1
|
Schukarucha Gomes A, Ellis CE, Spigelman AF, Dos Santos T, Maghera J, Suzuki K, MacDonald PE. Molecular correlates of glycine receptor activity in human β cells. Mol Metab 2025; 96:102156. [PMID: 40258441 DOI: 10.1016/j.molmet.2025.102156] [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: 12/03/2024] [Revised: 04/07/2025] [Accepted: 04/16/2025] [Indexed: 04/23/2025] Open
Abstract
OBJECTIVES Glycine acts in an autocrine positive feedback loop in human β cells through its ionotropic receptors (GlyRs). In type 2 diabetes (T2D), islet GlyR activity is impaired by unknown mechanisms. We sought to investigate if the GlyR dysfunction in T2D is replicated by hyperglycemia per se, and to further characterize its action in β cells and islets. METHODS GlyR-mediated currents were measured using whole-cell patch-clamp in human β cells from donors with or without T2D, or after high glucose (15 mM) culture. We also correlated glycine-induced current amplitude with transcript expression levels through patch-seq. The expression of the GlyR α1, α3, and β subunit mRNA splice variants was compared between islets from donors with and without T2D, and after high glucose culture. Insulin secretion from human islets was measured in the presence or absence of the GlyR antagonist strychnine. RESULTS Although gene expression of GlyRs was decreased in T2D islets, and β cell GlyR-mediated currents were smaller, we found no evidence for a shift in GlyR subunit splicing. Glycine-induced currents are also reduced after 48 h culture of islets from donors without diabetes in high glucose, where we also find the reduction of the α1 subunit expression, but an increase in the α3 subunit. We discovered that glycine-evoked currents are highly heterogeneous amongst β cells, inversely correlate with donor HbA1c, and are significantly correlated to the expression of 92 different transcripts and gene regulatory networks (GRNs) that include CREB3(+), RREB1(+) and ZNF697(+). Finally, glucose-stimulated insulin secretion is decreased in the presence of the GlyR antagonist strychnine. CONCLUSIONS We demonstrate that glucose can modulate GlyR expression, and that the current decrease in T2D is likely due to the receptor gene expression downregulation, and not a change in transcript splicing. Moreover, we define a previously unknown set of genes and regulons that are correlated to GlyR-mediated currents and could be involved in GlyR downregulation in T2D. Among those we validate the negative impact of EIF4EBP1 expression on GlyR activity.
Collapse
Affiliation(s)
- Amanda Schukarucha Gomes
- Alberta Diabetes Institute, Department of Pharmacology, University of Alberta, Edmonton, Alberta, T6G 2R3, Canada
| | - Cara E Ellis
- Alberta Diabetes Institute, Department of Pharmacology, University of Alberta, Edmonton, Alberta, T6G 2R3, Canada
| | - Aliya F Spigelman
- Alberta Diabetes Institute, Department of Pharmacology, University of Alberta, Edmonton, Alberta, T6G 2R3, Canada
| | - Theodore Dos Santos
- Alberta Diabetes Institute, Department of Pharmacology, University of Alberta, Edmonton, Alberta, T6G 2R3, Canada
| | - Jasmine Maghera
- Alberta Diabetes Institute, Department of Pharmacology, University of Alberta, Edmonton, Alberta, T6G 2R3, Canada
| | - Kunimasa Suzuki
- Alberta Diabetes Institute, Department of Pharmacology, University of Alberta, Edmonton, Alberta, T6G 2R3, Canada
| | - Patrick E MacDonald
- Alberta Diabetes Institute, Department of Pharmacology, University of Alberta, Edmonton, Alberta, T6G 2R3, Canada.
| |
Collapse
|
2
|
Chao KH, Mao A, Liu A, Salzberg SL, Pertea M. OpenSpliceAI: An efficient, modular implementation of SpliceAI enabling easy retraining on non-human species. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.20.644351. [PMID: 40166201 PMCID: PMC11957165 DOI: 10.1101/2025.03.20.644351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
The SpliceAI deep learning system is currently one of the most accurate methods for identifying splicing signals directly from DNA sequences. However, its utility is limited by its reliance on older software frameworks and human-centric training data. Here we introduce OpenSpliceAI, a trainable, open-source version of SpliceAI implemented in PyTorch to address these challenges. OpenSpliceAI supports both training from scratch and transfer learning, enabling seamless re-training on species-specific datasets and mitigating human-centric biases. Our experiments show that it achieves faster processing speeds and lower memory usage than the original SpliceAI code, allowing large-scale analyses of extensive genomic regions on a single GPU. Additionally, OpenSpliceAI's flexible architecture makes for easier integration with established machine learning ecosystems, simplifying the development of custom splicing models for different species and applications. We demonstrate that OpenSpliceAI's output is highly concordant with SpliceAI. In silico mutagenesis (ISM) analyses confirm that both models rely on similar sequence features, and calibration experiments demonstrate similar score probability estimates.
Collapse
Affiliation(s)
- Kuan-Hao Chao
- Department of Computer Science, Johns Hopkins University, Baltimore, MD 21218, USA
- Center for Computational Biology, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Alan Mao
- Department of Computer Science, Johns Hopkins University, Baltimore, MD 21218, USA
- Center for Computational Biology, Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Anqi Liu
- Department of Computer Science, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Steven L Salzberg
- Department of Computer Science, Johns Hopkins University, Baltimore, MD 21218, USA
- Center for Computational Biology, Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Biostatistics, Johns Hopkins University, Baltimore, MD 21211, USA
| | - Mihaela Pertea
- Department of Computer Science, Johns Hopkins University, Baltimore, MD 21218, USA
- Center for Computational Biology, Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| |
Collapse
|
3
|
Ojeda-Rodriguez A, Torres-Peña JD, Arenas-de Larriva AP, Rangel-Zuñiga OA, Podadera-Herreros A, Boughanem H, G-García ME, López-Moreno A, Katsiki N, Luque RM, Perez-Martinez P, Delgado-Lista J, Yubero-Serrano EM, Lopez-Miranda J. Differences in splicing factors may predict type 2 diabetes remission in the CORDIOPREV study. iScience 2025; 28:111527. [PMID: 39811651 PMCID: PMC11731613 DOI: 10.1016/j.isci.2024.111527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 10/22/2024] [Accepted: 12/02/2024] [Indexed: 01/16/2025] Open
Abstract
Alternative splicing is a post-transcriptional process resulting in multiple protein isoforms from a single gene. Abnormal splicing may lead to metabolic diseases, including type 2 diabetes mellitus (T2DM). To identify the splicing factor expression that predicts T2DM remission in coronary heart disease (CHD) patients, we identified newly diagnosed T2DM at baseline (n = 190) from the CORDIOPREV study. Patients were classified as Responders (T2DM remission during 5 years without antidiabetic drugs) or non-Responders. Baseline dysregulation in 5 splicing factors (MBNL1, RBM5, hnRNP G/RBMX, CD44, NT5E) distinguished Responders from non-Responders. Adding these factors to clinical variables [AUC = 0.67], insulin resistance, and beta-cell indexes [AUC = 0.76], improved T2DM remission prediction [AUC = 0.80]. Cox regression analysis showed those with higher remission scores had a 2.63-fold increased remission probability. To conclude, a set of splicing factors that contribute to predicting T2DM remission in patients with CHD has been identified. Further research is needed to elucidate these findings' clinical relevance.
Collapse
Affiliation(s)
- Ana Ojeda-Rodriguez
- Lipids and Atherosclerosis Unit, Internal Medicine Unit, Reina Sofia University Hospital, 14004 Cordoba, Spain
- Department of Medical and Surgical Science, University of Cordoba, 14004 Córdoba, Spain
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Av. Menendez Pidal, S/n, 14004 Cordoba, Spain
- CIBER Fisiopatologia de La Obesidad y Nutricion (CIBEROBN), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Jose D. Torres-Peña
- Lipids and Atherosclerosis Unit, Internal Medicine Unit, Reina Sofia University Hospital, 14004 Cordoba, Spain
- Department of Medical and Surgical Science, University of Cordoba, 14004 Córdoba, Spain
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Av. Menendez Pidal, S/n, 14004 Cordoba, Spain
- CIBER Fisiopatologia de La Obesidad y Nutricion (CIBEROBN), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Antonio Pablo Arenas-de Larriva
- Lipids and Atherosclerosis Unit, Internal Medicine Unit, Reina Sofia University Hospital, 14004 Cordoba, Spain
- Department of Medical and Surgical Science, University of Cordoba, 14004 Córdoba, Spain
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Av. Menendez Pidal, S/n, 14004 Cordoba, Spain
- CIBER Fisiopatologia de La Obesidad y Nutricion (CIBEROBN), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Oriol Alberto Rangel-Zuñiga
- Lipids and Atherosclerosis Unit, Internal Medicine Unit, Reina Sofia University Hospital, 14004 Cordoba, Spain
- Department of Medical and Surgical Science, University of Cordoba, 14004 Córdoba, Spain
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Av. Menendez Pidal, S/n, 14004 Cordoba, Spain
- CIBER Fisiopatologia de La Obesidad y Nutricion (CIBEROBN), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Alicia Podadera-Herreros
- Lipids and Atherosclerosis Unit, Internal Medicine Unit, Reina Sofia University Hospital, 14004 Cordoba, Spain
- Department of Medical and Surgical Science, University of Cordoba, 14004 Córdoba, Spain
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Av. Menendez Pidal, S/n, 14004 Cordoba, Spain
- CIBER Fisiopatologia de La Obesidad y Nutricion (CIBEROBN), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Hatim Boughanem
- Lipids and Atherosclerosis Unit, Internal Medicine Unit, Reina Sofia University Hospital, 14004 Cordoba, Spain
- Department of Medical and Surgical Science, University of Cordoba, 14004 Córdoba, Spain
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Av. Menendez Pidal, S/n, 14004 Cordoba, Spain
- CIBER Fisiopatologia de La Obesidad y Nutricion (CIBEROBN), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Miguel E. G-García
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Av. Menendez Pidal, S/n, 14004 Cordoba, Spain
- CIBER Fisiopatologia de La Obesidad y Nutricion (CIBEROBN), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Department of Cell Biology, Physiology and Immunology, University of Cordoba, 14004 Cordoba, Spain
| | - Alejandro López-Moreno
- Lipids and Atherosclerosis Unit, Internal Medicine Unit, Reina Sofia University Hospital, 14004 Cordoba, Spain
- Department of Medical and Surgical Science, University of Cordoba, 14004 Córdoba, Spain
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Av. Menendez Pidal, S/n, 14004 Cordoba, Spain
- CIBER Fisiopatologia de La Obesidad y Nutricion (CIBEROBN), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Niki Katsiki
- Department of Nutritional Sciences and Dietetics, International Hellenic University, 57400 Thessaloniki, Greece
- School of Medicine, European University Cyprus, 2404 Nicosia, Cyprus, Greece
| | - Raul M. Luque
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Av. Menendez Pidal, S/n, 14004 Cordoba, Spain
- CIBER Fisiopatologia de La Obesidad y Nutricion (CIBEROBN), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Department of Cell Biology, Physiology and Immunology, University of Cordoba, 14004 Cordoba, Spain
| | - Pablo Perez-Martinez
- Lipids and Atherosclerosis Unit, Internal Medicine Unit, Reina Sofia University Hospital, 14004 Cordoba, Spain
- Department of Medical and Surgical Science, University of Cordoba, 14004 Córdoba, Spain
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Av. Menendez Pidal, S/n, 14004 Cordoba, Spain
- CIBER Fisiopatologia de La Obesidad y Nutricion (CIBEROBN), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Javier Delgado-Lista
- Lipids and Atherosclerosis Unit, Internal Medicine Unit, Reina Sofia University Hospital, 14004 Cordoba, Spain
- Department of Medical and Surgical Science, University of Cordoba, 14004 Córdoba, Spain
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Av. Menendez Pidal, S/n, 14004 Cordoba, Spain
- CIBER Fisiopatologia de La Obesidad y Nutricion (CIBEROBN), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Elena M. Yubero-Serrano
- Lipids and Atherosclerosis Unit, Internal Medicine Unit, Reina Sofia University Hospital, 14004 Cordoba, Spain
- Department of Medical and Surgical Science, University of Cordoba, 14004 Córdoba, Spain
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Av. Menendez Pidal, S/n, 14004 Cordoba, Spain
- CIBER Fisiopatologia de La Obesidad y Nutricion (CIBEROBN), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Department of Food and Health, Instituto de La Grasa, Spanish National Research Council (CSIC), 41013 Seville, Spain
| | - Jose Lopez-Miranda
- Lipids and Atherosclerosis Unit, Internal Medicine Unit, Reina Sofia University Hospital, 14004 Cordoba, Spain
- Department of Medical and Surgical Science, University of Cordoba, 14004 Córdoba, Spain
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Av. Menendez Pidal, S/n, 14004 Cordoba, Spain
- CIBER Fisiopatologia de La Obesidad y Nutricion (CIBEROBN), Instituto de Salud Carlos III, 28029 Madrid, Spain
| |
Collapse
|
4
|
Dall'Agnese A, Zheng MM, Moreno S, Platt JM, Hoang AT, Kannan D, Dall'Agnese G, Overholt KJ, Sagi I, Hannett NM, Erb H, Corradin O, Chakraborty AK, Lee TI, Young RA. Proteolethargy is a pathogenic mechanism in chronic disease. Cell 2025; 188:207-221.e30. [PMID: 39610243 PMCID: PMC11724756 DOI: 10.1016/j.cell.2024.10.051] [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: 03/16/2024] [Revised: 08/07/2024] [Accepted: 10/31/2024] [Indexed: 11/30/2024]
Abstract
The pathogenic mechanisms of many diseases are well understood at the molecular level, but there are prevalent syndromes associated with pathogenic signaling, such as diabetes and chronic inflammation, where our understanding is more limited. Here, we report that pathogenic signaling suppresses the mobility of a spectrum of proteins that play essential roles in cellular functions known to be dysregulated in these chronic diseases. The reduced protein mobility, which we call proteolethargy, was linked to cysteine residues in the affected proteins and signaling-related increases in excess reactive oxygen species. Diverse pathogenic stimuli, including hyperglycemia, dyslipidemia, and inflammation, produce similar reduced protein mobility phenotypes. We propose that proteolethargy is an overlooked cellular mechanism that may account for various pathogenic features of diverse chronic diseases.
