1
|
Zhang M, Lu Z. tRNA modifications: greasing the wheels of translation and beyond. RNA Biol 2025; 22:1-25. [PMID: 39723662 DOI: 10.1080/15476286.2024.2442856] [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] [Revised: 11/29/2024] [Accepted: 12/11/2024] [Indexed: 12/28/2024] Open
Abstract
Transfer RNA (tRNA) is one of the most abundant RNA types in cells, acting as an adaptor to bridge the genetic information in mRNAs with the amino acid sequence in proteins. Both tRNAs and small fragments processed from them play many nonconventional roles in addition to translation. tRNA molecules undergo various types of chemical modifications to ensure the accuracy and efficiency of translation and regulate their diverse functions beyond translation. In this review, we discuss the biogenesis and molecular mechanisms of tRNA modifications, including major tRNA modifications, writer enzymes, and their dynamic regulation. We also summarize the state-of-the-art technologies for measuring tRNA modification, with a particular focus on 2'-O-methylation (Nm), and discuss their limitations and remaining challenges. Finally, we highlight recent discoveries linking dysregulation of tRNA modifications with genetic diseases.
Collapse
Affiliation(s)
- Minjie Zhang
- Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Tianjin, China
- Tianjin Key Laboratory of Medical Epigenetics, Department of Bioinformatics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Zhipeng Lu
- Department of Pharmacology and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA, USA
- Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, USA
- Eli and Edythe Broad CIRM Center for Regenerative Medicine and Stem Cell Research, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
2
|
Gao Y, Liu X, Li J. Targeting tRNA methyltransferases: from molecular mechanisms to drug discovery. SCIENCE CHINA. LIFE SCIENCES 2025:10.1007/s11427-024-2886-2. [PMID: 40347212 DOI: 10.1007/s11427-024-2886-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Accepted: 01/24/2025] [Indexed: 05/12/2025]
Abstract
Transfer RNA methyltransferases (tRNA MTases) catalyze site-specific methylation on tRNAs, a critical process that ensures the stability and functionality of tRNA molecules, thereby maintaining cellular homeostasis of tRNA methylation. Recent studies have illuminated the structural diversity, specific substrate recognition, and conserved catalytic mechanisms of tRNA MTases, revealing how their dysregulation contributes to various diseases, including cancers and neurodevelopmental disorders. This review integrates these advances, exploring the challenges of achieving precise substrate recognition and modification in the context of complex and specific tRNA modification landscape, while emphasizing the crucial role of tRNA MTases in disease pathogenesis. The identification of small-molecule inhibitors targeting specific tRNA MTases marks a promising step toward the development of novel therapies. With continued research into the broader biological functions and regulatory mechanisms of tRNA MTases, these insights hold great potential to drive clinical advancements and therapeutic innovations.
Collapse
Affiliation(s)
- Yanrong Gao
- Institutes of Biomedical Sciences, Inner Mongolia University, Hohhot, 010020, China
- School of Life Sciences, Inner Mongolia University, Hohhot, 010020, China
| | - Xinyu Liu
- Institutes of Biomedical Sciences, Inner Mongolia University, Hohhot, 010020, China
- School of Life Sciences, Inner Mongolia University, Hohhot, 010020, China
| | - Jiazhi Li
- Institutes of Biomedical Sciences, Inner Mongolia University, Hohhot, 010020, China.
- School of Life Sciences, Inner Mongolia University, Hohhot, 010020, China.
| |
Collapse
|
3
|
Li Z, Zhang T, Yang X, Peng Y. Role of noncoding RNA and protein interaction in pancreatic cancer. Chin Med J (Engl) 2025; 138:1019-1036. [PMID: 40205638 PMCID: PMC12068769 DOI: 10.1097/cm9.0000000000003587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2025] [Indexed: 04/11/2025] Open
Abstract
ABSTRACT Noncoding RNAs (ncRNAs) are a class of RNA molecules with little or no protein-coding potential. Emerging evidence indicates that ncRNAs are frequently dysregulated and play pivotal roles in the pathogenesis of pancreatic cancer. Their aberrant expression can arise from chromosomal abnormalities, dysregulated transcriptional control, and epigenetic modifications. ncRNAs function as protein scaffolds or molecular decoys to modulate interactions between proteins and other biomolecules, thereby regulating gene expression and contributing to pancreatic cancer progression. In this review, we summarize the mechanisms underlying ncRNA dysregulation in pancreatic cancer, emphasize the biological significance of ncRNA-protein interactions, and highlight their clinical relevance. A deeper understanding of ncRNA-protein interactions is essential to elucidate molecular mechanisms and advance translational research in pancreatic cancer.
Collapse
Affiliation(s)
- Zhang Li
- Center for Molecular Oncology, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Tingting Zhang
- Center for Molecular Oncology, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Xiaojuan Yang
- Center for Molecular Oncology, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Yong Peng
- Center for Molecular Oncology, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| |
Collapse
|
4
|
Bartusel M, Kim SX, Rehimi R, Darnell AM, Nikolić M, Heggemann J, Kolovos P, van Ijcken WFJ, Varineau J, Crispatzu G, Mangold E, Brugmann SA, Vander Heiden MG, Laugsch M, Ludwig KU, Rada-Iglesias A, Calo E. A non-syndromic orofacial cleft risk locus links tRNA splicing defects to neural crest cell pathologies. Am J Hum Genet 2025; 112:1097-1116. [PMID: 40250422 DOI: 10.1016/j.ajhg.2025.03.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2025] [Revised: 03/20/2025] [Accepted: 03/21/2025] [Indexed: 04/20/2025] Open
Abstract
Orofacial clefts are the most common form of congenital craniofacial malformation worldwide. The etiology of these birth defects is multifactorial, involving genetic and environmental factors. However, in most cases, the underlying causes remain unexplained, precluding a molecular understanding of disease mechanisms. Here, we integrated genome-wide association data, targeted resequencing of case and control cohorts, tissue- and cell-type-specific epigenomic profiling, and genome architecture analyses to molecularly dissect a genomic locus associated with an increased risk of non-syndromic orofacial cleft. We found that common and rare risk variants associated with orofacial cleft intersect with an enhancer (e2p24.2) that is active in human embryonic craniofacial tissue. We mapped e2p24.2 long-range interactions to a topologically associated domain harboring MYCN, DDX1, and CYRIA. We found that MYCN and DDX1, but not CYRIA, are required during craniofacial development in chicken embryos. We investigated the role of DDX1, a key component of the tRNA splicing complex, in cranial neural crest cells (cNCCs). The loss of DDX1 in cNCCs resulted in the accumulation of unspliced tRNA fragments, depletion of mature intron-containing tRNAs, and ribosome stalling at codons decoded by these tRNAs. This was accompanied by defects in both global protein synthesis and cNCC migration. We further showed that the induction of tRNA fragments is sufficient to disrupt craniofacial development. Together, these results uncovered a molecular mechanism in which impaired tRNA splicing affects cNCCs and craniofacial development and positioned MYCN, DDX1, and tRNA processing defects as risk factors in the pathogenesis of orofacial clefts.
Collapse
Affiliation(s)
- Michaela Bartusel
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA; Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany.
| | - Skylar X Kim
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Rizwan Rehimi
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany; Institute for Pharmacology and Toxicology (IPT), University Hospital Bonn, Venusberg-Campus 1, Biomedical Center 1 (Building 13), 53127 Bonn, Germany
| | - Alicia M Darnell
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Miloš Nikolić
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Julia Heggemann
- Institute of Human Genetics, University of Bonn, University Hospital Bonn, Medical Faculty, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Petros Kolovos
- Department of Molecular Biology and Genetics, Democritus University of Thrace, Alexandroupolis, Greece
| | | | - Jade Varineau
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Giuliano Crispatzu
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Elisabeth Mangold
- Institute of Human Genetics, University of Bonn, University Hospital Bonn, Medical Faculty, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Samantha A Brugmann
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, MLC 7007, Cincinnati, OH 45229, USA; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| | - Matthew G Vander Heiden
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA; Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA; Dana-Farber Cancer Institute, Boston, MA, USA
| | - Magdalena Laugsch
- Institute of Human Genetics, Heidelberg University, Heidelberg, Germany
| | - Kerstin U Ludwig
- Institute of Human Genetics, University of Bonn, University Hospital Bonn, Medical Faculty, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Alvaro Rada-Iglesias
- Institute of Biomedicine and Biotechnology of Cantabria (IBBTEC), CSIC/University of Cantabria, Santander, Spain
| | - Eliezer Calo
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA; Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA.
| |
Collapse
|
5
|
Yu J, Zhang X, Cai C, Zhou T, Chen Q. Small RNA and Toll-like receptor interactions: origins and disease mechanisms. Trends Biochem Sci 2025; 50:385-401. [PMID: 39956743 PMCID: PMC12048287 DOI: 10.1016/j.tibs.2025.01.004] [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: 11/04/2024] [Revised: 01/08/2025] [Accepted: 01/17/2025] [Indexed: 02/18/2025]
Abstract
Advances in small RNA sequencing have revealed diverse small noncoding RNAs (sncRNAs) beyond microRNAs (miRNAs), derived from transfer RNAs (tRNAs), ribosomal RNAs (rRNAs), small nuclear RNAs (snRNAs), and Y RNAs, carrying distinct RNA modifications. These emerging sncRNAs can function beyond RNA interference (RNAi), adopting aptamer-like roles by interacting with Toll-like receptors 7 and 8 (TLR7 and TLR8) via specific sequences, modifications, and structures. We propose a Sequential Activation Hypothesis where initial abnormal sncRNAs - triggered by infections or stresses - activate TLR7/8, leading to autoantibody production against autoantigens like RNA-binding proteins La and Ro. These autoantibody-antigen complexes further promote secondary immunogenic sncRNA production and repetitive TLR7/8 activation, perpetuating a vicious cycle sustaining autoimmunity. TLR7/8's X chromosome location and sex-biased expression contribute to female-dominant autoimmune diseases. Understanding sncRNA-TLR interactions is essential for designing novel therapeutic strategies.
Collapse
Affiliation(s)
- Jiancheng Yu
- Department of Human Genetics, University of Utah School of Medicine, Salt Lake City, UT, USA; Molecular Medicine Program, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Xudong Zhang
- Department of Human Genetics, University of Utah School of Medicine, Salt Lake City, UT, USA; Molecular Medicine Program, University of Utah School of Medicine, Salt Lake City, UT, USA; Division of Urology, Department of Surgery, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Chen Cai
- Department of Human Genetics, University of Utah School of Medicine, Salt Lake City, UT, USA; Molecular Medicine Program, University of Utah School of Medicine, Salt Lake City, UT, USA; Division of Urology, Department of Surgery, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Tong Zhou
- Department of Physiology and Cell Biology, University of Nevada, Reno School of Medicine, Reno, NV, USA.
| | - Qi Chen
- Department of Human Genetics, University of Utah School of Medicine, Salt Lake City, UT, USA; Molecular Medicine Program, University of Utah School of Medicine, Salt Lake City, UT, USA; Division of Urology, Department of Surgery, University of Utah School of Medicine, Salt Lake City, UT, USA.
| |
Collapse
|
6
|
Xiao Y, Liu C, Guan X, Fu Y, Fu M, Hong S, Wang C, Zhou Y, Li Y, He M, Zhang X, Guo H. Plasma tRNA-derived small RNAs mediated the association between benzo[a]pyrene exposure with lung cancer risk among Chinese adults from a case-control study. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2025; 295:118129. [PMID: 40187215 DOI: 10.1016/j.ecoenv.2025.118129] [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: 12/20/2024] [Revised: 02/28/2025] [Accepted: 03/28/2025] [Indexed: 04/07/2025]
Abstract
Exposure to benzo[a]pyrene (BaP) could lead to the development of lung cancer, but the potential mechanisms are complex and remain largely unclear. Plasma tRNA-derived small RNA (tsRNA), a new type epigenetic biomarker of cancer, might link environment exposure with lung cancer. Based on this case-control study of 272 participants recruited from China, we aim to reveal the plasma tsRNA signature of lung cancer and their mediation effects on BaP-induced lung cancer. We sequenced tsRNA profiles and detected the adducts of Benzo[a]pyrene diol epoxide-albumin (BPDE-Alb) in plasma samples. We applied linear regression models to estimate associations between the adducts of BPDE-Alb, normalized tsRNA and the risk of lung cancer. We used mediation analysis to explore the mediation roles of tsRNAs in BaP-associated lung cancer. We identified 104 tsRNAs significantly related to the risk of lung cancer (Bonferroni adjusted P < 0.05). Among these 104 tsRNAs, 17 tsRNAs were expressed at different concentrations between before and after lung cancer surgery groups with |fold change| > 2 at Bonferroni adjusted P < 0.05. In addition, of these 17 tsRNAs, the adducts of BPDE-Alb were associated with 5'-M-tRNA-Glu-CTC-1-1-L36, i-M-tRNA-Met-CAT-1-1-L24_pos29 and i-M-tRNA-Ala-AGC-2-2-L16_pos16, which could mediate a separate 40.8 %, 39.1 % and 36.8 % of the association between BaP exposure with lung cancer. Our study identified tsRNAs potentially mediating BaP-associated lung cancer development, and revealed tsRNAs as promising biomarkers of lung cancer.
