1
|
Xu Z, Asakawa S. Release and degradation of dissolved environmental RNAs from zebrafish cells. RNA Biol 2025; 22:1-12. [PMID: 40167163 PMCID: PMC12026185 DOI: 10.1080/15476286.2025.2486281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2024] [Revised: 02/27/2025] [Accepted: 03/21/2025] [Indexed: 04/02/2025] Open
Abstract
The sources and degradation profiles of dissolved environmental RNAs from fish in water remain unknown. In this study, laboratory experiments and mathematical modelling were conducted to investigate the permeability of RNA extracted from zebrafish cells through filters, the release of dissolved environmental RNAs from live and dying zebrafish cells, and the degradation of RNA extracted from zebrafish cells in a non-sterile aqueous environment. This research aimed to provide biological and ecological insights into fish RNAs dissolved in water. The results showed that most of the RNA extracted from zebrafish cells was detected in the filtrates after passage through 0.45 µm filters. Over the course of the 6-day experiment, dynamic levels of the RNAs in the liquid environment containing live or dying zebrafish cells were determined. The release and degradation rates of dissolved environmental RNA from zebrafish cells were calculated using mathematical modelling. RNA extracted from zebrafish cells degraded in non-sterile water in the tubes, and after 2 months, more than 15% of the RNAs in the water remained detectable. The half-life of the RNA in the tubes was approximately 20 ~ 43 days. The modelling results suggest that the levels of the dissolved environmental fish RNAs in natural waters or aquariums could be so low that it would be difficult to detect them using current techniques. The results obtained in this study will help develop new methods for measuring the dynamics of dissolved environmental fish RNAs in water and determining their significance.
Collapse
Affiliation(s)
- Zhongneng Xu
- Department of Ecology, Jinan University, Guangzhou, China
- Department of Aquatic Bioscience, Graduate School of Agricultural and Life Science, The University of Tokyo, Tokyo, Japan
| | - Shuichi Asakawa
- Department of Aquatic Bioscience, Graduate School of Agricultural and Life Science, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
2
|
Han L, Liu J, Zhang R, Cheng Y, Dong L, Wei L, Liu J, Wang K, Yu J. Insights From Nonsense-Mediated mRNA Decay for Prognosis in Homologous Recombination-Deficient Ovarian Cancer. Cancer Sci 2025; 116:1449-1463. [PMID: 40022542 PMCID: PMC12044663 DOI: 10.1111/cas.70034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 02/14/2025] [Accepted: 02/19/2025] [Indexed: 03/03/2025] Open
Abstract
Not all ovarian cancer patients with homologous recombination deficiency, especially those with germline BRCA mutations, can benefit from platinum-based and targeted therapy. Our study aimed to determine the value of nonsense-mediated mRNA decay, which targeted these mutations. The retrospective analysis of 797 ovarian cancer patients was performed using two public cohorts and one in-house cohort. We developed a prediction algorithm for nonsense-mediated mRNA decay to discriminate between trigger and escape status, finding that escape status indicated a better prognosis. Subsequently, we analyzed differential gene expression and functional pathways between the two statuses and filtered 8 genes associated with the cell cycle. Then the optimized key gene model was built using integrated machine learning algorithms (mean AUC > 0.89), which had a higher independent prognostic value for ovarian cancer with germline BRCA variants or homologous recombination deficiency than the nonsense-mediated mRNA decay algorithm. Furthermore, we classified patients into high- and low-risk groups by the machine learning model and found that the low-risk group had a better prognosis with higher drug response and immune levels of activated dendritic cells than the high-risk controls. Our findings provide a perspective based on nonsense-mediated mRNA decay and cell cycle pathways to distinguish subtypes of germline BRCA or homologous recombination deficiency.
Collapse
Affiliation(s)
- Lei Han
- Cancer Molecular Diagnostics CoreTianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Caner, Key Laboratory of Cancer Immunology and Biotherapy, Tianjin's Clinical Research Center for CancerTianjinChina
| | - Jialing Liu
- Cancer Molecular Diagnostics CoreTianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Caner, Key Laboratory of Cancer Immunology and Biotherapy, Tianjin's Clinical Research Center for CancerTianjinChina
| | - Runjiao Zhang
- Cancer Molecular Diagnostics CoreTianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Caner, Key Laboratory of Cancer Immunology and Biotherapy, Tianjin's Clinical Research Center for CancerTianjinChina
| | - Yanan Cheng
- Cancer Molecular Diagnostics CoreTianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Caner, Key Laboratory of Cancer Immunology and Biotherapy, Tianjin's Clinical Research Center for CancerTianjinChina
| | - Li Dong
- Cancer Molecular Diagnostics CoreTianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Caner, Key Laboratory of Cancer Immunology and Biotherapy, Tianjin's Clinical Research Center for CancerTianjinChina
| | - Lijuan Wei
- Cancer Prevention CenterTianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for CancerTianjinChina
| | - Juntian Liu
- Cancer Prevention CenterTianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for CancerTianjinChina
| | - Ke Wang
- Department of Gynecologic OncologyTianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for CancerTianjinChina
| | - Jinpu Yu
- Cancer Molecular Diagnostics CoreTianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Caner, Key Laboratory of Cancer Immunology and Biotherapy, Tianjin's Clinical Research Center for CancerTianjinChina
| |
Collapse
|
3
|
Lockbaum GJ, Lynes MM, Shen SA, Liu J, Holt N, Nayak SP, Knockenhauer KE, Yao S, Sickmier EA, Raman A, Wu J, Case A, Shehaj L, Buker SM, Grigoriu S, Ribich S, Blakemore SJ, Sparling BA, Duncan KW, Copeland RA, Silver SJ, Boriack-Sjodin PA. Characterization of exoribonuclease XRN1 as a cancer target and identification of adenosine-3',5'-bisphosphate as a potent enzyme inhibitor. Commun Biol 2025; 8:589. [PMID: 40205031 PMCID: PMC11982291 DOI: 10.1038/s42003-025-08005-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Accepted: 03/26/2025] [Indexed: 04/11/2025] Open
Abstract
XRN1 (5'-3' exoribonuclease 1) degrades RNA from the 5' → 3' direction and utilizes both single- and double-stranded RNA as substrates. XRN1 plays a critical role in mRNA turnover as well as regulating the cellular response to viral infection. XRN1 also protects the cell by preventing endogenous double-stranded RNA accumulation. XRN1 was identified as a putative vulnerability in a subset of cancer cell lines through analysis of publicly available CRISPR data. The role of XRN1 was explored using a set of non-small cell lung cancer cell lines with differential predicted XRN1 dependency to validate XRN1 as an oncology target. In predicted sensitive cell lines, XRN1 knockout reduced proliferation, increased apoptosis and activated the pPKR and MDA5 dsRNA sensing pathways. To facilitate drug discovery targeting XRN1, a suite of biochemical and biophysical assays was developed. These assays were used to characterize adenosine-3',5'-bisphosphate (pAp), a non-selective nuclease inhibitor, as a nanomolar inhibitor of XRN1. Additionally, the crystal structure of human XRN1 was solved with pAp bound, demonstrating distinct interactions for the compound in the XRN1 active site. These studies provide a strong foundation for the discovery of potent, selective inhibitors of XRN1 as a novel approach to cancer therapeutics.
Collapse
Affiliation(s)
| | | | | | - Julie Liu
- Accent Therapeutics, Lexington, MA, 02421, USA
| | | | | | | | - Shihua Yao
- Accent Therapeutics, Lexington, MA, 02421, USA
| | | | | | - Jie Wu
- Accent Therapeutics, Lexington, MA, 02421, USA
| | - April Case
- Accent Therapeutics, Lexington, MA, 02421, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
4
|
Wang X, Jin J, Yan H, Liu J, Huang S, Bai H, Guo M, Cheng X, Deng T, Ba Y, Gu Y, Gao X, Hu D. The mRNA export pathway licenses viral mimicry response and antitumor immunity by actively exporting nuclear retroelement transcripts. Sci Transl Med 2025; 17:eado4370. [PMID: 40203080 DOI: 10.1126/scitranslmed.ado4370] [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: 02/01/2024] [Revised: 08/07/2024] [Accepted: 03/06/2025] [Indexed: 04/11/2025]
Abstract
Nuclear retroelement transcripts (RTs), which can be elicited both transcriptionally and posttranscriptionally, form double-stranded RNA (dsRNA) in cytosol to trigger the viral mimicry response (VMR) and antitumor immunity. However, the strength of the induced VMR varies tremendously across tumor types, and the underlying mechanisms remain poorly understood. Here, we demonstrate that the mRNA export pathway modulates the VMR through actively exporting nuclear RTs for cytosolic dsRNA formation after their induction. Tumor cells hijack this process for immune evasion through aberrant coactivator-associated arginine methyltransferase 1 (CARM1) expression. Mechanistically, we show that the cytoplasmic transportation of RTs by the mRNA export pathway is counteracted by the RNA exosome, which cleaves multiple transcripts within this pathway, including those encoding the essential DExD-box helicase 39A (DDX39A) and the adaptor protein ALYREF. CARM1 enhances the RNA exosome activity to attenuate the nuclear export of RTs by the mRNA export pathway through two synergistic mechanisms: (i) transcriptionally activating several RNA exosome components and (ii) posttranslationally methylating arginine 6 of the RNA exosome subunit EXOSC1, which protects it from proteasome-mediated degradation. Collectively, our study highlights the critical active regulatory role of the mRNA export pathway in transporting nuclear RTs into the cytosol for triggering the VMR and tumor immunity. Furthermore, we propose that enhancing the mRNA export pathway activity, either through CARM1 inhibition or RNA exosome modulation, could reinforce the therapeutic agent-induced VMR, thus holding the promise for overcoming tumor immune evasion and immunotherapy resistance.
Collapse
Affiliation(s)
- Xiaoqiang Wang
- Key Laboratory of Breast Cancer Prevention and Therapy (Ministry of Education), State Key Laboratory of Experimental Hematology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Department of GI Medical Oncology, Tianjin Medical University Cancer Institute and Hospital, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Jiaxing Jin
- Key Laboratory of Breast Cancer Prevention and Therapy (Ministry of Education), State Key Laboratory of Experimental Hematology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Department of GI Medical Oncology, Tianjin Medical University Cancer Institute and Hospital, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Han Yan
- Key Laboratory of Breast Cancer Prevention and Therapy (Ministry of Education), State Key Laboratory of Experimental Hematology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Department of GI Medical Oncology, Tianjin Medical University Cancer Institute and Hospital, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Jinhua Liu
- Key Laboratory of Breast Cancer Prevention and Therapy (Ministry of Education), State Key Laboratory of Experimental Hematology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Department of GI Medical Oncology, Tianjin Medical University Cancer Institute and Hospital, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Shan Huang
- Key Laboratory of Breast Cancer Prevention and Therapy (Ministry of Education), State Key Laboratory of Experimental Hematology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Department of GI Medical Oncology, Tianjin Medical University Cancer Institute and Hospital, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Hui Bai
- Key Laboratory of Breast Cancer Prevention and Therapy (Ministry of Education), State Key Laboratory of Experimental Hematology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Department of GI Medical Oncology, Tianjin Medical University Cancer Institute and Hospital, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Mingrui Guo
- Key Laboratory of Breast Cancer Prevention and Therapy (Ministry of Education), State Key Laboratory of Experimental Hematology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Department of GI Medical Oncology, Tianjin Medical University Cancer Institute and Hospital, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Xinyue Cheng
- Key Laboratory of Breast Cancer Prevention and Therapy (Ministry of Education), State Key Laboratory of Experimental Hematology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Department of GI Medical Oncology, Tianjin Medical University Cancer Institute and Hospital, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Ting Deng
- Tianjin's Clinical Research Center for Cancer, Tianjin Key Laboratory of Digestive Cancer, Key Laboratory of Cancer Prevention and Therapy, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, China
| | - Yi Ba
- Tianjin's Clinical Research Center for Cancer, Tianjin Key Laboratory of Digestive Cancer, Key Laboratory of Cancer Prevention and Therapy, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, China
| | - Yong Gu
- Clinical Research Center, Hainan Hospital, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou University of Chinese Medicine, Hainan, 570203, China
| | - Xin Gao
- Key Laboratory of Breast Cancer Prevention and Therapy (Ministry of Education), State Key Laboratory of Experimental Hematology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Department of GI Medical Oncology, Tianjin Medical University Cancer Institute and Hospital, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Deqing Hu
- Key Laboratory of Breast Cancer Prevention and Therapy (Ministry of Education), State Key Laboratory of Experimental Hematology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Department of GI Medical Oncology, Tianjin Medical University Cancer Institute and Hospital, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
- Tianjin's Clinical Research Center for Cancer, Tianjin Key Laboratory of Digestive Cancer, Key Laboratory of Cancer Prevention and Therapy, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, China
| |
Collapse
|
5
|
Telkoparan-Akillilar P, Chichiarelli S, Tucci P, Saso L. Integration of MicroRNAs with nanomedicine: tumor targeting and therapeutic approaches. Front Cell Dev Biol 2025; 13:1569101. [PMID: 40260417 PMCID: PMC12009947 DOI: 10.3389/fcell.2025.1569101] [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: 01/31/2025] [Accepted: 03/12/2025] [Indexed: 04/23/2025] Open
Abstract
MicroRNAs (miRNAs) are small, non-coding RNA molecules that play a pivotal role in the post-transcriptional regulation of gene expression. Over the past decade, they have emerged as key regulators in cancer progression, influencing different cellular processes such as proliferation, apoptosis, metastasis, and immune evasion. Their unique ability to target multiple genes simultaneously makes miRNAs highly attractive as potential therapeutic agents in oncology. However, several challenges have hindered their direct clinical application, most notably their inherent instability in biological fluids, rapid degradation by nucleases, and inefficient delivery to specific tumor sites. Additionally, off-target effects and the potential for toxicity further complicate the therapeutic use of miRNAs. Nanomedicine offers a promising solution to these challenges by enabling the development of advanced platforms for the stable, safe, and targeted delivery of miRNAs. Nanoparticle-based delivery systems, such as liposomes, polymeric nanoparticles, and inorganic nanocarriers, can protect miRNAs from degradation, improve their bioavailability, and allow for precise tumor targeting through passive or active targeting mechanisms. These nanocarriers can also be engineered to release miRNAs in response to specific stimuli within the tumor microenvironment, enhancing therapeutic efficacy while minimizing side effects. This review will explore the integration of miRNAs with nanotechnology, focusing on various nanoparticle formulations and their roles in enhancing miRNA stability, specificity, and function in cancer treatment. In addition, we will discuss current advances in preclinical and clinical applications, highlight promising tumor-targeting strategies, and address the remaining challenges such as toxicity, immunogenicity, and scalability. Future research should focus on overcoming these barriers, ultimately paving the way for the widespread adoption of personalized miRNA-based nanomedicine in cancer therapy.
Collapse
Affiliation(s)
| | - Silvia Chichiarelli
- Department of Biochemical Sciences “A. Rossi-Fanelli”, Sapienza University of Rome, Rome, Italy
| | - Paolo Tucci
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Luciano Saso
- Department of Physiology and Pharmacology “Vittorio Erspamer”, La Sapienza University, Rome, Italy
| |
Collapse
|
6
|
Previderè C, Bonin S, Cuttaia C, Argentiero G, Livieri T, Cecchetto G, Oliva A, Fattorini P. Are pre-analytical factors fully considered in forensic FFPE molecular analyses? A systematic review reveals the need for standardised procedures. Int J Legal Med 2025:10.1007/s00414-025-03480-8. [PMID: 40172636 DOI: 10.1007/s00414-025-03480-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Accepted: 03/19/2025] [Indexed: 04/04/2025]
Abstract
The need for molecular analyses has become increasingly common in the forensic sciences, particularly in forensic pathology, to better shape the causes of death. This approach is called the "molecular autopsy," where conventional medico-legal findings are often enhanced with specific molecular tests to provide reliable clinical and forensic diagnoses. In this context, FFPE (Formalin-Fixed Paraffin-Embedded) tissue samples collected during forensic autopsies are the only available specimens in retrospective studies for molecular DNA and/or RNA analyses. It is well known that pre-analytical parameters such as the agonal time, the PMI (Post-Mortem Interval), the fixation procedures, and the FFPE ageing and storage conditions can deeply impact the quality and quantity of the recovered nucleic acids, thus influencing the reliability of the downstream molecular tests. In the present study, we reviewed the recent forensic literature to establish whether these parameters are reported. Our survey showed that up to 34.9% and 40.5% of the 50 selected studies on DNA and RNA, respectively, reported the pre-analytical parameters mentioned above. Many publications did not report the length of agony (if any), which is an important parameter in RNA-based studies and estimations of the PMI; in addition, even relevant information on formalin tissue fixation procedures was often missing, thus impairing any critical evaluation of the PCR-based results. To address these issues, we propose the use of a simple form we set up to be filled out by Forensic Pathologists, where each pre-analytical step concerning the tissue samples collected during autopsy is accurately described and reported. In our opinion, this standardization will help the forensic community compare and evaluate the results of different molecular tests, thus increasing the reliability of the molecular results in forensics.
