1
|
Embree CM, Stephanou A, Singh G. Direct and indirect effects of spliceosome disruption compromise gene regulation by Nonsense-Mediated mRNA Decay. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.27.630533. [PMID: 39763844 PMCID: PMC11703147 DOI: 10.1101/2024.12.27.630533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/11/2025]
Abstract
Pre-mRNA splicing, carried out in the nucleus by a large ribonucleoprotein machine known as the spliceosome, is functionally and physically coupled to the mRNA surveillance pathway in the cytoplasm called nonsense mediated mRNA decay (NMD). The NMD pathway monitors for premature translation termination signals, which can result from alternative splicing, by relying on the exon junction complex (EJC) deposited on exon-exon junctions by the spliceosome. Recently, multiple genetic screens in human cell lines have identified numerous spliceosome components as putative NMD factors. Using publicly available RNA-seq datasets from K562 and HepG2 cells depleted of 18 different spliceosome components, we find that natural NMD targeted mRNA isoforms are upregulated when members of the catalytic spliceosome are reduced. While some of this increase could be due to widespread pleiotropic effects of spliceosome dysfunction (e.g., reduced expression of NMD factors due to mis-splicing of their mRNAs), we identify that AQR, SF3B1, SF3B4 and CDC40 may have a more direct role in NMD. We also test the hypothesis that increased production of novel NMD substrates may overwhelm the pathway to find a direct correlation between the amount of novel NMD substrates detected and the degree of NMD inhibition observed. Finally, similar transcriptome alterations and NMD substrate upregulation are also observed in cells treated with spliceosome inhibitors and in cells derived from retinitis pigmentosa patients with mutations in PRPF8 and PRPF31. Overall, our results show that regardless of the cause, spliceosome disruption upregulates a broad set of NMD targets, which could contribute to cellular dysfunction in spliceosomopathies.
Collapse
Affiliation(s)
- Caleb M Embree
- Department of Molecular Genetics, Center for RNA Biology, The Ohio State University, Columbus, OH, 43210
| | - Andreas Stephanou
- Department of Molecular Genetics, Center for RNA Biology, The Ohio State University, Columbus, OH, 43210
| | - Guramrit Singh
- Department of Molecular Genetics, Center for RNA Biology, The Ohio State University, Columbus, OH, 43210
| |
Collapse
|
2
|
Cerna-Chavez R, Ortega-Gasco A, Baig HMA, Ehrenreich N, Metais T, Scandura MJ, Bujakowska K, Pierce EA, Garita-Hernandez M. Optimized Prime Editing of Human Induced Pluripotent Stem Cells to Efficiently Generate Isogenic Models of Mendelian Diseases. Int J Mol Sci 2024; 26:114. [PMID: 39795970 PMCID: PMC11719581 DOI: 10.3390/ijms26010114] [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: 12/06/2024] [Revised: 12/23/2024] [Accepted: 12/24/2024] [Indexed: 01/13/2025] Open
Abstract
Prime editing (PE) is a CRISPR-based tool for genome engineering that can be applied to generate human induced pluripotent stem cell (hiPSC)-based disease models. PE technology safely introduces point mutations, small insertions, and deletions (indels) into the genome. It uses a Cas9-nickase (nCas9) fused to a reverse transcriptase (RT) as an editor and a PE guide RNA (pegRNA), which introduces the desired edit with great precision without creating double-strand breaks (DSBs). PE leads to minimal off-targets or indels when introducing single-strand breaks (SSB) in the DNA. Low efficiency can be an obstacle to its use in hiPSCs, especially when the genetic context precludes the screening of multiple pegRNAs, and other strategies must be employed to achieve the desired edit. We developed a PE platform to efficiently generate isogenic models of Mendelian disorders. We introduced the c.25G>A (p.V9M) mutation in the NMNAT1 gene with over 25% efficiency by optimizing the PE workflow. Using our optimized system, we generated other isogenic models of inherited retinal diseases (IRDs), including the c.1481C>T (p.T494M) mutation in PRPF3 and the c.6926A>C (p.H2309P) mutation in PRPF8. We modified several determinants of the hiPSC PE procedure, such as plasmid concentrations, PE component ratios, and delivery method settings, showing that our improved workflow increased the hiPSC editing efficiency.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Marcela Garita-Hernandez
- Ocular Genomics Institute, Massachusetts Eye and Ear Infirmary, Department of Ophthalmology, Harvard Medical School, Boston, MA 02114, USA; (R.C.-C.); (A.O.-G.); (H.M.A.B.); (N.E.); (T.M.); (M.J.S.); (K.B.); (E.A.P.)