Collapse
Affiliation(s)
- Yang Xiao
- Department of Occupational and Environmental Health, State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Chenliang Liu
- Department of Occupational and Environmental Health, State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xin Guan
- Department of Occupational and Environmental Health, State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Ye Fu
- Department of Occupational and Environmental Health, State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Ming Fu
- Department of Occupational and Environmental Health, State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Shiru Hong
- Department of Occupational and Environmental Health, State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Chenming Wang
- Department of Occupational and Environmental Health, State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yuhan Zhou
- Department of Occupational and Environmental Health, State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yangkai Li
- Department of Thoracic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Meian He
- Department of Occupational and Environmental Health, State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xiaomin Zhang
- Department of Occupational and Environmental Health, State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Huan Guo
- Department of Occupational and Environmental Health, State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.
| |
Collapse
|
7
|
Vaknine Treidel S, Lobmaier SM, Sharma R, Madrer N, Dubnov S, Shulman D, Greenberg P, Bennett ER, Greenberg DS, Turjeman A, Zelgert C, Zimmermann P, Frasch MG, Carmel L, Antonelli MC, Soreq H. Maternal prenatal stress induces sex-dependent changes in tRNA fragment families and cholinergic pathways in newborns. Mol Psychiatry 2025:10.1038/s41380-025-03011-2. [PMID: 40188313 DOI: 10.1038/s41380-025-03011-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Revised: 03/05/2025] [Accepted: 03/31/2025] [Indexed: 04/07/2025]
Abstract
Maternal perceived prenatal stress (PPS) is a known risk factor for diverse developmental impairments in newborns, but the underlying molecular processes are incompletely understood. Here, we report that maternal PPS altered the birth profiles of blood transfer RNA fragments (tRFs), 16-50 nt long non-random cleavage products of tRNAs, in a sex-dependent manner. Importantly, comparing stressed versus control maternal and umbilical cord blood serum presented alterations that were not limited to individual tRFs, but rather reflected selective changes in particular tRF families grouped by their mitochondrial or nuclear genome origin, parental tRNA coded amino acid, and cleavage type. Specifically, tRF families that show stress- and sex-specific effects, revealed shared length and expression patterns which were strongest in the female newborns. Several of these tRFs carry complementary motifs to particular cholinergic mRNAs, suggesting possible translational regulation similar to microRNAs. Compatible with the cholinergic regulation of stress reactions, those "CholinotRFs" achieved an AUC of 95% when classifying female newborns according to maternal PPS. Moreover, we found altered catalytic activity of serum acetylcholinesterase, which was particularly elevated in male newborns, marking a second sex-specific effect. Our findings demonstrate an association of tRF families' patterns with newborns' sex-specific stress response to PPS and may lead to better diagnosis and therapeutic tools for these and other stressors.
Collapse
Affiliation(s)
- Shani Vaknine Treidel
- Department of Biological Chemistry and the Edmond and Lily Safra Center of Brain Science, the Hebrew University of Jerusalem, Jerusalem, Israel
- Department of Genetics, the Hebrew University of Jerusalem, Jerusalem, Israel
| | - Silvia M Lobmaier
- Department of Obstetrics and Gynecology, Technical University of Munich, Munich, Germany
| | - Ritika Sharma
- Department of Obstetrics and Gynecology, Technical University of Munich, Munich, Germany
- Helmholtz Centre Munich, German Research Center for Health and Environment (GmbH), Munich, Germany
| | - Nimrod Madrer
- Department of Biological Chemistry and the Edmond and Lily Safra Center of Brain Science, the Hebrew University of Jerusalem, Jerusalem, Israel
| | - Serafima Dubnov
- Department of Biological Chemistry and the Edmond and Lily Safra Center of Brain Science, the Hebrew University of Jerusalem, Jerusalem, Israel
| | - Dana Shulman
- Department of Biological Chemistry and the Edmond and Lily Safra Center of Brain Science, the Hebrew University of Jerusalem, Jerusalem, Israel
| | - Pnina Greenberg
- Department of Biological Chemistry and the Edmond and Lily Safra Center of Brain Science, the Hebrew University of Jerusalem, Jerusalem, Israel
| | - Estelle R Bennett
- Department of Biological Chemistry and the Edmond and Lily Safra Center of Brain Science, the Hebrew University of Jerusalem, Jerusalem, Israel
| | - David S Greenberg
- Department of Biological Chemistry and the Edmond and Lily Safra Center of Brain Science, the Hebrew University of Jerusalem, Jerusalem, Israel
| | - Adi Turjeman
- Genomic Center, the Hebrew University of Jerusalem, Jerusalem, Israel
| | - Camilla Zelgert
- Department of Obstetrics and Gynecology, Technical University of Munich, Munich, Germany
| | - Peter Zimmermann
- Department of Obstetrics and Gynecology, Technical University of Munich, Munich, Germany
| | - Martin G Frasch
- Department of Obstetrics and Gynecology and Institute on Human Development and Disability (IHDD), University of Washington, Seattle, WA, USA
| | - Liran Carmel
- Department of Genetics, the Hebrew University of Jerusalem, Jerusalem, Israel
| | - Marta C Antonelli
- Department of Obstetrics and Gynecology, Technical University of Munich, Munich, Germany
- Instituto de Biología Celular y Neurociencias "Prof. E. De Robertis", Facultad De Medicina, UBA, Buenos Aires, Argentina
| | - Hermona Soreq
- Department of Biological Chemistry and the Edmond and Lily Safra Center of Brain Science, the Hebrew University of Jerusalem, Jerusalem, Israel.
| |
Collapse
|
8
|
Shi J, Zhang Y, Li Y, Zhang L, Zhang X, Yan M, Chen Q, Zhang Y. Optimized identification and characterization of small RNAs with PANDORA-seq. Nat Protoc 2025:10.1038/s41596-025-01158-4. [PMID: 40181099 DOI: 10.1038/s41596-025-01158-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 02/11/2025] [Indexed: 04/05/2025]
Abstract
Small noncoding RNAs (sncRNAs) are a diverse group of RNAs including small interfering RNAs, microRNAs, PIWI-interacting RNAs and RNAs derived from structured RNAs such as transfer RNAs, ribosomal RNAs and others. These sncRNAs have varied termini and RNA modifications, which can interfere with adaptor ligation and reverse transcription during cDNA library construction, hindering detection of many types of sncRNA by standard small RNA sequencing methods. To address this limitation, PANDORA sequencing introduces a refined methodology. The procedure includes sequential enzymatic treatments of size-selected RNAs with T4PNK and AlkB, which effectively circumvent the challenges presented by the ligation-blocking termini and reverse transcription-blocking RNA modifications, followed by tailored small RNA library construction protocols and deep sequencing. The obtained datasets are analyzed with the SPORTS pipeline, which can comprehensively analyze various types of sncRNA beyond the traditionally studied classes, to include those derived from various parental RNAs (for example, from transfer RNA and ribosomal RNA), as well as output the locations on the parental RNA from which these sncRNAs are derived. The entire protocol takes ~7 d, depending on the sample size and sequencing turnaround time. PANDORA sequencing provides a transformative tool to further our understanding of the expanding small RNA universe and to explore the uncharted functions of sncRNAs.
Collapse
Affiliation(s)
- Junchao Shi
- China National Center for Bioinformation and Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China.
| | - Yunfang Zhang
- Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, China.
- Sycamore Research Institute of Life Sciences, Shanghai, China.
| | - Yun Li
- Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Liwen Zhang
- China National Center for Bioinformation and Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China
| | - Xudong Zhang
- Molecular Medicine Program, University of Utah School of Medicine, Salt Lake City, UT, USA
- Division of Urology, Department of Surgery, University of Utah School of Medicine, Salt Lake City, UT, USA
- Department of Human Genetics, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Menghong Yan
- Pudong Medical Center, Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Qi Chen
- Molecular Medicine Program, University of Utah School of Medicine, Salt Lake City, UT, USA
- Division of Urology, Department of Surgery, University of Utah School of Medicine, Salt Lake City, UT, USA
- Department of Human Genetics, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Ying Zhang
- The Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, Beijing Key Laboratory of Genetic Engineering Drug and Biotechnology, College of Life Sciences, Beijing Normal University, Beijing, China.
| |
Collapse
|
9
|
Nguyen NYT, Liu X, Dutta A, Su Z. The Secret Life of N 1-methyladenosine: A Review on its Regulatory Functions. J Mol Biol 2025:169099. [PMID: 40139310 DOI: 10.1016/j.jmb.2025.169099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 03/15/2025] [Accepted: 03/20/2025] [Indexed: 03/29/2025]
Abstract
N1-methyladenosine (m1A) is a conserved modification on house-keeping RNAs, including tRNAs and rRNAs. With recent advancement on m1A detection and mapping, m1A is revealed to have a secret life with regulatory functions. This includes the regulation of its canonical substrate tRNAs, and expands into new territories such as tRNA fragments, mRNAs and repeat RNAs. The dynamic regulation of m1A has been shown in different biological contexts, including stress response, diet, T cell activation and aging. Interestingly, m1A can also be installed by non-enzymatic mechanisms. However, technical challenges remain in m1A site mapping; as a result, controversies have been observed across different labs or different methods. In this review we will summarize the recent development of m1A detection, its dynamic regulation, and its biological functions on diverse RNA substrates.
Collapse
Affiliation(s)
- Nhi Yen Tran Nguyen
- Department of Genetics, University of Alabama at Birmingham, Birmingham, AL 35233, United States
| | - Xisheng Liu
- Department of Genetics, University of Alabama at Birmingham, Birmingham, AL 35233, United States
| | - Anindya Dutta
- Department of Genetics, University of Alabama at Birmingham, Birmingham, AL 35233, United States; O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL 35233, United States
| | - Zhangli Su
- Department of Genetics, University of Alabama at Birmingham, Birmingham, AL 35233, United States; O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL 35233, United States.
| |
Collapse
|
10
|
Wang F, Li P, Yan X, Yue A, Xu J, Shao Y, Zhang K, Zhang Q, Li Y, Sun K. Novel therapeutic insights into pathological cardiac hypertrophy: tRF-16-R29P4PE regulates PACE4 and metabolic pathways. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2025; 1872:119920. [PMID: 39947523 DOI: 10.1016/j.bbamcr.2025.119920] [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: 09/13/2024] [Revised: 01/06/2025] [Accepted: 02/07/2025] [Indexed: 02/21/2025]
Abstract
Pathological cardiac hypertrophy (PCH) is a complex condition with an incompletely understood pathogenesis. Emerging evidence suggests that transfer RNA-derived small RNAs (tsRNAs) may play a significant role in various cellular processes, yet their impact on PCH remains unexplored. In this study, we performed tsRNA sequencing on plasma samples from PCH patients and identified a marked decrease in the expression of tRNA-related fragment 16-R29P4PE (tRF-16-R29P4PE), a specific tsRNA fragment, with a diagnostic area under the curve value of 0.7750. Using Angiotensin II (Ang II)-stimulated H9c2 cardiomyocytes as an in vitro model and Sprague-Dawley rats as an in vivo model, we investigated the effects of tRF-16-R29P4PE minic/inhibitors and silencing of the paired basic amino acid cleaving system 4 (PACE4) gene. Our results demonstrated that modulating tRF-16-R29P4PE expression significantly reduced brain natriuretic peptide (BNP) and free fatty acid levels while enhancing ATP production, glucose levels, and mitochondrial membrane potential. These effects were accompanied by the downregulation of PACE4, hypoxia-inducible factor-1α (HIF-1α), glucose transporter-4 (GLUT-4), and medium-chain acyl-CoA dehydrogenase (MCAD), as well as the upregulation of peroxisome proliferator-activated receptor α (PPARα). Animal experiments revealed that tRF-16-R29P4PE minic improved cardiac function, reduced myocardial fibrosis, and mitigated metabolic disorders and mitochondrial damage. Furthermore, co-immunoprecipitation (Co-IP) and molecular docking assays confirmed a direct interaction between PACE4 and HIF-1α, and luciferase reporter assays identified PACE4 as a direct target of tRF-16-R29P4PE. By regulating the PACE4 and HIF-1α/PPARα signaling pathways, tRF-16-R29P4PE alleviates PCH, providing a promising molecular target for therapeutic intervention.