Collapse
Affiliation(s)
- Carlo Previderè
- Department of Public Health, Experimental and Forensic Medicine, Section of Legal Medicine and Forensic Sciences, University of Pavia, 27100, Pavia, Italy
| | - Serena Bonin
- Department of Medical Sciences, University of Trieste, 34149, Trieste, Italy
| | - Calogero Cuttaia
- Department of Medical Sciences, University of Trieste, 34149, Trieste, Italy
| | - Gianmarco Argentiero
- Department of Public Health, Experimental and Forensic Medicine, Section of Legal Medicine and Forensic Sciences, University of Pavia, 27100, Pavia, Italy
| | - Tommaso Livieri
- Department of Medical Sciences, University of Trieste, 34149, Trieste, Italy
| | - Giovanni Cecchetto
- Department of Public Health, Experimental and Forensic Medicine, Section of Legal Medicine and Forensic Sciences, University of Pavia, 27100, Pavia, Italy
| | - Antonio Oliva
- Department of Health Surveillance and Bioethics, Section of Legal Medicine, Fondazione Policlinico A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, 00168, Rome, Italy
| | - Paolo Fattorini
- Department of Medical Sciences, University of Trieste, 34149, Trieste, Italy.
| |
Collapse
|
7
|
Lee H, Massaro M, Abdelfattah N, Baudo G, Liu H, Yun K, Blanco E. Nuclear respiratory factor-1 (NRF1) induction as a powerful strategy to deter mitochondrial dysfunction and senescence in mesenchymal stem cells. Aging Cell 2025; 24:e14446. [PMID: 39720856 PMCID: PMC11984659 DOI: 10.1111/acel.14446] [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: 04/09/2024] [Revised: 11/27/2024] [Accepted: 12/02/2024] [Indexed: 12/26/2024] Open
Abstract
Mesenchymal stem cells (MSCs) are promising candidates for regenerative therapies due to their self-renewal and differentiation capabilities. Pathological microenvironments expose MSCs to senescence-inducing factors such as reactive oxygen species (ROS), resulting in MSC functional decline and loss of stemness. Oxidative stress leads to mitochondrial dysfunction, a hallmark of senescence, and is prevalent in aging tissues characterized by elevated ROS levels. We hypothesized that overexpression of nuclear respiratory factor-1 (NRF1), a driver of mitochondrial biogenesis, could metabolically potentiate MSCs and prevent MSC senescence. Single-cell RNA sequencing (scRNA-Seq) revealed that MSCs transfected with NRF1 messenger RNA (mRNA) exhibited upregulated expression of genes associated with oxidative phosphorylation (OXPHOS), decreased glycolytic markers, and suppression of senescence-related pathways. To test whether NRF1 induction could mitigate stress-induced premature senescence, we exposed MSCs to hydrogen peroxide (H2O2) and validated our findings in a replicative senescence model. NRF1 mRNA transfection significantly increased mitochondrial mass and improved aberrant mitochondrial processes associated with senescence, including reduced mitochondrial and intracellular total ROS production. Mitochondrial health and dynamics were preserved, and respiratory function was restored, as evidenced by enhanced OXPHOS, reduced glycolysis, and increased ATP production. Notably, NRF1 overexpression led to decreased senescence-associated β-galactosidase (SA-β-gal) activity and reduced expression of senescence markers p53, p21, and p16. Our findings demonstrate that NRF1 induction attenuates MSC senescence by enhancing mitochondrial function, suggesting potential translational applications for MSC-based therapies and senescence-targeted interventions.
Collapse
Affiliation(s)
- Hyunho Lee
- Department of NanomedicineHouston Methodist Research InstituteHoustonTexasUSA
| | - Matteo Massaro
- Department of NanomedicineHouston Methodist Research InstituteHoustonTexasUSA
| | | | - Gherardo Baudo
- Department of NanomedicineHouston Methodist Research InstituteHoustonTexasUSA
| | - Haoran Liu
- Department of NanomedicineHouston Methodist Research InstituteHoustonTexasUSA
| | - Kyuson Yun
- Department of NeurologyHouston Methodist Research InstituteHoustonTexasUSA
- Department of NeurologyWeill Cornell Medical CollegeNew YorkNew YorkUSA
| | - Elvin Blanco
- Department of NanomedicineHouston Methodist Research InstituteHoustonTexasUSA
- Department of MedicineWeill Cornell Medical CollegeNew YorkNew YorkUSA
- Department of CardiologyHouston Methodist DeBakey Heart and Vascular Center, Houston Methodist HospitalHoustonTexasUSA
| |
Collapse
|
8
|
Šupljika N, Paić A, Novačić A, Martinić Cezar T, Vallée B, Teparić R, Stuparević I, Žunar B. Saccharomyces cerevisiae Mub1, a substrate adaptor of E3 ubiquitin ligase Ubr2, modulates sensitivity to cell wall stressors through multiple transcription factors. FEBS J 2025. [PMID: 40165610 DOI: 10.1111/febs.70091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 12/20/2024] [Accepted: 03/25/2025] [Indexed: 04/02/2025]
Abstract
Yeasts evolved a complex regulatory programme to build and maintain their cell wall, the primary structure through which they interact with their environment. However, how this programme ties to essential cellular processes mostly remains unclear. Here, we focus on Saccharomyces cerevisiae MYND-type zinc finger protein MUB1 (Mub1), an adaptor protein of E3 ubiquitin-protein ligase Ubr2 that was previously associated with regulating proteasome genes through the transcription factor Rpn4. We show that S. cerevisiae cells lacking Mub1 become hyper-tolerant to standard cell wall stressors, outperforming wild-type cells. This protective mub1Δ phenotype stems from the activity of several transcription factors, leading to the inhibition of cell wall remodelling, a typically protective process that becomes maladaptive during chronic cell wall stress in laboratory conditions. Based on these results, we suggest that Mub1 regulates not only Rpn4 but a much broader range of transcription factors, and thus serves as an in-so-far unrecognised regulatory hub directly linking cell wall robustness with the ubiquitin-proteasome system.
Collapse
Affiliation(s)
- Nada Šupljika
- Laboratory for Biochemistry, Department of Chemistry and Biochemistry, University of Zagreb Faculty of Food Technology and Biotechnology, Pierottijeva 6, Zagreb, 10000, Croatia
| | - Antonia Paić
- Laboratory for Biochemistry, Department of Chemistry and Biochemistry, University of Zagreb Faculty of Food Technology and Biotechnology, Pierottijeva 6, Zagreb, 10000, Croatia
| | - Ana Novačić
- Laboratory for Biochemistry, Department of Chemistry and Biochemistry, University of Zagreb Faculty of Food Technology and Biotechnology, Pierottijeva 6, Zagreb, 10000, Croatia
| | - Tea Martinić Cezar
- Laboratory for Biochemistry, Department of Chemistry and Biochemistry, University of Zagreb Faculty of Food Technology and Biotechnology, Pierottijeva 6, Zagreb, 10000, Croatia
| | - Béatrice Vallée
- Centre de Biophysique Moléculaire (CBM), CNRS, UPR 4301, University of Orléans and INSERM, Orléans Cedex 2, France
| | - Renata Teparić
- Laboratory for Biochemistry, Department of Chemistry and Biochemistry, University of Zagreb Faculty of Food Technology and Biotechnology, Pierottijeva 6, Zagreb, 10000, Croatia
| | - Igor Stuparević
- Laboratory for Biochemistry, Department of Chemistry and Biochemistry, University of Zagreb Faculty of Food Technology and Biotechnology, Pierottijeva 6, Zagreb, 10000, Croatia
| | - Bojan Žunar
- Laboratory for Biochemistry, Department of Chemistry and Biochemistry, University of Zagreb Faculty of Food Technology and Biotechnology, Pierottijeva 6, Zagreb, 10000, Croatia
| |
Collapse
|
9
|
Shams M, Khadivi A. Mechanistic insights into DXO1 and XRN3: regulatory roles of RNA stability, transcription, and liquid-liquid phase separation in Arabidopsis thaliana (L.) Heynh. PLANT SCIENCE : AN INTERNATIONAL JOURNAL OF EXPERIMENTAL PLANT BIOLOGY 2025; 353:112413. [PMID: 39909287 DOI: 10.1016/j.plantsci.2025.112413] [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: 08/22/2024] [Revised: 01/29/2025] [Accepted: 01/31/2025] [Indexed: 02/07/2025]
Abstract
The regulation of RNA stability and transcription in eukaryotic organisms is a sophisticated process involving various complex mechanisms. This paper explores the regulatory functions of DXO1 and XRN3 proteins in RNA stability and transcription in the model plant Arabidopsis thaliana (L.) Heynh. DXO1 is noted for its roles in mRNA 5'-end quality control, removal of non-canonical NAD+ caps, and activation of RNA guanosine-7 methyltransferase. In contrast, XRN3 ensures RNA integrity through precise degradation. While current studies have identified various termination regions across genes influenced by XRN3, advanced RNA sequencing techniques have revealed that XRN3-mediated changes in gene expression often result from siRNA production, leading to gene silencing rather than direct effects on transcription termination. This review emphasizes the need to further explore the DXO1-XRN3 axis, their interactive mechanisms, and their potential involvement in liquid-liquid phase separation (LLPS) during transcription. It further suggests evaluating XRN proteins like XRN4 to assess potential redundancies in RNA degradation pathways. The advent of PSPredictor, a tool for identifying LLPS proteins, along with protein function prediction techniques, promises to advance our understanding of DXO1 and XRN3 in maintaining RNA equilibrium and the dynamics of LLPS in plant biology. The review concludes by calling for more studies on the plant-specific roles of the DXO1 N-terminal extension (NTE), predictive tools for LLPS-forming proteins, and the interplay of RNA Pol II CTD code modulation by transcription factors to enhance knowledge of plant stress adaptation and improve agricultural productivity.
Collapse
Affiliation(s)
- Mostafakamal Shams
- Department of Plant Experimental Biology and Biotechnology, Faculty of Biology, University of Gdansk, Gdansk, Poland.
| | - Ali Khadivi
- Department of Horticultural Sciences, Faculty of Agriculture and Natural Resources, Arak University, Arak 38156-8-8349, Iran.
| |
Collapse
|
10
|
Xie L, Jakutis G, Dooley CM, Guenther S, Kontarakis Z, Howard SP, Juan T, Stainier DYR. Induction of a transcriptional adaptation response by RNA destabilization events. EMBO Rep 2025:10.1038/s44319-025-00427-3. [PMID: 40128410 DOI: 10.1038/s44319-025-00427-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 03/03/2025] [Accepted: 03/10/2025] [Indexed: 03/26/2025] Open
Abstract
Transcriptional adaptation (TA) is a cellular process whereby mRNA-destabilizing mutations are associated with the transcriptional upregulation of so-called adapting genes. The nature of the TA-triggering factor(s) remains unclear, namely whether an mRNA-borne premature termination codon or the subsequent mRNA decay process, and/or its products, elicits TA. Here, working with mouse Actg1, we first establish two types of perturbations that lead to mRNA destabilization: Cas9-induced mutations predicted to lead to mutant mRNA decay, and Cas13d-mediated mRNA cleavage. We find that both types of perturbations are effective in degrading Actg1 mRNA, and that they both upregulate Actg2. Notably, increased chromatin accessibility at the Actg2 locus was observed only in the Cas9-induced mutant cells but not in the Cas13d-targeted cells, suggesting that chromatin remodeling is not required for Actg2 upregulation. We further show that ribozyme-mediated Actg1 pre-mRNA cleavage also leads to a robust upregulation of Actg2, and that this upregulation is again independent of chromatin remodeling. Together, these data highlight the critical role of RNA destabilization events as a trigger for TA, or at least a TA-like response.
Collapse
Affiliation(s)
- Lihan Xie
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Hessen, 61231, Germany
| | - Gabrielius Jakutis
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Hessen, 61231, Germany
| | - Christopher M Dooley
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Hessen, 61231, Germany
| | - Stefan Guenther
- ECCPS Bioinformatics and Deep Sequencing Platform, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Hessen, 61231, Germany
| | - Zacharias Kontarakis
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Hessen, 61231, Germany
- Genome Engineering and Measurement Laboratory (GEML), Eidgenössische Technische Hochschule (ETH) Zürich, Zürich, Switzerland
- Functional Genomics Center Zürich, ETH Zürich/University of Zürich, Zürich, 8057, Switzerland
| | - Sarah P Howard
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Hessen, 61231, Germany
| | - Thomas Juan
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Hessen, 61231, Germany.
- German Centre for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Bad Nauheim, Hessen, 61231, Germany.
- Excellence Cluster Cardio-Pulmonary Institute (CPI), Bad Nauheim, Giessen, Frankfurt, Germany.
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, 75 185, Sweden.
| | - Didier Y R Stainier
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Hessen, 61231, Germany.
- German Centre for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Bad Nauheim, Hessen, 61231, Germany.
- Excellence Cluster Cardio-Pulmonary Institute (CPI), Bad Nauheim, Giessen, Frankfurt, Germany.
| |
Collapse
|
11
|
Laturski AE, Dulay MT, Perry JL, DeSimone JM. Transfection via RNA-Based Nanoparticles: Comparing Encapsulation vs Adsorption Approaches of RNA Incorporation. Bioconjug Chem 2025; 36:367-376. [PMID: 39999074 DOI: 10.1021/acs.bioconjchem.5c00028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/27/2025]
Abstract
Historically, RNA delivery via nanoparticles has primarily relied on encapsulation, as demonstrated by lipid nanoparticles in SARS-CoV-2 vaccines. Concerns about RNA degradation on nanoparticle surfaces initially limited the exploration of adsorption-based approaches. However, recent advancements have renewed interest in adsorption as a viable alternative. This Viewpoint explores the approaches of RNA incorporation in nanoparticles, comparing encapsulation, adsorption, and the combination of encapsulation and adsorption, and presents a framework to guide the selection of the most suitable strategy based on general characteristics.
Collapse
Affiliation(s)
- Amy E Laturski
- Department of Chemistry, Stanford University, Stanford, California 94305, United States
| | - Maria T Dulay
- Department of Radiology, Stanford University, Stanford, California 94305, United States
| | - Jillian L Perry
- Center for Nanotechnology in Drug Delivery and Division of Pharmacoengineering and Molecular Pharmaceutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599-7575, United States
| | - Joseph M DeSimone
- Department of Chemical Engineering and Department of Radiology, Stanford University, Stanford, California 94305, United States
| |
Collapse
|
12
|
Zhaguparov D, Zhao M, Sekar RV, Woodside MT. Identifying the interactions conferring functional mechanical rigidity on RNase-resistant RNA from Zika virus. Proc Natl Acad Sci U S A 2025; 122:e2417234122. [PMID: 40063803 PMCID: PMC11929477 DOI: 10.1073/pnas.2417234122] [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: 08/23/2024] [Accepted: 01/14/2025] [Indexed: 03/25/2025] Open
Abstract
Some viruses counter host-cell efforts to digest invading viral RNA by using special structures resistant to host RNases, known as exoribonuclease-resistant RNAs (xrRNAs). xrRNAs typically form an unusual fold with the 5'-end threaded through a ring consisting of a multihelix junction closed by a pseudoknot. By using single-molecule force spectroscopy (SMFS), we previously showed that a Zika virus xrRNA is extremely rigid mechanically, withstanding very high forces, and that this mechanical resistance-not simply the knot-like fold topology-is essential for RNase resistance. Here, we have determined which interactions are most important for generating mechanical rigidity in the Zika virus xrRNA, by systematically mutating tertiary contacts. We found that removing any of the tertiary contacts involving the threaded 5' end was sufficient to abrogate mechanical resistance. In contrast, breaking a single pseudoknot base pair was not sufficient to do so: Two broken pairs were needed. This hierarchy of interaction importance for mechanical rigidity was supported by simulations mapping how mechanical tension was distributed within the xrRNA. For all mutants, RNase resistance varied in lock-step with mechanical resistance, confirming the primary role of mechanical rigidity in xrRNA function. This work reveals which interactions are most important for Zika xrRNA function, with implications for targeting the xrRNA therapeutically.