Collapse
Affiliation(s)
| | - Ming M Zheng
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA; Department of Physics, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Shannon Moreno
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA; Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Jesse M Platt
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA; Division of Gastroenterology, Department of Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
| | - An T Hoang
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA
| | - Deepti Kannan
- Department of Physics, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | | | - Kalon J Overholt
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA; Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Ido Sagi
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA
| | - Nancy M Hannett
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA
| | - Hailey Erb
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA
| | - Olivia Corradin
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA; Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Arup K Chakraborty
- Department of Physics, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Institute of Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Cambridge, MA 02139, USA
| | - Tong Ihn Lee
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA.
| | - Richard A Young
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA; Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.
| |
Collapse
|
5
|
Liu L, Zhang S, Xu J, Cao Y, Cui D, Liu C, Shen B, Wu Y, Zhang Q. Mass-spectrometry-based quantitative proteomic analysis reveals that methylglyoxal and carnosine influence oxidative stress and RNA-processing associated proteins in renal proximal tubule epithelial cells. Mol Biol Rep 2025; 52:100. [PMID: 39751919 DOI: 10.1007/s11033-024-10190-8] [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: 09/09/2024] [Accepted: 12/18/2024] [Indexed: 01/04/2025]
Abstract
BACKGROUND Tubular injury triggered by hyperglycemia is an important pathological characteristic in diabetic nephropathy (DN). Accumulated advanced glycation end products and their precursor methylglyoxal (MGO), contribute to the development of DN. Carnosine has been shown to prevent the development of DN but the underlying mechanism still needs to be studied in depth. In this study, we explored the potential proteins influenced by MGO and carnosine in tubule epithelial cells. METHODS AND RESULTS HK-2 cells were treated with MGO, carnosine, or a combination. Differentially expressed proteins (DEPs) between different groups were identified by isobaric tag for relative and absolute quantitation-based mass spectrometry. In the comparison between MGO and control, 29 DEPs were found to be associated with antioxidation and RNA methylation. In the comparison between carnosine and control, 10 DEPs were associated with ubiquitin protein ligase activity and RNA metabolism. In the comparison between MGO + carnosine and MGO, carnosine-induced DEPs in the presence of MGO were mainly related to RNA splicing and mRNA processing. MGO effects on OSTC expression was inversely correlated with that of carnosine. Some DEPs (OSTC, PRDX5, NEDD4L, NOP2, TRMT6, and GEMIN2) were validated by Western blotting. Additional experiments showed the 28 kD particle of Smith antigen was also influenced by MGO and carnosine. CONCLUSIONS Carnosine can influence RNA processing and spliceosome-related proteins, and change MGO's effect on HK-2 cells. This study helps to understand the mechanism by which MGO contributes to the development of DN and promotes further identification of carnosine downstream proteins as therapeutic targets for DN.
Collapse
Affiliation(s)
- Lei Liu
- Department of Endocrinology, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230022, People's Republic of China
| | - Shiqi Zhang
- Department of Endocrinology, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230022, People's Republic of China
| | - Juan Xu
- Department of Endocrinology, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230022, People's Republic of China
| | - Yadi Cao
- Department of Endocrinology, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230022, People's Republic of China
| | - Di Cui
- Department of Endocrinology, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230022, People's Republic of China
| | - Chao Liu
- Department of Endocrinology, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230022, People's Republic of China
| | - Bing Shen
- School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui, 230032, People's Republic of China
| | - Yonggui Wu
- Department of Nephrology, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230022, People's Republic of China.
| | - Qiu Zhang
- Department of Endocrinology, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230022, People's Republic of China.
| |
Collapse
|
6
|
Chimienti R, Torchio S, Siracusano G, Zamarian V, Monaco L, Lombardo MT, Pellegrini S, Manenti F, Cuozzo F, Rossi G, Carrera P, Sordi V, Broccoli V, Bonfanti R, Casari G, Frontino G, Piemonti L. A WFS1 variant disrupting acceptor splice site uncovers the impact of alternative splicing on beta cell apoptosis in a patient with Wolfram syndrome. Diabetologia 2025; 68:128-151. [PMID: 39520565 PMCID: PMC11663190 DOI: 10.1007/s00125-024-06307-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Accepted: 08/21/2024] [Indexed: 11/16/2024]
Abstract
AIMS/HYPOTHESIS Wolfram syndrome 1 (WS1) is an inherited condition mainly manifesting in childhood-onset diabetes mellitus and progressive optic nerve atrophy. The causative gene, WFS1, encodes wolframin, a master regulator of several cellular responses, and the gene's mutations associate with clinical variability. Indeed, nonsense/frameshift variants correlate with more severe symptoms than missense/in-frame variants. As achieving a genotype-phenotype correlation is crucial for dealing with disease outcome, works investigating the impact of transcriptional and translational landscapes stemming from such mutations are needed. Therefore, we sought to elucidate the molecular determinants behind the pathophysiological alterations in a WS1 patient carrying compound heterozygous mutations in WFS1: c.316-1G>A, affecting the acceptor splice site (ASS) upstream of exon 4; and c.757A>T, introducing a premature termination codon (PTC) in exon 7. METHODS Bioinformatic analysis was carried out to infer the alternative splicing events occurring after disruption of ASS, followed by RNA-seq and PCR to validate the transcriptional landscape. Patient-derived induced pluripotent stem cells (iPSCs) were used as an in vitro model of WS1 and to investigate the WFS1 alternative splicing isoforms in pancreatic beta cells. CRISPR/Cas9 technology was employed to correct ASS mutation and generate a syngeneic control for the endoplasmic reticulum stress induction and immunotoxicity assays. RESULTS We showed that patient-derived iPSCs retained the ability to differentiate into pancreatic beta cells. We demonstrated that the allele carrying the ASS mutation c.316-1G>A originates two PTC-containing alternative splicing transcripts (c.316del and c.316-460del), and two open reading frame-conserving mRNAs (c.271-513del and c.316-456del) leading to N-terminally truncated polypeptides. By retaining the C-terminal domain, these isoforms sustained the endoplasmic reticulum stress response in beta cells. Otherwise, PTC-carrying transcripts were regulated by the nonsense-mediated decay (NMD) in basal conditions. Exposure to cell stress inducers and proinflammatory cytokines affected expression levels of the NMD-related gene SMG7 (>twofold decrease; p<0.001) without eliciting a robust unfolded protein response in WFS1 beta cells. This resulted in a dramatic accumulation of the PTC-containing isoforms c.316del (>100-fold increase over basal; p<0.001) and c.316-460del (>20-fold increase over basal; p<0.001), predisposing affected beta cells to undergo apoptosis. Cas9-mediated recovery of ASS retrieved the canonical transcriptional landscape, rescuing the normal phenotype in patient-derived beta cells. CONCLUSIONS/INTERPRETATION This study represents a new model to study wolframin, highlighting how each single mutation of the WFS1 gene can determine dramatically different functional outcomes. Our data point to increased vulnerability of WFS1 beta cells to stress and inflammation and we postulate that this is triggered by escaping NMD and accumulation of mutated transcripts and truncated proteins. These findings pave the way for further studies on the molecular basis of genotype-phenotype relationship in WS1, to uncover the key determinants that might be targeted to ameliorate the clinical outcome of patients affected by this rare disease. DATA AVAILABILITY The in silico predicted N-terminal domain structure file of WT wolframin was deposited in the ModelArchive, together with procedures, ramachandran plots, inter-residue distance deviation and IDDT scores, and Gromacs configuration files (doi/10.5452/ma-cg3qd). The deep-sequencing data as fastq files used to generate consensus sequences of AS isoforms of WFS1 are available in the SRA database (BioProject PRJNA1109747).
Collapse
Affiliation(s)
- Raniero Chimienti
- Unit of β Cell Biology, Diabetes Research Institute, IRCCS San Raffaele Hospital, Milan, Italy.
- Vita-Salute San Raffaele University, Milan, Italy.
| | - Silvia Torchio
- Unit of β Cell Biology, Diabetes Research Institute, IRCCS San Raffaele Hospital, Milan, Italy.
- Vita-Salute San Raffaele University, Milan, Italy.
- Genetics and Developmental Biology (UMR3215 / U934), Institut Curie, Paris, France.
| | - Gabriel Siracusano
- Unit of β Cell Biology, Diabetes Research Institute, IRCCS San Raffaele Hospital, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Valentina Zamarian
- Unit of β Cell Biology, Diabetes Research Institute, IRCCS San Raffaele Hospital, Milan, Italy
| | - Laura Monaco
- Unit of β Cell Biology, Diabetes Research Institute, IRCCS San Raffaele Hospital, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Marta Tiffany Lombardo
- Unit of β Cell Biology, Diabetes Research Institute, IRCCS San Raffaele Hospital, Milan, Italy
| | - Silvia Pellegrini
- Unit of β Cell Biology, Diabetes Research Institute, IRCCS San Raffaele Hospital, Milan, Italy
| | - Fabio Manenti
- Unit of β Cell Biology, Diabetes Research Institute, IRCCS San Raffaele Hospital, Milan, Italy
| | - Federica Cuozzo
- Unit of β Cell Biology, Diabetes Research Institute, IRCCS San Raffaele Hospital, Milan, Italy
| | - Greta Rossi
- Vita-Salute San Raffaele University, Milan, Italy
- Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Paola Carrera
- Unit of Genomics for Human Disease Diagnosis, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Laboratory of Molecular Genetics, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Valeria Sordi
- Unit of β Cell Biology, Diabetes Research Institute, IRCCS San Raffaele Hospital, Milan, Italy
| | - Vania Broccoli
- Genetics and Developmental Biology (UMR3215 / U934), Institut Curie, Paris, France
- National Research Council of Italy, Institute of Neuroscience, Milan, Italy
| | | | - Giorgio Casari
- Vita-Salute San Raffaele University, Milan, Italy
- Unit of Genome-Phenome Relationship, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Giulio Frontino
- Department of Pediatrics, IRCCS San Raffaele Hospital, Milan, Italy
| | - Lorenzo Piemonti
- Unit of β Cell Biology, Diabetes Research Institute, IRCCS San Raffaele Hospital, Milan, Italy.