Collapse
Affiliation(s)
- Feng Wang
- Department of Pharmacy, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou 215008, China
| | - Ping Li
- Department of Central Laboratory, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou 215008, China
| | - Xinxin Yan
- Department of Pharmacy, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou 215008, China
| | - Anna Yue
- Department of Cardiology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou 215008, China
| | - Jingyi Xu
- Department of Central Laboratory, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou 215008, China
| | - Yaqing Shao
- Department of Cardiology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou 215008, China
| | - Kaiyu Zhang
- Department of Cardiology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou 215008, China
| | - Qian Zhang
- Department of Pharmacy, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou 215008, China
| | - Yuan Li
- Department of Cardiology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou 215008, China.
| | - Kangyun Sun
- Department of Cardiology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou 215008, China.
| |
Collapse
|
11
|
Bakowska-Zywicka K, Rzepczak A, Plawgo K, Sobanska D, Tyczewska A. tRNA-Derived Fragments in Age-Related Diseases: A Systematic Review. WILEY INTERDISCIPLINARY REVIEWS. RNA 2025; 16:e70013. [PMID: 40263934 DOI: 10.1002/wrna.70013] [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: 12/12/2024] [Revised: 03/27/2025] [Accepted: 03/31/2025] [Indexed: 04/24/2025]
Abstract
Aging is a progressive weakening of numerous functions of organisms resulting in diminished abilities to safeguard against environmental damage and augment physiological harmony. It is not a disease in itself; however, it is a main cause of debilitating and life-threatening chronic aging-related diseases (ARDs). tRNA-derived fragments (tDRs) are stable forms of tRNAs of 14-35 nt in length that function as regulatory small-RNA molecules. Here we aimed to perform a systematic review of original articles on the involvement of tDRs in the etiology of ARDs: their identification and characterization. The systematic review was conducted according to the Cochrane Handbook guidelines and the Preferred Reporting Items for Systematic Reviews and Meta-analyses (PRISMA) statement. Based on the eligibility criteria defined for the study, 21 original articles were included in this systematic review, covering 11 ARDs. The preferred research method used to study tDRs was high-throughput sequencing combined with RT-qPCR, and as a result, a number of tDRs were implicated in ARDs. Importantly, an in-depth analysis of the articles allowed us to identify several shortcomings: (i) the tDRs nomenclature varies between studies and articles, making it often difficult to precisely identify molecules differentiating in a given disease; (ii) the chosen tDRs have all been studied for a miRNA-like mechanism of action; however, tDRs also function in RNAi-independent ways, which need to be studied as well; (iii) to precisely identify tDRs, the sequencing techniques that overcome the issues of modifications harbored by tRNAs must be used.
Collapse
Affiliation(s)
| | - Alicja Rzepczak
- Institute of Bioorganic Chemistry Polish Academy of Sciences, Poznan, Poland
| | - Kinga Plawgo
- Institute of Bioorganic Chemistry Polish Academy of Sciences, Poznan, Poland
| | - Daria Sobanska
- Institute of Bioorganic Chemistry Polish Academy of Sciences, Poznan, Poland
| | - Agata Tyczewska
- Institute of Bioorganic Chemistry Polish Academy of Sciences, Poznan, Poland
| |
Collapse
|
12
|
Shigematsu M, Matsubara R, Gumas J, Kawamura T, Kirino Y. Angiogenin-catalyzed cleavage within tRNA anticodon-loops identified by cP-RNA-seq. Biosci Biotechnol Biochem 2025; 89:398-405. [PMID: 39658364 PMCID: PMC11840711 DOI: 10.1093/bbb/zbae192] [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: 10/23/2024] [Accepted: 12/02/2024] [Indexed: 12/12/2024]
Abstract
Angiogenin (Ang), an endoribonuclease belonging to the RNase A superfamily, cleaves the anticodon-loops of tRNAs to produce tRNA-half molecules. Although previous studies have demonstrated the involvement of Ang in the pathobiology of neurodegenerative disorders, the characterization of Ang-generated tRNA halves in neuronal cells remains limited. This is partly due to the technical limitations of standard RNA-seq methods, which cannot capture Ang-generated RNAs containing a 2',3'-cyclic phosphate (cP). In this report, we established an Ang-treatment model using SH-SY5Y, a human neuroblastoma cell line, and demonstrated Ang-dependent accumulation of tRNA halves. By performing cP-RNA-seq, which selectively captures cP-containing RNAs, we identified Ang-generated tRNA halves and the specific cleavage positions within tRNA anticodon-loops responsible for their generation. Our results provide insights into the anticodon-loop cleavage and the selective production of a specific subset of tRNA halves by Ang.
Collapse
Affiliation(s)
- Megumi Shigematsu
- Computational Medicine Center, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA
- Department of Biochemistry and Molecular Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA
| | - Ryuma Matsubara
- Computational Medicine Center, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA
- Department of Biochemistry and Molecular Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA
| | - Justin Gumas
- Computational Medicine Center, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA
- Department of Biochemistry and Molecular Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA
| | - Takuya Kawamura
- Computational Medicine Center, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA
- Department of Biochemistry and Molecular Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA
| | - Yohei Kirino
- Computational Medicine Center, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA
- Department of Biochemistry and Molecular Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA
| |
Collapse
|
13
|
Tan S, Yang W, Ren Z, Peng Q, Xu X, Jiang X, Wu Z, Oyang L, Luo X, Lin J, Xia L, Peng M, Wu N, Tang Y, Han Y, Liao Q, Zhou Y. Noncoding RNA-encoded peptides in cancer: biological functions, posttranslational modifications and therapeutic potential. J Hematol Oncol 2025; 18:20. [PMID: 39972384 PMCID: PMC11841355 DOI: 10.1186/s13045-025-01671-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Accepted: 02/07/2025] [Indexed: 02/21/2025] Open
Abstract
In the present era, noncoding RNAs (ncRNAs) have become a subject of considerable scientific interest, with peptides encoded by ncRNAs representing a particularly promising avenue of investigation. The identification of ncRNA-encoded peptides in human cancers is increasing. These peptides regulate cancer progression through multiple molecular mechanisms. Here, we delineate the patterns of diverse ncRNA-encoded peptides and provide a synopsis of the methodologies employed for the identification of ncRNAs that possess the capacity to encode these peptides. Furthermore, we discuss the impacts of ncRNA-encoded peptides on the biological behavior of cancer cells and the underlying molecular mechanisms. In conclusion, we describe the prospects of ncRNA-encoded peptides in cancer and the challenges that need to be overcome.
Collapse
Affiliation(s)
- Shiming Tan
- The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Hunan Key Laboratory of Cancer Metabolism, 283 Tongzipo Road, Changsha, 410013, Hunan, People's Republic of China
| | - Wenjuan Yang
- The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Hunan Key Laboratory of Cancer Metabolism, 283 Tongzipo Road, Changsha, 410013, Hunan, People's Republic of China
| | - Zongyao Ren
- The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Hunan Key Laboratory of Cancer Metabolism, 283 Tongzipo Road, Changsha, 410013, Hunan, People's Republic of China
- Hunan Engineering Research Center of Tumor Organoid Technology and Applications, Public Service Platform of Tumor Organoid Technology, 283 Tongzipo Road, Changsha, 410013, Hunan, People's Republic of China
| | - Qiu Peng
- The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Hunan Key Laboratory of Cancer Metabolism, 283 Tongzipo Road, Changsha, 410013, Hunan, People's Republic of China
- Hunan Engineering Research Center of Tumor Organoid Technology and Applications, Public Service Platform of Tumor Organoid Technology, 283 Tongzipo Road, Changsha, 410013, Hunan, People's Republic of China
| | - Xuemeng Xu
- The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Hunan Key Laboratory of Cancer Metabolism, 283 Tongzipo Road, Changsha, 410013, Hunan, People's Republic of China
- Hunan Engineering Research Center of Tumor Organoid Technology and Applications, Public Service Platform of Tumor Organoid Technology, 283 Tongzipo Road, Changsha, 410013, Hunan, People's Republic of China
| | - Xianjie Jiang
- The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Hunan Key Laboratory of Cancer Metabolism, 283 Tongzipo Road, Changsha, 410013, Hunan, People's Republic of China
- Hunan Engineering Research Center of Tumor Organoid Technology and Applications, Public Service Platform of Tumor Organoid Technology, 283 Tongzipo Road, Changsha, 410013, Hunan, People's Republic of China
| | - Zhu Wu
- The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Hunan Key Laboratory of Cancer Metabolism, 283 Tongzipo Road, Changsha, 410013, Hunan, People's Republic of China
- Hunan Engineering Research Center of Tumor Organoid Technology and Applications, Public Service Platform of Tumor Organoid Technology, 283 Tongzipo Road, Changsha, 410013, Hunan, People's Republic of China
| | - Linda Oyang
- The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Hunan Key Laboratory of Cancer Metabolism, 283 Tongzipo Road, Changsha, 410013, Hunan, People's Republic of China
- Hunan Engineering Research Center of Tumor Organoid Technology and Applications, Public Service Platform of Tumor Organoid Technology, 283 Tongzipo Road, Changsha, 410013, Hunan, People's Republic of China
| | - Xia Luo
- The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Hunan Key Laboratory of Cancer Metabolism, 283 Tongzipo Road, Changsha, 410013, Hunan, People's Republic of China
- Hunan Engineering Research Center of Tumor Organoid Technology and Applications, Public Service Platform of Tumor Organoid Technology, 283 Tongzipo Road, Changsha, 410013, Hunan, People's Republic of China
| | - Jinguan Lin
- The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Hunan Key Laboratory of Cancer Metabolism, 283 Tongzipo Road, Changsha, 410013, Hunan, People's Republic of China
- Hunan Engineering Research Center of Tumor Organoid Technology and Applications, Public Service Platform of Tumor Organoid Technology, 283 Tongzipo Road, Changsha, 410013, Hunan, People's Republic of China
| | - Longzheng Xia
- The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Hunan Key Laboratory of Cancer Metabolism, 283 Tongzipo Road, Changsha, 410013, Hunan, People's Republic of China
- Hunan Engineering Research Center of Tumor Organoid Technology and Applications, Public Service Platform of Tumor Organoid Technology, 283 Tongzipo Road, Changsha, 410013, Hunan, People's Republic of China
| | - Mingjing Peng
- The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Hunan Key Laboratory of Cancer Metabolism, 283 Tongzipo Road, Changsha, 410013, Hunan, People's Republic of China
- Hunan Engineering Research Center of Tumor Organoid Technology and Applications, Public Service Platform of Tumor Organoid Technology, 283 Tongzipo Road, Changsha, 410013, Hunan, People's Republic of China
| | - Nayiyuan Wu
- The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Hunan Key Laboratory of Cancer Metabolism, 283 Tongzipo Road, Changsha, 410013, Hunan, People's Republic of China
- Hunan Engineering Research Center of Tumor Organoid Technology and Applications, Public Service Platform of Tumor Organoid Technology, 283 Tongzipo Road, Changsha, 410013, Hunan, People's Republic of China
| | - Yanyan Tang
- The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Hunan Key Laboratory of Cancer Metabolism, 283 Tongzipo Road, Changsha, 410013, Hunan, People's Republic of China
- Hunan Engineering Research Center of Tumor Organoid Technology and Applications, Public Service Platform of Tumor Organoid Technology, 283 Tongzipo Road, Changsha, 410013, Hunan, People's Republic of China
| | - Yaqian Han
- The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Hunan Key Laboratory of Cancer Metabolism, 283 Tongzipo Road, Changsha, 410013, Hunan, People's Republic of China.
- Hunan Engineering Research Center of Tumor Organoid Technology and Applications, Public Service Platform of Tumor Organoid Technology, 283 Tongzipo Road, Changsha, 410013, Hunan, People's Republic of China.
| | - Qianjin Liao
- Department of Oncology, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, 410005, Hunan, People's Republic of China.
| | - Yujuan Zhou
- The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Hunan Key Laboratory of Cancer Metabolism, 283 Tongzipo Road, Changsha, 410013, Hunan, People's Republic of China.