Collapse
Affiliation(s)
- Daniiar Zhaguparov
- Department of Physics, University of Alberta, Edmonton, AB T6G2E1, Canada
| | - Meng Zhao
- Department of Physics, University of Alberta, Edmonton, AB T6G2E1, Canada
| | | | - Michael T Woodside
- Department of Physics, University of Alberta, Edmonton, AB T6G2E1, Canada
- Centre for Prions and Protein Folding Diseases, University of Alberta, Edmonton, AB T6G2E1, Canada
- Li Ka Shing Institute of Virology, University of Alberta, Edmonton, AB T6G2E1, Canada
| |
Collapse
|
13
|
Lancaster CL, Moberg KH, Corbett AH. Post-Transcriptional Regulation of Gene Expression and the Intricate Life of Eukaryotic mRNAs. WILEY INTERDISCIPLINARY REVIEWS. RNA 2025; 16:e70007. [PMID: 40059537 PMCID: PMC11949413 DOI: 10.1002/wrna.70007] [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: 08/22/2024] [Revised: 02/17/2025] [Accepted: 02/18/2025] [Indexed: 03/29/2025]
Abstract
In recent years, there has been a growing appreciation for how regulatory events that occur either co- or post-transcriptionally contribute to the control of gene expression. Messenger RNAs (mRNAs) are extensively regulated throughout their metabolism in a precise spatiotemporal manner that requires sophisticated molecular mechanisms for cell-type-specific gene expression, which dictates cell function. Moreover, dysfunction at any of these steps can result in a variety of human diseases, including cancers, muscular atrophies, and neurological diseases. This review summarizes the steps of the central dogma of molecular biology, focusing on the post-transcriptional regulation of gene expression.
Collapse
Affiliation(s)
- Carly L. Lancaster
- Department of Biology, Emory College of Arts and Sciences, Atlanta, Georgia, USA
- Department of Cell Biology Emory University School of Medicine, Atlanta, Georgia, USA
- Graduate Program in Biochemistry, Cell and Developmental Biology, Emory University Atlanta, Georgia, USA
| | - Kenneth H. Moberg
- Department of Cell Biology Emory University School of Medicine, Atlanta, Georgia, USA
| | - Anita H. Corbett
- Department of Biology, Emory College of Arts and Sciences, Atlanta, Georgia, USA
| |
Collapse
|
14
|
Xia B, Shaheen N, Chen H, Zhao J, Guo P, Zhao Y. RNA aptamer-mediated RNA nanotechnology for potential treatment of cardiopulmonary diseases. Pharmacol Res 2025; 213:107659. [PMID: 39978660 DOI: 10.1016/j.phrs.2025.107659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 01/14/2025] [Accepted: 02/14/2025] [Indexed: 02/22/2025]
Abstract
Ribonucleic acid (RNA) aptamers are single-stranded RNAs that bind to target proteins or other molecules with high specificity and affinity, modulating biological functions through distinct mechanisms. These aptamers can act n as antagonists to block pathological interactions, agonists to activate signaling pathways, or delivery vehicles for therapeutic cargos such as siRNAs and miRNAs. The advances in RNA nanotechnology further enhances the versatility of RNA aptamers, offering scalable platforms for engineering. In this review, we have summarized recent developments in RNA aptamer-mediated RNA nanotechnology and provide an overview of its potential in treating cardiovascular and respiratory disorders, including atherosclerosis, acute coronary syndromes, heart failure, lung cancer, pulmonary hypertension, asthma, chronic obstructive pulmonary disease (COPD), acute lung injury, viral respiratory infections, and pulmonary fibrosis. By integrating aptamer technologies with innovative delivery systems, RNA aptamers hold the potential to revolutionize the treatment landscape for cardiopulmonary diseases.
Collapse
Affiliation(s)
- Boyu Xia
- Department of Physiology and Cell Biology, College of Medicine, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA
| | - Nargis Shaheen
- Department of Physiology and Cell Biology, College of Medicine, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA
| | - Huilong Chen
- Department of Physiology and Cell Biology, College of Medicine, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA
| | - Jing Zhao
- Department of Physiology and Cell Biology, College of Medicine, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA
| | - Peixuan Guo
- Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH 43210, USA
| | - Yutong Zhao
- Department of Physiology and Cell Biology, College of Medicine, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA.
| |
Collapse
|
15
|
Zwolsman R, Darwish YB, Kluza E, van der Meel R. Engineering Lipid Nanoparticles for mRNA Immunotherapy. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2025; 17:e70007. [PMID: 40195623 PMCID: PMC11976204 DOI: 10.1002/wnan.70007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 03/11/2025] [Accepted: 03/15/2025] [Indexed: 04/09/2025]
Abstract
Over the last decades, messenger RNA (mRNA) has emerged as a promising therapeutic modality, enabling the delivery of genetic instructions to cells for producing therapeutic proteins or antigens. As such, mRNA-based therapies can be developed for a wide range of conditions, including infections, cancer, metabolic disorders, and genetic diseases. Nevertheless, using mRNA therapeutically requires chemical modifications to reduce immunostimulatory effects and nanotechnology to prevent degradation and ensure intracellular delivery. Lipid nanoparticles (LNPs) have become the most effective delivery platform for mRNA therapeutics, which are primarily employed for vaccine purposes following local administration and hepatic applications following systemic administration. Here, we review the state-of-the-art LNP-mRNA technology and discuss its potential for immunotherapy. We first outline the requirements for mRNA to be used therapeutically, including the role of LNP-mediated delivery. Next, we highlight LNP-mRNA immunotherapy approaches for vaccination, immuno-oncology, and autoimmune disorders. In addition, we discuss challenges that are limiting LNP-mRNA's widespread use, including tunable biodistribution and immunostimulatory effects. Finally, we provide an outlook on how implementing approaches such as library screening and machine learning will guide the development of next-generation mRNA therapeutics.
Collapse
Affiliation(s)
- Robby Zwolsman
- Laboratory of Chemical Biology, Department of Biomedical EngineeringEindhoven University of TechnologyEindhoventhe Netherlands
| | - Youssef B. Darwish
- Laboratory of Chemical Biology, Department of Biomedical EngineeringEindhoven University of TechnologyEindhoventhe Netherlands
| | - Ewelina Kluza
- Laboratory of Chemical Biology, Department of Biomedical EngineeringEindhoven University of TechnologyEindhoventhe Netherlands
| | - Roy van der Meel
- Laboratory of Chemical Biology, Department of Biomedical EngineeringEindhoven University of TechnologyEindhoventhe Netherlands
| |
Collapse
|
16
|
Vosoughi P, Naghib SM, Kangarshahi BM, Mozafari MR. A review of RNA nanoparticles for drug/gene/protein delivery in advanced therapies: Current state and future prospects. Int J Biol Macromol 2025; 295:139532. [PMID: 39765293 DOI: 10.1016/j.ijbiomac.2025.139532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2024] [Revised: 01/02/2025] [Accepted: 01/03/2025] [Indexed: 01/13/2025]
Abstract
Nanotechnology involves the utilization of materials with exceptional properties at the nanoscale. Over the past few years, nanotechnologies have demonstrated significant potential in improving human health, particularly in medical treatments. The self-assembly characteristic of RNA is a highly effective method for designing and constructing nanostructures using a combination of biological, chemical, and physical techniques from different fields. There is great potential for the application of RNA nanotechnology in therapeutics. This review explores various nano-based drug delivery systems and their unique features through the impressive progress of the RNA field and their significant therapeutic promises due to their unique performance in the COVID-19 pandemic. However, a significant hurdle in fully harnessing the power of RNA drugs lies in effectively delivering RNA to precise organs and tissues, a critical factor for achieving therapeutic effectiveness, minimizing side effects, and optimizing treatment outcomes. There have been many efforts to pursue targeting, but the clinical translation of RNA drugs has been hindered by the lack of clear guidelines and shared understanding. A comprehensive understanding of various principles is essential to develop vaccines using nucleic acids and nanomedicine successfully. These include mechanisms of immune responses, functions of nucleic acids, nanotechnology, and vaccinations. Regarding this matter, the aim of this review is to revisit the fundamental principles of the immune system's function, vaccination, nanotechnology, and drug delivery in relation to the creation and manufacturing of vaccines utilizing nanotechnology and nucleic acids. RNA drugs have demonstrated significant potential in treating a wide range of diseases in both clinical and preclinical research. One of the reasons is their capacity to regulate gene expression and manage protein production efficiently. Different methods, like modifying chemicals, connecting ligands, and utilizing nanotechnology, have been essential in enabling the effective use of RNA-based treatments in medical environments. The article reviews stimuli-responsive nanotechnologies for RNA delivery and their potential in RNA medicines. It emphasizes the notable benefits of these technologies in improving the effectiveness of RNA and targeting specific cells and organs. This review offers a comprehensive analysis of different RNA drugs and how they work to produce therapeutic benefits. Recent progress in using RNA-based drugs, especially mRNA treatments, has shown that targeted delivery methods work well in medical treatments.
Collapse
Affiliation(s)
- Pegah Vosoughi
- Nanotechnology Department, School of Advanced Technologies, Iran University of Science and Technology (IUST), Tehran 1684613114, Iran
| | - Seyed Morteza Naghib
- Nanotechnology Department, School of Advanced Technologies, Iran University of Science and Technology (IUST), Tehran 1684613114, Iran.
| | - Babak Mikaeeli Kangarshahi
- State Key Laboratory of Structure Analysis for Industrial Equipment, Department of Engineering Mechanics, Dalian University of Technology, Dalian, China
| | - M R Mozafari
- Australasian Nanoscience and Nanotechnology Initiative (ANNI), Monash University LPO, Clayton, VIC 3168, Australia
| |
Collapse
|
17
|
Olatunji M, Liu Y. RNA damage and its implications in genome stability. DNA Repair (Amst) 2025; 147:103821. [PMID: 40043352 DOI: 10.1016/j.dnarep.2025.103821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 02/19/2025] [Accepted: 02/25/2025] [Indexed: 03/17/2025]
Abstract
Endogenous and environmental stressors can damage DNA and RNA to compromise genome and transcriptome stability and integrity in cells, leading to genetic instability and diseases. Recent studies have demonstrated that RNA damage can also modulate genome stability via RNA-templated DNA synthesis, suggesting that it is essential to maintain RNA integrity for the sustainment of genome stability. However, little is known about RNA damage and repair and their roles in modulating genome stability. Current efforts have mainly focused on revealing RNA surveillance pathways that detect and degrade damaged RNA, while the critical role of RNA repair is often overlooked. Due to their abundance and susceptibility to nucleobase damaging agents, it is essential for cells to evolve robust RNA repair mechanisms that can remove RNA damage, maintaining RNA integrity during gene transcription. This is supported by the discovery of the alkylated RNA nucleobase repair enzyme human AlkB homolog 3 that can directly remove the methyl group on damaged RNA nucleobases, predominantly in the nucleus of human cells, thereby restoring the integrity of the damaged RNA nucleobases. This is further supported by the fact that several DNA repair enzymes can also process RNA damage. In this review, we discuss RNA damage and its effects on cellular function, DNA repair, genome instability, and potential RNA damage repair mechanisms. Our review underscores the necessity for future research on RNA damage and repair and their essential roles in modulating genome stability.
Collapse
Affiliation(s)
- Mustapha Olatunji
- Biochemistry Ph.D. Program, Florida International University, Miami, FL, USA
| | - Yuan Liu
- Biochemistry Ph.D. Program, Florida International University, Miami, FL, USA; Department of Chemistry and Biochemistry, and Florida International University, Miami, FL, USA; Biomolecular Sciences Institute, Florida International University, Miami, FL, USA.
| |
Collapse
|
18
|
Park W, Choi J, Hwang J, Kim S, Kim Y, Shim MK, Park W, Yu S, Jung S, Yang Y, Kweon DH. Apolipoprotein Fusion Enables Spontaneous Functionalization of mRNA Lipid Nanoparticles with Antibody for Targeted Cancer Therapy. ACS NANO 2025; 19:6412-6425. [PMID: 39908463 PMCID: PMC11841042 DOI: 10.1021/acsnano.4c16562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 01/24/2025] [Accepted: 01/24/2025] [Indexed: 02/07/2025]
Abstract
The mRNA-lipid nanoparticles (mRNA@LNPs) offer a novel opportunity to treat targets previously considered undruggable. Although antibody conjugation is crucial for enhancing the specificity, delivery efficiency, and minimizing the toxicity of mRNA therapeutics, current chemical conjugation methods are complex and produce heterogeneous particles with misoriented antibodies. In this work, we introduce a chemical-free approach to functionalize mRNA@LNPs with antibodies, mimicking protein corona formation for targeted mRNA delivery. By fusing apolipoprotein to the Fc domain of a targeting antibody, we enabled the antibody to spontaneously display on the surface of mRNA@LNPs without altering the existing LNP process or employing complex chemical conjugation techniques. We demonstrated precise protein expression using trastuzumab-bound mRNA@LNPs, facilitating specific mRNA expression in HER2-positive cancer cells. mRNA was efficiently delivered to the tumor site after intravenous administration. While the control LNPs lacking targeting antibodies caused acute liver toxicity, trastuzumab-displayed LNPs showed no systemic toxicity. The tumor-specific delivery of p53 tumor suppressor mRNA led to the complete regression of cancer cells. Thus, apolipoprotein fusion enables a straightforward and scalable production of antibody-functionalized mRNA@LNPs, offering significant therapeutic potential in gene therapy.
Collapse
Affiliation(s)
- Wonbeom Park
- Department
of Integrative Biotechnology, Sungkyunkwan
University, Suwon 16419, Republic
of Korea
| | - Jiwoong Choi
- Biomedical
Research Division, Korea Institute of Science
and Technology (KIST), Seoul 02792, Republic
of Korea
| | - Jaehyeon Hwang
- Department
of Integrative Biotechnology, Sungkyunkwan
University, Suwon 16419, Republic
of Korea
| | - Suhyun Kim
- Department
of Integrative Biotechnology, Sungkyunkwan
University, Suwon 16419, Republic
of Korea
| | - Yelee Kim
- Biomedical
Research Division, Korea Institute of Science
and Technology (KIST), Seoul 02792, Republic
of Korea
- Department
of Life Sciences, Korea University, Seoul 02841, Republic of Korea
| | - Man Kyu Shim
- Biomedical
Research Division, Korea Institute of Science
and Technology (KIST), Seoul 02792, Republic
of Korea
| | - Wooram Park
- Department
of Integrative Biotechnology, Sungkyunkwan
University, Suwon 16419, Republic
of Korea
| | - Seokhyeon Yu
- Research
Center, MVRIX, Anyang 14058, Republic of Korea
| | - Sangwon Jung
- Research
Center, MVRIX, Anyang 14058, Republic of Korea
| | - Yoosoo Yang
- Biomedical
Research Division, Korea Institute of Science
and Technology (KIST), Seoul 02792, Republic
of Korea
- Division
of Bio-Medical Science and Technology, KIST School, Korea University of Science and Technology, Seoul 02792, Republic of Korea
| | - Dae-Hyuk Kweon
- Department
of Integrative Biotechnology, Sungkyunkwan
University, Suwon 16419, Republic
of Korea
| |
Collapse
|
19
|
Ronemus M, Bradford D, Laster Z, Li S. Exploring genome-transcriptome correlations in cancer. Biochem Soc Trans 2025; 53:BST20240108. [PMID: 39910794 DOI: 10.1042/bst20240108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Revised: 12/16/2024] [Accepted: 12/23/2024] [Indexed: 02/07/2025]
Abstract
We examine the complex relationship between genomic copy number variation (CNV) and gene expression, highlighting the relevance to cancer biology and other biological contexts. By tracing the history of genometranscriptome correlations, we emphasize the complexity and challenges in understanding these interactions, particularly within the heterogeneous landscape of human cancers. Recent advances in computational algorithms and high-throughput single-cell multi-omic sequencing technologies are discussed, demonstrating their potential to refine our understanding of cancer biology and their limitations. The integration of genomic and transcriptomic analyses, which offers novel insights into tumor evolution and heterogeneity as well as therapeutic strategies, is presented as a crucial approach for advancing cancer research.