- Vita-Salute San Raffaele University, Milan, Italy.
| |
Collapse
|
7
|
Nishida Y, Watada H. A possible new biomarker for T1D? : RNA splicing in circulating blood. J Clin Endocrinol Metab 2024:dgae746. [PMID: 39531648 DOI: 10.1210/clinem/dgae746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 10/15/2024] [Accepted: 11/11/2024] [Indexed: 11/16/2024]
Affiliation(s)
- Yuya Nishida
- Department of Metabolism & Endocrinology, Juntendo University Graduate School of Medicine 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
| | - Hirotaka Watada
- Department of Metabolism & Endocrinology, Juntendo University Graduate School of Medicine 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
| |
Collapse
|
8
|
Wang J, Wen S, Chen M, Xie J, Lou X, Zhao H, Chen Y, Zhao M, Shi G. Regulation of endocrine cell alternative splicing revealed by single-cell RNA sequencing in type 2 diabetes pathogenesis. Commun Biol 2024; 7:778. [PMID: 38937540 PMCID: PMC11211498 DOI: 10.1038/s42003-024-06475-0] [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: 05/25/2023] [Accepted: 06/19/2024] [Indexed: 06/29/2024] Open
Abstract
The prevalent RNA alternative splicing (AS) contributes to molecular diversity, which has been demonstrated in cellular function regulation and disease pathogenesis. However, the contribution of AS in pancreatic islets during diabetes progression remains unclear. Here, we reanalyze the full-length single-cell RNA sequencing data from the deposited database to investigate AS regulation across human pancreatic endocrine cell types in non-diabetic (ND) and type 2 diabetic (T2D) individuals. Our analysis demonstrates the significant association between transcriptomic AS profiles and cell-type-specificity, which could be applied to distinguish the clustering of major endocrine cell types. Moreover, AS profiles are enabled to clearly define the mature subset of β-cells in healthy controls, which is completely lost in T2D. Further analysis reveals that RNA-binding proteins (RBPs), heterogeneous nuclear ribonucleoproteins (hnRNPs) and FXR1 family proteins are predicted to induce the functional impairment of β-cells through regulating AS profiles. Finally, trajectory analysis of endocrine cells suggests the β-cell identity shift through dedifferentiation and transdifferentiation of β-cells during the progression of T2D. Together, our study provides a mechanism for regulating β-cell functions and suggests the significant contribution of AS program during diabetes pathogenesis.
Collapse
Affiliation(s)
- Jin Wang
- Department of Endocrinology & Metabolism, Medical Center for Comprehensive Weight Control, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China.
| | - Shiyi Wen
- Department of Endocrinology & Metabolism, Medical Center for Comprehensive Weight Control, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Minqi Chen
- Key Laboratory of Stem Cells and Tissue Engineering, Zhongshan School of Medicine, Sun Yat-sen University, Ministry of Education, Guangzhou, Guangdong, China
| | - Jiayi Xie
- Key Laboratory of Stem Cells and Tissue Engineering, Zhongshan School of Medicine, Sun Yat-sen University, Ministry of Education, Guangzhou, Guangdong, China
| | - Xinhua Lou
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Haihan Zhao
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yanming Chen
- Department of Endocrinology & Metabolism, Medical Center for Comprehensive Weight Control, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Diabetology & Guangzhou Municipal Key Laboratory of Mechanistic and Translational Obesity Research, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Meng Zhao
- Key Laboratory of Stem Cells and Tissue Engineering, Zhongshan School of Medicine, Sun Yat-sen University, Ministry of Education, Guangzhou, Guangdong, China.
| | - Guojun Shi
- Department of Endocrinology & Metabolism, Medical Center for Comprehensive Weight Control, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China.
- Guangdong Provincial Key Laboratory of Diabetology & Guangzhou Municipal Key Laboratory of Mechanistic and Translational Obesity Research, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China.
- State Key Laboratory of Oncology in Southern China, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China.
| |
Collapse
|
9
|
Zhou AX, Jeansson M, He L, Wigge L, Tonelius P, Tati R, Cederblad L, Muhl L, Uhrbom M, Liu J, Björnson Granqvist A, Lerman LO, Betsholtz C, Hansen PBL. Renal Endothelial Single-Cell Transcriptomics Reveals Spatiotemporal Regulation and Divergent Roles of Differential Gene Transcription and Alternative Splicing in Murine Diabetic Nephropathy. Int J Mol Sci 2024; 25:4320. [PMID: 38673910 PMCID: PMC11050020 DOI: 10.3390/ijms25084320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 04/04/2024] [Accepted: 04/11/2024] [Indexed: 04/28/2024] Open
Abstract
Endothelial cell (EC) injury is a crucial contributor to the progression of diabetic kidney disease (DKD), but the specific EC populations and mechanisms involved remain elusive. Kidney ECs (n = 5464) were collected at three timepoints from diabetic BTBRob/ob mice and non-diabetic littermates. Their heterogeneity, transcriptional changes, and alternative splicing during DKD progression were mapped using SmartSeq2 single-cell RNA sequencing (scRNAseq) and elucidated through pathway, network, and gene ontology enrichment analyses. We identified 13 distinct transcriptional EC phenotypes corresponding to different kidney vessel subtypes, confirmed through in situ hybridization and immunofluorescence. EC subtypes along nephrons displayed extensive zonation related to their functions. Differential gene expression analyses in peritubular and glomerular ECs in DKD underlined the regulation of DKD-relevant pathways including EIF2 signaling, oxidative phosphorylation, and IGF1 signaling. Importantly, this revealed the differential alteration of these pathways between the two EC subtypes and changes during disease progression. Furthermore, glomerular and peritubular ECs also displayed aberrant and dynamic alterations in alternative splicing (AS), which is strongly associated with DNA repair. Strikingly, genes displaying differential transcription or alternative splicing participate in divergent biological processes. Our study reveals the spatiotemporal regulation of gene transcription and AS linked to DKD progression, providing insight into pathomechanisms and clues to novel therapeutic targets for DKD treatment.
Collapse
Affiliation(s)
- Alex-Xianghua Zhou
- Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, 43162 Mölndal, Sweden; (A.-X.Z.); (P.T.); (M.U.)
| | - Marie Jeansson
- Department of Medicine Huddinge, Karolinska Institutet, 141 52 Huddinge, Sweden; (M.J.); (J.L.)
- Department of Immunology, Genetics and Pathology, Uppsala University, 753 10 Uppsala, Sweden
| | - Liqun He
- Department of Medicine Huddinge, Karolinska Institutet, 141 52 Huddinge, Sweden; (M.J.); (J.L.)
- Department of Immunology, Genetics and Pathology, Uppsala University, 753 10 Uppsala, Sweden
| | - Leif Wigge
- Data Sciences and Quantitative Biology, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, 43162 Mölndal, Sweden
| | - Pernilla Tonelius
- Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, 43162 Mölndal, Sweden; (A.-X.Z.); (P.T.); (M.U.)
| | - Ramesh Tati
- Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, 43162 Mölndal, Sweden; (A.-X.Z.); (P.T.); (M.U.)
| | - Linda Cederblad
- Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, 43162 Mölndal, Sweden; (A.-X.Z.); (P.T.); (M.U.)
| | - Lars Muhl
- Department of Medicine Huddinge, Karolinska Institutet, 141 52 Huddinge, Sweden; (M.J.); (J.L.)
| | - Martin Uhrbom
- Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, 43162 Mölndal, Sweden; (A.-X.Z.); (P.T.); (M.U.)
- Department of Medicine Huddinge, Karolinska Institutet, 141 52 Huddinge, Sweden; (M.J.); (J.L.)
| | - Jianping Liu
- Department of Medicine Huddinge, Karolinska Institutet, 141 52 Huddinge, Sweden; (M.J.); (J.L.)
| | - Anna Björnson Granqvist
- Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, 43162 Mölndal, Sweden; (A.-X.Z.); (P.T.); (M.U.)
| | - Lilach O. Lerman
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN 55902, USA;
| | - Christer Betsholtz
- Department of Medicine Huddinge, Karolinska Institutet, 141 52 Huddinge, Sweden; (M.J.); (J.L.)
- Department of Immunology, Genetics and Pathology, Uppsala University, 753 10 Uppsala, Sweden
| | - Pernille B. L. Hansen
- Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, 43162 Mölndal, Sweden; (A.-X.Z.); (P.T.); (M.U.)
| |
Collapse
|
10
|
Lui A, Patel RS, Krause-Hauch M, Sparks RP, Patel NA. Regulation of Human Sortilin Alternative Splicing by Glucagon-like Peptide-1 (GLP1) in Adipocytes. Int J Mol Sci 2023; 24:14324. [PMID: 37762628 PMCID: PMC10531797 DOI: 10.3390/ijms241814324] [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: 09/01/2023] [Revised: 09/13/2023] [Accepted: 09/15/2023] [Indexed: 09/29/2023] Open
Abstract
Type 2 diabetes mellitus is a chronic metabolic disease with no cure. Adipose tissue is a major site of systemic insulin resistance. Sortilin is a central component of the glucose transporter -Glut4 storage vesicles (GSV) which translocate to the plasma membrane to uptake glucose from circulation. Here, using human adipocytes we demonstrate the presence of the alternatively spliced, truncated sortilin variant (Sort_T) whose expression is significantly increased in diabetic adipose tissue. Artificial-intelligence-based modeling, molecular dynamics, intrinsically disordered region analysis, and co-immunoprecipitation demonstrated association of Sort_T with Glut4 and decreased glucose uptake in adipocytes. The results show that glucagon-like peptide-1 (GLP1) hormone decreases Sort_T. We deciphered the molecular mechanism underlying GLP1 regulation of alternative splicing of human sortilin. Using splicing minigenes and RNA-immunoprecipitation assays, the results show that GLP1 regulates Sort_T alternative splicing via the splice factor, TRA2B. We demonstrate that targeted antisense oligonucleotide morpholinos reduces Sort_T levels and improves glucose uptake in diabetic adipocytes. Thus, we demonstrate that GLP1 regulates alternative splicing of sortilin in human diabetic adipocytes.
Collapse
Affiliation(s)
- Ashley Lui
- Department of Molecular Medicine, University of South Florida, Tampa, FL 33612, USA; (A.L.); (M.K.-H.)
| | - Rekha S. Patel
- Research Service, James A. Haley Veterans Hospital, Tampa, FL 33612, USA; (R.S.P.); (R.P.S.)
| | - Meredith Krause-Hauch
- Department of Molecular Medicine, University of South Florida, Tampa, FL 33612, USA; (A.L.); (M.K.-H.)
| | - Robert P. Sparks
- Research Service, James A. Haley Veterans Hospital, Tampa, FL 33612, USA; (R.S.P.); (R.P.S.)
- Department of Medicine, Division of Gastroenterology, UMass Chan Medical School, Worcester, MA 01655, USA
| | - Niketa A. Patel
- Department of Molecular Medicine, University of South Florida, Tampa, FL 33612, USA; (A.L.); (M.K.-H.)
- Research Service, James A. Haley Veterans Hospital, Tampa, FL 33612, USA; (R.S.P.); (R.P.S.)
| |
Collapse
|
11
|
De la Merced-García DS, Sánchez-Barrera Á, Hernández-Yonca J, Mancilla I, García-López G, Díaz NF, Terrazas LI, Molina-Hernández A. Increased Nuclear FOXP2 Is Related to Reduced Neural Stem Cell Number and Increased Neurogenesis in the Dorsal Telencephalon of Embryos of Diabetic Rats through Histamine H 1 Receptors. Cells 2023; 12:cells12030510. [PMID: 36766852 PMCID: PMC9914739 DOI: 10.3390/cells12030510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Accepted: 12/27/2022] [Indexed: 02/09/2023] Open
Abstract
Diabetic rat embryos have increased cortical neurogenesis and neuron maturation, and their offspring presented altered neuron polarity, lamination, and diminished neuron excitability. The FOXP2 overexpression results in higher cortical neurogenesis by increasing the transition of radial glia to the intermediate progenitor. Similarly, histamine through H1-receptor activation increases cortical neuron differentiation. Indeed, blocking the H1-receptor by the systemic administration of chlorpheniramine to diabetic pregnant rats prevents increased neurogenesis. Here, we explore the relationship between the H1-receptor and FOXP2 on embryo neurogenesis from diabetic dams. Through qRT-PCR, Western blot, immunohistofluorescence, and flow cytometry, we showed an increased FOXP2 expression and nuclear localization, a reduced Nestin expression and -positive cells number, and a higher PKCα expression in the cortical neuroepithelium of fourteen-day-old embryos from diabetic rats. Interestingly, this scenario was prevented by the chlorpheniramine systemic administration to diabetic pregnant rats at embryo day twelve. These data, together with the bioinformatic analysis, suggest that higher H1-receptor activity in embryos under high glucose increases FOXP2 nuclear translocation, presumably through PKCα phosphorylation, impairing the transition of radial glia to intermediate progenitor and increasing neuron differentiation in embryos of diabetic rats.