- Hunan Engineering Research Center of Tumor Organoid Technology and Applications, Public Service Platform of Tumor Organoid Technology, 283 Tongzipo Road, Changsha, 410013, Hunan, People's Republic of China.
| |
Collapse
|
14
|
Kompatscher M, Gonnella I, Erlacher M. Studying the Function of tRNA Modifications: Experimental Challenges and Opportunities. J Mol Biol 2025:168934. [PMID: 39756793 DOI: 10.1016/j.jmb.2024.168934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 12/19/2024] [Accepted: 12/31/2024] [Indexed: 01/07/2025]
Abstract
tRNAs are essential molecules in protein synthesis, responsible for translating the four-nucleotide genetic code into the corresponding amino acid sequence. RNA modifications play a crucial role in influencing tRNA folding, structure, and function. These modifications, ranging from simple methylations to complex hypermodified species, are distributed throughout the tRNA molecule. Depending on their type and position, they contribute to the accuracy and efficiency of decoding by participating in a complex network of interactions. The enzymatic processes introducing these modifications are equally intricate and diverse, adding further complexity. As a result, studying tRNA modifications faces limitations at multiple levels. This review addresses the challenges involved in manipulating and studying the function of tRNA modifications and discusses experimental strategies and possibilities to overcome these obstacles.
Collapse
Affiliation(s)
- Maria Kompatscher
- Institute of Genomics and RNomics, Medical University of Innsbruck, Innrain 80/82, 6020 Innsbruck, Austria
| | - Isabell Gonnella
- Institute of Genomics and RNomics, Medical University of Innsbruck, Innrain 80/82, 6020 Innsbruck, Austria
| | - Matthias Erlacher
- Institute of Genomics and RNomics, Medical University of Innsbruck, Innrain 80/82, 6020 Innsbruck, Austria.
| |
Collapse
|
15
|
Wittmann J. Overview of the Different Classes of Small RNAs During B-Cell Development. Methods Mol Biol 2025; 2883:1-29. [PMID: 39702702 DOI: 10.1007/978-1-0716-4290-0_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2024]
Abstract
B lymphocytes (B cells) are a type of white blood cell that play an essential role in the adaptive immune response. They are derived from pluripotent hematopoietic stem cells and undergo several developmental stages in the bone marrow and secondary lymphoid organs to become effector cells. B cells can act as antigen-presenting cells, secrete cytokines, generate immunological memory as memory B cells, and produce and secrete high-affinity antibodies as plasma B cells.B-cell development occurs in discontinuous steps within specific organs and niche environments, progressing through checkpoints controlled by the relative levels of numerous transcription factors, cytokines, and surface receptors. These complex interactions of distinct developmental programs operate through balanced control mechanisms rather than simple "on/off" signals.Over the past two decades, much has been learned about short non-coding RNA (ncRNA) molecules that play a critical role in fine-tuning gene expression by targeting specific messenger RNAs (mRNAs) for degradation or translational repression. In the intricate orchestration of B-cell development, ncRNAs contribute to the delicate balance between proliferation, differentiation, and apoptosis by influencing key checkpoints in the maturation process.Therefore, in this chapter, I will review the role of different classes of small ncRNAs, including microRNAs, glycoRNAs, tRNA-derived fragments, and ribosomal RNA-derived fragments, in modulating gene expression at the post-transcriptional level and their contribution to the intricate regulatory network that controls B-cell maturation.
Collapse
Affiliation(s)
- Jürgen Wittmann
- Division of Molecular Immunology, Department of Internal Medicine III, Nikolaus-Fiebiger-Center of Molecular Medicine, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany.
| |
Collapse
|
16
|
Janga H, Schmerer N, Aznaourova M, Schulte LN. Non-coding RNA Networks in Infection. Methods Mol Biol 2025; 2883:53-77. [PMID: 39702704 DOI: 10.1007/978-1-0716-4290-0_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2024]
Abstract
In the face of global health challenges posed by infectious diseases and the emergence of drug-resistant pathogens, the exploration of cellular non-coding RNA (ncRNA) networks has unveiled new dimensions in infection research. Particularly microRNAs (miRNAs) and long non-coding RNAs (lncRNAs) have emerged as instrumental players in ensuring a balance between protection against hyper-inflammatory conditions and the effective elimination of pathogens. Specifically, ncRNAs, such as the miRNA miR-155 or the lncRNAs MaIL1 (macrophage interferon-regulatory lncRNA 1), and LUCAT1 (lung cancer-associated transcript 1) have been recurrently linked to infectious and inflammatory diseases. Together with other ncRNAs, discussed in this chapter, they form a complex regulatory network shaping the host response to pathogens. Additionally, some pathogens exploit these ncRNAs to establish and sustain infections, underscoring their dual roles in host protection and colonization. Despite the substantial progress made, the vast majority of ncRNA loci remains unexplored, with ongoing research likely to reveal novel ncRNA categories and expand our understanding of their roles in infections. This chapter consolidates current insights into ncRNA-mediated regulatory networks, highlighting their contributions to severe diseases and their potential as targets and biomarkers for innovative therapeutic strategies.
Collapse
Affiliation(s)
| | - Nils Schmerer
- Institute for Lung Research, Philipps University, Marburg, Germany
| | | | - Leon N Schulte
- Institute for Lung Research, Philipps University, Marburg, Germany.
- German Center for Lung Research, Giessen, Germany.
| |
Collapse
|
17
|
Ouyang W, Sun H, Wang Y. Unlocking the small RNAs: local and systemic modulators for advancing agronomic enhancement. J Genet Genomics 2024:S1673-8527(24)00364-3. [PMID: 39716571 DOI: 10.1016/j.jgg.2024.12.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 12/17/2024] [Accepted: 12/17/2024] [Indexed: 12/25/2024]
Abstract
Small regulatory RNAs (sRNAs) are essential regulators of gene expression across a wide range of organisms to precisely modulate gene activity based on sequence-specific recognition. In model plants like Arabidopsis thaliana, extensive research has primarily concentrated on 21 to 24-nucleotide (nt) sRNAs, particularly microRNAs (miRNAs). Recent advancements in cell and tissue isolation techniques, coupled with advanced sequencing technologies, are revealing a diverse array of preciously uncharacterized sRNA species. These include previously novel structural RNA fragments as well as numerous cell- and tissue-specific sRNAs that are active during distinct developmental stages, thereby enhancing our understanding of the precise and dynamic regulatory roles of sRNAs in plant development regulation. Additionally, a notable feature of sRNAs is their capacity for amplification and movement between cells and tissues, which facilitates long-distance communication-an adaptation critical to plants due to their sessile nature. In this review, we will discuss the classification and mechanisms of action of sRNAs, using legumes as a primary example due to their essential engagement for the unique organ establishment of root nodules and long-distance signaling, and further illustrating the potential applications of sRNAs in modern agricultural breeding and environmentally sustainable plant protection strategies.
Collapse
Affiliation(s)
- Wenqi Ouyang
- Key Laboratory of Seed Innovation, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; College of Tropical Crops, Hainan University, Haikou, Hainan 570288, China
| | - Hongda Sun
- Key Laboratory of Seed Innovation, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing, China
| | - Yuan Wang
- Key Laboratory of Seed Innovation, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; CAS-JIC Centre of Excellence for Plant and Microbial Science (CEPAMS), Institute of Genetics and Developmental Biology, CAS, Beijing 100101, China.
| |
Collapse
|
18
|
Rosina A, Polacek N, Rauscher R. Ribosome-associated tDRs in yeast. Methods Enzymol 2024; 711:85-101. [PMID: 39952719 DOI: 10.1016/bs.mie.2024.11.006] [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: 02/17/2025]
Abstract
The regulation of gene expression in response to environmental stress is a key process that ensures cellular survival across all three domains of life. The adjustment of protein synthesis appears to be one of the initial steps toward the response and adaptation to stress. Ribosome-associated non-coding RNAs (rancRNAs) efficiently regulate translation as an immediate response to stress by directly targeting the ribosome and fine-tuning translation. tRNA-derived RNAs (tDRs) are part of the RNA species that constitute the functionally diverse class of rancRNAs. Here we report a new experimental approach for creating deep sequencing libraries of ribosome-associated small RNAs in yeast utilizing state-of-the-art technologies. Our new strategy is supported by validating previously identified rancRNAs and discovering novel tDRs interacting with the Saccharomyces cerevisiae ribosome.
Collapse
Affiliation(s)
- Alessia Rosina
- Department of Chemistry, Biochemistry and Pharmaceutical Sciences, University of Bern, Bern, Switzerland; Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Norbert Polacek
- Department of Chemistry, Biochemistry and Pharmaceutical Sciences, University of Bern, Bern, Switzerland.
| | - Robert Rauscher
- Department of Chemistry, Biochemistry and Pharmaceutical Sciences, University of Bern, Bern, Switzerland.
| |
Collapse
|
19
|
Zhu Z, Sun J, Xu W, Zeng Q, Feng H, Zang L, He Y, He X, Sheng N, Ren X, Liu G, Huang H, Huang R, Yan J. MGAT4A/Galectin9-Driven N-Glycosylation Aberration as a Promoting Mechanism for Poor Prognosis of Endometrial Cancer with TP53 Mutation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2409764. [PMID: 39527463 DOI: 10.1002/advs.202409764] [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: 08/16/2024] [Indexed: 11/16/2024]
Abstract
Emerging evidence recognizes aberrant glycosylation as the malignant characteristics of cancer cells, but little is known about glycogenes' roles in endometrial carcinoma (EC), especially the most aggressive subtype carrying TP53 mutations. Using unsupervised hierarchical clustering, an 11-glycogene cluster is identified to distinguish an EC subtype associated with frequent TP53 mutation and worse prognosis. Among them, MGAT4A (alpha-1,3-mannosyl-glycoprotein 4-β-N-acetylglucosaminyltransferase A) emerges as the most consistently overexpressed glycogene, contributing to EC aggressiveness. In the presence of galectin-9, MGAT4A increases EC cell proliferation and invasion via promoting glucose metabolism. N-glycoproteomics further revealed GLUT1, a glucose transporter, as a glycoprotein modified by MGAT4A. Binding of galectin-9 to the MGAT4A-branched N-glycan on GLUT1 enhances its cell membrane distribution, leading to glucose uptake increase. In addition, oncogenic mutations of TP53 gene in EC cells upregulate MGAT4A expression by disrupting the regulatory oversight exerted by wild-type p53 on tumor-suppressive miRNAs, including miR-34a and miR-449a/b. The findings highlight a new molecular mechanism involving MGAT4A-regulated N-glycosylation on the key regulator of glucose metabolism in p53 mutants-driven EC aggressiveness, which may provide a strategic avenue to combat advanced EC.
Collapse
Affiliation(s)
- Zhen Zhu
- Center for Medical Research and Innovation, Shanghai Pudong Hospital, Fudan University Pudong Medical Center;, Laboratory Animal Center, Fudan University, Shanghai, 200032, China
- Model Animal Research Center of Nanjing University, Nanjing, 210061, China
| | - Jingya Sun
- Center for Drug Safety Evaluation and Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Weiqing Xu
- Department of Pathology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Qinghe Zeng
- Center for Drug Safety Evaluation and Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Hanyi Feng
- Center for Drug Safety Evaluation and Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Lijuan Zang
- Department of Pathology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Yinyan He
- Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
- Department of Obstetrics and Gynecology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China
| | - Xiao He
- Center for Drug Safety Evaluation and Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Na Sheng
- Model Animal Research Center of Nanjing University, Nanjing, 210061, China
| | - Xuelian Ren
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Guobin Liu
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - He Huang
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China
| | - Ruimin Huang
- Center for Drug Safety Evaluation and Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, 210023, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Jun Yan
- Center for Medical Research and Innovation, Shanghai Pudong Hospital, Fudan University Pudong Medical Center;, Laboratory Animal Center, Fudan University, Shanghai, 200032, China
| |
Collapse
|
20
|
Li J, Wang X. Functional roles of conserved lncRNAs and circRNAs in eukaryotes. Noncoding RNA Res 2024; 9:1271-1279. [PMID: 39036601 PMCID: PMC11260338 DOI: 10.1016/j.ncrna.2024.06.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 06/14/2024] [Accepted: 06/24/2024] [Indexed: 07/23/2024] Open
Abstract
Long non-coding RNAs (lncRNAs) and circular RNAs (circRNAs) have emerged as critical regulators in essentially all biological processes across eukaryotes. They exert their functions through chromatin remodeling, transcriptional regulation, interacting with RNA-binding proteins (RBPs), serving as microRNA sponges, etc. Although non-coding RNAs are typically more species-specific than coding RNAs, a number of well-characterized lncRNA (such as XIST and NEAT1) and circRNA (such as CDR1as and ciRS-7) are evolutionarily conserved. The studies on conserved lncRNA and circRNAs across multiple species could facilitate a comprehensive understanding of their roles and mechanisms, thereby overcoming the limitations of single-species studies. In this review, we provide an overview of conserved lncRNAs and circRNAs, and summarize their conserved roles and mechanisms.