Collapse
Affiliation(s)
- Michael Ronemus
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, U.S.A
| | - Daniel Bradford
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, U.S.A
| | - Zachary Laster
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, U.S.A
| | - Siran Li
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, U.S.A
| |
Collapse
|
20
|
Stefanovic F, Brown LG, MacDonald J, Bammler T, Rinchai D, Nguyen S, Zeng Y, Shinkawa V, Adams K, Chaussabel D, Berthier E, Haack AJ, Theberge AB. Your Blood is Out for Delivery: Considerations of Shipping Time and Temperature on Degradation of RNA from Stabilized Whole Blood. Anal Chem 2025; 97:1635-1644. [PMID: 39818791 PMCID: PMC12036334 DOI: 10.1021/acs.analchem.4c04591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2025]
Abstract
Remote research studies are an invaluable tool for reaching populations with limited access to large medical centers or universities. To expand the remote study toolkit, we previously developed homeRNA, which allows for at-home self-collection and stabilization of blood and demonstrated the feasibility of using homeRNA in high temperature climates. Here, we expand upon this work through a systematic study exploring the effects of high temperature on RNA integrity (represented as RNA Integrity Number, RIN) through in-lab and field experiments. Compared to the frozen controls (overall mean RIN of 8.2, n = 8), samples kept at 37 °C for 2, 4, and 8 days had mean RINs of 7.6, 5.9, and 5.2 (n = 3), respectively, indicating that typical shipping conditions (∼2 days) yield samples suitable for downstream RNA sequencing. Shorter time intervals (6 h) resulted in minimal RNA degradation (median RIN of 6.4, n = 3) even at higher temperatures (50 °C) compared to the frozen control (mean RIN of 7.8, n = 3). Additionally, we shipped homeRNA-stabilized blood from a single donor to 14 states and back during the summer with continuous temperature probes (7.1 median RIN, n = 42). Samples from all locations were analyzed with 3' mRNA-seq to assess differences in gene counts, with the data suggesting that there was no preferential degradation of transcripts as a result of different shipping times, temperatures, and regions. Overall, our data support that homeRNA can be used in elevated temperature conditions, enabling decentralized sample collection for telemedicine, global health, and clinical research.
Collapse
Affiliation(s)
- Filip Stefanovic
- Department of Chemistry, University of Washington, Seattle, Washington 98195, United States
| | - Lauren G. Brown
- Department of Chemistry, University of Washington, Seattle, Washington 98195, United States
| | - James MacDonald
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, Washington 98195, United States
| | - Theo Bammler
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, Washington 98195, United States
| | - Darawan Rinchai
- Department of Infectious Diseases, St Jude’s Children Research Hospital, TN, Memphis 38105, United States
| | - Serena Nguyen
- Department of Chemistry, University of Washington, Seattle, Washington 98195, United States
| | - Yuting Zeng
- Department of Chemistry, University of Washington, Seattle, Washington 98195, United States
| | - Victoria Shinkawa
- Department of Chemistry, University of Washington, Seattle, Washington 98195, United States
| | - Karen Adams
- Department of Chemistry, University of Washington, Seattle, Washington 98195, United States
- Institute of Translational Health Sciences, School of Medicine, University of Washington, Seattle, Washington 98195, United States
| | - Damien Chaussabel
- Computer Sciences Department, The Jackson Laboratory, Farmington, CT, 06032, United States
| | - Erwin Berthier
- Department of Chemistry, University of Washington, Seattle, Washington 98195, United States
| | - Amanda J. Haack
- Department of Chemistry, University of Washington, Seattle, Washington 98195, United States
- School of Medicine, University of Washington, Seattle, Washington 98195, United States
| | - Ashleigh B. Theberge
- Department of Chemistry, University of Washington, Seattle, Washington 98195, United States
- Department of Urology, School of Medicine, University of Washington, Seattle, Washington 98195, United States
| |
Collapse
|
21
|
Vo GTT, Nguyen KKH, Kim BS. Evaluation of the Role of PnuC Gene in Enhancing Nicotinamide Mononucleotide Synthesis. Biotechnol Appl Biochem 2025. [PMID: 39865734 DOI: 10.1002/bab.2713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 12/22/2024] [Indexed: 01/28/2025]
Abstract
The PnuC gene plays a crucial role in the complex processes related to the absorption and synthesis of the nicotinamide mononucleotide (NMN) precursor. NMN, a nicotinamide adenine dinucleotide (NAD+) precursor, is important for cellular energy metabolism, DNA repair, and antiaging. This study focuses on elucidating the precursor absorption mechanism and the specific function of the PnuC gene in encoding membrane transport proteins, as well as its impact on the regulation and dynamics of NMN within the cell. This understanding aims to provide insights into its potential effects on metabolic balance, illustrated through two NAD+ biosynthesis pathways based on renewable and readily available cytoplasmic resources, assessing the potential of PnuC gene expression in clarifying complex interactions within regulation mechanisms. Enhanced expression analysis of the PnuC gene has initiated discussions on its potential applications in treating aging-related diseases and dysfunctions, contributing to cellular health maintenance.
Collapse
Affiliation(s)
- Giang Thi Thu Vo
- Department of Chemical Engineering, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea
| | - Khang Khoa Hoang Nguyen
- Department of Chemical Engineering, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea
| | - Beom Soo Kim
- Department of Chemical Engineering, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea
| |
Collapse
|
22
|
Kim Y, Jeon S, Kim B, Jeong YJ, Kim TH, Jeong S, Kim I, Oh J, Jung Y, Lee K, Choy YB, Kim SW, Chung JJ. Sticky Polyelectrolyte Shield for Enhancing Biological Half-Life of Growth Factors. ACS APPLIED MATERIALS & INTERFACES 2025; 17:445-466. [PMID: 39694662 DOI: 10.1021/acsami.4c16261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2024]
Abstract
Delivery of secretomes, which includes growth factors, cytokines, and mRNA, is critical in regenerative medicine for cell-to-cell communication. However, the harsh in vivo environment presents significant challenges for secretome delivery. Proteolytic enzymes shorten secretomes' half-lives, and secretomes tend to rapidly diffuse at defect sites. Therefore, a delivery system that ensures prolonged retention and enhanced therapeutic efficacy of secretomes is required. In this study, a Coating Optimized Drug Delivery Enhancement (COD2E) system, a coacervate composed of dopamine functionalized fucoidan and poly-l-lysine, was fabricated for secretome delivery. The dopamine modification significantly enhanced adhesive strength (>7-fold) compared to that of the neat coacervates, which enabled rapid (5 min) and uniform coating ability on collagen sponges. The COD2E system was able to encapsulate fibroblast growth factor (FGF2) and prolong the half-life of FGF2. Notably, its efficacy, demonstrated through a single application of FGF2 encapsulated COD2E on collagen sponge, in a wound model demonstrated a successful tissue repair. The COD2E system is an effective growth factor delivery vehicle since it can protect growth factors, has an antioxidant ability, adheres on various material surfaces, and is hemocompatible.
Collapse
Affiliation(s)
- Young Kim
- Department of Transdisciplinary Medicine, Seoul National University Hospital, Seoul 03080, Republic of Korea
- Innovative Medical Technology Research Institute, Seoul National University Hospital, Seoul 03080, Republic of Korea
- Department of Medicine, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
- Department of Clinical Medical Sciences, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Sungmi Jeon
- Division of Pediatric Plastic Surgery, Seoul National University Children's Hospital, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
- Department of Plastic and Reconstructive Surgery, Seoul National University Hospital, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Byulhana Kim
- Department of Transdisciplinary Medicine, Seoul National University Hospital, Seoul 03080, Republic of Korea
- Interdisciplinary Program in Bioengineering College of Engineering, Seoul National University, Seoul 08826, Republic of Korea
| | - Yu Jin Jeong
- Department of Plastic and Reconstructive Surgery, Seoul National University Hospital, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Tae Hee Kim
- Department of Fusion Research and Collaboration, Biomedical Research Institute, Seoul National University, Seoul 03080, Republic of Korea
| | - Subin Jeong
- Department of Transdisciplinary Medicine, Seoul National University Hospital, Seoul 03080, Republic of Korea
| | - Iljin Kim
- Department of Pharmacology and Program in Biomedical Science and Engineering, Inha University College of Medicine, Incheon 22212, South Korea
| | - Joomin Oh
- Department of Transdisciplinary Medicine, Seoul National University Hospital, Seoul 03080, Republic of Korea
- Innovative Medical Technology Research Institute, Seoul National University Hospital, Seoul 03080, Republic of Korea
- Department of Medicine, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
- Department of Clinical Medical Sciences, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Youngmee Jung
- Biomaterials Research Center, Biomedical Research Institute, Korea Institute of Science and Technology, Seoul 02792, Republic of Korea
- School of Electrical and Electronic Engineering, YU-KIST, Yonsei University, Seoul 03722, Republic of Korea
| | - Kangwon Lee
- Department of Applied Bioengineering, Graduate School of Convergence Science and Technology, Seoul National University, Seoul 08826, Republic of Korea
- Research Institute for Convergence Science, Seoul National University, Seoul 08826, Republic of Korea
| | - Young Bin Choy
- Innovative Medical Technology Research Institute, Seoul National University Hospital, Seoul 03080, Republic of Korea
- Department of Clinical Medical Sciences, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
- Interdisciplinary Program in Bioengineering College of Engineering, Seoul National University, Seoul 08826, Republic of Korea
- Institute of Medical and Biological Engineering, Medical Research Center, Seoul National University, Seoul 03080, Republic of Korea
- Department of Biomedical Engineering, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Sang Wha Kim
- Department of Plastic and Reconstructive Surgery, Seoul National University Hospital, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Justin J Chung
- Department of Transdisciplinary Medicine, Seoul National University Hospital, Seoul 03080, Republic of Korea
- Innovative Medical Technology Research Institute, Seoul National University Hospital, Seoul 03080, Republic of Korea
- Department of Medicine, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| |
Collapse
|
23
|
Mu K, Fei Y, Xu Y, Zhang QC. RASP v2.0: an updated atlas for RNA structure probing data. Nucleic Acids Res 2025; 53:D211-D219. [PMID: 39546630 PMCID: PMC11701657 DOI: 10.1093/nar/gkae1117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2024] [Revised: 10/16/2024] [Accepted: 11/14/2024] [Indexed: 11/17/2024] Open
Abstract
RNA molecules function in numerous biological processes by folding into intricate structures. Here we present RASP v2.0, an updated database for RNA structure probing data featuring a substantially expanded collection of datasets along with enhanced online structural analysis functionalities. Compared to the previous version, RASP v2.0 includes the following improvements: (i) the number of RNA structure datasets has increased from 156 to 438, comprising 216 transcriptome-wide RNA structure datasets, 141 target-specific RNA structure datasets, and 81 RNA-RNA interaction datasets, thereby broadening species coverage from 18 to 24, (ii) a deep learning-based model has been implemented to impute missing structural signals for 59 transcriptome-wide RNA structure datasets with low structure score coverage, significantly enhancing data quality, particularly for low-abundance RNAs, (iii) three new online analysis modules have been deployed to assist RNA structure studies, including missing structure score imputation, RNA secondary and tertiary structure prediction, and RNA binding protein (RBP) binding prediction. By providing a resource of much more comprehensive RNA structure data, RASP v2.0 is poised to facilitate the exploration of RNA structure-function relationships across diverse biological processes. RASP v2.0 is freely accessible at http://rasp2.zhanglab.net/.
Collapse
Affiliation(s)
- Kunting Mu
- MOE Key Laboratory of Bioinformatics, Center for Synthetic and Systems Biology, School of Life Sciences, Tsinghua University, Beijing 100084, China
- Beijing Advanced Innovation Center for Structural Biology & Frontier Research Center for Biological Structure, School of Life Sciences, Tsinghua University, Beijing 100084, China
- Tsinghua-Peking Center for Life Sciences, Beijing 100084, China
| | - Yuhan Fei
- MOE Key Laboratory of Bioinformatics, Center for Synthetic and Systems Biology, School of Life Sciences, Tsinghua University, Beijing 100084, China
- Beijing Advanced Innovation Center for Structural Biology & Frontier Research Center for Biological Structure, School of Life Sciences, Tsinghua University, Beijing 100084, China
- Tsinghua-Peking Center for Life Sciences, Beijing 100084, China
| | - Yiran Xu
- MOE Key Laboratory of Bioinformatics, Center for Synthetic and Systems Biology, School of Life Sciences, Tsinghua University, Beijing 100084, China
- Beijing Advanced Innovation Center for Structural Biology & Frontier Research Center for Biological Structure, School of Life Sciences, Tsinghua University, Beijing 100084, China
- Tsinghua-Peking Center for Life Sciences, Beijing 100084, China
| | - Qiangfeng Cliff Zhang
- MOE Key Laboratory of Bioinformatics, Center for Synthetic and Systems Biology, School of Life Sciences, Tsinghua University, Beijing 100084, China
- Beijing Advanced Innovation Center for Structural Biology & Frontier Research Center for Biological Structure, School of Life Sciences, Tsinghua University, Beijing 100084, China
- Tsinghua-Peking Center for Life Sciences, Beijing 100084, China
| |
Collapse
|
24
|
Dansereau SJ, Cui H, Dartawan RP, Sheng J. The Plethora of RNA-Protein Interactions Model a Basis for RNA Therapies. Genes (Basel) 2025; 16:48. [PMID: 39858595 PMCID: PMC11765398 DOI: 10.3390/genes16010048] [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: 11/30/2024] [Revised: 12/25/2024] [Accepted: 12/27/2024] [Indexed: 01/27/2025] Open
Abstract
The notion of RNA-based therapeutics has gained wide attractions in both academic and commercial institutions. RNA is a polymer of nucleic acids that has been proven to be impressively versatile, dating to its hypothesized RNA World origins, evidenced by its enzymatic roles in facilitating DNA replication, mRNA decay, and protein synthesis. This is underscored through the activities of riboswitches, spliceosomes, ribosomes, and telomerases. Given its broad range of interactions within the cell, RNA can be targeted by a therapeutic or modified as a pharmacologic scaffold for diseases such as nucleotide repeat disorders, infectious diseases, and cancer. RNA therapeutic techniques that have been researched include, but are not limited to, CRISPR/Cas gene editing, anti-sense oligonucleotides (ASOs), siRNA, small molecule treatments, and RNA aptamers. The knowledge gleaned from studying RNA-centric mechanisms will inevitably improve the design of RNA-based therapeutics. Building on this understanding, we explore the physiological diversity of RNA functions, examine specific dysfunctions, such as splicing errors and viral interactions, and discuss their therapeutic implications.
Collapse
Affiliation(s)
| | | | | | - Jia Sheng
- Department of Chemistry, The RNA Institute, University at Albany, SUNY, 1400 Washington Ave Extension, Albany, NY 12222, USA; (S.J.D.); (H.C.)
| |
Collapse
|
25
|
Fei J, Zhao D, Pang C, Li J, Li S, Qiao W, Tan J, Bi C, Zhang X. Mismatch prime editing gRNA increased efficiency and reduced indels. Nat Commun 2025; 16:139. [PMID: 39747083 PMCID: PMC11696010 DOI: 10.1038/s41467-024-55578-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Accepted: 12/17/2024] [Indexed: 01/04/2025] Open
Abstract
Prime editing enables precise and efficient genome editing, but its efficacy is hindered by pegRNA's 3' extension, forming secondary structures due to high complementarity with the protospacer. The continuous presence of the prime editing system also leads to unintended indel formation, raising safety concerns for therapeutic applications. To address these challenges, we develop a mismatched pegRNA (mpegRNA) strategy that introduces mismatched bases into the pegRNA protospacer, reducing complementarity and secondary structure formation, and preventing sustained activity. Our findings show that mpegRNA enhances editing efficiency by up to 2.3 times and reduces indel levels by 76.5% without compromising performance. Combining mpegRNA with epegRNA further increases efficiency up to 14-fold, or 2.4-fold in PE4max/PE5max systems, underscoring its potential in research and therapy. AlphaFold 3 analysis suggests that the optimal mpegRNA structure contributes significantly to improved editing outcomes. Overall, mpegRNA advances prime editing technology, improving efficiency while reducing indels.