Collapse
Affiliation(s)
- Diana Sarahi De la Merced-García
- Departamento de Fisiología y Desarrollo Celular, Instituto Nacional de Perinatología Isidro Espinosa de los Reyes, Montes Urales 800, Miguel Hidalgo, Ciudad de Mexico 11000, Mexico
| | - Ángel Sánchez-Barrera
- Unidad de Biomedicina, Facultad de Estudios Superiores (FES)-Iztacala, Universidad Nacional Autónoma de México (UNAM), Av. de los Barrios, Los Reyes Iztacala, Tlanepantla 54090, Mexico
| | - Juan Hernández-Yonca
- Departamento de Fisiología y Desarrollo Celular, Instituto Nacional de Perinatología Isidro Espinosa de los Reyes, Montes Urales 800, Miguel Hidalgo, Ciudad de Mexico 11000, Mexico
| | - Ismael Mancilla
- Departamento de Infectología, Instituto Nacional de Perinatología Isidro Espinosa de los Reyes, Montes Urales 800, Miguel Hidalgo, Ciudad de Mexico 11000, Mexico
| | - Guadalupe García-López
- Departamento de Fisiología y Desarrollo Celular, Instituto Nacional de Perinatología Isidro Espinosa de los Reyes, Montes Urales 800, Miguel Hidalgo, Ciudad de Mexico 11000, Mexico
| | - Néstor Fabián Díaz
- Departamento de Fisiología y Desarrollo Celular, Instituto Nacional de Perinatología Isidro Espinosa de los Reyes, Montes Urales 800, Miguel Hidalgo, Ciudad de Mexico 11000, Mexico
| | - Luis Ignacio Terrazas
- Departamento de Infectología, Instituto Nacional de Perinatología Isidro Espinosa de los Reyes, Montes Urales 800, Miguel Hidalgo, Ciudad de Mexico 11000, Mexico
- Laboratorio Nacional en Salud FES-Iztacala, Universidad Nacional Autónoma de México (UNAM), Av. de los Barrios, Los Reyes Iztacala, Tlanepantla 54090, Mexico
| | - Anayansi Molina-Hernández
- Departamento de Fisiología y Desarrollo Celular, Instituto Nacional de Perinatología Isidro Espinosa de los Reyes, Montes Urales 800, Miguel Hidalgo, Ciudad de Mexico 11000, Mexico
- Correspondence:
| |
Collapse
|
12
|
Zanini G, Selleri V, De Gaetano A, Gibellini L, Malerba M, Mattioli AV, Nasi M, Apostolova N, Pinti M. Differential Expression of Lonp1 Isoforms in Cancer Cells. Cells 2022; 11:cells11233940. [PMID: 36497197 PMCID: PMC9739308 DOI: 10.3390/cells11233940] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 11/26/2022] [Accepted: 12/05/2022] [Indexed: 12/12/2022] Open
Abstract
Lonp1 is a mitochondrial protease that degrades oxidized and damaged proteins, assists protein folding, and contributes to the maintenance of mitochondrial DNA. A higher expression of LonP1 has been associated with higher tumour aggressiveness. Besides the full-length isoform (ISO1), we identified two other isoforms of Lonp1 in humans, resulting from alternative splicing: Isoform-2 (ISO2) lacking aa 42-105 and isoform-3 (ISO3) lacking aa 1-196. An inspection of the public database TSVdb showed that ISO1 was upregulated in lung, bladder, prostate, and breast cancer, ISO2 in all the cancers analysed (including rectum, colon, cervical, bladder, prostate, breast, head, and neck), ISO3 did not show significant changes between cancer and normal tissue. We overexpressed ISO1, ISO2, and ISO3 in SW620 cells and found that the ISO1 isoform was exclusively mitochondrial, ISO2 was present in the organelle and in the cytoplasm, and ISO3 was exclusively cytoplasmatic. The overexpression of ISO1 and, at a letter extent, of ISO2 enhanced basal, ATP-linked, and maximal respiration without altering the mitochondria number or network, mtDNA amount. or mitochondrial dynamics. A higher extracellular acidification rate was observed in ISO1 and ISO2, overexpressing cells, suggesting an increase in glycolysis. Cells overexpressing the different isoforms did not show a difference in the proliferation rate but showed a great increase in anchorage-independent growth. ISO1 and ISO2, but not ISO3, determined an upregulation of EMT-related proteins, which appeared unrelated to higher mitochondrial ROS production, nor due to the activation of the MEK ERK pathway, but rather to global metabolic reprogramming of cells.
Collapse
Affiliation(s)
- Giada Zanini
- Department of Life Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Valentina Selleri
- Department of Life Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy
- National Institute for Cardiovascular Research (INRC), 40126 Bologna, Italy
| | - Anna De Gaetano
- Department of Life Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Lara Gibellini
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia, 41124 Modena, Italy
| | - Mara Malerba
- Department of Surgical, Medical, Dental and Morphological Sciences, University of Modena and Reggio Emilia, 41124 Modena, Italy
| | - Anna Vittoria Mattioli
- National Institute for Cardiovascular Research (INRC), 40126 Bologna, Italy
- Department of Surgical, Medical, Dental and Morphological Sciences, University of Modena and Reggio Emilia, 41124 Modena, Italy
| | - Milena Nasi
- Department of Surgical, Medical, Dental and Morphological Sciences, University of Modena and Reggio Emilia, 41124 Modena, Italy
| | - Nadezda Apostolova
- Department of Pharmacology, University of Valencia, 46010 Valencia, Spain
- FISABIO—Hospital Universitario Dr. Peset, 46017 Valencia, Spain
| | - Marcello Pinti
- Department of Life Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy
- Correspondence: ; Tel.: +39-059-205-5386
| |
Collapse
|
13
|
Szymczak F, Alvelos MI, Marín-Cañas S, Castela Â, Demine S, Colli ML, Op de Beeck A, Thomaidou S, Marselli L, Zaldumbide A, Marchetti P, Eizirik DL. Transcription and splicing regulation by NLRC5 shape the interferon response in human pancreatic β cells. SCIENCE ADVANCES 2022; 8:eabn5732. [PMID: 36103539 PMCID: PMC9473574 DOI: 10.1126/sciadv.abn5732] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 07/27/2022] [Indexed: 06/15/2023]
Abstract
IFNα is a key regulator of the dialogue between pancreatic β cells and the immune system in early type 1 diabetes (T1D). IFNα up-regulates HLA class I expression in human β cells, fostering autoantigen presentation to the immune system. We observed by bulk and single-cell RNA sequencing that exposure of human induced pluripotent-derived islet-like cells to IFNα induces expression of HLA class I and of other genes involved in antigen presentation, including the transcriptional activator NLRC5. We next evaluated the global role of NLRC5 in human insulin-producing EndoC-βH1 and human islet cells by RNA sequencing and targeted gene/protein determination. NLRC5 regulates expression of HLA class I, antigen presentation-related genes, and chemokines. NLRC5 also mediates the effects of IFNα on alternative splicing, a generator of β cell neoantigens, suggesting that it is a central player of the effects of IFNα on β cells that contribute to trigger and amplify autoimmunity in T1D.
Collapse
Affiliation(s)
- Florian Szymczak
- ULB Center for Diabetes Research, Medical Faculty, Université Libre De Bruxelles (ULB), Brussels, Belgium
- Interuniversity Institute of Bioinformatics in Brussels, Université Libre de Bruxelles-Vrije Universiteit Brussel, Brussels, Belgium
| | - Maria Inês Alvelos
- ULB Center for Diabetes Research, Medical Faculty, Université Libre De Bruxelles (ULB), Brussels, Belgium
| | - Sandra Marín-Cañas
- ULB Center for Diabetes Research, Medical Faculty, Université Libre De Bruxelles (ULB), Brussels, Belgium
| | - Ângela Castela
- ULB Center for Diabetes Research, Medical Faculty, Université Libre De Bruxelles (ULB), Brussels, Belgium
| | - Stéphane Demine
- Indiana Biosciences Research Institute, Indianapolis, IN, USA
| | - Maikel Luis Colli
- ULB Center for Diabetes Research, Medical Faculty, Université Libre De Bruxelles (ULB), Brussels, Belgium
| | - Anne Op de Beeck
- ULB Center for Diabetes Research, Medical Faculty, Université Libre De Bruxelles (ULB), Brussels, Belgium
| | - Sofia Thomaidou
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, Netherlands
| | - Lorella Marselli
- Department of Clinical and Experimental Medicine, Islet Cell Laboratory, University of Pisa, Pisa, Italy
| | - Arnaud Zaldumbide
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, Netherlands
| | - Piero Marchetti
- Department of Clinical and Experimental Medicine, Islet Cell Laboratory, University of Pisa, Pisa, Italy
| | - Décio L. Eizirik
- ULB Center for Diabetes Research, Medical Faculty, Université Libre De Bruxelles (ULB), Brussels, Belgium
- Welbio, Medical Faculty, Université Libre De Bruxelles (ULB), Brussels, Belgium
| |
Collapse
|
14
|
Ferragut Cardoso AP, Banerjee M, Al-Eryani L, Sayed M, Wilkey DW, Merchant ML, Park JW, States JC. Temporal Modulation of Differential Alternative Splicing in HaCaT Human Keratinocyte Cell Line Chronically Exposed to Arsenic for up to 28 Wk. ENVIRONMENTAL HEALTH PERSPECTIVES 2022; 130:17011. [PMID: 35072517 PMCID: PMC8785870 DOI: 10.1289/ehp9676] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 12/12/2021] [Accepted: 12/16/2021] [Indexed: 05/06/2023]
Abstract
BACKGROUND Chronic arsenic exposure via drinking water is associated with an increased risk of developing cancer and noncancer chronic diseases. Pre-mRNAs are often subject to alternative splicing, generating mRNA isoforms encoding functionally distinct protein isoforms. The resulting imbalance in isoform species can result in pathogenic changes in critical signaling pathways. Alternative splicing as a mechanism of arsenic-induced toxicity and carcinogenicity is understudied. OBJECTIVE This study aimed to accurately profile differential alternative splicing events in human keratinocytes induced by chronic arsenic exposure that might play a role in carcinogenesis. METHODS Independent quadruplicate cultures of immortalized human keratinocytes (HaCaT) were maintained continuously for 28 wk with 0 or 100 nM sodium arsenite. RNA-sequencing (RNA-Seq) was performed with poly(A) RNA isolated from cells harvested at 7, 19, and 28 wk with subsequent replicate multivariate analysis of transcript splicing (rMATS) analysis to detect and quantify differential alternative splicing events. Reverse transcriptase-polymerase chain reaction (RT-PCR) for selected alternative splicing events was performed to validate RNA-Seq predictions. Functional enrichment was performed by gene ontology (GO) analysis of the differential alternative splicing event data set at each time point. RESULTS At least 600 differential alternative splicing events were detected at each time point tested, comprising all the five main types of alternative splicing and occurring in both open reading frames (ORFs) and untranslated regions (UTRs). Based on functional relevance ELK4, SHC1, and XRRA1 were selected for validation of predicted alternative splicing events at 7 wk by RT-PCR. Densitometric analysis of RT-PCR data corroborated the rMATS predicted alternative splicing for all three events. Protein expression validation of the selected alternative splicing events was challenging given that very few isoform-specific antibodies are available. GO analysis demonstrated that the enriched terms in differential alternatively spliced mRNAs changed dynamically with the time of exposure. Notably, RNA metabolism and splicing regulation pathways were enriched at the 7-wk time point, when the greatest number of differentially alternatively spliced mRNAs are detected. Our preliminary proteomic analysis demonstrated that the expression of the canonical isoforms of the splice regulators DDX42, RMB25, and SRRM2 were induced upon chronic arsenic exposure, corroborating the splicing predictions. DISCUSSION These results using cultures of HaCaT cells suggest that arsenic exposure disrupted an alternative splice factor network and induced time-dependent genome-wide differential alternative splicing that likely contributed to the changing proteomic landscape in arsenic-induced carcinogenesis. However, significant challenges remain in corroborating alternative splicing data at the proteomic level. https://doi.org/10.1289/EHP9676.