| |
Collapse
|
3
|
Zheng N, Liao T, Zhang C, Zhang Z, Yan S, Xi X, Ruan F, Yang C, Zhao Q, Deng W, Huang J, Huang Z, Chen Z, Wang X, Qu Q, Zuo Z, He C. Quantum Dots-caused Retinal Degeneration in Zebrafish Regulated by Ferroptosis and Mitophagy in Retinal Pigment Epithelial Cells through Inhibiting Spliceosome. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2406343. [PMID: 39420512 PMCID: PMC11633537 DOI: 10.1002/advs.202406343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/08/2024] [Revised: 08/23/2024] [Indexed: 10/19/2024]
Abstract
Quantum dots (QDs) are widely used, but their health impact on the visual system is little known. This study aims to elucidate the effects and mechanisms of typical metallic QDs on retinas using zebrafish. Comprehensive histology, imaging, and bulk RNA sequencing reveal that InP/ZnS QDs cause retinal degeneration. Furthermore, single-cell RNA-seq reveals a reduction in the number of retinal pigment epithelial cells (RPE) and short-wave cone UV photoreceptor cells (PR(UV)), accompanied by an increase in middle- and long-wave cone red, green, and blue photoreceptor cells [PR(RGB)]. Mechanistically, after endocytosis by RPE, InP/ZnS QDs inhibit the expression of splicing factor prpf8, resulting in gpx4b mRNA unsplicing, which finally decrease glutathione and induce ferroptosis and mitophagy. The decrease of RPE fails to engulf the damaged outer segments of PR, possibly promoting the differentiation of PR(UV) to PR(RGB). Knockout prpf8 or gpx4b with CRISPR/Cas9 system, the retinal damage is also observed. Whereas, overexpression of prpf8 or gpx4b, or supplement of glutathione can rescue the retinal degenerative damage caused by InP/ZnS QDs. In conclusion, this study illustrates the potential health risks of InP/ZnS QDs on eye development and provides valuable insights into the underlying mechanisms of InP/ZnS QDs-caused retinal degeneration.
Collapse
Affiliation(s)
- Naying Zheng
- Department of Ophthalmology in Xiang'an Hospital of Xiamen UniversityState Key Laboratory of Cellular Stress BiologySchool of Life SciencesFaculty of Medicine and Life SciencesXiamen UniversityXiamenFujian361102China
| | - Tingting Liao
- Department of Ophthalmology in Xiang'an Hospital of Xiamen UniversityState Key Laboratory of Cellular Stress BiologySchool of Life SciencesFaculty of Medicine and Life SciencesXiamen UniversityXiamenFujian361102China
| | - Chuchu Zhang
- Department of Ophthalmology in Xiang'an Hospital of Xiamen UniversityState Key Laboratory of Cellular Stress BiologySchool of Life SciencesFaculty of Medicine and Life SciencesXiamen UniversityXiamenFujian361102China
| | - Zheyang Zhang
- Department of Ophthalmology in Xiang'an Hospital of Xiamen UniversityState Key Laboratory of Cellular Stress BiologySchool of Life SciencesFaculty of Medicine and Life SciencesXiamen UniversityXiamenFujian361102China
| | - Sen Yan
- Department of ChemistryState Key Laboratory of Physical Chemistry of Solid SurfacesCollaborative Innovation Center of Chemistry for Energy Materials (i‐ChEM)Innovation Laboratory for Sciences and Technologies of Energy Materials of Fujian Province (IKKEM)College of Chemistry and Chemical EngineeringXiamen UniversityXiamen361005China
| | - Xiaohan Xi
- Department of ChemistryState Key Laboratory of Physical Chemistry of Solid SurfacesCollaborative Innovation Center of Chemistry for Energy Materials (i‐ChEM)Innovation Laboratory for Sciences and Technologies of Energy Materials of Fujian Province (IKKEM)College of Chemistry and Chemical EngineeringXiamen UniversityXiamen361005China
| | - Fengkai Ruan
- Department of Ophthalmology in Xiang'an Hospital of Xiamen UniversityState Key Laboratory of Cellular Stress BiologySchool of Life SciencesFaculty of Medicine and Life SciencesXiamen UniversityXiamenFujian361102China
| | - Chunyan Yang
- Department of Ophthalmology in Xiang'an Hospital of Xiamen UniversityState Key Laboratory of Cellular Stress BiologySchool of Life SciencesFaculty of Medicine and Life SciencesXiamen UniversityXiamenFujian361102China
| | - Qingliang Zhao
- State Key Laboratory of Vaccines for Infectious DiseasesCenter for Molecular Imaging and Translational MedicineXiang An Biomedicine LaboratorySchool of Public HealthXiamen UniversityXiamenFujian361005China
| | - Wenbo Deng
- Key Laboratory of Reproductive Health ResearchFujian Province UniversitySchool of MedicineXiamen UniversityXiamenFujian361005China
| | - Jialiang Huang
- Department of Ophthalmology in Xiang'an Hospital of Xiamen UniversityState Key Laboratory of Cellular Stress BiologySchool of Life SciencesFaculty of Medicine and Life SciencesXiamen UniversityXiamenFujian361102China
| | - Zi‐Tao Huang
