, 48149, Germany
| | - Abhishek Goel
- CSIR-Centre for Cellular & Molecular Biology, Uppal Road, Habsiguda, Hyderabad, 500 007, India
| | - Niladri Koner
- CSIR-Centre for Cellular & Molecular Biology, Uppal Road, Habsiguda, Hyderabad, 500 007, India
| | - Gowthaman Gunasekaran
- CSIR-Centre for Cellular & Molecular Biology, Uppal Road, Habsiguda, Hyderabad, 500 007, India
- Department of Molecular Biology Laboratory of Chromatin Biology, Ariel University, Ariel, 40700, Israel
| | - Vegesna Radha
- CSIR-Centre for Cellular & Molecular Biology, Uppal Road, Habsiguda, Hyderabad, 500 007, India.
| |
Collapse
|
36
|
Pandi B, Brenman S, Black A, Ng DCM, Lau E, Lam MPY. Tissue Usage Preference and Intrinsically Disordered Region Remodeling of Alternative Splicing Derived Proteoforms in the Heart. J Proteome Res 2024; 23:3161-3173. [PMID: 38456420 PMCID: PMC11296937 DOI: 10.1021/acs.jproteome.3c00789] [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: 11/15/2023] [Revised: 02/08/2024] [Accepted: 02/27/2024] [Indexed: 03/09/2024]
Abstract
A computational analysis of mass spectrometry data was performed to uncover alternative splicing derived protein variants across chambers of the human heart. Evidence for 216 non-canonical isoforms was apparent in the atrium and the ventricle, including 52 isoforms not documented on SwissProt and recovered using an RNA sequencing derived database. Among non-canonical isoforms, 29 show signs of regulation based on statistically significant preferences in tissue usage, including a ventricular enriched protein isoform of tensin-1 (TNS1) and an atrium-enriched PDZ and LIM Domain 3 (PDLIM3) isoform 2 (PDLIM3-2/ALP-H). Examined variant regions that differ between alternative and canonical isoforms are highly enriched with intrinsically disordered regions. Moreover, over two-thirds of such regions are predicted to function in protein binding and RNA binding. The analysis here lends further credence to the notion that alternative splicing diversifies the proteome by rewiring intrinsically disordered regions, which are increasingly recognized to play important roles in the generation of biological function from protein sequences.
Collapse
Affiliation(s)
- Boomathi Pandi
- Department
of Medicine/Division of Cardiology, Department of Biochemistry &
Molecular Genetics, and Consortium for Fibrosis Research and Translation (CFReT), University of Colorado School of Medicine, Aurora, Colorado 80045, United States
| | - Stella Brenman
- Department
of Medicine/Division of Cardiology, Department of Biochemistry &
Molecular Genetics, and Consortium for Fibrosis Research and Translation (CFReT), University of Colorado School of Medicine, Aurora, Colorado 80045, United States
| | - Alexander Black
- Department
of Medicine/Division of Cardiology, Department of Biochemistry &
Molecular Genetics, and Consortium for Fibrosis Research and Translation (CFReT), University of Colorado School of Medicine, Aurora, Colorado 80045, United States
| | - Dominic C. M. Ng
- Department
of Medicine/Division of Cardiology, Department of Biochemistry &
Molecular Genetics, and Consortium for Fibrosis Research and Translation (CFReT), University of Colorado School of Medicine, Aurora, Colorado 80045, United States
| | - Edward Lau
- Department
of Medicine/Division of Cardiology, Department of Biochemistry &
Molecular Genetics, and Consortium for Fibrosis Research and Translation (CFReT), University of Colorado School of Medicine, Aurora, Colorado 80045, United States
| | - Maggie P. Y. Lam
- Department
of Medicine/Division of Cardiology, Department of Biochemistry &
Molecular Genetics, and Consortium for Fibrosis Research and Translation (CFReT), University of Colorado School of Medicine, Aurora, Colorado 80045, United States
| |
Collapse
|
37
|
Chung DEC, Deng X, Yalamanchili HK, Revelli JP, Han AL, Tadros B, Richman R, Dias M, Naini FA, Boeynaems S, Hyman BT, Zoghbi HY. The big tau splice isoform resists Alzheimer's-related pathological changes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.30.605685. [PMID: 39211086 PMCID: PMC11360890 DOI: 10.1101/2024.07.30.605685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
In Alzheimer's disease (AD), the microtubule-binding protein tau becomes abnormally hyperphosphorylated and aggregated in selective brain regions such as the cortex and hippocampus 1-3 . However, other brain regions like the cerebellum and brain stem remain largely intact despite the universal expression of tau throughout the brain. Here, we found that an understudied splice isoform of tau termed "big tau" is significantly more abundant in the brain regions less vulnerable to tau pathology compared to tau pathology-vulnerable regions. We used various cellular and animal models to demonstrate that big tau possesses multiple properties that can resist AD-related pathological changes. Importantly, human AD patients show a higher expression level of pathology-resisting big tau in the cerebellum, the brain region spared from tau pathology. Our study examines the unique properties of big tau, expanding our current understanding of tau pathophysiology. Altogether, our data suggest that alternative splicing to favor big tau is a viable strategy to modulate tau pathology.