Collapse
Affiliation(s)
- Jingxin Li
- Department of Clinical Laboratory, The First Affiliated Hospital of USTC, The RNA Institute, School of Basic Medical Sciences, Division of Life Science and Medicine, University of Science and Technology of China (UTSC), Hefei, 230027, Anhui, China
| | - Xiaolin Wang
- Department of Clinical Laboratory, The First Affiliated Hospital of USTC, The RNA Institute, School of Basic Medical Sciences, Division of Life Science and Medicine, University of Science and Technology of China (UTSC), Hefei, 230027, Anhui, China
| |
Collapse
|
21
|
Johnston AG, Anam M, Dutta A, Su Z. TGIRT-seq to profile tRNA-derived RNAs and associated RNA modifications. Methods Enzymol 2024; 711:223-240. [PMID: 39952707 PMCID: PMC11890191 DOI: 10.1016/bs.mie.2024.11.001] [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: 02/17/2025]
Abstract
RNA modifications are key regulators for RNA processes. tRNA-derived RNAs are small RNAs with size between 15 and 50 bases long that are processed from mature or precursor tRNAs. Despite their more recent discovery, tRNA-derived RNAs have been found to play regulatory roles in many cellular processes including gene silencing, protein synthesis, stress response, and transgenerational inheritance. Furthermore, tRNA-derived RNAs are highly abundant in bodily fluids, posing as potential biomarkers. A unique feature of tRNA-derived RNAs is that they are rich in RNA modifications. Many of the RNA modifications on tRNA-derived RNAs disrupt Watson-Crick base pairing and will thus stall reverse transcriptase, such as N1-methyladenosine (m1A), N1-methylguanosine (m1G) and N2, N2-dimethylguanosine (m22G). These RNA modifications add another layer of regulation onto tRNA-derived RNAs' functions and are of interests for future research. However, these RNA modifications could also lead to lower detection of modification-containing RNAs in genome-wide small RNA sequencing analysis due to reverse transcriptase stall. To circumvent this bias, TGIRT (Thermostable Group II Intron Reverse Transcriptase) has been used to readthrough RNA modifications inserting mismatches. These mismatch signatures can then be used to precisely map the modification sites at base resolution. Here we describe the step-by-step experimental protocol to start with purified RNAs from cells or tissues and use TGIRT to make small RNA sequencing library for Illumina sequencing to profile the abundance of tRNA-derived RNAs and the associated RNA modifications.
Collapse
Affiliation(s)
- Abigail Grace Johnston
- Department of Genetics, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Monima Anam
- Department of Genetics, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Anindya Dutta
- Department of Genetics, University of Alabama at Birmingham, Birmingham, AL, United States; O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Zhangli Su
- Department of Genetics, University of Alabama at Birmingham, Birmingham, AL, United States; O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, United States.
| |
Collapse
|
22
|
Feng X, Guang S. Functions and applications of RNA interference and small regulatory RNAs. Acta Biochim Biophys Sin (Shanghai) 2024; 57:119-130. [PMID: 39578714 PMCID: PMC11802346 DOI: 10.3724/abbs.2024196] [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: 06/23/2024] [Accepted: 09/03/2024] [Indexed: 11/24/2024] Open
Abstract
Small regulatory RNAs play a variety of crucial roles in eukaryotes, influencing gene regulation, developmental timing, antiviral defense, and genome integrity via a process termed RNA interference (RNAi). This process involves Argonaute/small RNA (AGO/sRNA) complexes that target transcripts via sequence complementarity and modulate gene expression and epigenetic modifications. RNAi is a highly conserved gene regulatory phenomenon that recognizes self- and non-self nucleic acids, thereby defending against invasive sequences. Since its discovery, RNAi has been widely applied in functional genomic studies and a range of practical applications. In this review, we focus on the current understanding of the biological roles of the RNAi pathway in transposon silencing, fertility, developmental regulation, immunity, stress responses, and acquired transgenerational inheritance. Additionally, we provide an overview of the applications of RNAi technology in biomedical research, agriculture, and therapeutics.
Collapse
Affiliation(s)
- Xuezhu Feng
- School of Basic Medical SciencesAnhui Medical UniversityHefei230032China
| | - Shouhong Guang
- Department of Obstetrics and Gynecologythe First Affiliated Hospital of USTCThe USTC RNA InstituteMinistry of Education Key Laboratory for Membraneless Organelles & Cellular DynamicsHefei National Research Center for Physical Sciences at the MicroscaleCenter for Advanced Interdisciplinary Science and Biomedicine of IHMSchool of Life SciencesDivision of Life Sciences and MedicineBiomedical Sciences and Health Laboratory of Anhui ProvinceUniversity of Science and Technology of ChinaHefei230027China
| |
Collapse
|
23
|
A L, Qu L, He J, Ge L, Gao H, Huang X, You T, Gong H, Liang Q, Chen S, Xie J, Xu H. Exosomes derived from IFNγ-stimulated mesenchymal stem cells protect photoreceptors in RCS rats by restoring immune homeostasis through tsRNAs. Cell Commun Signal 2024; 22:543. [PMID: 39538308 PMCID: PMC11562488 DOI: 10.1186/s12964-024-01920-3] [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: 07/22/2024] [Accepted: 11/01/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND Retinitis pigmentosa is a neurodegenerative disease with major pathologies of photoreceptor apoptosis and immune imbalance. Mesenchymal stem cells (MSCs) have been approved for clinical application for treating various immune-related or neurodegenerative diseases. The objective of this research was to investigate the mechanisms underlying the safeguarding effects of MSC-derived exosomes in a retinal degenerative disease model. METHODS Interferon gamma-stimulated exosomes (IFNγ-Exos) secreted from MSCs were isolated, purified, and injected into the vitreous body of RCS rats on postnatal day (P) 21. Morphological and functional changes in the retina were examined at P28, P35, P42, and P49 in Royal College of Surgeons (RCS) rats. The mechanism was explored using high-throughput sequencing technology and confirmed in vitro. RESULTS Treatment with IFNγ-Exo produced better protective effects on photoreceptors and improved visual function in RCS rats. IFNγ-Exo significantly suppressed the activated microglia and inhibited the inflammatory responses in the retina of RCS rats, which was also confirmed in the lipopolysaccharide-activated microglia cell line BV2. Furthermore, through tRNA-derived small RNA (tsRNA) sequencing, we found that IFNγ-Exos from MSCs contained higher levels of Other-1_17-tRNA-Phe-GAA-1-M3, Other-6_23-tRNA-Lys-TTT-3, and TRF-57:75-GLN-CGG-2-m2 than native exosomes, which mainly regulated inflammatory and immune-related pathways, including the mTOR signaling pathway and EGFR tyrosine kinase inhibitor resistance. CONCLUSIONS IFNγ stimulation enhanced the neuroprotective effects of MSC-derived exosomes on photoreceptors of the degenerative retina, which may be mediated by immune regulatory tsRNAs acting on microglia. In conclusion, IFNγ-Exo is a promising nanotherapeutic agent for the treatment of retinitis pigmentosa.
Collapse
Affiliation(s)
- Luodan A
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Army Medical University, Chongqing, 400038, China
- Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing, 400038, China
| | - Linghui Qu
- Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing, 400038, China
- Department of Ophthalmology, The 74th Army Group Hospital, Guangzhou, 510318, China
| | - Juncai He
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Army Medical University, Chongqing, 400038, China
- Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing, 400038, China
| | - Lingling Ge
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Army Medical University, Chongqing, 400038, China
- Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing, 400038, China
| | - Hui Gao
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Army Medical University, Chongqing, 400038, China
- Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing, 400038, China
- Shigatse Branch of Xinqiao Hospital, 953th Hospital, Army Medical University (Third Military Medical University), Shigatse, 857000, China
| | - Xiaona Huang
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Army Medical University, Chongqing, 400038, China
- Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing, 400038, China
| | - Tianjing You
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Army Medical University, Chongqing, 400038, China
- Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing, 400038, China
| | - Hong Gong
- Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing, 400038, China
- Department of Military Cognitive Psychology, School of Psychology, Army Medical University, Chongqing, 400038, China
| | - Qingle Liang
- Department of Clinical Laboratory Medicine, First Affiliated Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Siyu Chen
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Army Medical University, Chongqing, 400038, China
- Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing, 400038, China
| | - Jing Xie
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Army Medical University, Chongqing, 400038, China.
- Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing, 400038, China.
| | - Haiwei Xu
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Army Medical University, Chongqing, 400038, China.
- Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing, 400038, China.
| |
Collapse
|
24
|
Baindoor S, Gibriel HAY, Venø MT, Su J, Morrissey EP, Jirström E, Woods I, Kenny A, Alves M, Halang L, Fabbrizio P, Bilen M, Engel T, Hogg MC, Bendotti C, Nardo G, Slack RS, Kjems J, Prehn JHM. Distinct fingerprints of tRNA-derived small non-coding RNA in animal models of neurodegeneration. Dis Model Mech 2024; 17:dmm050870. [PMID: 39552337 PMCID: PMC11603119 DOI: 10.1242/dmm.050870] [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: 04/30/2024] [Accepted: 10/17/2024] [Indexed: 11/19/2024] Open
Abstract
Transfer RNA-derived small RNAs (tsRNAs) - categorized as tRNA-derived fragments (tRFs), tRNA-derived stress-induced RNAs (tiRNAs) and internal tRF (itRF) - are small non-coding RNAs that participate in various cellular processes such as translation inhibition and responses to cellular stress. We here identified tsRNA profiles within susceptible tissues in animal models of amyotrophic lateral sclerosis (ALS), frontotemporal dementia (FTD) and Parkinson's disease (PD) to pinpoint disease-specific tsRNAs and those shared across neurodegenerative diseases. We performed small RNA sequencing in the SOD1G93A and TDP43A315T mouse models of ALS (spinal cord), the TauP301S model of FTD (hippocampus), and the parkin/POLG model of PD (substantia nigra). Bioinformatic analysis showed higher expression of 5' tiRNAs selectively in the two ALS models, lower expression of 3' tRFs in both the ALS and FTD mouse models, and lower expression of itRF Arg in the PD model. Experimental validation confirmed the expression of tsRNAs. Gene Ontology analysis of targets associated with validated 3' tRFs indicated functions in the regulation of synaptic and neuronal pathways. Our profiling of tsRNAs indicates disease-specific fingerprints in animal models of neurodegeneration, which require validation in human disease.