- Guangdong Key Laboratory of Environmental Catalysis and Health Risk ControlGuangdong‐Hong Kong‐Macao Joint Laboratory for Contaminants Exposure and HealthSchool of Environmental Science and EngineeringGuangdong University of TechnologyGuangzhou510006China
| | - Zhi‐Feng Chen
- Guangdong Key Laboratory of Environmental Catalysis and Health Risk ControlGuangdong‐Hong Kong‐Macao Joint Laboratory for Contaminants Exposure and HealthSchool of Environmental Science and EngineeringGuangdong University of TechnologyGuangzhou510006China
| | - Xiang Wang
- Department of ChemistryState Key Laboratory of Physical Chemistry of Solid SurfacesCollaborative Innovation Center of Chemistry for Energy Materials (i‐ChEM)Innovation Laboratory for Sciences and Technologies of Energy Materials of Fujian Province (IKKEM)College of Chemistry and Chemical EngineeringXiamen UniversityXiamen361005China
| | - Qingming Qu
- Department of Ophthalmology in Xiang'an Hospital of Xiamen UniversityState Key Laboratory of Cellular Stress BiologySchool of Life SciencesFaculty of Medicine and Life SciencesXiamen UniversityXiamenFujian361102China
| | - Zhenghong Zuo
- Department of Ophthalmology in Xiang'an Hospital of Xiamen UniversityState Key Laboratory of Cellular Stress BiologySchool of Life SciencesFaculty of Medicine and Life SciencesXiamen UniversityXiamenFujian361102China
| | - Chengyong He
- Department of Ophthalmology in Xiang'an Hospital of Xiamen UniversityState Key Laboratory of Cellular Stress BiologySchool of Life SciencesFaculty of Medicine and Life SciencesXiamen UniversityXiamenFujian361102China
| |
Collapse
|
4
|
Alors‐Pérez E, Blázquez‐Encinas R, Moreno‐Montilla MT, García‐Vioque V, Jiménez‐Vacas JM, Mafficini A, González‐Borja I, Luchini C, Sánchez‐Hidalgo JM, Sánchez‐Frías ME, Pedraza‐Arevalo S, Romero‐Ruiz A, Lawlor RT, Viúdez A, Gahete MD, Scarpa A, Arjona‐Sánchez Á, Luque RM, Ibáñez‐Costa A, Castaño JP. Spliceosomic dysregulation in pancreatic cancer uncovers splicing factors PRPF8 and RBMX as novel candidate actionable targets. Mol Oncol 2024; 18:2524-2540. [PMID: 38790138 PMCID: PMC11459039 DOI: 10.1002/1878-0261.13658] [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: 04/14/2023] [Revised: 03/28/2024] [Accepted: 04/22/2024] [Indexed: 05/26/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly lethal cancer, characterized by late diagnosis and poor treatment response. Surgery is the only curative approach, only available to early-diagnosed patients. Current therapies have limited effects, cause severe toxicities, and minimally improve overall survival. Understanding of splicing machinery alterations in PDAC remains incomplete. Here, we comprehensively examined 59 splicing machinery components, uncovering dysregulation in pre-mRNA processing factor 8 (PRPF8) and RNA-binding motif protein X-linked (RBMX). Their downregulated expression was linked to poor prognosis and malignancy features, including tumor stage, invasion and metastasis, and associated with poorer survival and the mutation of key PDAC genes. Experimental modulation of these splicing factors in pancreatic cancer cell lines reverted their expression to non-tumor levels and resulted in decreased key tumor-related features. These results provide evidence that the splicing machinery is altered in PDAC, wherein PRPF8 and RBMX emerge as candidate actionable therapeutic targets.
Collapse
Affiliation(s)
- Emilia Alors‐Pérez
- Maimonides Biomedical Research Institute of Córdoba (IMIBIC)Spain
- Department of Cell Biology, Physiology, and ImmunologyUniversity of CórdobaSpain
- Reina Sofia University HospitalCórdobaSpain
| | - Ricardo Blázquez‐Encinas
- Maimonides Biomedical Research Institute of Córdoba (IMIBIC)Spain
- Department of Cell Biology, Physiology, and ImmunologyUniversity of CórdobaSpain
- Reina Sofia University HospitalCórdobaSpain
| | - María Trinidad Moreno‐Montilla
- Maimonides Biomedical Research Institute of Córdoba (IMIBIC)Spain
- Department of Cell Biology, Physiology, and ImmunologyUniversity of CórdobaSpain
- Reina Sofia University HospitalCórdobaSpain
| | - Víctor García‐Vioque
- Maimonides Biomedical Research Institute of Córdoba (IMIBIC)Spain
- Department of Cell Biology, Physiology, and ImmunologyUniversity of CórdobaSpain
- Reina Sofia University HospitalCórdobaSpain
| | - Juan Manuel Jiménez‐Vacas
- Maimonides Biomedical Research Institute of Córdoba (IMIBIC)Spain
- Department of Cell Biology, Physiology, and ImmunologyUniversity of CórdobaSpain
- Reina Sofia University HospitalCórdobaSpain
| | - Andrea Mafficini
- ARC‐Net Research Centre and Section of Pathology of Department of Diagnostics and Public HealthUniversity and Hospital Trust of VeronaItaly
| | - Iranzu González‐Borja
- OncobionaTras Lab, Navarrabiomed, Hospital Universitario de Navarra, Instituto de Investigación Sanitaria de Navarra‐IDISNAUniversidad Pública de NavarraPamplonaSpain