Collapse
Affiliation(s)
- Ana P. Ferragut Cardoso
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, Kentucky, USA
| | - Mayukh Banerjee
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, Kentucky, USA
| | - Laila Al-Eryani
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, Kentucky, USA
| | - Mohammed Sayed
- Computer Science and Engineering, University of Louisville, Louisville, Kentucky, USA
| | - Daniel W. Wilkey
- Division of Nephrology & Hypertension, Department of Medicine, University of Louisville, Louisville, Kentucky, USA
| | - Michael L. Merchant
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, Kentucky, USA
- Division of Nephrology & Hypertension, Department of Medicine, University of Louisville, Louisville, Kentucky, USA
| | - Juw W. Park
- Computer Science and Engineering, University of Louisville, Louisville, Kentucky, USA
- KY INBRE Bioinformatics Core, University of Louisville, Louisville, Kentucky, USA
| | - J. Christopher States
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, Kentucky, USA
| |
Collapse
|
15
|
Liang J, Hong Z, Sun B, Guo Z, Wang C, Zhu J. The Alternatively Spliced Isoforms of Key Molecules in the cGAS-STING Signaling Pathway. Front Immunol 2021; 12:771744. [PMID: 34868032 PMCID: PMC8636596 DOI: 10.3389/fimmu.2021.771744] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 11/03/2021] [Indexed: 01/04/2023] Open
Abstract
Alternative splicing of pre-mRNA increases transcriptome and proteome diversity by generating distinct isoforms that encode functionally diverse proteins, thus affecting many biological processes, including innate immunity. cGAS-STING signaling pathway, whose key molecules also undergo alternative splicing, plays a crucial role in regulating innate immunity. Protein isoforms of key components in the cGAS-STING-TBK1-IRF3 axis have been detected in a variety of species. A chain of evidence showed that these protein isoforms exhibit distinct functions compared to their normal counterparts. The mentioned isoforms act as positive or negative modulators in interferon response via distinct mechanisms. Particularly, we highlight that alternative splicing serves a vital function for the host to avoid the overactivation of the cGAS-STING signaling pathway and that viruses can utilize alternative splicing to resist antiviral response by the host. These findings could provide insights for potential alternative splicing-targeting therapeutic applications.
Collapse
Affiliation(s)
- Jiaqian Liang
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Ze Hong
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Boyue Sun
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, Israel
| | - Zhaoxi Guo
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Chen Wang
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Juanjuan Zhu
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
16
|
Wang S, Xu G, Chao F, Zhang C, Han D, Chen G. HNRNPC Promotes Proliferation, Metastasis and Predicts Prognosis in Prostate Cancer. Cancer Manag Res 2021; 13:7263-7276. [PMID: 34584453 PMCID: PMC8464311 DOI: 10.2147/cmar.s330713] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 09/13/2021] [Indexed: 12/22/2022] Open
Abstract
INTRODUCTION The incidence of prostate cancer remains high worldwide, while exploring new therapeutic targets for prostate cancer is essential. Heterogeneous nuclear ribonucleoproteins have been proved to regulate tumorigeneses in various cancers. This study aimed to explore the role of HNRNPC in prostate cancer progression. METHODS HNRNPC expression and its correlation with clinical features and immune infiltration were analyzed by bioinformatics analysis. The effects of HNRNPC on prostate cell proliferation, migration, and invasion were accessed by EdU, colony formation, transwell, and wound-healing assays. RESULTS The expression level of HNRNPC was significantly increased in prostate cancer tissues and was correlated with the T stage, N stage, Gleason score, PSA level, residual tumors, overall survival, disease-specific survival, and progression-free interval of prostate cancer patients. Silencing HNRNPC inhibited the proliferation and metastasis of prostate cancer cells. The expression of HNRNPC was negatively correlated with the infiltration level of most immune cells in prostate cancer. Mechanistically, HNRNPC may function through regulating gene expression at the posttranscriptional level. CONCLUSION HNRNPC could be a potential marker for the treatment and prognosis prediction of prostate cancer.
Collapse
Affiliation(s)
- Shiyu Wang
- Department of Urology, Jinshan Hospital, Fudan University, Shanghai, 201508, People’s Republic of China
- Department of Surgery, Shanghai Medical College, Fudan University, Shanghai, 200032, People’s Republic of China
| | - Guoxiong Xu
- Research Center for Clinical Medicine, Jinshan Hospital, Fudan University, Shanghai, 201508, People’s Republic of China
| | - Fan Chao
- Department of Urology, Jinshan Hospital, Fudan University, Shanghai, 201508, People’s Republic of China
- Department of Surgery, Shanghai Medical College, Fudan University, Shanghai, 200032, People’s Republic of China
| | - Cong Zhang
- Department of Urology, Jinshan Hospital, Fudan University, Shanghai, 201508, People’s Republic of China
- Department of Surgery, Shanghai Medical College, Fudan University, Shanghai, 200032, People’s Republic of China
| | - Dunsheng Han
- Department of Urology, Jinshan Hospital, Fudan University, Shanghai, 201508, People’s Republic of China
- Department of Surgery, Shanghai Medical College, Fudan University, Shanghai, 200032, People’s Republic of China
| | - Gang Chen
- Department of Urology, Jinshan Hospital, Fudan University, Shanghai, 201508, People’s Republic of China
- Department of Surgery, Shanghai Medical College, Fudan University, Shanghai, 200032, People’s Republic of China
| |
Collapse
|
17
|
Cornelius VA, Fulton JR, Margariti A. Alternative Splicing: A Key Mediator of Diabetic Vasculopathy. Genes (Basel) 2021; 12:1332. [PMID: 34573314 PMCID: PMC8469645 DOI: 10.3390/genes12091332] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 08/24/2021] [Accepted: 08/24/2021] [Indexed: 12/25/2022] Open
Abstract
Cardiovascular disease is the leading cause of death amongst diabetic individuals. Atherosclerosis is the prominent driver of diabetic vascular complications, which is triggered by the detrimental effects of hyperglycemia and oxidative stress on the vasculature. Research has extensively shown diabetes to result in the malfunction of the endothelium, the main component of blood vessels, causing severe vascular complications. The pathogenic mechanism in which diabetes induces vascular dysfunction, however, remains largely unclear. Alternative splicing of protein coding pre-mRNAs is an essential regulatory mechanism of gene expression and is accepted to be intertwined with cellular physiology. Recently, a role for alternative splicing has arisen within vascular health, with aberrant mis-splicing having a critical role in disease development, including in atherosclerosis. This review focuses on the current knowledge of alternative splicing and the roles of alternatively spliced isoforms within the vasculature, with a particular focus on disease states. Furthermore, we explore the recent elucidation of the alternatively spliced QKI gene within vascular cell physiology and the onset of diabetic vasculopathy. Potential therapeutic strategies to restore aberrant splicing are also discussed.
Collapse
Affiliation(s)
| | | | - Andriana Margariti
- The Wellcome-Wolfson Institute of Experimental Medicine, Belfast BT9 7BL, UK; (V.A.C.); (J.R.F.)
| |
Collapse
|
18
|
A deep learning approach to identify gene targets of a therapeutic for human splicing disorders. Nat Commun 2021; 12:3332. [PMID: 34099697 PMCID: PMC8185002 DOI: 10.1038/s41467-021-23663-2] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Accepted: 05/07/2021] [Indexed: 01/16/2023] Open
Abstract
Pre-mRNA splicing is a key controller of human gene expression. Disturbances in splicing due to mutation lead to dysregulated protein expression and contribute to a substantial fraction of human disease. Several classes of splicing modulator compounds (SMCs) have been recently identified and establish that pre-mRNA splicing represents a target for therapy. We describe herein the identification of BPN-15477, a SMC that restores correct splicing of ELP1 exon 20. Using transcriptome sequencing from treated fibroblast cells and a machine learning approach, we identify BPN-15477 responsive sequence signatures. We then leverage this model to discover 155 human disease genes harboring ClinVar mutations predicted to alter pre-mRNA splicing as targets for BPN-15477. Splicing assays confirm successful correction of splicing defects caused by mutations in CFTR, LIPA, MLH1 and MAPT. Subsequent validations in two disease-relevant cellular models demonstrate that BPN-15477 increases functional protein, confirming the clinical potential of our predictions.
Collapse
|
19
|
Rehman SU, Schallschmidt T, Rasche A, Knebel B, Stermann T, Altenhofen D, Herwig R, Schürmann A, Chadt A, Al-Hasani H. Alternative exon splicing and differential expression in pancreatic islets reveals candidate genes and pathways implicated in early diabetes development. Mamm Genome 2021; 32:153-172. [PMID: 33880624 PMCID: PMC8128753 DOI: 10.1007/s00335-021-09869-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Accepted: 04/03/2021] [Indexed: 12/29/2022]
Abstract
Type 2 diabetes (T2D) has a strong genetic component. Most of the gene variants driving the pathogenesis of T2D seem to target pancreatic β-cell function. To identify novel gene variants acting at early stage of the disease, we analyzed whole transcriptome data to identify differential expression (DE) and alternative exon splicing (AS) transcripts in pancreatic islets collected from two metabolically diverse mouse strains at 6 weeks of age after three weeks of high-fat-diet intervention. Our analysis revealed 1218 DE and 436 AS genes in islets from NZO/Hl vs C3HeB/FeJ. Whereas some of the revealed genes present well-established markers for β-cell failure, such as Cd36 or Aldh1a3, we identified numerous DE/AS genes that have not been described in context with β-cell function before. The gene Lgals2, previously associated with human T2D development, was DE as well as AS and localizes in a quantitative trait locus (QTL) for blood glucose on Chr.15 that we reported recently in our N2(NZOxC3H) population. In addition, pathway enrichment analysis of DE and AS genes showed an overlap of only half of the revealed pathways, indicating that DE and AS in large parts influence different pathways in T2D development. PPARG and adipogenesis pathways, two well-established metabolic pathways, were overrepresented for both DE and AS genes, probably as an adaptive mechanism to cope for increased cellular stress. Our results provide guidance for the identification of novel T2D candidate genes and demonstrate the presence of numerous AS transcripts possibly involved in islet function and maintenance of glucose homeostasis.