Collapse
Affiliation(s)
- Jidong Fei
- College of Life Science, Tianjin Normal University, Tianjin, China
- Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin, China
- University of Chinese Academy of Sciences, Beijing, China
- National Technology Innovation Center of Synthetic Biology, Tianjin, China
- Westlake Laboratory of Life Sciences and Biomedicine, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
| | - Dongdong Zhao
- Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin, China.
- National Technology Innovation Center of Synthetic Biology, Tianjin, China.
| | - Caiyi Pang
- Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin, China
- National Technology Innovation Center of Synthetic Biology, Tianjin, China
- Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, College of Life Sciences, Nankai University, Tianjin, China
| | - Ju Li
- College of Life Science, Tianjin Normal University, Tianjin, China
| | - Siwei Li
- Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin, China
- National Technology Innovation Center of Synthetic Biology, Tianjin, China
| | - Wentao Qiao
- Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, College of Life Sciences, Nankai University, Tianjin, China
| | - Juan Tan
- Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, College of Life Sciences, Nankai University, Tianjin, China
| | - Changhao Bi
- Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin, China.
- National Technology Innovation Center of Synthetic Biology, Tianjin, China.
| | - Xueli Zhang
- Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin, China.
- National Technology Innovation Center of Synthetic Biology, Tianjin, China.
| |
Collapse
|
26
|
Jin W, Li X, Argandona SM, Ray RM, Lin MKTH, Melle F, Clergeaud G, Lars Andresen T, Nielsen M, Fairen-Jimenez D, Astakhova K, Qvortrup K. Surface engineering of metal-organic framework nanoparticles-based miRNA carrier: Boosting RNA stability, intracellular delivery and synergistic therapy. J Colloid Interface Sci 2025; 677:429-440. [PMID: 39153246 DOI: 10.1016/j.jcis.2024.08.074] [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: 05/09/2024] [Revised: 08/01/2024] [Accepted: 08/11/2024] [Indexed: 08/19/2024]
Abstract
MicroRNAs (miRNAs) are small noncoding RNAs that are critical for the regulation of multiple physiological and pathological processes, thus holding great clinical potential. However, the therapeutic applications of miRNAs are severely limited by their biological instability and poor intracellular delivery. Herein, we describe a dual-layers surface engineering strategy to design an efficient miRNA delivery nanosystem based on metal-organic frameworks (MOFs) incorporating lipid coating. The resulting nanoparticle system was demonstrated to protect miRNA from ribonuclease degradation, enhance cellular uptake and facilitate lysosomal escape. These ensured effective miRNA mediated gene therapy, which synergized with MOF-specific photodynamic therapy and pre-encapsulated doxorubicin (Dox) chemotherapy to provide a multifunctional with therapeutic effectiveness against cencer cells The mechanisms of miRNA binding and Dox loading were revealed, demonstrating the potential of the present MOFs surface-engineered strategy to overcome their inherent pore-size restriction for macromolecular miRNA carrying, enableefficient co-delivery. In vitro studies revealed the potential of our multifunctional system for miRNA delivery and the demonstrated the therapeutic effectiveness against cancer cells, thereby providing a versatile all-in-one MOFs strategy for delivery of nucleic acids and diverse therapeutic molecules in synergistic therapy.
Collapse
Affiliation(s)
- Weiguang Jin
- Department of Chemistry, Technical University of Denmark, 2800 Kongens Lyngby, Denmark.
| | - Xin Li
- Department of Health Technology, Technical University of Denmark, 2800 Kongens Lyngby, Denmark.
| | - Sergio Mercado Argandona
- The Adsorption and Advanced Laboratory, Department of Chemical Engineering & Biotechnology, University of Cambridge, Cambridge CB3 0AS, UK.
| | - Roslyn M Ray
- Center for Gene Therapy, City of Hope-Beckman Research Institute, Duarte, CA 91010, USA.
| | - Marie Karen Tracy Hong Lin
- National Center for Nanofabrication and Characterization, Technical University of Denmark, 2800 Kongens Lyngby, Denmark.
| | - Francesca Melle
- The Adsorption and Advanced Laboratory, Department of Chemical Engineering & Biotechnology, University of Cambridge, Cambridge CB3 0AS, UK.
| | - Gael Clergeaud
- Department of Health Technology, Technical University of Denmark, 2800 Kongens Lyngby, Denmark.
| | - Thomas Lars Andresen
- Department of Health Technology, Technical University of Denmark, 2800 Kongens Lyngby, Denmark.
| | - Martin Nielsen
- Department of Chemistry, Technical University of Denmark, 2800 Kongens Lyngby, Denmark.
| | - David Fairen-Jimenez
- The Adsorption and Advanced Laboratory, Department of Chemical Engineering & Biotechnology, University of Cambridge, Cambridge CB3 0AS, UK.
| | - Kira Astakhova
- Department of Chemistry, Technical University of Denmark, 2800 Kongens Lyngby, Denmark.
| | - Katrine Qvortrup
- Department of Chemistry, Technical University of Denmark, 2800 Kongens Lyngby, Denmark.
| |
Collapse
|
27
|
Ibrahim F, Mourelatos Z. Defining the True Native Ends of RNAs at Single-Molecule Level with TERA-Seq. Methods Mol Biol 2025; 2863:359-372. [PMID: 39535720 PMCID: PMC12013618 DOI: 10.1007/978-1-0716-4176-7_21] [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: 11/16/2024]
Abstract
Turnover of messenger RNAs (mRNAs) is a highly regulated process and serves to control expression of RNA molecules and to eliminate aberrant transcripts. Profiling mRNA decay using short-read sequencing methods that target either the 5' or 3' ends of RNAs, overlooks valuable information about the other end, which could provide significant insights into biological aspects and mechanisms of RNA decay. Oxford Nanopore Technology (ONT) is rapidly emerging as a powerful platform for direct sequencing of native, single-RNA molecules. However, as currently designed, the existing ONT platform is unable to sequence the very 5' ends of RNAs and is limited to polyadenylated molecules. Here, we present a detailed step-by-step experimental protocol for True End-to-end RNA Sequencing (TERA-Seq), a new method that addresses ONT's limitations, allowing accurate representation and characterization of RNAs at the level of single molecules. TERA-Seq describes both poly- and non-polyadenylated RNA molecules and accurately identifies their native ends by ligating uniquely designed adapters to the 5' ends (5TERA), the 3' ends (TERA3), or both ends (5TERA3) that are sequenced along with the transcripts.
Collapse
Affiliation(s)
- Fadia Ibrahim
- Department of Biochemistry and Molecular Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA.
| | - Zissimos Mourelatos
- Department of Pathology and Laboratory Medicine, Division of Neuropathology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
28
|
Jin L, Zhou Y, Zhang S, Chen SJ. mRNA vaccine sequence and structure design and optimization: Advances and challenges. J Biol Chem 2025; 301:108015. [PMID: 39608721 PMCID: PMC11728972 DOI: 10.1016/j.jbc.2024.108015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 11/13/2024] [Accepted: 11/16/2024] [Indexed: 11/30/2024] Open
Abstract
Messenger RNA (mRNA) vaccines have emerged as a powerful tool against communicable diseases and cancers, as demonstrated by their huge success during the coronavirus disease 2019 (COVID-19) pandemic. Despite the outstanding achievements, mRNA vaccines still face challenges such as stringent storage requirements, insufficient antigen expression, and unexpected immune responses. Since the intrinsic properties of mRNA molecules significantly impact vaccine performance, optimizing mRNA design is crucial in preclinical development. In this review, we outline four key principles for optimal mRNA sequence design: enhancing ribosome loading and translation efficiency through untranslated region (UTR) optimization, improving translation efficiency via codon optimization, increasing structural stability by refining global RNA sequence and extending in-cell lifetime and expression fidelity by adjusting local RNA structures. We also explore recent advancements in computational models for designing and optimizing mRNA vaccine sequences following these principles. By integrating current mRNA knowledge, addressing challenges, and examining advanced computational methods, this review aims to promote the application of computational approaches in mRNA vaccine development and inspire novel solutions to existing obstacles.
Collapse
Affiliation(s)
- Lei Jin
- Department of Physics and Astronomy, University of Missouri, Columbia, Missouri, USA
| | - Yuanzhe Zhou
- Department of Physics and Astronomy, University of Missouri, Columbia, Missouri, USA
| | - Sicheng Zhang
- Department of Physics and Astronomy, University of Missouri, Columbia, Missouri, USA
| | - Shi-Jie Chen
- Department of Physics and Astronomy, University of Missouri, Columbia, Missouri, USA; Department of Biochemistry, MU Institute for Data Science and Informatics, University of Missouri, Columbia, Missouri, USA.
| |
Collapse
|
29
|
Giraldo-Ocampo S, Valiente-Echeverría F, Soto-Rifo R. Host RNA-Binding Proteins as Regulators of HIV-1 Replication. Viruses 2024; 17:43. [PMID: 39861832 PMCID: PMC11768693 DOI: 10.3390/v17010043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 12/26/2024] [Accepted: 12/27/2024] [Indexed: 01/27/2025] Open
Abstract
RNA-binding proteins (RBPs) are cellular factors involved in every step of RNA metabolism. During HIV-1 infection, these proteins are key players in the fine-tuning of viral and host cellular and molecular pathways, including (but not limited to) viral entry, transcription, splicing, RNA modification, translation, decay, assembly, and packaging, as well as the modulation of the antiviral response. Targeted studies have been of paramount importance in identifying and understanding the role of RNA-binding proteins that bind to HIV-1 RNAs. However, novel approaches aimed at identifying all the proteins bound to specific RNAs (RBPome), such as RNA interactome capture, have also contributed to expanding our understanding of the HIV-1 replication cycle, allowing the identification of RBPs with functions not only in viral RNA metabolism but also in cellular metabolism. Strikingly, several of the RBPs found through interactome capture are not canonical RBPs, meaning that they do not have conventional RNA-binding domains and are therefore not readily predicted as being RBPs. Further studies on the different cellular targets of HIV-1, such as subtypes of T cells or myeloid cells, or on the context (active replication versus reactivation from latency) are needed to fully elucidate the host RBPome bound to the viral RNA, which will allow researchers and clinicians to discover new therapeutic targets during active replication and provirus reactivation from latency.
Collapse
Affiliation(s)
- Sebastian Giraldo-Ocampo
- Laboratory of Molecular and Cellular Virology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago 8380453, Chile; (S.G.-O.); (F.V.-E.)
- Center for HIV/AIDS Integral Research (CHAIR), Faculty of Medicine, Universidad de Chile, Santiago 8380453, Chile
- Millennium Institute in Immunology and Immunotherapy, Santiago 8380453, Chile
| | - Fernando Valiente-Echeverría
- Laboratory of Molecular and Cellular Virology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago 8380453, Chile; (S.G.-O.); (F.V.-E.)
- Center for HIV/AIDS Integral Research (CHAIR), Faculty of Medicine, Universidad de Chile, Santiago 8380453, Chile
- Millennium Institute in Immunology and Immunotherapy, Santiago 8380453, Chile
| | - Ricardo Soto-Rifo
- Laboratory of Molecular and Cellular Virology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago 8380453, Chile; (S.G.-O.); (F.V.-E.)
- Center for HIV/AIDS Integral Research (CHAIR), Faculty of Medicine, Universidad de Chile, Santiago 8380453, Chile
- Millennium Institute in Immunology and Immunotherapy, Santiago 8380453, Chile
| |
Collapse
|
30
|
Portelli C, Seria E, Attard R, Barzine M, Esquinas-Roman EM, Borg Carbott F, Cassar K, Vella M, Scicluna BP, Ebejer JP, Farrugia R, Bezzina Wettinger S. Isolating high-quality RNA for RNA-Seq from 10-year-old blood samples. Sci Rep 2024; 14:30716. [PMID: 39730418 DOI: 10.1038/s41598-024-80287-4] [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: 02/09/2024] [Accepted: 11/18/2024] [Indexed: 12/29/2024] Open
Abstract
There is much interest in analysing RNA, particularly with RNA Sequencing, across both research and diagnostic domains. However, its inherent instability renders it susceptible to degradation. Given the imperative for RNA integrity in such applications, proper storage and biobanking of blood samples and successful subsequent RNA isolation is essential to guarantee optimal integrity for downstream analyses. Especially for larger collections, it would be particularly beneficial if these methods would additionally offer affordability, minimal blood volume requirements and also long-term storage. In this study, RNA of high quality, suitable for transcriptomics, has been successfully isolated from 400 µL of EDTA and citrated whole blood samples in Boom's lysis buffer stored at -85 °C for 10 years. Isolation was carried out using a modified Zymo Research Quick-RNA kit protocol. This isolation method showed significant improvement in RNA integrity when compared to RNA extracted using the original Boom method. RNA Sequencing provided high-quality data comparable to that of other studies using recently frozen blood in RNA stabilisation tubes. Additionally, sequencing data from blood collected in citrate and EDTA anticoagulants also showed excellent correlation.
Collapse
Affiliation(s)
- Charlene Portelli
- Department of Applied Biomedical Science, Faculty of Health Sciences, University of Malta, Msida, 2080, MSD, Malta
| | - Elisa Seria
- Department of Applied Biomedical Science, Faculty of Health Sciences, University of Malta, Msida, 2080, MSD, Malta
| | - Ritienne Attard
- Department of Applied Biomedical Science, Faculty of Health Sciences, University of Malta, Msida, 2080, MSD, Malta
| | - Mitra Barzine
- Department of Applied Biomedical Science, Faculty of Health Sciences, University of Malta, Msida, 2080, MSD, Malta
| | - Eva M Esquinas-Roman
- Department of Applied Biomedical Science, Faculty of Health Sciences, University of Malta, Msida, 2080, MSD, Malta
| | - Francesca Borg Carbott
- Department of Applied Biomedical Science, Faculty of Health Sciences, University of Malta, Msida, 2080, MSD, Malta
| | - Karen Cassar
- Department of Medicine, Faculty of Medicine and Surgery, University of Malta, Msida, 2080, MSD, Malta
| | - Matthew Vella
- Department of Applied Biomedical Science, Faculty of Health Sciences, University of Malta, Msida, 2080, MSD, Malta
| | - Brendon P Scicluna
- Department of Applied Biomedical Science, Faculty of Health Sciences, University of Malta, Msida, 2080, MSD, Malta
- Centre for Molecular Medicine and Biobanking, University of Malta, Msida, 2080, MSD, Malta
| | - Jean-Paul Ebejer
- Centre for Molecular Medicine and Biobanking, University of Malta, Msida, 2080, MSD, Malta
| | - Rosienne Farrugia
- Department of Applied Biomedical Science, Faculty of Health Sciences, University of Malta, Msida, 2080, MSD, Malta
- Centre for Molecular Medicine and Biobanking, University of Malta, Msida, 2080, MSD, Malta
| | - Stephanie Bezzina Wettinger
- Department of Applied Biomedical Science, Faculty of Health Sciences, University of Malta, Msida, 2080, MSD, Malta.
- Centre for Molecular Medicine and Biobanking, University of Malta, Msida, 2080, MSD, Malta.
| |
Collapse
|
31
|
Kornienko IV, Aramova OY, Tishchenko AA, Rudoy DV, Chikindas ML. RNA Stability: A Review of the Role of Structural Features and Environmental Conditions. Molecules 2024; 29:5978. [PMID: 39770066 PMCID: PMC11676819 DOI: 10.3390/molecules29245978] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 12/05/2024] [Accepted: 12/16/2024] [Indexed: 01/11/2025] Open
Abstract
The stability of RNA is a critical factor in determining its functionality and degradation in the cell. In recent years, it has been shown that the stability of RNA depends on a complex interaction of external and internal factors. External conditions, such as temperature fluctuations, the level of acidity of the environment, the presence of various substances and ions, as well as the effects of oxidative stress, can change the structure of RNA and affect its stability. Internal factors, including the specific structural features of RNA and its interactions with protein molecules, also have a significant impact on the regulation of the stability of these molecules. In this article, we review the main factors influencing RNA stability, since understanding the factors influencing this extremely complex process is important not only for understanding the regulation of expression at the RNA level but also for developing new methods for isolating and stabilizing RNA in preparation for creating biobanks of genetic material. We reviewed a modern solution to this problem and formulated basic recommendations for RNA storage aimed at minimizing degradation and damage to the molecule.