Collapse
|
38
|
Xu T, Wang Q, Wang Q, Sun L. Mass spectrometry-intensive top-down proteomics: an update on technology advancements and biomedical applications. ANALYTICAL METHODS : ADVANCING METHODS AND APPLICATIONS 2024; 16:4664-4682. [PMID: 38973469 PMCID: PMC11257149 DOI: 10.1039/d4ay00651h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 06/25/2024] [Indexed: 07/09/2024]
Abstract
Proteoforms are all forms of protein molecules from the same gene because of variations at the DNA, RNA, and protein levels, e.g., alternative splicing and post-translational modifications (PTMs). Delineation of proteins in a proteoform-specific manner is crucial for understanding their biological functions. Mass spectrometry (MS)-intensive top-down proteomics (TDP) is promising for comprehensively characterizing intact proteoforms in complex biological systems. It has achieved substantial progress in technological development, including sample preparation, proteoform separations, MS instrumentation, and bioinformatics tools. In a single TDP study, thousands of proteoforms can be identified and quantified from a cell lysate. It has also been applied to various biomedical research to better our understanding of protein function in regulating cellular processes and to discover novel proteoform biomarkers of diseases for early diagnosis and therapeutic development. This review covers the most recent technological development and biomedical applications of MS-intensive TDP.
Collapse
Affiliation(s)
- Tian Xu
- Department of Chemistry, Michigan State University, 578 S Shaw Lane, East Lansing, MI 48824, USA.
| | - Qianjie Wang
- Department of Chemistry, Michigan State University, 578 S Shaw Lane, East Lansing, MI 48824, USA.
| | - Qianyi Wang
- Department of Chemistry, Michigan State University, 578 S Shaw Lane, East Lansing, MI 48824, USA.
| | - Liangliang Sun
- Department of Chemistry, Michigan State University, 578 S Shaw Lane, East Lansing, MI 48824, USA.
| |
Collapse
|
39
|
Xiao MS, Damodaran AP, Kumari B, Dickson E, Xing K, On TA, Parab N, King HE, Perez AR, Guiblet WM, Duncan G, Che A, Chari R, Andresson T, Vidigal JA, Weatheritt RJ, Aregger M, Gonatopoulos-Pournatzis T. Genome-scale exon perturbation screens uncover exons critical for cell fitness. Mol Cell 2024; 84:2553-2572.e19. [PMID: 38917794 PMCID: PMC11246229 DOI: 10.1016/j.molcel.2024.05.024] [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: 10/01/2023] [Revised: 04/04/2024] [Accepted: 05/24/2024] [Indexed: 06/27/2024]
Abstract
CRISPR-Cas technology has transformed functional genomics, yet understanding of how individual exons differentially shape cellular phenotypes remains limited. Here, we optimized and conducted massively parallel exon deletion and splice-site mutation screens in human cell lines to identify exons that regulate cellular fitness. Fitness-promoting exons are prevalent in essential and highly expressed genes and commonly overlap with protein domains and interaction interfaces. Conversely, fitness-suppressing exons are enriched in nonessential genes, exhibiting lower inclusion levels, and overlap with intrinsically disordered regions and disease-associated mutations. In-depth mechanistic investigation of the screen-hit TAF5 alternative exon-8 revealed that its inclusion is required for assembly of the TFIID general transcription initiation complex, thereby regulating global gene expression output. Collectively, our orthogonal exon perturbation screens established a comprehensive repository of phenotypically important exons and uncovered regulatory mechanisms governing cellular fitness and gene expression.
Collapse
Affiliation(s)
- Mei-Sheng Xiao
- RNA Biology Laboratory, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH), Frederick, MD 21702, USA
| | - Arun Prasath Damodaran
- RNA Biology Laboratory, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH), Frederick, MD 21702, USA.
| | - Bandana Kumari
- RNA Biology Laboratory, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH), Frederick, MD 21702, USA
| | - Ethan Dickson
- RNA Biology Laboratory, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH), Frederick, MD 21702, USA
| | - Kun Xing
- RNA Biology Laboratory, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH), Frederick, MD 21702, USA
| | - Tyler A On
- Molecular Targets Program, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH), Frederick, MD 21702, USA
| | - Nikhil Parab
- RNA Biology Laboratory, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH), Frederick, MD 21702, USA
| | - Helen E King
- EMBL Australia and Garvan Institute of Medical Research, Sydney, NSW 2010, Australia
| | - Alexendar R Perez
- Laboratory of Biochemistry and Molecular Biology, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD 20892, USA; Department of Anesthesia and Perioperative Care, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Wilfried M Guiblet
- RNA Biology Laboratory, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH), Frederick, MD 21702, USA
| | - Gerard Duncan
- Protein Characterization Laboratory, Frederick National Laboratory for Cancer Research (FNLCR), Frederick, MD 21701, USA
| | - Anney Che
- Advanced Biomedical Computational Science, Frederick National Laboratory for Cancer Research (FNLCR), Frederick, MD 21701, USA
| | - Raj Chari
- Genome Modification Core, Frederick National Laboratory for Cancer Research (FNLCR), Frederick, MD 21702, USA
| | - Thorkell Andresson
- Protein Characterization Laboratory, Frederick National Laboratory for Cancer Research (FNLCR), Frederick, MD 21701, USA
| | - Joana A Vidigal
- Laboratory of Biochemistry and Molecular Biology, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Robert J Weatheritt
- EMBL Australia and Garvan Institute of Medical Research, Sydney, NSW 2010, Australia; School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, NSW 2010, Australia
| | - Michael Aregger
- Molecular Targets Program, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH), Frederick, MD 21702, USA.
| | - Thomas Gonatopoulos-Pournatzis
- RNA Biology Laboratory, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH), Frederick, MD 21702, USA.