| | - Claudio Luchini
- ARC‐Net Research Centre and Section of Pathology of Department of Diagnostics and Public HealthUniversity and Hospital Trust of VeronaItaly
| | - Juan M. Sánchez‐Hidalgo
- Maimonides Biomedical Research Institute of Córdoba (IMIBIC)Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn)CórdobaSpain
- Surgery ServiceReina Sofia University HospitalCórdobaSpain
| | - Marina E. Sánchez‐Frías
- Maimonides Biomedical Research Institute of Córdoba (IMIBIC)Spain
- Pathology ServiceReina Sofia University HospitalCórdobaSpain
| | - Sergio Pedraza‐Arevalo
- Maimonides Biomedical Research Institute of Córdoba (IMIBIC)Spain
- Department of Cell Biology, Physiology, and ImmunologyUniversity of CórdobaSpain
- Reina Sofia University HospitalCórdobaSpain
| | | | - Rita T. Lawlor
- ARC‐Net Research Centre and Section of Pathology of Department of Diagnostics and Public HealthUniversity and Hospital Trust of VeronaItaly
| | - Antonio Viúdez
- OncobionaTras Lab, Navarrabiomed, Hospital Universitario de Navarra, Instituto de Investigación Sanitaria de Navarra‐IDISNAUniversidad Pública de NavarraPamplonaSpain
- ICON plcPamplonaSpain
| | - Manuel D. Gahete
- Maimonides Biomedical Research Institute of Córdoba (IMIBIC)Spain
- Department of Cell Biology, Physiology, and ImmunologyUniversity of CórdobaSpain
- Reina Sofia University HospitalCórdobaSpain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn)CórdobaSpain
| | - Aldo Scarpa
- ARC‐Net Research Centre and Section of Pathology of Department of Diagnostics and Public HealthUniversity and Hospital Trust of VeronaItaly
| | - Álvaro Arjona‐Sánchez
- Maimonides Biomedical Research Institute of Córdoba (IMIBIC)Spain
- Surgery ServiceReina Sofia University HospitalCórdobaSpain
| | - Raúl M. Luque
- Maimonides Biomedical Research Institute of Córdoba (IMIBIC)Spain
- Department of Cell Biology, Physiology, and ImmunologyUniversity of CórdobaSpain
- Reina Sofia University HospitalCórdobaSpain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn)CórdobaSpain
| | - Alejandro Ibáñez‐Costa
- Maimonides Biomedical Research Institute of Córdoba (IMIBIC)Spain
- Department of Cell Biology, Physiology, and ImmunologyUniversity of CórdobaSpain
- Reina Sofia University HospitalCórdobaSpain
| | - Justo P. Castaño
- Maimonides Biomedical Research Institute of Córdoba (IMIBIC)Spain
- Department of Cell Biology, Physiology, and ImmunologyUniversity of CórdobaSpain
- Reina Sofia University HospitalCórdobaSpain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn)CórdobaSpain
| |
Collapse
|
5
|
Koljonen L, Salonen P, Rusanen S, Mäyränpää MK, Pekkinen M, Mäkitie O. A de novo PRPF8 Pathogenic Variant in Transient Severe Hypophosphatemia with Delayed Puberty and Growth Failure. Horm Res Paediatr 2024:1-11. [PMID: 38976971 DOI: 10.1159/000540249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 07/02/2024] [Indexed: 07/10/2024] Open
Abstract
INTRODUCTION Childhood hypophosphatemia is a rare condition and may be caused by malabsorption, malignancies, or genetic factors. Prolonged hypophosphatemia leads to impaired growth and radiographic signs of rickets. METHODS We performed a detailed clinical and genetic evaluation of an adolescent boy with repeatedly low plasma phosphate concentrations (below 0.60 mmol/L) and growth failure. RESULTS At 14 years, the patient presented with decelerating growth and delayed puberty. Biochemistry showed hypophosphatemia due to increased urinary phosphate loss; kidney function and vitamin D status were normal. Radiographs showed mild metaphyseal changes. A gene panel for known genetic hypophosphatemia was negative. Trio exome analysis followed by Sanger sequencing identified a pathogenic heterozygous de novo stop-gain variant in PRPF8 gene, c.5548C>T p.(Arg1850*), in the conserved RNase H homology domain. PRPF8 encodes the pre-RNA protein 8, which has a role in RNA processing. Heterozygous PRPF8 variants have been associated with retinitis pigmentosa and neurodevelopmental disorders but not with phosphate metabolism. The patient underwent growth hormone (GH) stimulation tests which confirmed GH deficiency. Head MRI indicated partially empty sella. GH treatment was started at 15 years. Surprisingly, phosphate metabolism normalized during GH treatment, suggesting that hypophosphatemia was at least partly secondary to GH deficiency. CONCLUSION The evaluation of an adolescent with profound long-term hypophosphatemia revealed a pituitary developmental defect associated with a stop-gain variant in PRPF8. Hypophosphatemia alleviated with GH treatment. The pathological PRPF8 variant may contribute to abnormal pituitary development; however, its role in phosphate metabolism remains uncertain.