Collapse
Affiliation(s)
- Igor V. Kornienko
- Center for Agrobiotechnology, Don State Technical University, Gagarina Sq. 1, Rostov-on-Don 344003, Russia; (I.V.K.); (D.V.R.); (M.L.C.)
- Federal Research Centre Southern Scientific Centre of the Russian Academy of Sciences, Chekhov Ave. 41, Rostov-on-Don 344006, Russia
- Department of Genetics Academy of Biology and Biotechnology, Southern Federal University, Stachki Ave. 194/1, Rostov-on-Don 344090, Russia
| | - Olga Yu. Aramova
- Federal Research Centre Southern Scientific Centre of the Russian Academy of Sciences, Chekhov Ave. 41, Rostov-on-Don 344006, Russia
- Department of Genetics Academy of Biology and Biotechnology, Southern Federal University, Stachki Ave. 194/1, Rostov-on-Don 344090, Russia
| | - Anna A. Tishchenko
- Department of Big Data and Machine Learning, St. Petersburg National Research University of Information Technologies, Mechanics and Optics, Kronverksky Pr. 49, St. Petersburg 197101, Russia;
| | - Dmitriy V. Rudoy
- Center for Agrobiotechnology, Don State Technical University, Gagarina Sq. 1, Rostov-on-Don 344003, Russia; (I.V.K.); (D.V.R.); (M.L.C.)
| | - Michael Leonidas Chikindas
- Center for Agrobiotechnology, Don State Technical University, Gagarina Sq. 1, Rostov-on-Don 344003, Russia; (I.V.K.); (D.V.R.); (M.L.C.)
- Health Promoting Naturals Laboratory, School of Environmental and Biological Sciences, Rutgers State University, 65 Dudley Road, New Brunswick, NJ 08901-8525, USA
- Department of General Hygiene, I.M. Sechenov First Moscow State Medical University, Trubetskaya Str. 8, Bldg 2, Moscow 119048, Russia
| |
Collapse
|
32
|
Martins C, Carvalho LM, Cabral IM, Saúde L, Dreij K, Costa PM. A mechanistic study on the interaction effects between legacy and pollutants of emerging concern: A case study with B[a]P and diclofenac. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2024; 363:125189. [PMID: 39454814 DOI: 10.1016/j.envpol.2024.125189] [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: 07/08/2024] [Revised: 09/09/2024] [Accepted: 10/22/2024] [Indexed: 10/28/2024]
Abstract
To study the intricate toxicological mechanisms triggered by exposure to mixed pollutants, we exposed zebrafish embryos to legacy and emerging pollutants through binary mixtures of benzo[a]pyrene (B[a]P) and diclofenac (DFC). The combination of next-generation transcriptomics and toxicopathology disclosed instances where exposure to mixtures did not attain the expected sum of acute effects of individual toxicants, indicating potential antagonism. Despite overall higher mortality in DFC treatments, the same antagonistic trend was noted in genotoxicity and molecular pathways related to RNA turnover, cell proliferation, apoptosis and cell-cycle control. The formation of oedemas in the heart cavity and yolk sac can be an adverse outcome (AO) resulting from exposure to DFC isolated or combined, whose potential key events (KEs) may involve cell cycle arrest and apoptosis via p53 and MAPK pathways. From the findings it can be hypothesised that, rather than genotoxicity, the molecular initiating event (MIE) maybe inflammation triggered by oxidative stress. Nonetheless, the exact role of ROS in the process needs further clarification. Impaired eye function by action of DFC and B[a]P combined may be another AO, in the case caused by ocular degeneration following the suppression of biologic processes and molecular functions involved in eye development and its functionalities, possibly linked to hindered regulation of the expression of hsf4 and cryaa. Altogether, toxicopathology suggests predominance of antagonistic effects, but its integration with mechanism suggests that interactions between DFC and B[a]P in environmentally-relevant concentrations that may lead to hindrance of key functions such as the control of inflammation and cell cycle. These outcomes suggest potentially severe implications for health and survival, in case of prolonged chronic exposure to combined toxicants.
Collapse
Affiliation(s)
- Carla Martins
- Associate Laboratory I4HB Institute for Health and Bioeconomy, NOVA School of Science and Technology, NOVA University of Lisbon, 2829-516, Caparica, Portugal; UCIBIO Applied Molecular Biosciences Unit, Department of Life Sciences, NOVA School of Science and Technology, NOVA University of Lisbon, 2829-516, Caparica, Portugal.
| | - Lara M Carvalho
- GIMM - Gulbenkian Institute for Molecular Medicine, 1649-035 Lisboa, Portugal; Faculdade de Medicina, Universidade de Lisboa, 1649-035 Lisboa, Portugal
| | - Inês Moutinho Cabral
- Associate Laboratory I4HB Institute for Health and Bioeconomy, NOVA School of Science and Technology, NOVA University of Lisbon, 2829-516, Caparica, Portugal; UCIBIO Applied Molecular Biosciences Unit, Department of Life Sciences, NOVA School of Science and Technology, NOVA University of Lisbon, 2829-516, Caparica, Portugal
| | - Leonor Saúde
- GIMM - Gulbenkian Institute for Molecular Medicine, 1649-035 Lisboa, Portugal; Faculdade de Medicina, Universidade de Lisboa, 1649-035 Lisboa, Portugal
| | - Kristian Dreij
- Institute of Environmental Medicine, Karolinska Institutet, Box 210, SE-171 77 Stockholm, Sweden
| | - Pedro M Costa
- Associate Laboratory I4HB Institute for Health and Bioeconomy, NOVA School of Science and Technology, NOVA University of Lisbon, 2829-516, Caparica, Portugal; UCIBIO Applied Molecular Biosciences Unit, Department of Life Sciences, NOVA School of Science and Technology, NOVA University of Lisbon, 2829-516, Caparica, Portugal.
| |
Collapse
|
33
|
Wada Y, Naito T, Fukushima T, Saito M. Evaluation of ALKBH2 and ALKBH3 gene regulation in patients with adult T-cell leukemia/lymphoma. Virol J 2024; 21:316. [PMID: 39633427 PMCID: PMC11619432 DOI: 10.1186/s12985-024-02590-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Accepted: 11/26/2024] [Indexed: 12/07/2024] Open
Abstract
BACKGROUND Human T-cell leukemia virus type 1 (HTLV-1) is an oncogenic virus that causes malignant adult T-cell leukemia/lymphoma (ATL). Patients infected with HTLV-1 are considered HTLV-1 carriers, and a small proportion of patients progress to life-threatening ATL after a long asymptomatic phase. No antiviral agent or preventive vaccine specific for HTLV-1 infection is established in current situation. For development of countermeasures to combat HTLV-1 infection and ATL, it is essential to expand our knowledge about their pathogenesis. Recently, AlkB homolog (ALKBH) family have been shown to participate in the oncogenesis of various cancer types. METHODS To investigate the potential role of ALKBH family members in the pathogenesis of ATL, we analyzed their gene expression dynamics in HTLV-1-infected T-cell lines and peripheral blood mononuclear cell-derived clinical specimens obtained from asymptomatic HTLV-1 carriers and patients with acute-type ATL. Epigenetic analysis was performed to dissect the mechanisms of ALKBH3 gene regulation using cultivated cells and a public dataset. RESULTS The mRNA expression levels of ALKBH2 and ALKBH3 were significantly or suggestively decreased in asymptomatic HTLV-1 carriers, but reverted in acute-type ATL patients, correlating with HTLV-1 basic leucine zipper factor gene expression. Intriguingly, the pre-mRNA expression of ALKBH2 and ALKBH3 was significantly suppressed in patients infected with HTLV-1, but not in healthy controls. Epigenetic analysis was performed to dissect the mechanisms of ALKBH3 gene regulation. In vitro analysis suggested a possible relationship between DNA methylation and ALKBH3 gene expression. Investigation of a public dataset revealed that specific CpG sites exhibited characteristically regulated methylation states in HTLV-1-infected T-cell subsets. CONCLUSION We discovered dynamically regulated patterns of ALKBH2 and ALKBH3 gene expression in patients infected with HTLV-1, and specific CpG sites epigenetically regulated by HTLV-1 infection. This study provides novel insights into HTLV-1 infection and contributes to the elucidation of ATL pathogenesis.
Collapse
Affiliation(s)
- Yuji Wada
- Department of Microbiology, Kawasaki Medical School, Kurashiki, Okayama, Japan
| | - Tadasuke Naito
- Department of Microbiology, Kawasaki Medical School, Kurashiki, Okayama, Japan
| | - Takuya Fukushima
- Laboratory of Hematoimmunology, Graduate School of Health Sciences, Faculty of Medicine, University of the Ryukyus, Nishihara, Okinawa, Japan
| | - Mineki Saito
- Department of Microbiology, Kawasaki Medical School, Kurashiki, Okayama, Japan.
| |
Collapse
|
34
|
Regényi E, Mashreghi MF, Schütte C, Sunkara V. Exploring transcription modalities from bimodal, single-cell RNA sequencing data. NAR Genom Bioinform 2024; 6:lqae179. [PMID: 39703422 PMCID: PMC11655292 DOI: 10.1093/nargab/lqae179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 11/23/2024] [Accepted: 12/06/2024] [Indexed: 12/21/2024] Open
Abstract
There is a growing interest in generating bimodal, single-cell RNA sequencing (RNA-seq) data for studying biological pathways. These data are predominantly utilized in understanding phenotypic trajectories using RNA velocities; however, the shape information encoded in the two-dimensional resolution of such data is not yet exploited. In this paper, we present an elliptical parametrization of two-dimensional RNA-seq data, from which we derived statistics that reveal four different modalities. These modalities can be interpreted as manifestations of the changes in the rates of splicing, transcription or degradation. We performed our analysis on a cell cycle and a colorectal cancer dataset. In both datasets, we found genes that are not picked up by differential gene expression analysis (DGEA), and are consequently unnoticed, yet visibly delineate phenotypes. This indicates that, in addition to DGEA, searching for genes that exhibit the discovered modalities could aid recovering genes that set phenotypes apart. For communities studying biomarkers and cellular phenotyping, the modalities present in bimodal RNA-seq data broaden the search space of genes, and furthermore, allow for incorporating cellular RNA processing into regulatory analyses.
Collapse
Affiliation(s)
- Enikő Regényi
- Systems Rheumatology, German Rheumatism Research Centre Berlin, Virchowweg 12, 10117 Berlin, Germany
- Visual and Data-Centric Computing, Zuse Institute Berlin, Takustraße 7, 14195 Berlin, Germany
| | - Mir-Farzin Mashreghi
- Systems Rheumatology, German Rheumatism Research Centre Berlin, Virchowweg 12, 10117 Berlin, Germany
| | - Christof Schütte
- Modeling and Simulation of Complex Processes, Zuse Institute Berlin, Takustraße 7, 14195 Berlin, Germany
| | - Vikram Sunkara
- Systems Rheumatology, German Rheumatism Research Centre Berlin, Virchowweg 12, 10117 Berlin, Germany
- Visual and Data-Centric Computing, Zuse Institute Berlin, Takustraße 7, 14195 Berlin, Germany
| |
Collapse
|
35
|
Yilmaz Demirel N, Weber M, Höfer K. Bridging the gap: RNAylation conjugates RNAs to proteins. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2024; 1871:119826. [PMID: 39182583 DOI: 10.1016/j.bbamcr.2024.119826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Revised: 08/04/2024] [Accepted: 08/19/2024] [Indexed: 08/27/2024]
Abstract
In nature, the majority of known RNA-protein interactions are transient. Our recent study has depicted a novel mechanism known as RNAylation, which covalently links proteins and RNAs. This novel modification bridges the realms of RNA and protein modifications. This review specifically explores RNAylation catalyzed by bacteriophage T4 ADP-ribosyltransferase ModB, with a focus on its protein targets and RNA substrates in the context of Escherichia coli-bacteriophage T4 interaction. Additionally, we discuss the biological significance of RNAylation and present perspectives on RNAylation as a versatile bioconjugation strategy for RNAs and proteins.
Collapse
Affiliation(s)
- Nurseda Yilmaz Demirel
- Max-Planck-Institute for Terrestrial Microbiology and Center for Synthetic Microbiology, 35043 Marburg, Germany
| | - Moritz Weber
- Max-Planck-Institute for Terrestrial Microbiology and Center for Synthetic Microbiology, 35043 Marburg, Germany
| | - Katharina Höfer
- Max-Planck-Institute for Terrestrial Microbiology and Center for Synthetic Microbiology, 35043 Marburg, Germany; Center for Synthetic Microbiology (SYNMIKRO), Philipps-Universität Marburg, Marburg, Germany.
| |
Collapse
|
36
|
Zhang J, Sun T, Zhang W, Chen L. Identification of acidic stress-responsive genes and acid tolerance engineering in Synechococcus elongatus PCC 7942. Appl Microbiol Biotechnol 2024; 108:115. [PMID: 38204133 PMCID: PMC10781874 DOI: 10.1007/s00253-023-12984-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 12/11/2023] [Accepted: 12/21/2023] [Indexed: 01/12/2024]
Abstract
Cyanobacteria are excellent autotrophic photosynthetic chassis employed in synthetic biology, and previous studies have suggested that they have alkaline tolerance but low acid tolerance, significantly limiting their productivity as photosynthetic chassis and necessitating investigations into the acid stress resistance mechanism. In this study, differentially expressed genes were obtained by RNA sequencing-based comparative transcriptomic analysis under long-term acidic stress conditions and acidic shock treatment, in the model cyanobacterium Synechococcus elongatus PCC 7942. A pathway enrichment analysis revealed the upregulated and downregulated pathways during long-term acidic and shock stress treatment. The subsequent single gene knockout and phenotype analysis showed that under acidic stress conditions, the strains with chlL, chlN, pex, synpcc7942_2038, synpcc7942_1890, or synpcc7942_2547 knocked out grew worse than the wild type, suggesting their involvement in acid tolerance. This finding was further confirmed by introducing the corresponding genes back into the knockout mutant individually. Moreover, individual overexpression of the chlL and chlN genes in the wild type successfully improved the tolerance of S. elongatus PCC 7942 to acidic stress. This work successfully identified six genes involved in acidic stress responses, and overexpressing chIL or chIN individually successfully improved acid tolerance in S. elongatus PCC 7942, providing valuable information to better understand the acid resistance mechanism in S. elongatus PCC 7942 and novel insights into the robustness and tolerance engineering of cyanobacterial chassis. KEY POINTS: • DEGs were identified by RNA-seq based transcriptomics analysis in response to acidic stress in S. elongatus PCC 7942. • Six genes were identified to be involved in acid tolerance in S. elongatus PCC 7942. • Overexpression of chIL or chIN individually successfully improved the acid tolerance of S. elongatus PCC 7942.
Collapse
Affiliation(s)
- Jie Zhang
- Laboratory of Synthetic Microbiology, School of Chemical Engineering & Technology, Tianjin University, Tianjin, 300072, People's Republic of China
- Frontiers Science Center for Synthetic Biology, Tianjin University, Tianjin, 300072, People's Republic of China
- Key Laboratory of Systems Bioengineering (MOE), Tianjin University, Tianjin, 300072, People's Republic of China
| | - Tao Sun
- Laboratory of Synthetic Microbiology, School of Chemical Engineering & Technology, Tianjin University, Tianjin, 300072, People's Republic of China
- Frontiers Science Center for Synthetic Biology, Tianjin University, Tianjin, 300072, People's Republic of China
- Key Laboratory of Systems Bioengineering (MOE), Tianjin University, Tianjin, 300072, People's Republic of China
- Center for Biosafety Research and Strategy, Tianjin University, Tianjin, 300072, People's Republic of China
| | - Weiwen Zhang
- Laboratory of Synthetic Microbiology, School of Chemical Engineering & Technology, Tianjin University, Tianjin, 300072, People's Republic of China
- Frontiers Science Center for Synthetic Biology, Tianjin University, Tianjin, 300072, People's Republic of China
- Key Laboratory of Systems Bioengineering (MOE), Tianjin University, Tianjin, 300072, People's Republic of China
- Center for Biosafety Research and Strategy, Tianjin University, Tianjin, 300072, People's Republic of China
| | - Lei Chen
- Laboratory of Synthetic Microbiology, School of Chemical Engineering & Technology, Tianjin University, Tianjin, 300072, People's Republic of China.