| |
Collapse
|
40
|
Kopell BH, Kaji DA, Liharska LE, Vornholt E, Valentine A, Lund A, Hashemi A, Thompson RC, Lohrenz T, Johnson JS, Bussola N, Cheng E, Park YJ, Shah P, Ma W, Searfoss R, Qasim S, Miller GM, Chand NM, Aristel A, Humphrey J, Wilkins L, Ziafat K, Silk H, Linares LM, Sullivan B, Feng C, Batten SR, Bang D, Barbosa LS, Twomey T, White JP, Vannucci M, Hadj-Amar B, Cohen V, Kota P, Moya E, Rieder MK, Figee M, Nadkarni GN, Breen MS, Kishida KT, Scarpa J, Ruderfer DM, Narain NR, Wang P, Kiebish MA, Schadt EE, Saez I, Montague PR, Beckmann ND, Charney AW. Multiomic foundations of human prefrontal cortex tissue function. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.05.17.24307537. [PMID: 38798344 PMCID: PMC11118644 DOI: 10.1101/2024.05.17.24307537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
The prefrontal cortex (PFC) is a region of the brain that in humans is involved in the production of higher-order functions such as cognition, emotion, perception, and behavior. Neurotransmission in the PFC produces higher-order functions by integrating information from other areas of the brain. At the foundation of neurotransmission, and by extension at the foundation of higher-order brain functions, are an untold number of coordinated molecular processes involving the DNA sequence variants in the genome, RNA transcripts in the transcriptome, and proteins in the proteome. These "multiomic" foundations are poorly understood in humans, perhaps in part because most modern studies that characterize the molecular state of the human PFC use tissue obtained when neurotransmission and higher-order brain functions have ceased (i.e., the postmortem state). Here, analyses are presented on data generated for the Living Brain Project (LBP) to investigate whether PFC tissue from individuals with intact higher-order brain function has characteristic multiomic foundations. Two complementary strategies were employed towards this end. The first strategy was to identify in PFC samples obtained from living study participants a signature of RNA transcript expression associated with neurotransmission measured intracranially at the time of PFC sampling, in some cases while participants performed a task engaging higher-order brain functions. The second strategy was to perform multiomic comparisons between PFC samples obtained from individuals with intact higher-order brain function at the time of sampling (i.e., living study participants) and PFC samples obtained in the postmortem state. RNA transcript expression within multiple PFC cell types was associated with fluctuations of dopaminergic, serotonergic, and/or noradrenergic neurotransmission in the substantia nigra measured while participants played a computer game that engaged higher-order brain functions. A subset of these associations - termed the "transcriptional program associated with neurotransmission" (TPAWN) - were reproduced in analyses of brain RNA transcript expression and intracranial neurotransmission data obtained from a second LBP cohort and from a cohort in an independent study. RNA transcripts involved in TPAWN were found to be (1) enriched for RNA transcripts associated with measures of neurotransmission in rodent and cell models, (2) enriched for RNA transcripts encoded by evolutionarily constrained genes, (3) depleted of RNA transcripts regulated by common DNA sequence variants, and (4) enriched for RNA transcripts implicated in higher-order brain functions by human population genetic studies. In PFC excitatory neurons of living study participants, higher expression of the genes in TPAWN tracked with higher expression of RNA transcripts that in rodent PFC samples are markers of a class of excitatory neurons that connect the PFC to deep brain structures. TPAWN was further reproduced by RNA transcript expression patterns differentiating living PFC samples from postmortem PFC samples, and significant differences between living and postmortem PFC samples were additionally observed with respect to (1) the expression of most primary RNA transcripts, mature RNA transcripts, and proteins, (2) the splicing of most primary RNA transcripts into mature RNA transcripts, (3) the patterns of co-expression between RNA transcripts and proteins, and (4) the effects of some DNA sequence variants on RNA transcript and protein expression. Taken together, this report highlights that studies of brain tissue obtained in a safe and ethical manner from large cohorts of living individuals can help advance understanding of the multiomic foundations of brain function.
Collapse
|
41
|
Kurgan N, Kjærgaard Larsen J, Deshmukh AS. Harnessing the power of proteomics in precision diabetes medicine. Diabetologia 2024; 67:783-797. [PMID: 38345659 DOI: 10.1007/s00125-024-06097-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 12/20/2023] [Indexed: 03/21/2024]
Abstract
Precision diabetes medicine (PDM) aims to reduce errors in prevention programmes, diagnosis thresholds, prognosis prediction and treatment strategies. However, its advancement and implementation are difficult due to the heterogeneity of complex molecular processes and environmental exposures that influence an individual's disease trajectory. To address this challenge, it is imperative to develop robust screening methods for all areas of PDM. Innovative proteomic technologies, alongside genomics, have proven effective in precision cancer medicine and are showing promise in diabetes research for potential translation. This narrative review highlights how proteomics is well-positioned to help improve PDM. Specifically, a critical assessment of widely adopted affinity-based proteomic technologies in large-scale clinical studies and evidence of the benefits and feasibility of using MS-based plasma proteomics is presented. We also present a case for the use of proteomics to identify predictive protein panels for type 2 diabetes subtyping and the development of clinical prediction models for prevention, diagnosis, prognosis and treatment strategies. Lastly, we discuss the importance of plasma and tissue proteomics and its integration with genomics (proteogenomics) for identifying unique type 2 diabetes intra- and inter-subtype aetiology. We conclude with a call for action formed on advancing proteomics technologies, benchmarking their performance and standardisation across sites, with an emphasis on data sharing and the inclusion of diverse ancestries in large cohort studies. These efforts should foster collaboration with key stakeholders and align with ongoing academic programmes such as the Precision Medicine in Diabetes Initiative consortium.