Collapse
Affiliation(s)
- Laura Koljonen
- Folkhälsan Research Center, Helsinki, Finland
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Pia Salonen
- Department of Pediatrics, Central Hospital of Päijät-Häme, Lahti, Finland
| | | | - Mervi K Mäyränpää
- Department of Radiology, HUS Diagnostic Center, Children's Hospital, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Minna Pekkinen
- Folkhälsan Research Center, Helsinki, Finland
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Pediatric Research Center, Children's Hospital, University of Helsinki, and Helsinki University Hospital, Helsinki, Finland
| | - Outi Mäkitie
- Folkhälsan Research Center, Helsinki, Finland
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Pediatric Research Center, Children's Hospital, University of Helsinki, and Helsinki University Hospital, Helsinki, Finland
- Department of Molecular Medicine and Surgery, Karolinska Institutet, and Clinical Genetics, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
6
|
Kurzawa-Akanbi M, Tzoumas N, Corral-Serrano JC, Guarascio R, Steel DH, Cheetham ME, Armstrong L, Lako M. Pluripotent stem cell-derived models of retinal disease: Elucidating pathogenesis, evaluating novel treatments, and estimating toxicity. Prog Retin Eye Res 2024; 100:101248. [PMID: 38369182 DOI: 10.1016/j.preteyeres.2024.101248] [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: 12/07/2023] [Revised: 02/13/2024] [Accepted: 02/14/2024] [Indexed: 02/20/2024]
Abstract
Blindness poses a growing global challenge, with approximately 26% of cases attributed to degenerative retinal diseases. While gene therapy, optogenetic tools, photosensitive switches, and retinal prostheses offer hope for vision restoration, these high-cost therapies will benefit few patients. Understanding retinal diseases is therefore key to advance effective treatments, requiring in vitro models replicating pathology and allowing quantitative assessments for drug discovery. Pluripotent stem cells (PSCs) provide a unique solution given their limitless supply and ability to differentiate into light-responsive retinal tissues encompassing all cell types. This review focuses on the history and current state of photoreceptor and retinal pigment epithelium (RPE) cell generation from PSCs. We explore the applications of this technology in disease modelling, experimental therapy testing, biomarker identification, and toxicity studies. We consider challenges in scalability, standardisation, and reproducibility, and stress the importance of incorporating vasculature and immune cells into retinal organoids. We advocate for high-throughput automation in data acquisition and analyses and underscore the value of advanced micro-physiological systems that fully capture the interactions between the neural retina, RPE, and choriocapillaris.
Collapse
|
7
|
Varineau JE, Calo E. A common cellular response to broad splicing perturbations is characterized by metabolic transcript downregulation driven by the Mdm2-p53 axis. Dis Model Mech 2024; 17:dmm050356. [PMID: 38426258 PMCID: PMC10924232 DOI: 10.1242/dmm.050356] [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: 06/16/2023] [Accepted: 01/09/2024] [Indexed: 03/02/2024] Open
Abstract
Disruptions in core cellular processes elicit stress responses that drive cell-state changes leading to organismal phenotypes. Perturbations in the splicing machinery cause widespread mis-splicing, resulting in p53-dependent cell-state changes that give rise to cell-type-specific phenotypes and disease. However, a unified framework for how cells respond to splicing perturbations, and how this response manifests itself in nuanced disease phenotypes, has yet to be established. Here, we show that a p53-stabilizing Mdm2 alternative splicing event and the resulting widespread downregulation of metabolic transcripts are common events that arise in response to various splicing perturbations in both cellular and organismal models. Together, our results classify a common cellular response to splicing perturbations, put forth a new mechanism behind the cell-type-specific phenotypes that arise when splicing is broadly disrupted, and lend insight into the pleiotropic nature of the effects of p53 stabilization in disease.
Collapse
Affiliation(s)
- Jade E. Varineau
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Eliezer Calo
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| |
Collapse
|
8
|
Cui H, Srinivasan S, Gao Z, Korkin D. The Extent of Edgetic Perturbations in the Human Interactome Caused by Population-Specific Mutations. Biomolecules 2023; 14:40. [PMID: 38254640 PMCID: PMC11154503 DOI: 10.3390/biom14010040] [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: 08/11/2023] [Revised: 11/30/2023] [Accepted: 12/03/2023] [Indexed: 01/24/2024] Open
Abstract
Until recently, efforts in population genetics have been focused primarily on people of European ancestry. To attenuate this bias, global population studies, such as the 1000 Genomes Project, have revealed differences in genetic variation across ethnic groups. How many of these differences can be attributed to population-specific traits? To answer this question, the mutation data must be linked with functional outcomes. A new "edgotype" concept has been proposed, which emphasizes the interaction-specific, "edgetic", perturbations caused by mutations in the interacting proteins. In this work, we performed systematic in silico edgetic profiling of ~50,000 non-synonymous SNVs (nsSNVs) from the 1000 Genomes Project by leveraging our semi-supervised learning approach SNP-IN tool on a comprehensive set of over 10,000 protein interaction complexes. We interrogated the functional roles of the variants and their impact on the human interactome and compared the results with the pathogenic variants disrupting PPIs in the same interactome. Our results demonstrated that a considerable number of nsSNVs from healthy populations could rewire the interactome. We also showed that the proteins enriched with interaction-disrupting mutations were associated with diverse functions and had implications in a broad spectrum of diseases. Further analysis indicated that distinct gene edgetic profiles among major populations could shed light on the molecular mechanisms behind the population phenotypic variances. Finally, the network analysis revealed that the disease-associated modules surprisingly harbored a higher density of interaction-disrupting mutations from healthy populations. The variation in the cumulative network damage within these modules could potentially account for the observed disparities in disease susceptibility, which are distinctly specific to certain populations. Our work demonstrates the feasibility of a large-scale in silico edgetic study, and reveals insights into the orchestrated play of population-specific mutations in the human interactome.