- Frontiers Science Center for Synthetic Biology, Tianjin University, Tianjin, 300072, People's Republic of China.
- Key Laboratory of Systems Bioengineering (MOE), Tianjin University, Tianjin, 300072, People's Republic of China.
| |
Collapse
|
37
|
Solyga M, Majumdar A, Besse F. Regulating translation in aging: from global to gene-specific mechanisms. EMBO Rep 2024; 25:5265-5276. [PMID: 39562712 PMCID: PMC11624266 DOI: 10.1038/s44319-024-00315-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 10/23/2024] [Accepted: 10/29/2024] [Indexed: 11/21/2024] Open
Abstract
Aging is characterized by a decline in various biological functions that is associated with changes in gene expression programs. Recent transcriptome-wide integrative studies in diverse organisms and tissues have revealed a gradual uncoupling between RNA and protein levels with aging, which highlights the importance of post-transcriptional regulatory processes. Here, we provide an overview of multi-omics analyses that show the progressive uncorrelation of transcriptomes and proteomes during the course of healthy aging. We then describe the molecular changes leading to global downregulation of protein synthesis with age and review recent work dissecting the mechanisms involved in gene-specific translational regulation in complementary model organisms. These mechanisms include the recognition of regulated mRNAs by trans-acting factors such as miRNA and RNA-binding proteins, the condensation of mRNAs into repressive cytoplasmic RNP granules, and the pausing of ribosomes at specific residues. Lastly, we mention future challenges of this emerging field, possible buffering functions as well as potential links with disease.
Collapse
Affiliation(s)
- Mathilde Solyga
- Université Côte d'Azur, CNRS, Inserm, Institut de Biologie Valrose, Nice, France
| | - Amitabha Majumdar
- National Centre for Cell Science, Savitribai Phule Pune University Campus, Pune, Maharashtra, India
| | - Florence Besse
- Université Côte d'Azur, CNRS, Inserm, Institut de Biologie Valrose, Nice, France.
| |
Collapse
|
38
|
Hashmi MATS, Fatima H, Ahmad S, Rehman A, Safdar F. The interplay between epitranscriptomic RNA modifications and neurodegenerative disorders: Mechanistic insights and potential therapeutic strategies. IBRAIN 2024; 10:395-426. [PMID: 39691424 PMCID: PMC11649393 DOI: 10.1002/ibra.12183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Revised: 10/16/2024] [Accepted: 10/19/2024] [Indexed: 12/19/2024]
Abstract
Neurodegenerative disorders encompass a group of age-related conditions characterized by the gradual decline in both the structure and functionality of the central nervous system (CNS). RNA modifications, arising from the epitranscriptome or RNA-modifying protein mutations, have recently been observed to contribute significantly to neurodegenerative disorders. Specific modifications like N6-methyladenine (m6A), N1-methyladenine (m1A), 5-methylcytosine (m5C), pseudouridine and adenosine-to-inosine (A-to-I) play key roles, with their regulators serving as crucial therapeutic targets. These epitranscriptomic changes intricately control gene expression, influencing cellular functions and contributing to disease pathology. Dysregulation of RNA metabolism, affecting mRNA processing and noncoding RNA biogenesis, is a central factor in these diseases. This review underscores the complex relationship between RNA modifications and neurodegenerative disorders, emphasizing the influence of RNA modification and the epitranscriptome, exploring the function of RNA modification enzymes in neurodegenerative processes, investigating the functional consequences of RNA modifications within neurodegenerative pathways, and evaluating the potential therapeutic advancements derived from assessing the epitranscriptome.
Collapse
Affiliation(s)
| | | | - Sadia Ahmad
- Institute of ZoologyUniversity of PunjabLahorePakistan
| | - Amna Rehman
- Institute of ZoologyUniversity of PunjabLahorePakistan
| | - Fiza Safdar
- Department of BiochemistryUniversity of NarowalNarowalPakistan
| |
Collapse
|
39
|
Gu Y, Chen J, Wang Z, Liu C, Wang T, Kim CJ, Durikova H, Fernandes S, Johnson DN, De Rose R, Cortez-Jugo C, Caruso F. mRNA delivery enabled by metal-organic nanoparticles. Nat Commun 2024; 15:9664. [PMID: 39511206 PMCID: PMC11544223 DOI: 10.1038/s41467-024-53969-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 10/25/2024] [Indexed: 11/15/2024] Open
Abstract
mRNA therapeutics are set to revolutionize disease prevention and treatment, inspiring the development of platforms for safe and effective mRNA delivery. However, current mRNA delivery platforms face some challenges, including limited organ tropism for nonvaccine applications and inflammation induced by cationic nanoparticle components. Herein, we address these challenges through a versatile, noncationic nanoparticle platform whereby mRNA is assembled into a poly(ethylene glycol)-polyphenol network stabilized by metal ions. Screening a range of components and relative compositional ratios affords a library of stable, noncationic, and highly biocompatible metal-organic nanoparticles with robust mRNA transfection in vitro and in mice. Intravenous administration of the lead mRNA-containing metal-organic nanoparticles enables predominant protein expression and gene editing in the brain, liver, and kidney, while organ tropism is tuned by varying nanoparticle composition. This study opens an avenue for realizing metal-organic nanoparticle-enabled mRNA delivery, offering a modular approach to assembling mRNA therapeutics for health applications.
Collapse
Affiliation(s)
- Yuang Gu
- Department of Chemical Engineering, The University of Melbourne, Parkville, VIC, Australia
| | - Jingqu Chen
- Department of Chemical Engineering, The University of Melbourne, Parkville, VIC, Australia
| | - Zhaoran Wang
- Department of Chemical Engineering, The University of Melbourne, Parkville, VIC, Australia
| | - Chang Liu
- Department of Chemical Engineering, The University of Melbourne, Parkville, VIC, Australia
| | - Tianzheng Wang
- Department of Chemical Engineering, The University of Melbourne, Parkville, VIC, Australia
| | - Chan-Jin Kim
- Department of Chemical Engineering, The University of Melbourne, Parkville, VIC, Australia
| | - Helena Durikova
- Department of Chemical Engineering, The University of Melbourne, Parkville, VIC, Australia
| | - Soraia Fernandes
- Department of Chemical Engineering, The University of Melbourne, Parkville, VIC, Australia
| | - Darryl N Johnson
- Materials Characterisation and Fabrication Platform, The University of Melbourne, Parkville, VIC, Australia
| | - Robert De Rose
- Department of Chemical Engineering, The University of Melbourne, Parkville, VIC, Australia
| | - Christina Cortez-Jugo
- Department of Chemical Engineering, The University of Melbourne, Parkville, VIC, Australia
| | - Frank Caruso
- Department of Chemical Engineering, The University of Melbourne, Parkville, VIC, Australia.
| |
Collapse
|
40
|
Xu Z, Asakawa S. The Definition of RNA Age Related to RNA Sequence Changes. WILEY INTERDISCIPLINARY REVIEWS. RNA 2024; 15:e1876. [PMID: 39628136 DOI: 10.1002/wrna.1876] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 10/27/2024] [Accepted: 11/06/2024] [Indexed: 04/10/2025]
Abstract
Ribonucleic acid (RNA) undergoes dynamic changes in its structure and function under various intracellular and extracellular conditions over time. However, there is a lack of research on the concept of the RNA age to describe its diverse fates. This study proposes a definition of RNA age to address this issue. RNA age was defined as a sequence of numbers wherein the elements in the sequence were the nucleotide ages of the ribonucleotide residues in the RNA. Mean nucleotide age was used to represent RNA age. This definition describes the temporal properties of RNAs that have undergone diverse life histories and reflects the dynamic state of each ribonucleotide residue, which can be expressed mathematically. Notably, events (including base insertions, base deletions, and base substitutions) are likely to cause RNA to become younger or older when using mean nucleotide ages to represent the RNA age. Although information, including the presence of added markers in RNA, chemical modification structure of the RNA, and the excision of introns in the mRNA in cells, may provide a basis for identifying RNA age, little is known about determining the RNA age of extracellular RNA in the wild. Nonetheless, we believe that RNA age has an important relationship with the diverse biological properties of RNA under intracellular and extracellular conditions. Therefore, our proposed definition of RNA age offers new perspectives for studying dynamic changes in RNA function, RNA aging, ancient RNA, environmental RNA, and the ages of other biomolecules.
Collapse
Affiliation(s)
- Zhongneng Xu
- Department of Ecology, Jinan University, Guangzhou, China
- Department of Aquatic Bioscience, Graduate School of Agricultural and Life Science, The University of Tokyo, Tokyo, Japan
| | - Shuichi Asakawa
- Department of Aquatic Bioscience, Graduate School of Agricultural and Life Science, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
41
|
He X, Yan C, Yang Y, Wang W, Liu X, Wu C, Zhou Z, Huang X, Fu W, Hu J, Yang P, Wang J, Zhu M, Liu Y, Zhang W, Li S, Dong G, Yuan X, Lin Y, Jing H, Zhang W. Prognostic significance and biological implications of SM-like genes in mantle cell lymphoma. Blood Res 2024; 59:33. [PMID: 39417944 PMCID: PMC11486876 DOI: 10.1007/s44313-024-00037-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Accepted: 10/07/2024] [Indexed: 10/19/2024] Open
Abstract
BACKGROUND SM-like (LSM) genes a family of RNA-binding proteins, are involved in mRNA regulation and can function as oncogenes by altering mRNA stability. However, their roles in B-cell progression and tumorigenesis remain poorly understood. METHODS We analyzed gene expression profiles and overall survival data of 123 patients with mantle cell lymphoma (MCL). The LSM index was developed to assess its potential as a prognostic marker of MCL survival. RESULTS Five of the eight LSM genes were identified as potential prognostic markers for survival in MCL, with particular emphasis on the LSM.index. The expression levels of these LSM genes demonstrated their potential utility as classifiers of MCL. The LSM.index-high group exhibited both poorer survival rates and lower RNA levels than did the overall transcript profile. Notably, LSM1 and LSM8 were overexpressed in the LSM.index-high group, with LSM1 showing 2.5-fold increase (p < 0.001) and LSM8 depicting 1.8-fold increase (p < 0.01) than those in the LSM.index-low group. Furthermore, elevated LSM gene expression was associated with increased cell division and RNA splicing pathway activity. CONCLUSIONS The LSM.index demonstrates potential as a prognostic marker for survival in patients with MCL. Elevated expression of LSM genes, particularly LSM1 and LSM8, may be linked to poor survival outcomes through their involvement in cell division and RNA splicing pathways. These findings suggest that LSM genes may contribute to the aggressive behavior of MCL and represent potential targets for therapeutic interventions.
Collapse
Affiliation(s)
- Xue He
- Department of Pathology, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China
| | - Changjian Yan
- Department of Hematology, Lymphoma Research Center, Peking University Third Hospital, Beijing, 100191, China
- The Second Affiliated Hospital of Fujian Medical University, Quanzhou, 362000, China
| | - Yaru Yang
- The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Weijia Wang
- Department of Pathology, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| | - Xiaoni Liu
- Department of Respiratory Medicine, The First Affiliated Hospital of Gannan Medical University, Ganzhou, 341000, China
| | - Chaoling Wu
- Department of Respiratory Medicine, Affiliated Hospital of Jiujiang University, Jiujiang, 332000, China
| | - Zimu Zhou
- Gannan Medical University, Ganzhou, 341000, China
| | - Xin Huang
- Department of Hematology, Lymphoma Research Center, Peking University Third Hospital, Beijing, 100191, China
| | - Wei Fu
- Department of Hematology, Lymphoma Research Center, Peking University Third Hospital, Beijing, 100191, China
| | - Jing Hu
- Department of Hematology, Lymphoma Research Center, Peking University Third Hospital, Beijing, 100191, China
| | - Ping Yang
- Department of Hematology, Lymphoma Research Center, Peking University Third Hospital, Beijing, 100191, China
| | - Jing Wang
- Department of Hematology, Lymphoma Research Center, Peking University Third Hospital, Beijing, 100191, China
| | - Mingxia Zhu
- Department of Hematology, Lymphoma Research Center, Peking University Third Hospital, Beijing, 100191, China
| | - Yan Liu
- Department of Hematology, Lymphoma Research Center, Peking University Third Hospital, Beijing, 100191, China
| | - Wei Zhang
- Department of Pathology, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China
| | - Shaoxiang Li
- Department of Pathology, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China
| | - Gehong Dong
- Department of Pathology, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China
| | - Xiaoliang Yuan
- Department of Respiratory Medicine, The First Affiliated Hospital of Gannan Medical University, Ganzhou, 341000, China
| | - Yuansheng Lin
- Department of Intensive Care Unit, Suzhou Research Center of Medical School, Suzhou Hospital, Affiliated Hospital of Medical School, Nanjing University, Suzhou, 215000, China.
| | - Hongmei Jing
- Department of Hematology, Lymphoma Research Center, Peking University Third Hospital, Beijing, 100191, China.
| | - Weilong Zhang
- Department of Hematology, Lymphoma Research Center, Peking University Third Hospital, Beijing, 100191, China.
| |
Collapse
|
42
|
Wu L, Yi W, Yao S, Xie S, Peng R, Zhang J, Tan W. mRNA-Based Cancer Vaccines: Advancements and Prospects. NANO LETTERS 2024. [PMID: 39375146 DOI: 10.1021/acs.nanolett.4c03296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/09/2024]
Abstract
The success of mRNA COVID-19 vaccines has reinvigorated research and interest in mRNA-based cancer vaccines. Despite promising results in clinical trials, therapeutic mRNA-based cancer vaccines have not yet been approved for human use. These vaccines are designed to trigger tumor regression, establish enduring antitumor memory, and mitigate adverse reactions. However, challenges such as tumor-induced immunosuppression and immunoresistance significantly hinder their application. Here, we provide an overview of the recent advances of neoantigen discovery and delivery systems for mRNA vaccines, focusing on improving clinical efficacy. Additionally, we summarize the recent clinical advances involving mRNA cancer vaccines and discuss prospective strategies for overcoming immuneresistance.
Collapse
Affiliation(s)
- Lijin Wu
- The Key Laboratory of Zhejiang Province for Aptamers and Theranostics, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
- School of Molecular Medicine, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China
- University of Chinese Academy of Sciences, No.19 A Yuquan Road, Beijing 100049, China
| | - Weicheng Yi
- The Key Laboratory of Zhejiang Province for Aptamers and Theranostics, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Shiyu Yao
- The Key Laboratory of Zhejiang Province for Aptamers and Theranostics, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan 410082, China
| | - Sitao Xie
- The Key Laboratory of Zhejiang Province for Aptamers and Theranostics, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Ruizi Peng
- The Key Laboratory of Zhejiang Province for Aptamers and Theranostics, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Jing Zhang
- The Key Laboratory of Zhejiang Province for Aptamers and Theranostics, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Weihong Tan
- The Key Laboratory of Zhejiang Province for Aptamers and Theranostics, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan 410082, China
| |
Collapse
|
43
|
Reitemeier J, Metro J, Bohn PW. Detection of aldehydes from degradation of lipid nanoparticle formulations using a hierarchically-organized nanopore electrochemical biosensor. Biosens Bioelectron 2024; 261:116457. [PMID: 38850733 DOI: 10.1016/j.bios.2024.116457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 05/20/2024] [Accepted: 05/30/2024] [Indexed: 06/10/2024]
Abstract
Degradation of ionizable lipids in mRNA-based vaccines was recently found to deactivate the payload, demanding rigorous monitoring of impurities in lipid nanoparticle (LNP) formulations. However, parallel screening for lipid degradation in customized delivery systems for next-generation therapeutics maintains a challenging and unsolved problem. Here, we describe a nanopore electrochemical sensor to detect ppb-levels of aldehydes arising from lipid degradation in LNP formulations that can be deployed in massively parallel fashion. Specifically, we combine nanopore electrodes with a block copolymer (BCP) membrane capable of hydrophobic gating of analyte transport between the bulk solution and the nanopore volume. By incorporating aldehyde dehydrogenase (ALDH), enzymatic oxidation of aldehydes generates NADH to enable ultrasensitive voltammetric detection with limits-of-detection (LOD) down to 1.2 ppb. Sensor utility was demonstrated by detecting degradation of N-oxidized SM-102, the ionizable lipid in Moderna's SpikeVax™ vaccine, in mRNA-1273 LNP formulation. This work should be of significant use in the pharmaceutical industry, paving the way for automated on-line quality assessments of next-generation therapeutics.