Collapse
Affiliation(s)
- Nigel Kurgan
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Jeppe Kjærgaard Larsen
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Atul S Deshmukh
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
42
|
Nakamura M. Lipotoxicity as a therapeutic target in obesity and diabetic cardiomyopathy. JOURNAL OF PHARMACY & PHARMACEUTICAL SCIENCES : A PUBLICATION OF THE CANADIAN SOCIETY FOR PHARMACEUTICAL SCIENCES, SOCIETE CANADIENNE DES SCIENCES PHARMACEUTIQUES 2024; 27:12568. [PMID: 38706718 PMCID: PMC11066298 DOI: 10.3389/jpps.2024.12568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 04/09/2024] [Indexed: 05/07/2024]
Abstract
Unhealthy sources of fats, ultra-processed foods with added sugars, and a sedentary lifestyle make humans more susceptible to developing overweight and obesity. While lipids constitute an integral component of the organism, excessive and abnormal lipid accumulation that exceeds the storage capacity of lipid droplets disrupts the intracellular composition of fatty acids and results in the release of deleterious lipid species, thereby giving rise to a pathological state termed lipotoxicity. This condition induces endoplasmic reticulum stress, mitochondrial dysfunction, inflammatory responses, and cell death. Recent advances in omics technologies and analytical methodologies and clinical research have provided novel insights into the mechanisms of lipotoxicity, including gut dysbiosis, epigenetic and epitranscriptomic modifications, dysfunction of lipid droplets, post-translational modifications, and altered membrane lipid composition. In this review, we discuss the recent knowledge on the mechanisms underlying the development of lipotoxicity and lipotoxic cardiometabolic disease in obesity, with a particular focus on lipotoxic and diabetic cardiomyopathy.
Collapse
Affiliation(s)
- Michinari Nakamura
- Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School, Rutgers Biomedical and Health Sciences, Newark, United States
| |
Collapse
|
43
|
Lambourne L, Mattioli K, Santoso C, Sheynkman G, Inukai S, Kaundal B, Berenson A, Spirohn-Fitzgerald K, Bhattacharjee A, Rothman E, Shrestha S, Laval F, Yang Z, Bisht D, Sewell JA, Li G, Prasad A, Phanor S, Lane R, Campbell DM, Hunt T, Balcha D, Gebbia M, Twizere JC, Hao T, Frankish A, Riback JA, Salomonis N, Calderwood MA, Hill DE, Sahni N, Vidal M, Bulyk ML, Fuxman Bass JI. Widespread variation in molecular interactions and regulatory properties among transcription factor isoforms. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.12.584681. [PMID: 38617209 PMCID: PMC11014633 DOI: 10.1101/2024.03.12.584681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/16/2024]
Abstract
Most human Transcription factors (TFs) genes encode multiple protein isoforms differing in DNA binding domains, effector domains, or other protein regions. The global extent to which this results in functional differences between isoforms remains unknown. Here, we systematically compared 693 isoforms of 246 TF genes, assessing DNA binding, protein binding, transcriptional activation, subcellular localization, and condensate formation. Relative to reference isoforms, two-thirds of alternative TF isoforms exhibit differences in one or more molecular activities, which often could not be predicted from sequence. We observed two primary categories of alternative TF isoforms: "rewirers" and "negative regulators", both of which were associated with differentiation and cancer. Our results support a model wherein the relative expression levels of, and interactions involving, TF isoforms add an understudied layer of complexity to gene regulatory networks, demonstrating the importance of isoform-aware characterization of TF functions and providing a rich resource for further studies.
Collapse
Affiliation(s)
- Luke Lambourne
- Center for Cancer Systems Biology (CCSB), Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Kaia Mattioli
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Clarissa Santoso
- Department of Biology, Boston University, Boston, MA, USA
- Bioinformatics Program, Boston University, Boston, MA, USA
| | - Gloria Sheynkman
- Center for Cancer Systems Biology (CCSB), Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Sachi Inukai
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Babita Kaundal
- Department of Epigenetics and Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Anna Berenson
- Molecular Biology, Cell Biology & Biochemistry Program, Boston University, Boston, MA, USA
| | - Kerstin Spirohn-Fitzgerald
- Center for Cancer Systems Biology (CCSB), Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Anukana Bhattacharjee
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
- Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Elisabeth Rothman
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | | | - Florent Laval
- Center for Cancer Systems Biology (CCSB), Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
- TERRA Teaching and Research Centre, University of Liège, Gembloux, Belgium
- Laboratory of Viral Interactomes, GIGA Institute, University of Liège, Liège, Belgium
| | - Zhipeng Yang
- Center for Cancer Systems Biology (CCSB), Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Deepa Bisht
- Department of Epigenetics and Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jared A Sewell
- Department of Biology, Boston University, Boston, MA, USA
| | - Guangyuan Li
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
- Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Anisa Prasad
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Harvard College, Cambridge MA, USA
| | - Sabrina Phanor
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Ryan Lane
- Department of Biology, Boston University, Boston, MA, USA
| | | | - Toby Hunt
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Dawit Balcha