Collapse
Affiliation(s)
- Sayeed Ur Rehman
- Institute for Clinical Biochemistry and Pathobiochemistry, German Diabetes Center (DDZ), Leibniz Center for Diabetes Research at Heinrich Heine University, Medical Faculty, Duesseldorf, Germany.,German Center for Diabetes Research (DZD), München-Neuherberg, Germany.,Department of Biochemistry, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi, 110062, India
| | - Tanja Schallschmidt
- Institute for Clinical Biochemistry and Pathobiochemistry, German Diabetes Center (DDZ), Leibniz Center for Diabetes Research at Heinrich Heine University, Medical Faculty, Duesseldorf, Germany.,German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | - Axel Rasche
- Department of Computational Molecular Biology, Max Planck Institute for Molecular Genetics, Berlin, Germany
| | - Birgit Knebel
- Institute for Clinical Biochemistry and Pathobiochemistry, German Diabetes Center (DDZ), Leibniz Center for Diabetes Research at Heinrich Heine University, Medical Faculty, Duesseldorf, Germany.,German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | - Torben Stermann
- Institute for Clinical Biochemistry and Pathobiochemistry, German Diabetes Center (DDZ), Leibniz Center for Diabetes Research at Heinrich Heine University, Medical Faculty, Duesseldorf, Germany.,German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | - Delsi Altenhofen
- Institute for Clinical Biochemistry and Pathobiochemistry, German Diabetes Center (DDZ), Leibniz Center for Diabetes Research at Heinrich Heine University, Medical Faculty, Duesseldorf, Germany.,German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | - Ralf Herwig
- Department of Computational Molecular Biology, Max Planck Institute for Molecular Genetics, Berlin, Germany
| | - Annette Schürmann
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany.,German Institute of Human Nutrition, Potsdam, Germany
| | - Alexandra Chadt
- Institute for Clinical Biochemistry and Pathobiochemistry, German Diabetes Center (DDZ), Leibniz Center for Diabetes Research at Heinrich Heine University, Medical Faculty, Duesseldorf, Germany.,German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | - Hadi Al-Hasani
- Institute for Clinical Biochemistry and Pathobiochemistry, German Diabetes Center (DDZ), Leibniz Center for Diabetes Research at Heinrich Heine University, Medical Faculty, Duesseldorf, Germany. .,German Center for Diabetes Research (DZD), München-Neuherberg, Germany.
| |
Collapse
|
20
|
Ghiasi SM, Rutter GA. Consequences for Pancreatic β-Cell Identity and Function of Unregulated Transcript Processing. Front Endocrinol (Lausanne) 2021; 12:625235. [PMID: 33763030 PMCID: PMC7984428 DOI: 10.3389/fendo.2021.625235] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Accepted: 01/26/2021] [Indexed: 12/25/2022] Open
Abstract
Mounting evidence suggests a role for alternative splicing (AS) of transcripts in the normal physiology and pathophysiology of the pancreatic β-cell. In the apparent absence of RNA repair systems, RNA decay pathways are likely to play an important role in controlling the stability, distribution and diversity of transcript isoforms in these cells. Around 35% of alternatively spliced transcripts in human cells contain premature termination codons (PTCs) and are targeted for degradation via nonsense-mediated decay (NMD), a vital quality control process. Inflammatory cytokines, whose levels are increased in both type 1 (T1D) and type 2 (T2D) diabetes, stimulate alternative splicing events and the expression of NMD components, and may or may not be associated with the activation of the NMD pathway. It is, however, now possible to infer that NMD plays a crucial role in regulating transcript processing in normal and stress conditions in pancreatic β-cells. In this review, we describe the possible role of Regulated Unproductive Splicing and Translation (RUST), a molecular mechanism embracing NMD activity in relationship to AS and translation of damaged transcript isoforms in these cells. This process substantially reduces the abundance of non-functional transcript isoforms, and its dysregulation may be involved in pancreatic β-cell failure in diabetes.
Collapse
Affiliation(s)
- Seyed M. Ghiasi
- Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Guy A. Rutter
- Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, London, United Kingdom
| |
Collapse
|
21
|
Zhang Y, Qian J, Gu C, Yang Y. Alternative splicing and cancer: a systematic review. Signal Transduct Target Ther 2021; 6:78. [PMID: 33623018 PMCID: PMC7902610 DOI: 10.1038/s41392-021-00486-7] [Citation(s) in RCA: 222] [Impact Index Per Article: 55.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2020] [Revised: 09/24/2020] [Accepted: 09/28/2020] [Indexed: 01/31/2023] Open
Abstract
The abnormal regulation of alternative splicing is usually accompanied by the occurrence and development of tumors, which would produce multiple different isoforms and diversify protein expression. The aim of the present study was to conduct a systematic review in order to describe the regulatory mechanisms of alternative splicing, as well as its functions in tumor cells, from proliferation and apoptosis to invasion and metastasis, and from angiogenesis to metabolism. The abnormal splicing events contributed to tumor progression as oncogenic drivers and/or bystander factors. The alterations in splicing factors detected in tumors and other mis-splicing events (i.e., long non-coding and circular RNAs) in tumorigenesis were also included. The findings of recent therapeutic approaches targeting splicing catalysis and splicing regulatory proteins to modulate pathogenically spliced events (including tumor-specific neo-antigens for cancer immunotherapy) were introduced. The emerging RNA-based strategies for the treatment of cancer with abnormally alternative splicing isoforms were also discussed. However, further studies are still required to address the association between alternative splicing and cancer in more detail.
Collapse
Affiliation(s)
- Yuanjiao Zhang
- The Third Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Jinjun Qian
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Chunyan Gu
- The Third Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China.
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China.
| | - Ye Yang
- The Third Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China.
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China.
| |
Collapse
|
22
|
Identification of Sortilin Alternatively Spliced Variants in Mouse 3T3L1 Adipocytes. Int J Mol Sci 2021; 22:ijms22030983. [PMID: 33498179 PMCID: PMC7863940 DOI: 10.3390/ijms22030983] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 01/06/2021] [Accepted: 01/13/2021] [Indexed: 12/13/2022] Open
Abstract
Type 2 diabetes mellitus is a metabolic disorder defined by systemic insulin resistance. Insulin resistance in adipocytes, an important regulator of glucose metabolism, results in impaired glucose uptake. The trafficking protein, sortilin, regulates major glucose transporter 4 (Glut4) movement, thereby promoting glucose uptake in adipocytes. Here, we demonstrate the presence of an alternatively spliced sortilin variant (Sort17b), whose levels increase with insulin resistance in mouse 3T3L1 adipocytes. Using a splicing minigene, we show that inclusion of alternative exon 17b results in the expression of Sort17b splice variant. Bioinformatic analysis indicated a novel intrinsic disorder region (IDR) encoded by exon 17b of Sort17b. Root mean square deviation (RMSD) and root mean square fluctuation (RMSF) measurements using molecular dynamics demonstrated increased flexibility of the protein backbone within the IDR. Using protein–protein docking and co-immunoprecipitation assays, we show robust binding of Glut4 to Sort17b. Further, results demonstrate that over-expression of Sort17b correlates with reduced Glut4 translocation and decreased glucose uptake in adipocytes. The study demonstrates that insulin resistance in 3T3L1 adipocytes promotes expression of a novel sortilin splice variant with thus far unknown implications in glucose metabolism. This knowledge may be used to develop therapeutics targeting sortilin variants in the management of type 2 diabetes and metabolic syndrome.
Collapse
|
23
|
del Río-Moreno M, Luque RM, Rangel-Zúñiga OA, Alors-Pérez E, Alcalá-Diaz JF, Roncero-Ramos I, Camargo A, Gahete MD, López-Miranda J, Castaño JP. Dietary Intervention Modulates the Expression of Splicing Machinery in Cardiovascular Patients at High Risk of Type 2 Diabetes Development: From the CORDIOPREV Study. Nutrients 2020; 12:E3528. [PMID: 33212780 PMCID: PMC7696699 DOI: 10.3390/nu12113528] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 11/03/2020] [Accepted: 11/12/2020] [Indexed: 12/11/2022] Open
Abstract
Type-2 diabetes mellitus (T2DM) has become a major health problem worldwide. T2DM risk can be reduced with healthy dietary interventions, but the precise molecular underpinnings behind this association are still incompletely understood. We recently discovered that the expression profile of the splicing machinery is associated with the risk of T2DM development. Thus, the aim of this work was to evaluate the influence of 3-year dietary intervention in the expression pattern of the splicing machinery components in peripheral blood mononuclear cells (PBMCs) from patients within the CORDIOPREV study. Expression of splicing machinery components was determined in PBMCs, at baseline and after 3 years of follow-up, from all patients who developed T2DM (Incident-T2DM, n = 107) and 108 randomly selected non-T2DM subjects, who were randomly enrolled in two healthy dietary patterns (Mediterranean or low-fat diets). Dietary intervention modulated the expression of key splicing machinery components (i.e., up-regulation of SPFQ/RMB45/RNU6, etc., down-regulation of RNU2/SRSF6) after three years, independently of the type of healthy diet. Some of these changes (SPFQ/RMB45/SRSF6) were associated with key clinical features and were differentially induced in Incident-T2DM patients and non-T2DM subjects. This study reveals that splicing machinery can be modulated by long-term dietary intervention, and could become a valuable tool to screen the progression of T2DM.
Collapse
Grants
- PIE14/00005 Instituto de Salud Carlos III
- PIE14/00031 Instituto de Salud Carlos III
- PI16/00264 Instituto de Salud Carlos III
- CP15/00156 Instituto de Salud Carlos III
- PI17/002287 Instituto de Salud Carlos III
- BFU2016-80360-R Ministerio de Economía, Industria y Competitividad, Gobierno de España
- TIN2017-83445-P Ministerio de Economía, Industria y Competitividad, Gobierno de España
- PI13/00023 Ministerio de Economía, Industria y Competitividad, Gobierno de España
- AGL2012/39615 Ministerio de Economía, Industria y Competitividad, Gobierno de España
- AGL2015-67896-P Ministerio de Economía, Industria y Competitividad, Gobierno de España
- BIO-0139 Junta de Andalucía
- CTS-1406 Junta de Andalucía
- CTS-525 Junta de Andalucía
- PI-0541-2013 Junta de Andalucía
- CVI-7450 Junta de Andalucía
Collapse
Affiliation(s)
- Mercedes del Río-Moreno
- Maimonides Institute for Biomedical Research of Cordoba (IMIBIC), 14004 Córdoba, Spain; (M.d.R.-M.); (O.A.R.-Z.); (E.A.-P.); (J.F.A.-D.); (I.R.-R.); (A.C.)
- Department of Cell Biology, University of Córdoba, 14004 Córdoba, Spain
- Reina Sofia University Hospital, 14004 Córdoba, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), 14004 Córdoba, Spain
| | - Raúl M. Luque
- Maimonides Institute for Biomedical Research of Cordoba (IMIBIC), 14004 Córdoba, Spain; (M.d.R.-M.); (O.A.R.-Z.); (E.A.-P.); (J.F.A.-D.); (I.R.-R.); (A.C.)
- Department of Cell Biology, University of Córdoba, 14004 Córdoba, Spain
- Reina Sofia University Hospital, 14004 Córdoba, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), 14004 Córdoba, Spain
| | - Oriol A. Rangel-Zúñiga
- Maimonides Institute for Biomedical Research of Cordoba (IMIBIC), 14004 Córdoba, Spain; (M.d.R.-M.); (O.A.R.-Z.); (E.A.-P.); (J.F.A.-D.); (I.R.-R.); (A.C.)
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), 14004 Córdoba, Spain
- Lipid and Atherosclerosis Unit, Department of Medicine, Reina Sofia University Hospital, University of Córdoba, 14004 Córdoba, Spain
| | - Emilia Alors-Pérez
- Maimonides Institute for Biomedical Research of Cordoba (IMIBIC), 14004 Córdoba, Spain; (M.d.R.-M.); (O.A.R.-Z.); (E.A.-P.); (J.F.A.-D.); (I.R.-R.); (A.C.)
- Department of Cell Biology, University of Córdoba, 14004 Córdoba, Spain
- Reina Sofia University Hospital, 14004 Córdoba, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), 14004 Córdoba, Spain
| | - Juan F. Alcalá-Diaz
- Maimonides Institute for Biomedical Research of Cordoba (IMIBIC), 14004 Córdoba, Spain; (M.d.R.-M.); (O.A.R.-Z.); (E.A.-P.); (J.F.A.-D.); (I.R.-R.); (A.C.)
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), 14004 Córdoba, Spain
- Lipid and Atherosclerosis Unit, Department of Medicine, Reina Sofia University Hospital, University of Córdoba, 14004 Córdoba, Spain
| | - Irene Roncero-Ramos
- Maimonides Institute for Biomedical Research of Cordoba (IMIBIC), 14004 Córdoba, Spain; (M.d.R.-M.); (O.A.R.-Z.); (E.A.-P.); (J.F.A.-D.); (I.R.-R.); (A.C.)
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), 14004 Córdoba, Spain
- Lipid and Atherosclerosis Unit, Department of Medicine, Reina Sofia University Hospital, University of Córdoba, 14004 Córdoba, Spain
| | - Antonio Camargo
- Maimonides Institute for Biomedical Research of Cordoba (IMIBIC), 14004 Córdoba, Spain; (M.d.R.-M.); (O.A.R.-Z.); (E.A.-P.); (J.F.A.-D.); (I.R.-R.); (A.C.)
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), 14004 Córdoba, Spain
- Lipid and Atherosclerosis Unit, Department of Medicine, Reina Sofia University Hospital, University of Córdoba, 14004 Córdoba, Spain
| | - Manuel D. Gahete
- Maimonides Institute for Biomedical Research of Cordoba (IMIBIC), 14004 Córdoba, Spain; (M.d.R.-M.); (O.A.R.-Z.); (E.A.-P.); (J.F.A.-D.); (I.R.-R.); (A.C.)