Collapse
Affiliation(s)
- Julius Reitemeier
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN 46556, United States
| | - Jarek Metro
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN 46556, United States
| | - Paul W Bohn
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN 46556, United States; Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, IN 46556, United States.
| |
Collapse
|
44
|
Querl L, Krebber H. Defenders of the Transcriptome: Guard Protein-Mediated mRNA Quality Control in Saccharomyces cerevisiae. Int J Mol Sci 2024; 25:10241. [PMID: 39408571 PMCID: PMC11476243 DOI: 10.3390/ijms251910241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 09/17/2024] [Accepted: 09/19/2024] [Indexed: 10/20/2024] Open
Abstract
Cell survival depends on precise gene expression, which is controlled sequentially. The guard proteins surveil mRNAs from their synthesis in the nucleus to their translation in the cytoplasm. Although the proteins within this group share many similarities, they play distinct roles in controlling nuclear mRNA maturation and cytoplasmic translation by supporting the degradation of faulty transcripts. Notably, this group is continuously expanding, currently including the RNA-binding proteins Npl3, Gbp2, Hrb1, Hrp1, and Nab2 in Saccharomyces cerevisiae. Some of the human serine-arginine (SR) splicing factors (SRSFs) show remarkable similarities to the yeast guard proteins and may be considered as functional homologues. Here, we provide a comprehensive summary of their crucial mRNA surveillance functions and their implications for cellular health.
Collapse
Affiliation(s)
| | - Heike Krebber
- Abteilung für Molekulare Genetik, Institut für Mikrobiologie und Genetik, Göttinger Zentrum für Molekulare Biowissenschaften (GZMB), Georg-August Universität Göttingen, 37077 Göttingen, Germany;
| |
Collapse
|
45
|
An W, Yan Y, Ye K. High resolution landscape of ribosomal RNA processing and surveillance. Nucleic Acids Res 2024; 52:10630-10644. [PMID: 38994562 PMCID: PMC11417381 DOI: 10.1093/nar/gkae606] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 06/21/2024] [Accepted: 06/28/2024] [Indexed: 07/13/2024] Open
Abstract
Ribosomal RNAs are processed in a complex pathway. We profiled rRNA processing intermediates in yeast at single-molecule and single-nucleotide levels with circularization, targeted amplification and deep sequencing (CircTA-seq), gaining significant mechanistic insights into rRNA processing and surveillance. The long form of the 5' end of 5.8S rRNA is converted to the short form and represents an intermediate of a unified processing pathway. The initial 3' end processing of 5.8S rRNA involves trimming by Rex1 and Rex2 and Trf4-mediated polyadenylation. The 3' end of 25S rRNA is formed by sequential digestion by four Rex proteins. Intermediates with an extended A1 site are generated during 5' degradation of aberrant 18S rRNA precursors. We determined precise polyadenylation profiles for pre-rRNAs and show that the degradation efficiency of polyadenylated 20S pre-rRNA critically depends on poly(A) lengths and degradation intermediates released from the exosome are often extensively re-polyadenylated.
Collapse
MESH Headings
- RNA Processing, Post-Transcriptional
- Saccharomyces cerevisiae/genetics
- Saccharomyces cerevisiae/metabolism
- RNA, Ribosomal/metabolism
- RNA, Ribosomal/genetics
- RNA, Ribosomal/chemistry
- RNA, Ribosomal, 5.8S/genetics
- RNA, Ribosomal, 5.8S/metabolism
- Saccharomyces cerevisiae Proteins/metabolism
- Saccharomyces cerevisiae Proteins/genetics
- RNA Precursors/metabolism
- RNA Precursors/genetics
- RNA, Ribosomal, 18S/metabolism
- RNA, Ribosomal, 18S/genetics
- Polyadenylation
- RNA, Fungal/metabolism
- RNA, Fungal/chemistry
- RNA, Fungal/genetics
- Exosome Multienzyme Ribonuclease Complex/metabolism
- Exosome Multienzyme Ribonuclease Complex/genetics
- High-Throughput Nucleotide Sequencing
- RNA Stability
Collapse
Affiliation(s)
- Weidong An
- Key Laboratory of RNA Science and Engineering, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Yunxiao Yan
- Key Laboratory of RNA Science and Engineering, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Keqiong Ye
- Key Laboratory of RNA Science and Engineering, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
46
|
Huang L, Liang S, Luo L, Wu M, Fu H, Zhong Z. Transcriptomic analysis reveals effects of fertilization towards growth and quality of Fritillariae thunbergii bulbus. PLoS One 2024; 19:e0309978. [PMID: 39302908 DOI: 10.1371/journal.pone.0309978] [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: 04/07/2024] [Accepted: 08/17/2024] [Indexed: 09/22/2024] Open
Abstract
Fritillariae thunbergii Bulbus (FTB) is a traditional Chinese medicine that has been widely cultivated for its expectorant, antitussive, antiasthmatic, antiviral, and anticancer properties. The yield and quality of F. thunbergii are influenced by cultivation conditions, such as the use of fertilizers. However, the optimal type of fertilizers for maximum quality and yield and underlying mechanisms are not clear. We collected F. thunbergii using raw chicken manure (RC), organic fertilizer (OF), and plant ash (PA) as the base fertilizer in Pan'an County, Jinhua City, Zhejiang Province as experimental materials. The combined results of HPLC-ELSD detection and yield statistics showed that the F. thunbergii with OF application was the best, with the content of peimine and peiminine reaching 0.0603% and 0.0502%, respectively. In addition, the yield was 2.70 kg/m2. Transcriptome analysis indicated that up-regulation of the ABA signaling pathway might promote bulb yield. Furthermore, putative key genes responsible for steroidal alkaloid accumulation were identified. These results provided guiding significance for the rational fertilization conditions of F. thunbergii as well as the basis for the exploration of functional genes related to the alkaloid biosynthesis pathway.
Collapse
Affiliation(s)
- Luman Huang
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, P.R. China
| | - Shuang Liang
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, P.R. China
| | - Lei Luo
- Zhejiang Institute for Food and Drug Control, Hangzhou, P.R. China
| | - Mengmin Wu
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, P.R. China
| | - Hongwei Fu
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, P.R. China
| | - Zhuoheng Zhong
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, P.R. China
| |
Collapse
|
47
|
Dutta S, Srivatsan SG. Enzymatic Functionalization of RNA Oligonucleotides by Terminal Uridylyl Transferase Using Fluorescent and Clickable Nucleotide Analogs. Chem Asian J 2024; 19:e202400475. [PMID: 38949615 DOI: 10.1002/asia.202400475] [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: 04/26/2024] [Revised: 06/27/2024] [Accepted: 07/01/2024] [Indexed: 07/02/2024]
Abstract
We report a systematic study on controlling the enzyme activity of a terminal uridylyl transferase (TUTase) called SpCID1, which provides methods to effect site-specific incorporation of a single modified nucleotide analog at the 3'-end of an RNA oligonucleotide (ON). Responsive heterocycle-modified fluorescent UTP probes that are useful in analyzing non-canonical nucleic acid structures and azide- and alkyne-modified UTP analogs that are compatible for chemoenzymatic functionalization were used as study systems. In the first strategy, we balanced the concentration of essential metal ion cofactors (Mg2+ and Mn2+ ions) to restrict the processivity of the enzyme, which gave a very good control on the incorporation of clickable nucleotide analogs. In the second approach, borate that complexes with 2' and 3' oxygen atoms of a ribose sugar was used as a reversibly binding chelator to block repeated addition of nucleotide analogs. Notably, in the presence of heterocycle-modified fluorescent UTPs, we obtained single-nucleotide incorporated RNA products in reasonable yields, while with clickable nucleotides yields were very good. Further, 3'-end azide- and alkyne-labeled RNA ONs were post-enzymatically functionalized by CuAAC and SPAAC reactions with fluorescent probes. These strategies broaden the scope of TUTase in site-specifically installing modifications of different types onto RNA for various applications.
Collapse
Affiliation(s)
- Swagata Dutta
- Department of Chemistry, Indian Institute of Science Education and Research (IISER), Pune Dr. Homi Bhabha Road, Pune, 411008, India
| | - Seergazhi G Srivatsan
- Department of Chemistry, Indian Institute of Science Education and Research (IISER), Pune Dr. Homi Bhabha Road, Pune, 411008, India
| |
Collapse
|
48
|
Hulscher N, McCullough PA, Marotta DE. Strategic deactivation of mRNA COVID-19 vaccines: New applications for siRNA therapy and RIBOTACs. J Gene Med 2024; 26:e3733. [PMID: 39183706 DOI: 10.1002/jgm.3733] [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: 06/02/2024] [Revised: 07/19/2024] [Accepted: 08/13/2024] [Indexed: 08/27/2024] Open
Abstract
The rapid development and authorization of messenger ribonucleic acid (mRNA) vaccines by Pfizer-BioNTech (BNT162b2) and Moderna (mRNA-1273) in 2020 marked a significant milestone in human mRNA product application, overcoming previous obstacles such as mRNA instability and immunogenicity. This paper reviews the strategic modifications incorporated into these vaccines to enhance mRNA stability and translation efficiency, such as the inclusion of nucleoside modifications and optimized mRNA design elements including the 5' cap and poly(A) tail. We highlight emerging concerns regarding the wide systemic biodistribution of these mRNA vaccines leading to prolonged inflammatory responses and other safety concerns. The regulatory framework guiding the biodistribution studies is pivotal in assessing the safety profiles of new mRNA formulations in use today. The stability of mRNA vaccines, their pervasive distribution, and the longevity of the encapsulated mRNA along with unlimited production of the damaging and potentially lethal spike (S) protein call for strategies to mitigate potential adverse effects. Here, we explore the potential of small interfering RNA (siRNA) and ribonuclease targeting chimeras (RIBOTACs) as promising solutions to target, inactivate, and degrade residual and persistent vaccine mRNA, thereby potentially preventing uncontrolled S protein production and reducing toxicity. The targeted nature of siRNA and RIBOTACs allows for precise intervention, offering a path to prevent and mitigate adverse events of mRNA-based therapies. This review calls for further research into siRNA and RIBOTAC applications as antidotes and detoxication products for mRNA vaccine technology.
Collapse
|
49
|
Li A, Zhu Z, Yang J, Liu Y, Zhang Y, Liu J. Precise Insertion of AttB Sequences in Goat Genome Using Enhanced Prime Editor. Int J Mol Sci 2024; 25:9486. [PMID: 39273433 PMCID: PMC11395042 DOI: 10.3390/ijms25179486] [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: 06/12/2024] [Revised: 08/15/2024] [Accepted: 08/26/2024] [Indexed: 09/15/2024] Open
Abstract
Prime editor, an editing tool based on the CRISPR/Cas9 system, allows for all 12 types of nucleotide exchanges and arbitrary indels in genomic sequences without the need for inducing DNA double-strand breaks. Despite its flexibility and precision, prime editing efficiency is still low and hindered by various factors such as target sites, editing types, and the length of the primer binding site. In this study, we developed a prime editing system by incorporating an RNA motif at the 3' terminal of the pegRNA and integrating all twin prime editor factors into a single plasmid. These two strategies enhanced prime editing efficiency at target sites by up to 3.58-fold and 2.19-fold, respectively. Subsequently, enhanced prime editor was employed in goat cells and embryos to efficiently insert a 38 bp attB sequence into the Gt(ROSA)26Sor (Rosa26) and C-C motif chemokine receptor 5 (CCR5) loci. The enhanced prime editor can mediate 11.9% and 6.8% editing efficiency in parthenogenetic activation of embryos through embryo microinjection. In summary, our study introduces a modified prime editing system with improved editing and transfection efficiency, making it more suitable for inserting foreign sequences into primary cells and embryos. These results broaden the potential applications of prime editing technologies in the production of transgenic animals.
Collapse
Affiliation(s)
- Aicong Li
- Key Laboratory of Livestock Biology, Northwest A&F University, Yangling 712100, China
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China
| | - Zhenliang Zhu
- Key Laboratory of Livestock Biology, Northwest A&F University, Yangling 712100, China
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China
| | - Jing Yang
- Key Laboratory of Livestock Biology, Northwest A&F University, Yangling 712100, China
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China
| | - Yayi Liu
- Key Laboratory of Livestock Biology, Northwest A&F University, Yangling 712100, China
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China
| | - Yong Zhang
- Key Laboratory of Livestock Biology, Northwest A&F University, Yangling 712100, China
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China
| | - Jun Liu
- Key Laboratory of Livestock Biology, Northwest A&F University, Yangling 712100, China
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China
| |
Collapse
|
50
|
Stefanovic F, Brown LG, MacDonald J, Bammler T, Rinchai D, Nguyen S, Zeng Y, Shinkawa V, Adams K, Chausabel D, Berthier E, Haack AJ, Theberge AB. Your Blood is Out for Delivery: Considerations of Shipping Time and Temperature on Degradation of RNA from Stabilized Whole Blood. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.24.609519. [PMID: 39229214 PMCID: PMC11370555 DOI: 10.1101/2024.08.24.609519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
Remote research studies are an invaluable tool for reaching populations in geographical regions with limited access to large medical centers or universities. To expand the remote study toolkit, we have previously developed homeRNA, which allows for at-home self-collection and stabilization of blood and demonstrated the feasibility of using homeRNA in high temperature climates. Here, we expand upon this work through a systematic study exploring the effects of high temperature on RNA integrity through in-lab and field experiments. Compared to the frozen controls (overall mean RIN of 8.2, n = 8), samples kept at 37°C for 2, 4, and 8 days had mean RINs of 7.6, 5.9, and 5.2 (n = 3), respectively, indicating that typical shipping conditions (~2 days) yield samples suitable for downstream RNA sequencing. Shorter time intervals (6 hours) resulted in minimal RNA degradation (median RIN of 6.4, n = 3) even at higher temperatures (50°C) compared to the frozen control (mean RIN of 7.8, n = 3). Additionally, we shipped homeRNA-stabilized blood from a single donor to 14 different states and back during the summer with continuous temperature probes (7.1 median RIN, n = 42). Samples from all locations were analyzed with 3' mRNA-seq to assess differences in gene counts, with the transcriptomic data suggesting that there was no preferential degradation of transcripts as a result of different shipping times, temperatures, and regions. Overall, our data support that homeRNA can be used in elevated temperature conditions, enabling decentralized sample collection for telemedicine, global health, and clinical research.
Collapse
Affiliation(s)
- Filip Stefanovic
- Department of Chemistry, University of Washington, Seattle, Washington 98195, United States
| | - Lauren G. Brown
- Department of Chemistry, University of Washington, Seattle, Washington 98195, United States
| | - James MacDonald
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, Washington 98195, United States
| | - Theo Bammler
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, Washington 98195, United States
| | - Darawan Rinchai
- Department of Infectious Diseases, St Jude’s Children Research Hospital, TN, Memphis 38105, United States
| | - Serena Nguyen
- Department of Chemistry, University of Washington, Seattle, Washington 98195, United States
| | - Yuting Zeng
- Department of Chemistry, University of Washington, Seattle, Washington 98195, United States
| | - Victoria Shinkawa
- Department of Chemistry, University of Washington, Seattle, Washington 98195, United States
| | - Karen Adams
- Department of Chemistry, University of Washington, Seattle, Washington 98195, United States
- Institute of Translational Health Sciences, School of Medicine, University of Washington, Seattle, Washington 98195, United States
| | - Damien Chausabel
- Computer Sciences Department, The Jackson Laboratory, Farmington, CT, 06032, United States
| | - Erwin Berthier
- Department of Chemistry, University of Washington, Seattle, Washington 98195, United States
| | - Amanda J. Haack
- Department of Chemistry, University of Washington, Seattle, Washington 98195, United States
- School of Medicine, University of Washington, Seattle, Washington 98195, United States
| | - Ashleigh B. Theberge
- Department of Chemistry, University of Washington, Seattle, Washington 98195, United States
- Department of Urology, School of Medicine, University of Washington, Seattle, Washington 98195, United States
| |
Collapse
|