- Center for Cancer Systems Biology (CCSB), Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Marinella Gebbia
- Center for Cancer Systems Biology (CCSB), Dana-Farber Cancer Institute, Boston, MA, USA
- The Donnelly Centre, University of Toronto, Toronto, Ontario, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
- Lunenfeld-Tanenbaum Research Institute (LTRI), Sinai Health System, Toronto, Ontario, Canada
| | - Jean-Claude Twizere
- TERRA Teaching and Research Centre, University of Liège, Gembloux, Belgium
- Laboratory of Viral Interactomes, GIGA Institute, University of Liège, Liège, Belgium
| | - Tong Hao
- Center for Cancer Systems Biology (CCSB), Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Adam Frankish
- Laboratory of Viral Interactomes, GIGA Institute, University of Liège, Liège, Belgium
| | - Josh A Riback
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Nathan Salomonis
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
- Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Michael A Calderwood
- Center for Cancer Systems Biology (CCSB), Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - David E Hill
- Center for Cancer Systems Biology (CCSB), Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Nidhi Sahni
- Department of Epigenetics and Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Marc Vidal
- Center for Cancer Systems Biology (CCSB), Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Martha L Bulyk
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Juan I Fuxman Bass
- Department of Biology, Boston University, Boston, MA, USA
- Bioinformatics Program, Boston University, Boston, MA, USA
- Molecular Biology, Cell Biology & Biochemistry Program, Boston University, Boston, MA, USA
| |
Collapse
|
44
|
Bai Y, Cui G, Sun X, Wei M, Liu Y, Guo J, Yang Y. Angiopoietin-Related Protein 4-Transcript 3 Increases the Proliferation, Invasion, and Migration of Hepatocellular Carcinoma Cells and Inhibits Apoptosis. DNA Cell Biol 2024; 43:175-184. [PMID: 38466955 DOI: 10.1089/dna.2023.0392] [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: 03/13/2024] Open
Abstract
To investigate the functional differences of angiopoietin-related protein 4 (ANGPTL4) transcripts in hepatocellular carcinoma (HCC) cells. By transfecting ANGPTL4-Transcript 1 and ANGPTL4-Transcript 3 overexpression vectors into HepG2 and Huh7 cell lines with ANGPTL4 knockdown, the effects of overexpression of two transcripts on cell viability, invasion, migration, and apoptosis were analyzed. The expression of two transcripts was compared in human liver cancer tissue, and their effects on tumor development were validated in vivo experiments in mice. Compared with control, the overexpression of ANGPTL4-Transcript 1 had no significant effect on viability, invasion, healing, and apoptosis of HepG2 and Huh7 cells. However, these two cell lines overexpressing ANGPTL4-Transcript 3 showed remarkably enhanced cell viability, invasive and healing ability, and decreased apoptosis ability. Furthermore, the mRNA level of ANGPTL4-Transcript 3 was significantly increased in human HCC tissues and promoted tumor growth compared with Transcript 1. Different transcripts of gene ANGPTL4 have distinct effects on HCC. The abnormally elevated Transcript 3 with the specific ability of promoting HCC proliferation, infiltration, and migration is expected to become a new biological marker and more precise intervention target for HCC.
Collapse
Affiliation(s)
- Yun Bai
- Department of Oncology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Guanghua Cui
- Department of Oncology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xiaoke Sun
- Department of Oncology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Meiqi Wei
- Department of Oncology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yanying Liu
- Department of Oncology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jialu Guo
- Department of Oncology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yu Yang
- Department of Oncology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
45
|
Sinha IR, Sandal PS, Burns GD, Mallika AP, Irwin KE, Cruz ALF, Wang V, Rodríguez JL, Wong PC, Ling JP. Large-scale RNA-seq mining reveals ciclopirox triggers TDP-43 cryptic exons. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.27.587011. [PMID: 38585725 PMCID: PMC10996699 DOI: 10.1101/2024.03.27.587011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
Nuclear clearance and cytoplasmic aggregation of TDP-43 in neurons, initially identified in ALS-FTD, are hallmark pathological features observed across a spectrum of neurodegenerative diseases. We previously found that TDP-43 loss-of-function leads to the transcriptome-wide inclusion of deleterious cryptic exons in brains and biofluids post-mortem as well as during the presymptomatic stage of ALS-FTD, but upstream mechanisms that lead to TDP-43 dysregulation remain unclear. Here, we developed a web-based resource (SnapMine) to determine the levels of TDP-43 cryptic exon inclusion across hundreds of thousands of publicly available RNA sequencing datasets. We established cryptic exon inclusion across a variety of human cells and tissues to provide ground truth references for future studies on TDP-43 dysregulation. We then explored studies that were entirely unrelated to TDP-43 or neurodegeneration and found that ciclopirox olamine (CPX), an FDA-approved antifungal, can trigger the inclusion of TDP-43-associated cryptic exons in a variety of mouse and human primary cells. CPX induction of cryptic exon occurs via heavy metal toxicity and oxidative stress, suggesting that similar vulnerabilities could play a role in neurodegeneration. Our work demonstrates how diverse datasets can be linked through common biological features and underscores that public archives of sequencing data represent a vastly underutilized resource with tremendous potential for uncovering novel insights into complex biological mechanisms and diseases.