Collapse
Affiliation(s)
- Sharada Baindoor
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, 123 St Stephen's Green, Dublin D02 YN77, Ireland
- FutureNeuro SFI Research Centre for Chronic and Rare Neurological Diseases, Royal College of Surgeons in Ireland, Dublin D02 YN77, Ireland
| | - Hesham A. Y. Gibriel
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, 123 St Stephen's Green, Dublin D02 YN77, Ireland
- FutureNeuro SFI Research Centre for Chronic and Rare Neurological Diseases, Royal College of Surgeons in Ireland, Dublin D02 YN77, Ireland
| | | | - Junyi Su
- Omiics ApS, DK-8200 Aarhus N, Denmark
- Interdisciplinary Nanoscience Centre, Department of Molecular Biology and Genetics, Aarhus University, DK-8000 Aarhus C, Denmark
| | - Elena Perez Morrissey
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, 123 St Stephen's Green, Dublin D02 YN77, Ireland
- FutureNeuro SFI Research Centre for Chronic and Rare Neurological Diseases, Royal College of Surgeons in Ireland, Dublin D02 YN77, Ireland
| | - Elisabeth Jirström
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, 123 St Stephen's Green, Dublin D02 YN77, Ireland
- FutureNeuro SFI Research Centre for Chronic and Rare Neurological Diseases, Royal College of Surgeons in Ireland, Dublin D02 YN77, Ireland
| | - Ina Woods
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, 123 St Stephen's Green, Dublin D02 YN77, Ireland
- FutureNeuro SFI Research Centre for Chronic and Rare Neurological Diseases, Royal College of Surgeons in Ireland, Dublin D02 YN77, Ireland
| | - Aidan Kenny
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, 123 St Stephen's Green, Dublin D02 YN77, Ireland
- FutureNeuro SFI Research Centre for Chronic and Rare Neurological Diseases, Royal College of Surgeons in Ireland, Dublin D02 YN77, Ireland
| | - Mariana Alves
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, 123 St Stephen's Green, Dublin D02 YN77, Ireland
- FutureNeuro SFI Research Centre for Chronic and Rare Neurological Diseases, Royal College of Surgeons in Ireland, Dublin D02 YN77, Ireland
| | - Luise Halang
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, 123 St Stephen's Green, Dublin D02 YN77, Ireland
| | - Paola Fabbrizio
- Laboratory of Neurobiology and Preclinical Therapeutics, Department of Neuroscience, IRCCS - Mario Negri Institute for Pharmacological Research, Via Mario Negri, 2, 20156 Milan, Italy
| | - Maria Bilen
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Tobias Engel
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, 123 St Stephen's Green, Dublin D02 YN77, Ireland
- FutureNeuro SFI Research Centre for Chronic and Rare Neurological Diseases, Royal College of Surgeons in Ireland, Dublin D02 YN77, Ireland
| | - Marion C. Hogg
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, 123 St Stephen's Green, Dublin D02 YN77, Ireland
- FutureNeuro SFI Research Centre for Chronic and Rare Neurological Diseases, Royal College of Surgeons in Ireland, Dublin D02 YN77, Ireland
- Department of Biosciences, Nottingham Trent University, Nottingham NG11 8NS, UK
| | - Caterina Bendotti
- Laboratory of Neurobiology and Preclinical Therapeutics, Department of Neuroscience, IRCCS - Mario Negri Institute for Pharmacological Research, Via Mario Negri, 2, 20156 Milan, Italy
| | - Giovanni Nardo
- Laboratory of Neurobiology and Preclinical Therapeutics, Department of Neuroscience, IRCCS - Mario Negri Institute for Pharmacological Research, Via Mario Negri, 2, 20156 Milan, Italy
| | - Ruth S. Slack
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Jørgen Kjems
- Interdisciplinary Nanoscience Centre, Department of Molecular Biology and Genetics, Aarhus University, DK-8000 Aarhus C, Denmark
| | - Jochen H. M. Prehn
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, 123 St Stephen's Green, Dublin D02 YN77, Ireland
- FutureNeuro SFI Research Centre for Chronic and Rare Neurological Diseases, Royal College of Surgeons in Ireland, Dublin D02 YN77, Ireland
| |
Collapse
|
25
|
Wang M, Guo J, Chen W, Wang H, Hou X. Emerging roles of tRNA-derived small RNAs in injuries. PeerJ 2024; 12:e18348. [PMID: 39465146 PMCID: PMC11512806 DOI: 10.7717/peerj.18348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Accepted: 09/27/2024] [Indexed: 10/29/2024] Open
Abstract
tRNA-derived small RNAs (tsRNAs) are a novel class of small noncoding RNAs, precisely cleaved from tRNA, functioning as regulatory molecules. The topic of tsRNAs in injuries has not been extensively discussed, and studies on tsRNAs are entering a new era. Here, we provide a fresh perspective on this topic. We systematically reviewed the classification, generation, and biological functions of tsRNAs in response to stress, as well as their potential as biomarkers and therapeutic targets in various injuries, including lung injury, liver injury, renal injury, cardiac injury, neuronal injury, vascular injury, skeletal muscle injury, and skin injury. We also provided a fresh perspective on the association between stress-induced tsRNAs and organ injury from a clinical perspective.
Collapse
Affiliation(s)
- Mengjun Wang
- Center for Cardiac Intensive Care, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Junfeng Guo
- Center for Cardiac Intensive Care, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Wei Chen
- Center for Cardiac Intensive Care, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Hong Wang
- Center for Cardiac Intensive Care, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Xiaotong Hou
- Center for Cardiac Intensive Care, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
26
|
Li Y, Yu Z, Jiang W, Lyu X, Guo A, Sun X, Yang Y, Zhang Y. tRNA and tsRNA: From Heterogeneity to Multifaceted Regulators. Biomolecules 2024; 14:1340. [PMID: 39456272 PMCID: PMC11506809 DOI: 10.3390/biom14101340] [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: 08/16/2024] [Revised: 10/01/2024] [Accepted: 10/04/2024] [Indexed: 10/28/2024] Open
Abstract
As the most ancient RNA, transfer RNAs (tRNAs) play a more complex role than their constitutive function as amino acid transporters in the protein synthesis process. The transcription and maturation of tRNA in cells are subject to stringent regulation, resulting in the formation of tissue- and cell-specific tRNA pools with variations in tRNA overall abundance, composition, modification, and charging levels. The heterogeneity of tRNA pools contributes to facilitating the formation of histocyte-specific protein expression patterns and is involved in diverse biological processes. Moreover, tRNAs can be recognized by various RNase under physiological and pathological conditions to generate tRNA-derived small RNAs (tsRNAs) and serve as small regulatory RNAs in various biological processes. Here, we summarize these recent insights into the heterogeneity of tRNA and highlight the advances in the regulation of tRNA function and tsRNA biogenesis by tRNA modifications. We synthesize diverse mechanisms of tRNA and tsRNA in embryonic development, cell fate determination, and epigenetic inheritance regulation. We also discuss the potential clinical applications based on the new knowledge of tRNA and tsRNA as diagnostic and prognostic biomarkers and new therapeutic strategies for multiple diseases.
Collapse
Affiliation(s)
- Yun Li
- Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China; (Y.L.); (Z.Y.); (W.J.); (X.L.); (A.G.); (X.S.)
| | - Zongyu Yu
- Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China; (Y.L.); (Z.Y.); (W.J.); (X.L.); (A.G.); (X.S.)
| | - Wenlin Jiang
- Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China; (Y.L.); (Z.Y.); (W.J.); (X.L.); (A.G.); (X.S.)
| | - Xinyi Lyu
- Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China; (Y.L.); (Z.Y.); (W.J.); (X.L.); (A.G.); (X.S.)
| | - Ailian Guo
- Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China; (Y.L.); (Z.Y.); (W.J.); (X.L.); (A.G.); (X.S.)
| | - Xiaorui Sun
- Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China; (Y.L.); (Z.Y.); (W.J.); (X.L.); (A.G.); (X.S.)
| | - Yiting Yang
- Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China; (Y.L.); (Z.Y.); (W.J.); (X.L.); (A.G.); (X.S.)
- NHC Key Laboratory of Reproduction Regulation, Shanghai-MOST Key Laboratory of Health and Disease Genomics, Shanghai Institute for Biomedical and Pharmaceutical Technologies (SIBPT), Shanghai 200032, China
| | - Yunfang Zhang
- Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China; (Y.L.); (Z.Y.); (W.J.); (X.L.); (A.G.); (X.S.)
| |
Collapse
|
27
|
Wu D, Li X, Khan FA, Yuan C, Pandupuspitasari NS, Huang C, Sun F, Guan K. tRNA modifications and tRNA-derived small RNAs: new insights of tRNA in human disease. Cell Biol Toxicol 2024; 40:76. [PMID: 39276283 PMCID: PMC11401796 DOI: 10.1007/s10565-024-09919-9] [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: 06/10/2024] [Accepted: 09/04/2024] [Indexed: 09/16/2024]
Abstract
tRNAs are codon decoders that convert the transcriptome into the proteome. The field of tRNA research is excited by the increasing discovery of specific tRNA modifications that are installed at specific, evolutionarily conserved positions by a set of specialized tRNA-modifying enzymes and the biogenesis of tRNA-derived regulatory fragments (tsRNAs) which exhibit copious activities through multiple mechanisms. Dysregulation of tRNA modification usually has pathological consequences, a phenomenon referred to as "tRNA modopathy". Current evidence suggests that certain tRNA-modifying enzymes and tsRNAs may serve as promising diagnostic biomarkers and therapeutic targets, particularly for chemoresistant cancers. In this review, we discuss the latest discoveries that elucidate the molecular mechanisms underlying the functions of clinically relevant tRNA modifications and tsRNAs, with a focus on malignancies. We also discuss the therapeutic potential of tRNA/tsRNA-based therapies, aiming to provide insights for the development of innovative therapeutic strategies. Further efforts to unravel the complexities inherent in tRNA biology hold the promise of yielding better biomarkers for the diagnosis and prognosis of diseases, thereby advancing the development of precision medicine for health improvement.
Collapse
Affiliation(s)
- Di Wu
- Institute of Reproductive Medicine, School of Medicine, Nantong University, Nantong, 226001, China
| | - Xiuling Li
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, 450046, China
| | - Faheem Ahmed Khan
- Research Center for Animal Husbandry, National Research and Innovation Agency, Jakarta Pusat, 10340, Indonesia
| | - Chenyang Yuan
- College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, China
| | | | - Chunjie Huang
- Institute of Reproductive Medicine, School of Medicine, Nantong University, Nantong, 226001, China.
| | - Fei Sun
- Institute of Reproductive Medicine, School of Medicine, Nantong University, Nantong, 226001, China.
| | - Kaifeng Guan
- School of Advanced Agricultural Sciences, Peking University, Beijing, 100871, China.
| |
Collapse
|
28
|
Davis DV, Choi EJ, Ismail D, Hernandez ML, Choi JM, Zhang K, Khatkar K, Jung SY, Wu W, Bao X. Role of Poly(A)-Binding Protein Cytoplasmic 1, a tRNA-Derived RNA Fragment-Bound Protein, in Respiratory Syncytial Virus Infection. Pathogens 2024; 13:791. [PMID: 39338982 PMCID: PMC11434780 DOI: 10.3390/pathogens13090791] [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: 07/30/2024] [Revised: 09/07/2024] [Accepted: 09/11/2024] [Indexed: 09/30/2024] Open
Abstract
Respiratory Syncytial Virus (RSV) is a significant cause of lower respiratory tract infections (LRTI) across all demographics, with increasing mortality and morbidity among high-risk groups such as infants under two years old, the elderly, and immunocompromised individuals. Although newly approved vaccines and treatments have substantially reduced RSV hospitalizations, accessibility remains limited, and response to treatment varies. This underscores the importance of comprehensive studies on host-RSV interactions. tRNA-derived RNA fragments (tRFs) are recently discovered non-coding RNAs, notable for their regulatory roles in diseases, including viral infections. Our prior work demonstrated that RSV infection induces tRFs, primarily derived from the 5'-end of a limited subset of tRNAs (tRF5), to promote RSV replication by partially targeting the mRNA of antiviral genes. This study found that tRFs could also use their bound proteins to regulate replication. Our proteomics data identified that PABPC1 (poly(A)-binding protein cytoplasmic 1) is associated with tRF5-GluCTC, an RSV-induced tRF. Western blot experimentally confirmed the presence of PABPC1 in the tRF5-GluCTC complex. In addition, tRF5-GluCTC is in the anti-PABPC1-precipitated immune complex. This study also discovered that suppressing PABPC1 with its specific siRNA increased RSV (-) genome copies without impacting viral gene transcription, but led to less infectious progeny viruses, suggesting the importance of PABPC1 in virus assembly, which was supported by its interaction with the RSV matrix protein. Additionally, PABPC1 knockdown decreased the production of the cytokines MIP-1α, MIP-1β, MCP-1, and TNF-α. This is the first observation suggesting that tRFs may regulate viral infection via their bound proteins.
Collapse
Affiliation(s)
- Devin V. Davis
- Department of Pediatrics, University of Texas Medical Branch, Galveston, TX 77555, USA; (D.V.D.); (E.-J.C.); (D.I.); (M.L.H.); (K.Z.); (K.K.)
| | - Eun-Jin Choi
- Department of Pediatrics, University of Texas Medical Branch, Galveston, TX 77555, USA; (D.V.D.); (E.-J.C.); (D.I.); (M.L.H.); (K.Z.); (K.K.)
| | - Deena Ismail
- Department of Pediatrics, University of Texas Medical Branch, Galveston, TX 77555, USA; (D.V.D.); (E.-J.C.); (D.I.); (M.L.H.); (K.Z.); (K.K.)
| | - Miranda L. Hernandez
- Department of Pediatrics, University of Texas Medical Branch, Galveston, TX 77555, USA; (D.V.D.); (E.-J.C.); (D.I.); (M.L.H.); (K.Z.); (K.K.)
| | - Jong Min Choi
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030, USA;
| | - Ke Zhang
- Department of Pediatrics, University of Texas Medical Branch, Galveston, TX 77555, USA; (D.V.D.); (E.-J.C.); (D.I.); (M.L.H.); (K.Z.); (K.K.)
| | - Kashish Khatkar
- Department of Pediatrics, University of Texas Medical Branch, Galveston, TX 77555, USA; (D.V.D.); (E.-J.C.); (D.I.); (M.L.H.); (K.Z.); (K.K.)
| | - Sung Yun Jung
- Verna and Marrs McLean Department of Biochemistry and Molecular Pharmacology, Baylor College of Medicine, Houston, TX 77030, USA;
| | - Wenzhe Wu
- Department of Pediatrics, University of Texas Medical Branch, Galveston, TX 77555, USA; (D.V.D.); (E.-J.C.); (D.I.); (M.L.H.); (K.Z.); (K.K.)
| | - Xiaoyong Bao
- Department of Pediatrics, University of Texas Medical Branch, Galveston, TX 77555, USA; (D.V.D.); (E.-J.C.); (D.I.); (M.L.H.); (K.Z.); (K.K.)