Collapse
Affiliation(s)
- Hongzhu Cui
- Bioinformatics and Computational Biology Program, Worcester Polytechnic Institute, Worcester, MA 01609, USA;
- Chromatography and Mass Spectrometry Division, Thermo Fisher Scientific, San Jose, CA 95134, USA
| | - Suhas Srinivasan
- Data Science Program, Worcester Polytechnic Institute, Worcester, MA 01609, USA;
- Program in Epithelial Biology, Stanford School of Medicine, Stanford, CA 94305, USA
- Center for Personal Dynamic Regulomes, Stanford School of Medicine, Stanford, CA 94305, USA
| | - Ziyang Gao
- Bioinformatics and Computational Biology Program, Worcester Polytechnic Institute, Worcester, MA 01609, USA;
| | - Dmitry Korkin
- Bioinformatics and Computational Biology Program, Worcester Polytechnic Institute, Worcester, MA 01609, USA;
- Data Science Program, Worcester Polytechnic Institute, Worcester, MA 01609, USA;
- Computer Science Department, Worcester Polytechnic Institute, Worcester, MA 01609, USA
| |
Collapse
|
9
|
Liang Y, Sun X, Duan C, Tang S, Chen J. Application of patient-derived induced pluripotent stem cells and organoids in inherited retinal diseases. Stem Cell Res Ther 2023; 14:340. [PMID: 38012786 PMCID: PMC10683306 DOI: 10.1186/s13287-023-03564-5] [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: 08/29/2023] [Accepted: 11/06/2023] [Indexed: 11/29/2023] Open
Abstract
Inherited retinal diseases (IRDs) can induce severe sight-threatening retinal degeneration and impose a considerable economic burden on patients and society, making efforts to cure blindness imperative. Transgenic animals mimicking human genetic diseases have long been used as a primary research tool to decipher the underlying pathogenesis, but there are still some obvious limitations. As an alternative strategy, patient-derived induced pluripotent stem cells (iPSCs), particularly three-dimensional (3D) organoid technology, are considered a promising platform for modeling different forms of IRDs, including retinitis pigmentosa, Leber congenital amaurosis, X-linked recessive retinoschisis, Batten disease, achromatopsia, and best vitelliform macular dystrophy. Here, this paper focuses on the status of patient-derived iPSCs and organoids in IRDs in recent years concerning disease modeling and therapeutic exploration, along with potential challenges for translating laboratory research to clinical application. Finally, the importance of human iPSCs and organoids in combination with emerging technologies such as multi-omics integration analysis, 3D bioprinting, or microfluidic chip platform are highlighted. Patient-derived retinal organoids may be a preferred choice for more accurately uncovering the mechanisms of human retinal diseases and will contribute to clinical practice.
Collapse
Affiliation(s)
- Yuqin Liang
- Aier Eye Institute, Changsha, 410015, China
- Eye Center of Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Xihao Sun
- Aier Eye Institute, Changsha, 410015, China
- Eye Center of Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Chunwen Duan
- Aier Eye Institute, Changsha, 410015, China
- Changsha Aier Eye Hospital, Changsha, 410015, China
| | - Shibo Tang
- Aier Eye Institute, Changsha, 410015, China.
- Changsha Aier Eye Hospital, Changsha, 410015, China.
| | - Jiansu Chen
- Aier Eye Institute, Changsha, 410015, China.
- Changsha Aier Eye Hospital, Changsha, 410015, China.
- Key Laboratory for Regenerative Medicine, Ministry of Education, Jinan University, Guangzhou, 510632, China.