Collapse
Affiliation(s)
- Irika R Sinha
- Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Parker S Sandal
- Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Grace D Burns
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | | | - Katherine E Irwin
- Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Anna Lourdes F Cruz
- Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Vania Wang
- Department of Oncology, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
- Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | | | - Philip C Wong
- Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Jonathan P Ling
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
46
|
Murali M, Saquing J, Lu S, Gao Z, Jordan B, Wakefield ZP, Fiszbein A, Cooper DR, Castaldi PJ, Korkin D, Sheynkman G. Biosurfer for systematic tracking of regulatory mechanisms leading to protein isoform diversity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.15.585320. [PMID: 38559226 PMCID: PMC10980011 DOI: 10.1101/2024.03.15.585320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Long-read RNA sequencing has shed light on transcriptomic complexity, but questions remain about the functionality of downstream protein products. We introduce Biosurfer, a computational approach for comparing protein isoforms, while systematically tracking the transcriptional, splicing, and translational variations that underlie differences in the sequences of the protein products. Using Biosurfer, we analyzed the differences in 32,799 pairs of GENCODE annotated protein isoforms, finding a majority (70%) of variable N-termini are due to the alternative transcription start sites, while only 9% arise from 5' UTR alternative splicing. Biosurfer's detailed tracking of nucleotide-to-residue relationships helped reveal an uncommonly tracked source of single amino acid residue changes arising from the codon splits at junctions. For 17% of internal sequence changes, such split codon patterns lead to single residue differences, termed "ragged codons". Of variable C-termini, 72% involve splice- or intron retention-induced reading frameshifts. We found an unusual pattern of reading frame changes, in which the first frameshift is closely followed by a distinct second frameshift that restores the original frame, which we term a "snapback" frameshift. We analyzed long read RNA-seq-predicted proteome of a human cell line and found similar trends as compared to our GENCODE analysis, with the exception of a higher proportion of isoforms predicted to undergo nonsense-mediated decay. Biosurfer's comprehensive characterization of long-read RNA-seq datasets should accelerate insights of the functional role of protein isoforms, providing mechanistic explanation of the origins of the proteomic diversity driven by the alternative splicing. Biosurfer is available as a Python package at https://github.com/sheynkman-lab/biosurfer.
Collapse
Affiliation(s)
- Mayank Murali
- Broad Institute of MIT and Harvard University, Cambridge, MA, USA
| | - Jamie Saquing
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, VA, USA
| | - Senbao Lu
- Bioinformatics and Computational Biology Program, Worcester Polytechnic Institute, Worcester, MA, USA
- Computer Science Department, Worcester Polytechnic Institute, Worcester, MA, USA
| | - Ziyang Gao
- Bioinformatics and Computational Biology Program, Worcester Polytechnic Institute, Worcester, MA, USA
- Computer Science Department, Worcester Polytechnic Institute, Worcester, MA, USA
| | - Ben Jordan
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, VA, USA
| | - Zachary Peters Wakefield
- Bioinformatics Program, Boston University, Boston, MA, USA
- Department of Biology, Boston University, Boston, MA, USA
| | - Ana Fiszbein
- Bioinformatics Program, Boston University, Boston, MA, USA
- Department of Biology, Boston University, Boston, MA, USA
| | - David R. Cooper
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, VA, USA
| | - Peter J. Castaldi
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital, Boston, MA, USA
- Division of General Medicine and Primary Care, Department of Medicine, Brigham and Women’s Hospital, Boston, MA, USA
| | - Dmitry Korkin
- Bioinformatics and Computational Biology Program, Worcester Polytechnic Institute, Worcester, MA, USA
- Computer Science Department, Worcester Polytechnic Institute, Worcester, MA, USA
| | - Gloria Sheynkman
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, VA, USA
- Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, VA, USA
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA, USA
- UVA Cancer Center, University of Virginia, Charlottesville, VA, USA
| |
Collapse
|
47
|
Jablonowski CM, Quarni W, Singh S, Tan H, Bostanthirige DH, Jin H, Fang J, Chang TC, Finkelstein D, Cho JH, Hu D, Pagala V, Sakurada SM, Pruett-Miller SM, Wang R, Murphy A, Freeman K, Peng J, Davidoff AM, Wu G, Yang J. Metabolic reprogramming of cancer cells by JMJD6-mediated pre-mRNA splicing associated with therapeutic response to splicing inhibitor. eLife 2024; 12:RP90993. [PMID: 38488852 PMCID: PMC10942784 DOI: 10.7554/elife.90993] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/17/2024] Open
Abstract
Dysregulated pre-mRNA splicing and metabolism are two hallmarks of MYC-driven cancers. Pharmacological inhibition of both processes has been extensively investigated as potential therapeutic avenues in preclinical and clinical studies. However, how pre-mRNA splicing and metabolism are orchestrated in response to oncogenic stress and therapies is poorly understood. Here, we demonstrate that jumonji domain containing 6, arginine demethylase, and lysine hydroxylase, JMJD6, acts as a hub connecting splicing and metabolism in MYC-driven human neuroblastoma. JMJD6 cooperates with MYC in cellular transformation of murine neural crest cells by physically interacting with RNA binding proteins involved in pre-mRNA splicing and protein homeostasis. Notably, JMJD6 controls the alternative splicing of two isoforms of glutaminase (GLS), namely kidney-type glutaminase (KGA) and glutaminase C (GAC), which are rate-limiting enzymes of glutaminolysis in the central carbon metabolism in neuroblastoma. Further, we show that JMJD6 is correlated with the anti-cancer activity of indisulam, a 'molecular glue' that degrades splicing factor RBM39, which complexes with JMJD6. The indisulam-mediated cancer cell killing is at least partly dependent on the glutamine-related metabolic pathway mediated by JMJD6. Our findings reveal a cancer-promoting metabolic program is associated with alternative pre-mRNA splicing through JMJD6, providing a rationale to target JMJD6 as a therapeutic avenue for treating MYC-driven cancers.