- Institute of Translational Science, University of Texas Medical Branch, Galveston, TX 77555, USA
- Institute for Human Infections & Immunity, University of Texas Medical Branch, Galveston, TX 77555, USA
| |
Collapse
|
29
|
Hernandez R, Li X, Shi J, Dave TR, Zhou T, Chen Q, Zhou C. Paternal hypercholesterolemia elicits sex-specific exacerbation of atherosclerosis in offspring. JCI Insight 2024; 9:e179291. [PMID: 39253968 PMCID: PMC11385100 DOI: 10.1172/jci.insight.179291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 07/18/2024] [Indexed: 09/11/2024] Open
Abstract
Emerging studies suggest that various parental exposures affect offspring cardiovascular health, yet the specific mechanisms, particularly the influence of paternal cardiovascular disease (CVD) risk factors on offspring cardiovascular health, remain elusive. The present study explores how paternal hypercholesterolemia affects offspring atherosclerosis development using the LDL receptor-deficient (LDLR-/-) mouse model. We found that paternal high-cholesterol diet feeding led to significantly increased atherosclerosis in F1 female, but not male, LDLR-/- offspring. Transcriptomic analysis highlighted that paternal hypercholesterolemia stimulated proatherogenic genes, including Ccn1 and Ccn2, in the intima of female offspring. Sperm small noncoding RNAs (sncRNAs), particularly transfer RNA-derived (tRNA-derived) small RNAs (tsRNAs) and rRNA-derived small RNAs (rsRNAs), contribute to the intergenerational transmission of paternally acquired metabolic phenotypes. Using a newly developed PANDORA-Seq method, we identified that high-cholesterol feeding elicited changes in sperm tsRNA/rsRNA profiles that were undetectable by traditional RNA-Seq, and these altered sperm sncRNAs were potentially key factors mediating paternal hypercholesterolemia-elicited atherogenesis in offspring. Interestingly, high-cholesterol feeding altered sncRNA biogenesis-related gene expression in the epididymis but not testis of LDLR-/- sires; this may have led to the modified sperm sncRNA landscape. Our results underscore the sex-specific intergenerational effect of paternal hypercholesterolemia on offspring cardiovascular health and contribute to the understanding of chronic disease etiology originating from parental exposures.
Collapse
Affiliation(s)
- Rebecca Hernandez
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, California, USA
| | - Xiuchun Li
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, California, USA
| | - Junchao Shi
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, California, USA
- Molecular Medicine Program, Department of Human Genetics, and
- Division of Urology, Department of Surgery, School of Medicine, University of Utah, Salt Lake City, Utah, USA
| | - Tejasvi R. Dave
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, California, USA
| | - Tong Zhou
- Department of Physiology and Cell Biology, University of Nevada, Reno School of Medicine, Reno, Nevada, USA
| | - Qi Chen
- Molecular Medicine Program, Department of Human Genetics, and
- Division of Urology, Department of Surgery, School of Medicine, University of Utah, Salt Lake City, Utah, USA
| | - Changcheng Zhou
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, California, USA
| |
Collapse
|
30
|
Caporali A, Anwar M, Devaux Y, Katare R, Martelli F, Srivastava PK, Pedrazzini T, Emanueli C. Non-coding RNAs as therapeutic targets and biomarkers in ischaemic heart disease. Nat Rev Cardiol 2024; 21:556-573. [PMID: 38499868 DOI: 10.1038/s41569-024-01001-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/19/2024] [Indexed: 03/20/2024]
Abstract
The adult heart is a complex, multicellular organ that is subjected to a series of regulatory stimuli and circuits and has poor reparative potential. Despite progress in our understanding of disease mechanisms and in the quality of health care, ischaemic heart disease remains the leading cause of death globally, owing to adverse cardiac remodelling, leading to ischaemic cardiomyopathy and heart failure. Therapeutic targets are urgently required for the protection and repair of the ischaemic heart. Moreover, personalized clinical biomarkers are necessary for clinical diagnosis, medical management and to inform the individual response to treatment. Non-coding RNAs (ncRNAs) deeply influence cardiovascular functions and contribute to communication between cells in the cardiac microenvironment and between the heart and other organs. As such, ncRNAs are candidates for translation into clinical practice. However, ncRNA biology has not yet been completely deciphered, given that classes and modes of action have emerged only in the past 5 years. In this Review, we discuss the latest discoveries from basic research on ncRNAs and highlight both the clinical value and the challenges underscoring the translation of these molecules as biomarkers and therapeutic regulators of the processes contributing to the initiation, progression and potentially the prevention or resolution of ischaemic heart disease and heart failure.
Collapse
Affiliation(s)
- Andrea Caporali
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
| | - Maryam Anwar
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Yvan Devaux
- Cardiovascular Research Unit, Department of Precision Health, Luxembourg Institute of Health, Luxembourg, Luxemburg
| | - Rajesh Katare
- Department of Physiology, HeartOtago, University of Otago, Dunedin, New Zealand
| | - Fabio Martelli
- Molecular Cardiology Laboratory, IRCCS Policlinico San Donato, Milan, Italy
| | | | - Thierry Pedrazzini
- Experimental Cardiology Unit, Division of Cardiology, Department of Cardiovascular Medicine, University of Lausanne Medical School, Lausanne, Switzerland
- School of Cardiovascular and Metabolic Medicine & Sciences, King's College London, London, UK
- British Heart Foundation Centre of Research Excellence, King's College London, London, UK
| | - Costanza Emanueli
- National Heart and Lung Institute, Imperial College London, London, UK.
| |
Collapse
|
31
|
Schultz SK, Kothe U. RNA modifying enzymes shape tRNA biogenesis and function. J Biol Chem 2024; 300:107488. [PMID: 38908752 PMCID: PMC11301382 DOI: 10.1016/j.jbc.2024.107488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 06/11/2024] [Accepted: 06/12/2024] [Indexed: 06/24/2024] Open
Abstract
Transfer RNAs (tRNAs) are the most highly modified cellular RNAs, both with respect to the proportion of nucleotides that are modified within the tRNA sequence and with respect to the extraordinary diversity in tRNA modification chemistry. However, the functions of many different tRNA modifications are only beginning to emerge. tRNAs have two general clusters of modifications. The first cluster is within the anticodon stem-loop including several modifications essential for protein translation. The second cluster of modifications is within the tRNA elbow, and roles for these modifications are less clear. In general, tRNA elbow modifications are typically not essential for cell growth, but nonetheless several tRNA elbow modifications have been highly conserved throughout all domains of life. In addition to forming modifications, many tRNA modifying enzymes have been demonstrated or hypothesized to also play an important role in folding tRNA acting as tRNA chaperones. In this review, we summarize the known functions of tRNA modifying enzymes throughout the lifecycle of a tRNA molecule, from transcription to degradation. Thereby, we describe how tRNA modification and folding by tRNA modifying enzymes enhance tRNA maturation, tRNA aminoacylation, and tRNA function during protein synthesis, ultimately impacting cellular phenotypes and disease.
Collapse
Affiliation(s)
- Sarah K Schultz
- Department of Chemistry, University of Manitoba, Winnipeg, Manitoba, Canada; Alberta RNA Research and Training Institute (ARRTI), Department of Chemistry and Biochemistry, University of Lethbridge, Lethbridge, Alberta, Canada.
| | - Ute Kothe
- Department of Chemistry, University of Manitoba, Winnipeg, Manitoba, Canada; Alberta RNA Research and Training Institute (ARRTI), Department of Chemistry and Biochemistry, University of Lethbridge, Lethbridge, Alberta, Canada.
| |
Collapse
|
32
|
Wang Q, Huang Q, Ying X, Zhou Y, Duan S. Exploring the regulatory role of tsRNAs in the TNF signaling pathway: Implications for cancer and non-cancer diseases. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2024; 191:1-10. [PMID: 38971324 DOI: 10.1016/j.pbiomolbio.2024.07.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 05/31/2024] [Accepted: 07/03/2024] [Indexed: 07/08/2024]
Abstract
Transfer RNA-derived small RNAs (tsRNAs), a recently identified subclass of small non-coding RNAs (sncRNAs), emerge through the cleavage of mature transfer RNA (tRNA) or tRNA precursors mediated by specific enzymes. The tumor necrosis factor (TNF) protein, a signaling molecule produced by activated macrophages, plays a pivotal role in systemic inflammation. Its multifaceted functions include the capacity to eliminate or hinder tumor cells, enhance the phagocytic capabilities of neutrophils, confer resistance against infections, induce fever, and prompt the production of acute phase proteins. Notably, four TNF-related tsRNAs have been conclusively linked to distinct diseases. Examples include 5'tiRNA-Gly in skeletal muscle injury, tsRNA-21109 in systemic lupus erythematosus (SLE), tRF-Leu-AAG-001 in endometriosis (EMs), and tsRNA-04002 in intervertebral disk degeneration (IDD). These tsRNAs exhibit the ability to suppress the expression of TNF-α. Additionally, KEGG analysis has identified seven tsRNAs potentially involved in modulating the TNF pathway, exerting their influence across a spectrum of non-cancerous diseases. Noteworthy instances include aberrant tiRNA-Ser-TGA-001 and tRF-Val-AAC-034 in intrauterine growth restriction (IUGR), irregular tRF-Ala-AGC-052 and tRF-Ala-TGC-027 in obesity, and deviant tiRNA-His-GTG-001, tRF-Ser-GCT-113, and tRF-Gln-TTG-035 in irritable bowel syndrome with diarrhea (IBS-D). This comprehensive review explores the biological functions and mechanisms of tsRNAs associated with the TNF signaling pathway in both cancer and other diseases, offering novel insights for future translational medical research.
Collapse
Affiliation(s)
- Qurui Wang
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, Zhejiang, China.
| | - Qinyuan Huang
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, Zhejiang, China.
| | - Xiaowei Ying
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, Zhejiang, China.
| | - Yang Zhou
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, Zhejiang, China.
| | - Shiwei Duan
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, Zhejiang, China; Department of Clinical Medicine, School of Medicine, Hangzhou City University, Hangzhou, Zhejiang, China.
| |
Collapse
|
33
|
Wang X, Zhang ZY, Zhao S, Liu MF. New insights into small non-coding RNAs during spermatogenesis. Sci Bull (Beijing) 2024; 69:1581-1585. [PMID: 38423874 DOI: 10.1016/j.scib.2024.02.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/02/2024]
Affiliation(s)
- Xin Wang
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China.
| | - Zhen-Yi Zhang
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China
| | - Shuang Zhao
- New Cornerstone Science Laboratory, State Key Laboratory of Molecular Biology, State Key Laboratory of Cell Biology, Shanghai Key Laboratory of Molecular Andrology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Mo-Fang Liu
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China; New Cornerstone Science Laboratory, State Key Laboratory of Molecular Biology, State Key Laboratory of Cell Biology, Shanghai Key Laboratory of Molecular Andrology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China; School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China.
| |
Collapse
|
34
|
Cai C, Chen Q. Father's diet influences son's metabolic health through sperm RNA. Nature 2024; 630:571-573. [PMID: 38839997 PMCID: PMC11573352 DOI: 10.1038/d41586-024-01502-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] [Indexed: 06/07/2024]
Abstract
DNA from organelles called mitochondria is not inherited from the father. But mitochondrial RNAs that sense paternal diet and mitochondrial quality are delivered from sperm to egg, affecting offspring metabolism.