- Department of Cell Biology, University of Córdoba, 14004 Córdoba, Spain
- Reina Sofia University Hospital, 14004 Córdoba, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), 14004 Córdoba, Spain
| | - José López-Miranda
- Maimonides Institute for Biomedical Research of Cordoba (IMIBIC), 14004 Córdoba, Spain; (M.d.R.-M.); (O.A.R.-Z.); (E.A.-P.); (J.F.A.-D.); (I.R.-R.); (A.C.)
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), 14004 Córdoba, Spain
- Lipid and Atherosclerosis Unit, Department of Medicine, Reina Sofia University Hospital, University of Córdoba, 14004 Córdoba, Spain
| | - Justo P. Castaño
- Maimonides Institute for Biomedical Research of Cordoba (IMIBIC), 14004 Córdoba, Spain; (M.d.R.-M.); (O.A.R.-Z.); (E.A.-P.); (J.F.A.-D.); (I.R.-R.); (A.C.)
- Department of Cell Biology, University of Córdoba, 14004 Córdoba, Spain
- Reina Sofia University Hospital, 14004 Córdoba, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), 14004 Córdoba, Spain
| |
Collapse
|
24
|
Paz S, Ritchie A, Mauer C, Caputi M. The RNA binding protein SRSF1 is a master switch of gene expression and regulation in the immune system. Cytokine Growth Factor Rev 2020; 57:19-26. [PMID: 33160830 DOI: 10.1016/j.cytogfr.2020.10.008] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 10/26/2020] [Accepted: 10/28/2020] [Indexed: 12/22/2022]
Abstract
Serine/Arginine splicing factor 1 (SRSF1) is an RNA binding protein abundantly expressed in most tissues. The pleiotropic functions of SRSF1 exert multiple roles in gene expression by regulating major steps in transcription, processing, export through the nuclear pores and translation of nascent RNA transcripts. The aim of this review is to highlight recent findings in the functions of this protein and to describe its role in immune system development, functions and regulation.
Collapse
Affiliation(s)
- Sean Paz
- Charles E. Schmidt College of Medicine, Florida Atlantic University, 777 Glades Rd, Boca Raton, FL, 33431, United States
| | - Anastasia Ritchie
- Charles E. Schmidt College of Medicine, Florida Atlantic University, 777 Glades Rd, Boca Raton, FL, 33431, United States
| | - Christopher Mauer
- Charles E. Schmidt College of Medicine, Florida Atlantic University, 777 Glades Rd, Boca Raton, FL, 33431, United States
| | - Massimo Caputi
- Charles E. Schmidt College of Medicine, Florida Atlantic University, 777 Glades Rd, Boca Raton, FL, 33431, United States.
| |
Collapse
|
25
|
Moss ND, Sussel L. mRNA Processing: An Emerging Frontier in the Regulation of Pancreatic β Cell Function. Front Genet 2020; 11:983. [PMID: 33088281 PMCID: PMC7490333 DOI: 10.3389/fgene.2020.00983] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Accepted: 08/03/2020] [Indexed: 01/04/2023] Open
Abstract
Robust endocrine cell function, particularly β cell function, is required to maintain blood glucose homeostasis. Diabetes can result from the loss or dysfunction of β cells. Despite decades of clinical and basic research, the precise regulation of β cell function and pathogenesis in diabetes remains incompletely understood. In this review, we highlight RNA processing of mRNAs as a rapidly emerging mechanism regulating β cell function and survival. RNA-binding proteins (RBPs) and RNA modifications are primed to be the next frontier to explain many of the poorly understood molecular processes that regulate β cell formation and function, and provide an exciting potential for the development of novel therapeutics. Here we outline the current understanding of β cell specific functions of several characterized RBPs, alternative splicing events, and transcriptome wide changes in RNA methylation. We also highlight several RBPs that are dysregulated in both Type 1 and Type 2 diabetes, and discuss remaining knowledge gaps in the field.
Collapse
Affiliation(s)
- Nicole D Moss
- Cell, Stem Cells, and Development Graduate Program, Department of Pediatrics, Barbara Davis Center, University of Colorado Denver Anschutz Medical Campus, Aurora, CO, United States
| | - Lori Sussel
- Cell, Stem Cells, and Development Graduate Program, Department of Pediatrics, Barbara Davis Center, University of Colorado Denver Anschutz Medical Campus, Aurora, CO, United States
| |
Collapse
|
26
|
Qi J, Wang D, Yin X, Zhang Q, Gao JM. New Metabolite With Inhibitory Activity Against α-Glucosidase and α-Amylase From Endophytic Chaetomium globosum. Nat Prod Commun 2020. [DOI: 10.1177/1934578x20941338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
Abstract
An efficient bioactive tracking separation strategy based on liquid-liquid extraction and high-speed counter-current chromatography (HSCCC) was developed and used to isolate bioactive natural products from the endophytic fungus Chaetomium globosum residing in Ginkgo biloba. Using HSCCC, the novel metabolite chaetoglobol acid (1) as well as 11 known compounds (2-12), including 6 chlorinated azaphilones and 3 cytochalasans, were successfully isolated. The structure of compound 1 was elucidated through spectroscopic analyses and HRESIMS data. Compound 1 possesses a rare C11-polyketide skeleton. All isolates were evaluated for their α-glucosidase and α-amylase inhibitory activities in vitro. Compound 1 showed high inhibition against α-glucosidase (IC50 = 3.04 μM), 18-fold higher than that of acarbose (IC50 = 54.74 μM), and also displayed moderate inhibitory activity against α-amylase (IC50 = 22.18 μM). As the results indicated that 1 has inhibitory effects against both α-glucosidase and α-amylase, 1 may be a promising candidate for mediating type 2 diabetes.
Collapse
Affiliation(s)
- Jianzhao Qi
- Shaanxi Key Laboratory of Natural Products & Chemical Biology, College of Chemistry and Pharmacy, Northwest A&F University, Yangling, 712100, China
| | - Dacheng Wang
- Shaanxi Key Laboratory of Natural Products & Chemical Biology, College of Chemistry and Pharmacy, Northwest A&F University, Yangling, 712100, China
| | - Xia Yin
- Shaanxi Key Laboratory of Natural Products & Chemical Biology, College of Chemistry and Pharmacy, Northwest A&F University, Yangling, 712100, China
| | - Qiang Zhang
- Shaanxi Key Laboratory of Natural Products & Chemical Biology, College of Chemistry and Pharmacy, Northwest A&F University, Yangling, 712100, China
| | - Jin-Ming Gao
- Shaanxi Key Laboratory of Natural Products & Chemical Biology, College of Chemistry and Pharmacy, Northwest A&F University, Yangling, 712100, China
| |
Collapse
|
27
|
Martinez BA, Reis Rodrigues P, Nuñez Medina RM, Mondal P, Harrison NJ, Lone MA, Webster A, Gurkar AU, Grill B, Gill MS. An alternatively spliced, non-signaling insulin receptor modulates insulin sensitivity via insulin peptide sequestration in C. elegans. eLife 2020; 9:49917. [PMID: 32096469 PMCID: PMC7041946 DOI: 10.7554/elife.49917] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Accepted: 01/10/2020] [Indexed: 01/05/2023] Open
Abstract
In the nematode C. elegans, insulin signaling regulates development and aging in response to the secretion of numerous insulin peptides. Here, we describe a novel, non-signaling isoform of the nematode insulin receptor (IR), DAF-2B, that modulates insulin signaling by sequestration of insulin peptides. DAF-2B arises via alternative splicing and retains the extracellular ligand binding domain but lacks the intracellular signaling domain. A daf-2b splicing reporter revealed active regulation of this transcript through development, particularly in the dauer larva, a diapause stage associated with longevity. CRISPR knock-in of mScarlet into the daf-2b genomic locus confirmed that DAF-2B is expressed in vivo and is likely secreted. Genetic studies indicate that DAF-2B influences dauer entry, dauer recovery and adult lifespan by altering insulin sensitivity according to the prevailing insulin milieu. Thus, in C. elegans alternative splicing at the daf-2 locus generates a truncated IR that fine-tunes insulin signaling in response to the environment.
Collapse
Affiliation(s)
- Bryan A Martinez
- Department of Molecular Medicine, The Scripps Research Institute - Scripps Florida, Jupiter, United States
| | - Pedro Reis Rodrigues
- Department of Molecular Medicine, The Scripps Research Institute - Scripps Florida, Jupiter, United States
| | - Ricardo M Nuñez Medina
- Department of Molecular Medicine, The Scripps Research Institute - Scripps Florida, Jupiter, United States
| | - Prosenjit Mondal
- Department of Molecular Medicine, The Scripps Research Institute - Scripps Florida, Jupiter, United States
| | - Neale J Harrison
- Department of Molecular Medicine, The Scripps Research Institute - Scripps Florida, Jupiter, United States
| | - Museer A Lone
- Department of Molecular Medicine, The Scripps Research Institute - Scripps Florida, Jupiter, United States
| | - Amanda Webster
- Department of Molecular Medicine, The Scripps Research Institute - Scripps Florida, Jupiter, United States
| | - Aditi U Gurkar
- Department of Molecular Medicine, The Scripps Research Institute - Scripps Florida, Jupiter, United States
| | - Brock Grill
- Department of Neuroscience, The Scripps Research Institute - Scripps Florida, Jupiter, United States
| | - Matthew S Gill
- Department of Molecular Medicine, The Scripps Research Institute - Scripps Florida, Jupiter, United States
| |
Collapse
|
28
|
Xu C, Vitone GJ, Inoue K, Ng C, Zhao B. Identification of a Novel Role for Foxo3 Isoform2 in Osteoclastic Inhibition. THE JOURNAL OF IMMUNOLOGY 2019; 203:2141-2149. [PMID: 31541023 DOI: 10.4049/jimmunol.1900707] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Accepted: 08/20/2019] [Indexed: 01/08/2023]
Abstract
Foxo3 acts as an important central regulator that integrates signaling pathways and coordinates cellular responses to environmental changes. Recent studies show the involvement of Foxo3 in osteoclastogenesis and rheumatoid arthritis, which prompted us to further investigate the FOXO3 locus. Several databases document FOXO3 isoform2, an N-terminal truncated mutation of the full-length FOXO3 However, the biological function of FOXO3 isoform2 is unclear. In this study, we established a conditional allele of Foxo3 in mice that deletes the full-length Foxo3 except isoform2, a close ortholog of the human FOXO3 isoform2. Expression of Foxo3 isoform2 specifically in macrophage/osteoclast lineage suppresses osteoclastogenesis and leads to the osteopetrotic phenotype in mice. Mechanistically, Foxo3 isoform2 enhances the expression of type I IFN response genes to RANKL stimulation and thus inhibits osteoclastogenesis via endogenous IFN-β-mediated feedback inhibition. Our findings identify, to our knowledge, the first known biological function of Foxo3 isoform2 that acts as a novel osteoclastic inhibitor in bone remodeling.