| |
Collapse
|
10
|
Machado HE, Øbro NF, Williams N, Tan S, Boukerrou AZ, Davies M, Belmonte M, Mitchell E, Baxter EJ, Mende N, Clay A, Ancliff P, Köglmeier J, Killick SB, Kulasekararaj A, Meyer S, Laurenti E, Campbell PJ, Kent DG, Nangalia J, Warren AJ. Convergent somatic evolution commences in utero in a germline ribosomopathy. Nat Commun 2023; 14:5092. [PMID: 37608017 PMCID: PMC10444798 DOI: 10.1038/s41467-023-40896-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Accepted: 08/14/2023] [Indexed: 08/24/2023] Open
Abstract
Clonal tracking of cells using somatic mutations permits exploration of clonal dynamics in human disease. Here, we perform whole genome sequencing of 323 haematopoietic colonies from 10 individuals with the inherited ribosomopathy Shwachman-Diamond syndrome to reconstruct haematopoietic phylogenies. In ~30% of colonies, we identify mutually exclusive mutations in TP53, EIF6, RPL5, RPL22, PRPF8, plus chromosome 7 and 15 aberrations that increase SBDS and EFL1 gene dosage, respectively. Target gene mutations commence in utero, resulting in a profusion of clonal expansions, with only a few haematopoietic stem cell lineages (mean 8, range 1-24) contributing ~50% of haematopoietic colonies across 8 individuals (range 4-100% clonality) by young adulthood. Rapid clonal expansion during disease transformation is associated with biallelic TP53 mutations and increased mutation burden. Our study highlights how convergent somatic mutation of the p53-dependent nucleolar surveillance pathway offsets the deleterious effects of germline ribosomopathy but increases opportunity for TP53-mutated cancer evolution.
Collapse
Affiliation(s)
| | - Nina F Øbro
- Wellcome MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
- Department of Haematology, University of Cambridge, Cambridge, UK
- Department of Clinical Immunology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | | | - Shengjiang Tan
- Wellcome MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
- Department of Haematology, University of Cambridge, Cambridge, UK
- Cambridge Institute for Medical Research, Keith Peters Building, Cambridge, UK
| | - Ahmed Z Boukerrou
- Wellcome MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
- Department of Haematology, University of Cambridge, Cambridge, UK
- Cambridge Institute for Medical Research, Keith Peters Building, Cambridge, UK
| | - Megan Davies
- Wellcome MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
- Department of Haematology, University of Cambridge, Cambridge, UK
| | - Miriam Belmonte
- Wellcome MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
- Department of Haematology, University of Cambridge, Cambridge, UK
| | - Emily Mitchell
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
- Wellcome MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
| | - E Joanna Baxter
- Department of Haematology, University of Cambridge, Cambridge, UK
| | - Nicole Mende
- Wellcome MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
- Department of Haematology, University of Cambridge, Cambridge, UK
| | - Anna Clay
- Wellcome MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
- Department of Haematology, University of Cambridge, Cambridge, UK
| | - Philip Ancliff
- Department of Haematology, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Jutta Köglmeier
- Department of Haematology, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Sally B Killick
- University Hospitals Dorset NHS Foundation Trust, The Royal Bournemouth Hospital, Bournemouth, UK
| | - Austin Kulasekararaj
- Department of Haematological Medicine, King's College Hospital NHS Foundation Trust and King's College London, London, UK
| | - Stefan Meyer
- Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Cancer Research Centre, Wilmslow Road, Manchester, UK
- Department of Paediatric Haematology and Oncology, Royal Manchester Children's Hospital, Manchester Foundation Trust, Manchester, Oxford Road, Manchester, UK
- Teenage and Adolescent Oncology, The Christie NHS Foundation Trust, Wilmslow Road, Manchester, UK
| | - Elisa Laurenti
- Wellcome MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
- Department of Haematology, University of Cambridge, Cambridge, UK
| | | | - David G Kent
- Wellcome MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK.
- Department of Haematology, University of Cambridge, Cambridge, UK.
- York Biomedical Research Institute, Department of Biology, University of York, York, UK.
| | - Jyoti Nangalia
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK.
- Wellcome MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK.
- Department of Haematology, University of Cambridge, Cambridge, UK.
| | - Alan J Warren
- Wellcome MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK.
- Department of Haematology, University of Cambridge, Cambridge, UK.
- Cambridge Institute for Medical Research, Keith Peters Building, Cambridge, UK.
| |
Collapse
|
11
|
Modeling PRPF31 retinitis pigmentosa using retinal pigment epithelium and organoids combined with gene augmentation rescue. NPJ Regen Med 2022; 7:39. [PMID: 35974011 PMCID: PMC9381579 DOI: 10.1038/s41536-022-00235-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 07/20/2022] [Indexed: 11/17/2022] Open
Abstract
Mutations in the ubiquitously expressed pre-mRNA processing factor (PRPF) 31 gene, one of the most common causes of dominant form of Retinitis Pigmentosa (RP), lead to a retina-specific phenotype. It is uncertain which retinal cell types are affected and animal models do not clearly present the RP phenotype observed in PRPF31 patients. Retinal organoids and retinal pigment epithelial (RPE) cells derived from human-induced pluripotent stem cells (iPSCs) provide potential opportunities for studying human PRPF31-related RP. We demonstrate here that RPE cells carrying PRPF31 mutations present important morphological and functional changes and that PRPF31-mutated retinal organoids recapitulate the human RP phenotype, with a rod photoreceptor cell death followed by a loss of cones. The low level of PRPF31 expression may explain the defective phenotypes of PRPF31-mutated RPE and photoreceptor cells, which were not observed in cells derived from asymptomatic patients or after correction of the pathogenic mutation by CRISPR/Cas9. Transcriptome profiles revealed differentially expressed and mis-spliced genes belonging to pathways in line with the observed defective phenotypes. The rescue of RPE and photoreceptor defective phenotypes by PRPF31 gene augmentation provide the proof of concept for future therapeutic strategies.