Collapse
Affiliation(s)
| | - Waise Quarni
- Department of Surgery, St Jude Children’s Research HospitalMemphisUnited States
| | - Shivendra Singh
- Department of Surgery, St Jude Children’s Research HospitalMemphisUnited States
| | - Haiyan Tan
- Center for Proteomics and Metabolomics, St Jude Children's Research HospitalMemphisUnited States
| | | | - Hongjian Jin
- Center for Applied Bioinformatics, St Jude Children’s Research HospitalMemphisUnited States
| | - Jie Fang
- Department of Surgery, St Jude Children’s Research HospitalMemphisUnited States
| | - Ti-Cheng Chang
- Center for Applied Bioinformatics, St Jude Children’s Research HospitalMemphisUnited States
| | - David Finkelstein
- Center for Applied Bioinformatics, St Jude Children’s Research HospitalMemphisUnited States
| | - Ji-Hoon Cho
- Center for Proteomics and Metabolomics, St Jude Children's Research HospitalMemphisUnited States
| | - Dongli Hu
- Department of Surgery, St Jude Children’s Research HospitalMemphisUnited States
| | - Vishwajeeth Pagala
- Center for Proteomics and Metabolomics, St Jude Children's Research HospitalMemphisUnited States
| | - Sadie Miki Sakurada
- Department of Cell and Molecular Biology, St Jude Children's Research HospitalMemphisUnited States
| | - Shondra M Pruett-Miller
- Department of Cell and Molecular Biology, St Jude Children's Research HospitalMemphisUnited States
| | - Ruoning Wang
- Center for Childhood Cancer and Blood Disease, Abigail Wexner Research Institute, Nationwide Children’s HospitalColumbusUnited States
| | - Andrew Murphy
- Department of Surgery, St Jude Children’s Research HospitalMemphisUnited States
| | - Kevin Freeman
- Genetics, Genomics & Informatics, The University of Tennessee Health Science Center (UTHSC)MemphisUnited States
| | - Junmin Peng
- Department of Structural Biology, St Jude Children’s Research HospitalMemphisUnited States
| | - Andrew M Davidoff
- Department of Surgery, St Jude Children’s Research HospitalMemphisUnited States
- St Jude Graduate School of Biomedical Sciences, St Jude Children’s Research HospitalMemphisUnited States
- Department of Pathology and Laboratory Medicine, College of Medicine, The University of Tennessee Health Science CenterMemphisUnited States
| | - Gang Wu
- Center for Applied Bioinformatics, St Jude Children’s Research HospitalMemphisUnited States
| | - Jun Yang
- Department of Surgery, St Jude Children’s Research HospitalMemphisUnited States
- St Jude Graduate School of Biomedical Sciences, St Jude Children’s Research HospitalMemphisUnited States
- Department of Pathology and Laboratory Medicine, College of Medicine, The University of Tennessee Health Science CenterMemphisUnited States
- College of Graduate Health Sciences, University of Tennessee Health Science CenterMemphisUnited States
| |
Collapse
|
48
|
Levillayer L, Brighelli C, Demeret C, Sakuntabhai A, Bureau JF. Role of two modules controlling the interaction between SKAP1 and SRC kinases comparison with SKAP2 architecture and consequences for evolution. PLoS One 2024; 19:e0296230. [PMID: 38483858 PMCID: PMC10939263 DOI: 10.1371/journal.pone.0296230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 02/28/2024] [Indexed: 03/17/2024] Open
Abstract
SRC kinase associated phosphoprotein 1 (SKAP1), an adaptor for protein assembly, plays an important role in the immune system such as stabilizing immune synapses. Understanding how these functions are controlled at the level of the protein-protein interactions is necessary to describe these processes and to develop therapeutics. Here, we dissected the SKAP1 modular organization to recognize SRC kinases and compared it to that of its paralog SRC kinase associated phosphoprotein 2 (SKAP2). Different conserved motifs common to either both proteins or specific to SKAP2 were found using this comparison. Two modules harboring different binding properties between SKAP1 and SKAP2 were identified: one composed of two conserved motifs located in the second interdomain interacting at least with the SH2 domain of SRC kinases and a second one composed of the DIM domain modulated by the SH3 domain and the activation of SRC kinases. This work suggests a convergent evolution of the binding properties of some SRC kinases interacting specifically with either SKAP1 or SKAP2.