Collapse
|
35
|
Siemers M, Lippegaus A, Papenfort K. ChimericFragments: computation, analysis and visualization of global RNA networks. NAR Genom Bioinform 2024; 6:lqae035. [PMID: 38633425 PMCID: PMC11023125 DOI: 10.1093/nargab/lqae035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 03/08/2024] [Accepted: 03/28/2024] [Indexed: 04/19/2024] Open
Abstract
RNA-RNA interactions are a key feature of post-transcriptional gene regulation in all domains of life. While ever more experimental protocols are being developed to study RNA duplex formation on a genome-wide scale, computational methods for the analysis and interpretation of the underlying data are lagging behind. Here, we present ChimericFragments, an analysis framework for RNA-seq experiments that produce chimeric RNA molecules. ChimericFragments implements a novel statistical method based on the complementarity of the base-pairing RNAs around their ligation site and provides an interactive graph-based visualization for data exploration and interpretation. ChimericFragments detects true RNA-RNA interactions with high precision and is compatible with several widely used experimental procedures such as RIL-seq, LIGR-seq or CLASH. We further demonstrate that ChimericFragments enables the systematic detection of novel RNA regulators and RNA-target pairs with crucial roles in microbial physiology and virulence. ChimericFragments is written in Julia and available at: https://github.com/maltesie/ChimericFragments.
Collapse
Affiliation(s)
- Malte Siemers
- Friedrich Schiller University, Institute of Microbiology, 07745 Jena, Germany
- Microverse Cluster, Friedrich Schiller University Jena, 07743 Jena, Germany
| | - Anne Lippegaus
- Friedrich Schiller University, Institute of Microbiology, 07745 Jena, Germany
| | - Kai Papenfort
- Friedrich Schiller University, Institute of Microbiology, 07745 Jena, Germany
- Microverse Cluster, Friedrich Schiller University Jena, 07743 Jena, Germany
| |
Collapse
|
36
|
Elder JJH, Papadopoulos R, Hayne CK, Stanley RE. The making and breaking of tRNAs by ribonucleases. Trends Genet 2024; 40:511-525. [PMID: 38641471 PMCID: PMC11152995 DOI: 10.1016/j.tig.2024.03.007] [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: 02/08/2024] [Revised: 03/21/2024] [Accepted: 03/21/2024] [Indexed: 04/21/2024]
Abstract
Ribonucleases (RNases) play important roles in supporting canonical and non-canonical roles of tRNAs by catalyzing the cleavage of the tRNA phosphodiester backbone. Here, we highlight how recent advances in cryo-electron microscopy (cryo-EM), protein structure prediction, reconstitution experiments, tRNA sequencing, and other studies have revealed new insight into the nucleases that process tRNA. This represents a very diverse group of nucleases that utilize distinct mechanisms to recognize and cleave tRNA during different stages of a tRNA's life cycle including biogenesis, fragmentation, surveillance, and decay. In this review, we provide a synthesis of the structure, mechanism, regulation, and modes of tRNA recognition by tRNA nucleases, along with open questions for future investigation.
Collapse
Affiliation(s)
- Jessica J H Elder
- Signal Transduction Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, USA
| | - Ry Papadopoulos
- Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, IL, USA; Department of Chemistry, University of Chicago, Chicago, IL, USA
| | - Cassandra K Hayne
- Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, IL, USA.
| | - Robin E Stanley
- Signal Transduction Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, USA.
| |
Collapse
|
37
|
Pawar K, Kawamura T, Kirino Y. The tRNA Val half: A strong endogenous Toll-like receptor 7 ligand with a 5'-terminal universal sequence signature. Proc Natl Acad Sci U S A 2024; 121:e2319569121. [PMID: 38683985 PMCID: PMC11087793 DOI: 10.1073/pnas.2319569121] [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: 11/07/2023] [Accepted: 03/24/2024] [Indexed: 05/02/2024] Open
Abstract
Toll-like receptors (TLRs) are crucial components of the innate immune system. Endosomal TLR7 recognizes single-stranded RNAs, yet its endogenous ssRNA ligands are not fully understood. We previously showed that extracellular (ex-) 5'-half molecules of tRNAHisGUG (the 5'-tRNAHisGUG half) in extracellular vesicles (EVs) of human macrophages activate TLR7 when delivered into endosomes of recipient macrophages. Here, we fully explored immunostimulatory ex-5'-tRNA half molecules and identified the 5'-tRNAValCAC/AAC half, the most abundant tRNA-derived RNA in macrophage EVs, as another 5'-tRNA half molecule with strong TLR7 activation capacity. Levels of the ex-5'-tRNAValCAC/AAC half were highly up-regulated in macrophage EVs upon exposure to lipopolysaccharide and in the plasma of patients infected with Mycobacterium tuberculosis. The 5'-tRNAValCAC/AAC half-mediated activation of TLR7 effectively eradicated bacteria infected in macrophages. Mutation analyses of the 5'-tRNAValCAC/AAC half identified the terminal GUUU sequence as a determinant for TLR7 activation. We confirmed that GUUU is the optimal ratio of guanosine and uridine for TLR7 activation; microRNAs or other RNAs with the terminal GUUU motif can indeed stimulate TLR7, establishing the motif as a universal signature for TLR7 activation. These results advance our understanding of endogenous ssRNA ligands of TLR7 and offer insights into diverse TLR7-involved pathologies and their therapeutic strategies.
Collapse
Affiliation(s)
- Kamlesh Pawar
- Computational Medicine Center, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA19107
- Department of Life Sciences, School of Natural Science, Shiv Nadar Institution of Eminence Deemed to be University, Delhi National Capital Region, Greater Noida201314, India
| | - Takuya Kawamura
- Computational Medicine Center, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA19107
| | - Yohei Kirino
- Computational Medicine Center, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA19107
| |
Collapse
|
38
|
Zhou M, He X, Zhang J, Mei C, Zhong B, Ou C. tRNA-derived small RNAs in human cancers: roles, mechanisms, and clinical application. Mol Cancer 2024; 23:76. [PMID: 38622694 PMCID: PMC11020452 DOI: 10.1186/s12943-024-01992-2] [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: 01/03/2024] [Accepted: 04/02/2024] [Indexed: 04/17/2024] Open
Abstract
Transfer RNA (tRNA)-derived small RNAs (tsRNAs) are a new type of non-coding RNAs (ncRNAs) produced by the specific cleavage of precursor or mature tRNAs. tsRNAs are involved in various basic biological processes such as epigenetic, transcriptional, post-transcriptional, and translation regulation, thereby affecting the occurrence and development of various human diseases, including cancers. Recent studies have shown that tsRNAs play an important role in tumorigenesis by regulating biological behaviors such as malignant proliferation, invasion and metastasis, angiogenesis, immune response, tumor resistance, and tumor metabolism reprogramming. These may be new potential targets for tumor treatment. Furthermore, tsRNAs can exist abundantly and stably in various bodily fluids (e.g., blood, serum, and urine) in the form of free or encapsulated extracellular vesicles, thereby affecting intercellular communication in the tumor microenvironment (TME). Meanwhile, their abnormal expression is closely related to the clinicopathological features of tumor patients, such as tumor staging, lymph node metastasis, and poor prognosis of tumor patients; thus, tsRNAs can be served as a novel type of liquid biopsy biomarker. This review summarizes the discovery, production, and expression of tsRNAs and analyzes their molecular mechanisms in tumor development and potential applications in tumor therapy, which may provide new strategies for early diagnosis and targeted therapy of tumors.
Collapse
Affiliation(s)
- Manli Zhou
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- Department of Clinical Laboratory, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Xiaoyun He
- Departments of Ultrasound Imaging, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Jing Zhang
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Cheng Mei
- Department of Blood Transfusion, Xiangya Hospital, Clinical Transfusion Research Center, Central South University, Changsha, Hunan, 410008, China.
| | - Baiyun Zhong
- Department of Clinical Laboratory, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.
| | - Chunlin Ou
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.
| |
Collapse
|
39
|
Wang Q, Huang Q, Ying X, Shen J, Duan S. Unveiling the role of tRNA-derived small RNAs in MAPK signaling pathway: implications for cancer and beyond. Front Genet 2024; 15:1346852. [PMID: 38596214 PMCID: PMC11002130 DOI: 10.3389/fgene.2024.1346852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 03/13/2024] [Indexed: 04/11/2024] Open
Abstract
tRNA-derived small RNAs (tsRNAs) are novel small non-coding RNAs originating from mature or precursor tRNAs (pre-tRNA), typically spanning 14 to 30 nt. The Mitogen-activated protein kinases (MAPK) pathway orchestrates cellular responses, influencing proliferation, differentiation, apoptosis, and transformation. tsRNAs influence the expression of the MAPK signaling pathway by targeting specific proteins within the pathway. Presently, four MAPK-linked tsRNAs have implications in gastric cancer (GC) and high-grade serous ovarian cancer (HGSOC). Notably, tRF-Glu-TTC-027 and tRF-Val-CAC-016 modulate MAPK-related protein expression, encompassing p38, Myc, ERK, CyclinD1, CyclinB, and c-Myc, hindering GC progression via MAPK pathway inhibition. Moreover, tRF-24-V29K9UV3IU and tRF-03357 remain unexplored in specific mechanisms. KEGG analysis posits varied tsRNAs in MAPK pathway modulation for diverse non-cancer maladies. Notably, high tRF-36-F900BY4D84KRIME and tRF-23-87R8WP9IY expression relates to varicose vein (VV) risk. Elevated tiRNA-Gly-GCC-001, tRF-Gly-GCC-012, tRF-Gly-GCC-013, and tRF-Gly-GCC-016 target spinal cord injury (SCI)-related brain-derived neurotrophic factor (BDNF), influencing MAPK expression. tRF-Gly-CCC-039 associates with diabetes foot sustained healing, while tRF-5014a inhibits autophagy-linked ATG5 in diabetic cardiomyopathy (DCM). Additionally, tsRNA-14783 influences keloid formation by regulating M2 macrophage polarization. Upregulation of tRF-Arg-ACG-007 and downregulation of tRF-Ser-GCT-008 are associated with diabetes. tsRNA-04002 alleviates Intervertebral disk degeneration (IDD) by targeting PRKCA. tsRNA-21109 alleviates Systemic lupus erythematosus (SLE) by inhibiting macrophage M1 polarization. The upregulated tiNA-Gly-GCC-002 and the downregulated tRF-Ala-AGC-010, tRF-Gln-CTG-005 and tRF-Leu-AAG-001 may be involved in the pathogenesis of Lupus nephritis (LN) by affecting the expression of MAPK pathway. Downregulation of tsRNA-1018, tsRNA-3045b, tsRNA-5021a and tsRNA-1020 affected the expression of MAPK pathway, thereby improving Acute lung injury (ALI). This review comprehensively dissects tsRNA roles in MAPK signaling across cancers and other diseases, illuminating a novel avenue for translational medical exploration.
Collapse
Affiliation(s)
- Qurui Wang
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, Zhejiang, China
| | - Qinyuan Huang
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, Zhejiang, China
| | - Xiaowei Ying
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, Zhejiang, China
| | - Jinze Shen
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, Zhejiang, China
| | - Shiwei Duan
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, Zhejiang, China
- Department of Clinical Medicine, School of Medicine, Hangzhou City University, Hangzhou, Zhejiang, China
| |
Collapse
|
40
|
Zhang J. Recognition of the tRNA structure: Everything everywhere but not all at once. Cell Chem Biol 2024; 31:36-52. [PMID: 38159570 PMCID: PMC10843564 DOI: 10.1016/j.chembiol.2023.12.008] [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: 08/27/2023] [Revised: 12/02/2023] [Accepted: 12/11/2023] [Indexed: 01/03/2024]
Abstract
tRNAs are among the most abundant and essential biomolecules in cells. These spontaneously folding, extensively structured yet conformationally flexible anionic polymers literally bridge the worlds of RNAs and proteins, and serve as Rosetta stones that decipher and interpret the genetic code. Their ubiquitous presence, functional irreplaceability, and privileged access to cellular compartments and ribosomes render them prime targets for both endogenous regulation and exogenous manipulation. There is essentially no part of the tRNA that is not touched by another interaction partner, either as programmed or imposed by an external adversary. Recent progresses in genetic, biochemical, and structural analyses of the tRNA interactome produced a wealth of new knowledge into their interaction networks, regulatory functions, and molecular interfaces. In this review, I describe and illustrate the general principles of tRNA recognition by proteins and other RNAs, and discuss the underlying molecular mechanisms that deliver affinity, specificity, and functional competency.
Collapse
Affiliation(s)
- Jinwei Zhang
- Laboratory of Molecular Biology, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD 20892, USA.
| |
Collapse
|