Collapse
|
12
|
Liang Y, Tan F, Sun X, Cui Z, Gu J, Mao S, Chan HF, Tang S, Chen J. Aberrant Retinal Pigment Epithelial Cells Derived from Induced Pluripotent Stem Cells of a Retinitis Pigmentosa Patient with the PRPF6 Mutation. Int J Mol Sci 2022; 23:ijms23169049. [PMID: 36012314 PMCID: PMC9409096 DOI: 10.3390/ijms23169049] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 08/08/2022] [Accepted: 08/11/2022] [Indexed: 11/16/2022] Open
Abstract
Pre-mRNA processing factors (PRPFs) are vital components of the spliceosome and are involved in the physiological process necessary for pre-mRNA splicing to mature mRNA. As an important member, PRPF6 mutation resulting in autosomal dominant retinitis pigmentosa (adRP) is not common. Recently, we reported the establishment of an induced pluripotent stem cells (iPSCs; CSUASOi004-A) model by reprogramming the peripheral blood mononuclear cells of a PRPF6-related adRP patient, which could recapitulate a consistent disease-specific genotype. In this study, a disease model of retinal pigment epithelial (RPE) cells was generated from the iPSCs of this patient to further investigate the underlying molecular and pathological mechanisms. The results showed the irregular morphology, disorganized apical microvilli and reduced expressions of RPE-specific genes in the patient’s iPSC-derived RPE cells. In addition, RPE cells carrying the PRPF6 mutation displayed a decrease in the phagocytosis of fluorescein isothiocyanate-labeled photoreceptor outer segments and exhibited impaired cell polarity and barrier function. This study will benefit the understanding of PRPF6-related RPE cells and future cell therapy.
Collapse
Affiliation(s)
- Yuqin Liang
- Aier School of Ophthalmology, Central South University, Changsha 410015, China
- Aier Eye Institute, Changsha 410015, China
| | - Feng Tan
- Aier School of Ophthalmology, Central South University, Changsha 410015, China
- Aier Eye Institute, Changsha 410015, China
| | - Xihao Sun
- Aier School of Ophthalmology, Central South University, Changsha 410015, China
- Aier Eye Institute, Changsha 410015, China
| | - Zekai Cui
- Aier School of Ophthalmology, Central South University, Changsha 410015, China
- Aier Eye Institute, Changsha 410015, China
| | - Jianing Gu
- Aier Eye Institute, Changsha 410015, China
| | | | - Hon Fai Chan
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong 999077, China
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong 999077, China
| | - Shibo Tang
- Aier School of Ophthalmology, Central South University, Changsha 410015, China
- Aier Eye Institute, Changsha 410015, China
- Correspondence: (S.T.); (J.C.); Tel.: +86-139-2510-0123 (S.T.); +86-186-7583-9029 (J.C.)
| | - Jiansu Chen
- Aier School of Ophthalmology, Central South University, Changsha 410015, China
- Aier Eye Institute, Changsha 410015, China
- Key Laboratory for Regenerative Medicine, Ministry of Education, Jinan University, Guangzhou 510632, China
- Correspondence: (S.T.); (J.C.); Tel.: +86-139-2510-0123 (S.T.); +86-186-7583-9029 (J.C.)
| |
Collapse
|
13
|
The Clinical Role of SRSF1 Expression in Cancer: A Review of the Current Literature. APPLIED SCIENCES-BASEL 2022. [DOI: 10.3390/app12052268] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Background: SFRS1 is a member of the splicing factor protein family. Through a specific sequence of alteration, SRSF1 can move from the cytoplasm to the nucleus where it can work autonomously as a splicing activator, or as a silencer when interacting with other regulators. Alternative splicing (AS) is a fundamental biological process that ensures protein diversity. In fact, different proteins, produced by alternative splicing, can gain different and even antagonistic biological functions. Methods: Our review is based on English articles published in the MEDLINE/PubMed medical library between 2000 and 2021. We retrieved articles that were specifically related to SRSF1 and cancers, and we excluded other reviews and meta-analyses. We included in vitro studies, animal studies and clinical studies, evaluated using the Medical Education Research Study Quality Instrument (MERSQI) and the Newcastle–Ottawa Scale-Education (NOSE). Result: SRSF1 is related to various genes and plays a role in cell cycle, ubiquitin-mediated proteolysis, nucleotide excision repair, p53 pathway, apoptosis, DNA replication and RNA degradation. In most cases, SRSF1 carries out its cancer-related function via abnormal alternative splicing (AS). However, according to the most recent literature, SRSF1 may also be involved in mRNA translation and cancer chemoresistance or radio-sensitivity. Conclusion: Our results showed that SRSF1 plays a key clinical role in tumorigenesis and tumor progression in several types of cancer (such as Prostate, Lung, Breast, Colon, Glioblastoma), through various mechanisms of action and different cellular pathways. This review could be a starting point for several studies regarding the biology of and therapies for cancer.
Collapse
|