Collapse
Affiliation(s)
- Laurine Levillayer
- Institut Pasteur, Institut National de Recherche pour l’Agriculture, Université de Paris-Cité, CNRS UMR 2000, l’Alimentation et l’Environnement (INRAE) USC 1510, Unité Écologie et Émergence des Pathogènes Transmis par les Arthropodes (EEPTA), Paris, France
| | - Camille Brighelli
- Institut Pasteur, Institut National de Recherche pour l’Agriculture, Université de Paris-Cité, CNRS UMR 2000, l’Alimentation et l’Environnement (INRAE) USC 1510, Unité Écologie et Émergence des Pathogènes Transmis par les Arthropodes (EEPTA), Paris, France
| | - Caroline Demeret
- Institut Pasteur, Université de Paris-Cité, Laboratoire Interactomique, ARN et Immunité ‐ Interactomics, RNA and Immunity, Paris, France
| | - Anavaj Sakuntabhai
- Institut Pasteur, Institut National de Recherche pour l’Agriculture, Université de Paris-Cité, CNRS UMR 2000, l’Alimentation et l’Environnement (INRAE) USC 1510, Unité Écologie et Émergence des Pathogènes Transmis par les Arthropodes (EEPTA), Paris, France
| | - Jean-François Bureau
- Institut Pasteur, Institut National de Recherche pour l’Agriculture, Université de Paris-Cité, CNRS UMR 2000, l’Alimentation et l’Environnement (INRAE) USC 1510, Unité Écologie et Émergence des Pathogènes Transmis par les Arthropodes (EEPTA), Paris, France
| |
Collapse
|
49
|
Sun L, Hui F, Tang GY, Shen HL, Cao XL, Gao JX, Li LF. Selective degradation of PL2L60 by metabolic stresses‑induced autophagy suppresses multi‑cancer growth. Oncol Rep 2024; 51:41. [PMID: 38624021 PMCID: PMC10823339 DOI: 10.3892/or.2024.8700] [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: 06/22/2023] [Accepted: 11/08/2023] [Indexed: 04/17/2024] Open
Abstract
It has been reported that PL2L60 proteins, a product of PIWIL2 gene which might be activated by an intragenic promoter, could mediate a common pathway specifically for tumorigenesis. In the present study, it was further identified by using western blot assay that the PL2L60 proteins could be degraded in cancer cells through a mechanism of selective autophagy in response to oxidative stress. The PL2L60 was downregulated in various types of cancer cells under the hypoxic condition independently of HIF‑1α, resulting in apoptosis of cancer cells. Inhibition of autophagy by small interfering RNA targeting of either Beclin‑1 (BECN1) or Atg5 resulted in restoration of PL2L60 expression in hypoxic cancer cell. The hypoxic degradation of PL2L60 was also blocked by the attenuation of the autophagosome membrane protein Atg8/microtubule‑associated protein 1 light chain 3 (LC3) or autophagy cargo protein p62 expression. Surprisingly, Immunofluorescence analysis demonstrated that LC3 could be directly bound to PL2L60 and was required for the transport of PL2L60 from the nucleus to the cytoplasm for lysosomal flux under basal or activated autophagy in cancer cells. Moreover, flow cytometric analysis displayed that knocking down of PL2L60 mRNA but not PIWIL2 mRNA effectively inhibited cancer cell proliferation and promoted apoptosis of cancer cells. The similar results were obtained from in vivo tumorigenic experiment, in which PL2L60 downregulation in necroptosis areas was confirmed by immunohistochemistry. These results suggested that various cancer could be suppressed by promoting autophagy. The present study revealed a key role of autophagic degradation of PL2L60 in hypoxia‑induced cancer cell death, which could be used as a novel therapeutic target of cancer.
Collapse
Affiliation(s)
- Lei Sun
- The State Key Laboratory of Oncogenes and Related Genes, and The Laboratory of Tumorigenesis and Immunity, Renji-Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, PuDong, Shanghai 200127, P.R. China
- Department of Oncology, First Affiliated Hospital of Weifang Medical University, Weifang, Shandong 261000, P.R. China
| | - Fu Hui
- Department of Oncology, First Affiliated Hospital of Weifang Medical University, Weifang, Shandong 261000, P.R. China
| | - Gao-Yan Tang
- Department of Oncology, First Affiliated Hospital of Weifang Medical University, Weifang, Shandong 261000, P.R. China
| | - Hai-Lian Shen
- Sam and Ann Barshop Institute for Longevity of Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, TX 78292, USA
| | - Xue-Lei Cao
- Department of Clinical Laboratory, Qi Lu Hospital of Shandong University, Jinan, Shandong 250012, P.R. China
| | - Jian-Xin Gao
- The State Key Laboratory of Oncogenes and Related Genes, and The Laboratory of Tumorigenesis and Immunity, Renji-Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, PuDong, Shanghai 200127, P.R. China
| | - Lin-Feng Li
- The State Key Laboratory of Oncogenes and Related Genes, and The Laboratory of Tumorigenesis and Immunity, Renji-Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, PuDong, Shanghai 200127, P.R. China
| |
Collapse
|
50
|
Figueroa-Navedo AM, Ivanov AR. Experimental and data analysis advances in thermal proteome profiling. CELL REPORTS METHODS 2024; 4:100717. [PMID: 38412830 PMCID: PMC10921035 DOI: 10.1016/j.crmeth.2024.100717] [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: 11/06/2023] [Revised: 12/17/2023] [Accepted: 02/05/2024] [Indexed: 02/29/2024]
Abstract
Method development for mass spectrometry (MS)-based thermal shift proteomic assays have advanced to probe small molecules with known and unknown protein-ligand interaction mechanisms and specificity, which is predominantly used in characterization of drug-protein interactions. In the discovery of target and off-target protein-ligand interactions, a thorough investigation of method development and their impact on the sensitivity and accuracy of protein-small molecule and protein-protein interactions is warranted. In this review, we discuss areas of improvement at each stage of thermal proteome profiling data analysis that includes processing of MS-based data, method development, and their effect on the overall quality of thermal proteome profiles. We also overview the optimization of experimental strategies and prioritization of an increased number of independent biological replicates over the number of evaluated temperatures.
Collapse
Affiliation(s)
- Amanda M Figueroa-Navedo
- Barnett Institute of Chemical and Biological Analysis, Department of Chemistry and Chemical Biology, Northeastern University, Boston, MA 02115, USA
| | - Alexander R Ivanov
- Barnett Institute of Chemical and Biological Analysis, Department of Chemistry and Chemical Biology, Northeastern University, Boston, MA 02115, USA.
| |
Collapse
|