Collapse
Affiliation(s)
- Cheng Xu
- Arthritis and Tissue Degeneration Program, Hospital for Special Surgery, New York, NY 10021.,David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, NY 10021
| | - Gregory J Vitone
- Arthritis and Tissue Degeneration Program, Hospital for Special Surgery, New York, NY 10021.,David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, NY 10021
| | - Kazuki Inoue
- Arthritis and Tissue Degeneration Program, Hospital for Special Surgery, New York, NY 10021.,David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, NY 10021.,Department of Medicine, Weill Cornell Medical College, New York, NY 10065; and
| | - Courtney Ng
- Arthritis and Tissue Degeneration Program, Hospital for Special Surgery, New York, NY 10021.,David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, NY 10021
| | - Baohong Zhao
- Arthritis and Tissue Degeneration Program, Hospital for Special Surgery, New York, NY 10021; .,David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, NY 10021.,Department of Medicine, Weill Cornell Medical College, New York, NY 10065; and.,Graduate Program in Cell and Development Biology, Weill Cornell Graduate School of Medical Sciences, New York, NY 10065
| |
Collapse
|
29
|
Liu HC, Peng YS, Lee HC. miRDRN-miRNA disease regulatory network: a tool for exploring disease and tissue-specific microRNA regulatory networks. PeerJ 2019; 7:e7309. [PMID: 31404401 PMCID: PMC6688598 DOI: 10.7717/peerj.7309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 06/17/2019] [Indexed: 11/20/2022] Open
Abstract
BACKGROUND MicroRNA (miRNA) regulates cellular processes by acting on specific target genes, and cellular processes proceed through multiple interactions often organized into pathways among genes and gene products. Hundreds of miRNAs and their target genes have been identified, as are many miRNA-disease associations. These, together with huge amounts of data on gene annotation, biological pathways, and protein-protein interactions are available in public databases. Here, using such data we built a database and web service platform, miRNA disease regulatory network (miRDRN), for users to construct disease and tissue-specific miRNA-protein regulatory networks, with which they may explore disease related molecular and pathway associations, or find new ones, and possibly discover new modes of drug action. METHODS Data on disease-miRNA association, miRNA-target association and validation, gene-tissue association, gene-tumor association, biological pathways, human protein interaction, gene ID, gene ontology, gene annotation, and product were collected from publicly available databases and integrated. A large set of miRNA target-specific regulatory sub-pathways (RSPs) having the form (T, G 1, G 2) was built from the integrated data and stored, where T is a miRNA-associated target gene, G 1 (G 2) is a gene/protein interacting with T (G 1). Each sequence (T, G 1, G 2) was assigned a p-value weighted by the participation of the three genes in molecular interactions and reaction pathways. RESULTS A web service platform, miRDRN (http://mirdrn.ncu.edu.tw/mirdrn/), was built. The database part of miRDRN currently stores 6,973,875 p-valued RSPs associated with 116 diseases in 78 tissue types built from 207 diseases-associated miRNA regulating 389 genes. miRDRN also provides facilities for the user to construct disease and tissue-specific miRNA regulatory networks from RSPs it stores, and to download and/or visualize parts or all of the product. User may use miRDRN to explore a single disease, or a disease-pair to gain insights on comorbidity. As demonstrations, miRDRN was applied: to explore the single disease colorectal cancer (CRC), in which 26 novel potential CRC target genes were identified; to study the comorbidity of the disease-pair Alzheimer's disease-Type 2 diabetes, in which 18 novel potential comorbid genes were identified; and, to explore possible causes that may shed light on recent failures of late-phase trials of anti-AD, BACE1 inhibitor drugs, in which genes downstream to BACE1 whose suppression may affect signal transduction were identified.
Collapse
Affiliation(s)
- Hsueh-Chuan Liu
- Department of Biomedical Sciences and Engineering, National Central University, Taoyuan City, Taiwan
| | - Yi-Shian Peng
- Department of Biomedical Sciences and Engineering, National Central University, Taoyuan City, Taiwan
| | - Hoong-Chien Lee
- Department of Biomedical Sciences and Engineering, National Central University, Taoyuan City, Taiwan
- Department of Physics, Chung Yuan Christian University, Zhongli District, Taoyuan City, Taiwan
| |
Collapse
|
30
|
Dan WJ, Zhang Q, Zhang F, Wang WW, Gao JM. Benzonate derivatives of acetophenone as potent α-glucosidase inhibitors: synthesis, structure-activity relationship and mechanism. J Enzyme Inhib Med Chem 2019; 34:937-945. [PMID: 31072245 PMCID: PMC6522914 DOI: 10.1080/14756366.2019.1604519] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
In this article, 23 compounds (6 and 7a–7v) were prepared and evaluated for their in vitro α-glucosidase inhibitory activity. The compounds 7d, 7f, 7i, 7n, 7o, 7r, 7s, 7u, and 7v displayed the α-glucosidase inhibition activity with IC50 values ranging from 1.68 to 7.88 µM. Among all tested compounds, 7u was found to be the most efficient, being 32-fold more active than the standard drug acarbose, which significantly attenuated postprandial blood glucose in mice. In addition, the compound 7u also induced the fluorescence quenching and conformational changes of enzyme, by forming α-glucosidase–7u complex in a mixed inhibition type. The thermodynamic constants recognised the interaction between 7u and α-glucosidase and was an enthalpy-driven spontaneous exothermic reaction. The synchronous fluorescence and CD spectra also indicate that the compound 7u changed the enzyme conformation. The findings identify the binding interactions between new ligands and α-glucosidase and reveal the compound 7u as a potent α-glucosidase inhibitor.
Collapse
Affiliation(s)
- Wen-Jia Dan
- a Shaanxi Key Laboratory of Natural Products and Chemical Biology, College of Chemistry and Pharmacy , Northwest A&F University , Yangling , Shaanxi , China
| | - Qiang Zhang
- a Shaanxi Key Laboratory of Natural Products and Chemical Biology, College of Chemistry and Pharmacy , Northwest A&F University , Yangling , Shaanxi , China
| | - Fan Zhang
- a Shaanxi Key Laboratory of Natural Products and Chemical Biology, College of Chemistry and Pharmacy , Northwest A&F University , Yangling , Shaanxi , China
| | - Wei-Wei Wang
- a Shaanxi Key Laboratory of Natural Products and Chemical Biology, College of Chemistry and Pharmacy , Northwest A&F University , Yangling , Shaanxi , China
| | - Jin-Ming Gao
- a Shaanxi Key Laboratory of Natural Products and Chemical Biology, College of Chemistry and Pharmacy , Northwest A&F University , Yangling , Shaanxi , China
| |
Collapse
|
31
|
Zhao S. Alternative splicing, RNA-seq and drug discovery. Drug Discov Today 2019; 24:1258-1267. [PMID: 30953866 DOI: 10.1016/j.drudis.2019.03.030] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Revised: 02/14/2019] [Accepted: 03/28/2019] [Indexed: 12/27/2022]
Abstract
Alternative splicing, hereafter referred to as AS, is an essential component of gene expression regulation that contributes to the diversity of proteomes. Recent developments in RNA sequencing (RNA-seq) technologies, combined with the advent of computational tools, have enabled transcriptome-wide studies of AS at an unprecedented scale and resolution. RNA mis-splicing can cause human disease, and to target alternative splicing has led to the development of novel therapeutics. Splice variants diversify the repertoire of biomarkers and functionally contribute to drug resistance. Our expanding knowledge of AS variation in human populations holds great promise for improving disease diagnoses and ultimately patient care in the era of sequencing and precision medicine.
Collapse
Affiliation(s)
- Shanrong Zhao
- Pfizer Worldwide Research and Development, Cambridge, MA 02139, USA.
| |
Collapse
|
32
|
Gahete MD, Del Rio-Moreno M, Camargo A, Alcala-Diaz JF, Alors-Perez E, Delgado-Lista J, Reyes O, Ventura S, Perez-Martínez P, Castaño JP, Lopez-Miranda J, Luque RM. Changes in Splicing Machinery Components Influence, Precede, and Early Predict the Development of Type 2 Diabetes: From the CORDIOPREV Study. EBioMedicine 2018; 37:356-365. [PMID: 30446432 PMCID: PMC6286259 DOI: 10.1016/j.ebiom.2018.10.056] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Revised: 10/18/2018] [Accepted: 10/24/2018] [Indexed: 12/20/2022] Open
Abstract
Background Type-2 diabetes mellitus (T2DM) is a major health problem with increasing incidence, which severely impacts cardiovascular disease. Because T2DM is associated with altered gene expression and aberrant splicing, we hypothesized that dysregulations in splicing machinery could precede, contribute to, and predict T2DM development. Methods A cohort of patients with cardiovascular disease (CORDIOPREV study) and without T2DM at baseline (at the inclusion of the study) was used (n = 215). We determined the expression of selected splicing machinery components in fasting and 4 h-postprandial peripheral blood mononuclear cells (PBMCs, obtained at baseline) from all the patients who developed T2DM during 5-years of follow-up (n = 107 incident-T2DM cases) and 108 randomly selected non-T2DM patients (controls). Serum from incident-T2DM and control patients was used to analyze in vitro the modulation of splicing machinery expression in control PBMCs from an independent cohort of healthy subjects. Findings Expression of key splicing machinery components (e.g. RNU2, RNU4 or RNU12) from fasting and 4 h-postprandial PBMCs of incident-T2DM patients was markedly altered compared to non-T2DM controls. Moreover, in vitro treatment of healthy individuals PBMCs with serum from incident-T2DM patients (compared to non-T2DM controls) reduced the expression of splicing machinery elements found down-regulated in incident-T2DM patients PBMCs. Finally, fasting/postprandial levels of several splicing machinery components in the PBMCs of CORDIOPREV patients were associated to higher risk of T2DM (Odds Ratio > 4) and could accurately predict (AUC > 0.85) T2DM development. Interpretation Our results reveal the existence of splicing machinery alterations that precede and predict T2DM development in patients with cardiovascular disease. Fund ISCIII, MINECO, CIBERObn.
Collapse
Affiliation(s)
- Manuel D Gahete
- Maimonides Institute for Biomedical Research of Cordoba (IMIBIC), Córdoba, Spain.; Universidad de Córdoba, Córdoba, Spain.; Reina Sofia University Hospital, Córdoba, Spain.; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Córdoba, Spain
| | - Mercedes Del Rio-Moreno
- Maimonides Institute for Biomedical Research of Cordoba (IMIBIC), Córdoba, Spain.; Universidad de Córdoba, Córdoba, Spain.; Reina Sofia University Hospital, Córdoba, Spain.; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Córdoba, Spain
| | - Antonio Camargo
- Maimonides Institute for Biomedical Research of Cordoba (IMIBIC), Córdoba, Spain.; Universidad de Córdoba, Córdoba, Spain.; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Córdoba, Spain.; Lipid and Atherosclerosis Unit, Department of Medicine, Reina Sofia University Hospital, University of Córdoba, Córdoba, Spain
| | - Juan F Alcala-Diaz
- Maimonides Institute for Biomedical Research of Cordoba (IMIBIC), Córdoba, Spain.; Universidad de Córdoba, Córdoba, Spain.; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Córdoba, Spain.; Lipid and Atherosclerosis Unit, Department of Medicine, Reina Sofia University Hospital, University of Córdoba, Córdoba, Spain
| | - Emilia Alors-Perez
- Maimonides Institute for Biomedical Research of Cordoba (IMIBIC), Córdoba, Spain.; Universidad de Córdoba, Córdoba, Spain.; Reina Sofia University Hospital, Córdoba, Spain.; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Córdoba, Spain
| | - Javier Delgado-Lista
- Maimonides Institute for Biomedical Research of Cordoba (IMIBIC), Córdoba, Spain.; Universidad de Córdoba, Córdoba, Spain.; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Córdoba, Spain.; Lipid and Atherosclerosis Unit, Department of Medicine, Reina Sofia University Hospital, University of Córdoba, Córdoba, Spain
| | - Oscar Reyes
- Maimonides Institute for Biomedical Research of Cordoba (IMIBIC), Córdoba, Spain.; Universidad de Córdoba, Córdoba, Spain.; Reina Sofia University Hospital, Córdoba, Spain.; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Córdoba, Spain.; Department of Computer Sciences, University of Cordoba, Córdoba, Spain
| | - Sebastian Ventura
- Maimonides Institute for Biomedical Research of Cordoba (IMIBIC), Córdoba, Spain.; Universidad de Córdoba, Córdoba, Spain.; Reina Sofia University Hospital, Córdoba, Spain.; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Córdoba, Spain.; Department of Computer Sciences, University of Cordoba, Córdoba, Spain
| | - Pablo Perez-Martínez
- Maimonides Institute for Biomedical Research of Cordoba (IMIBIC), Córdoba, Spain.; Universidad de Córdoba, Córdoba, Spain.; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Córdoba, Spain.; Lipid and Atherosclerosis Unit, Department of Medicine, Reina Sofia University Hospital, University of Córdoba, Córdoba, Spain
| | - Justo P Castaño
- Maimonides Institute for Biomedical Research of Cordoba (IMIBIC), Córdoba, Spain.; Universidad de Córdoba, Córdoba, Spain.; Reina Sofia University Hospital, Córdoba, Spain.; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Córdoba, Spain
| | - José Lopez-Miranda
- Maimonides Institute for Biomedical Research of Cordoba (IMIBIC), Córdoba, Spain.; Universidad de Córdoba, Córdoba, Spain.; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Córdoba, Spain.; Lipid and Atherosclerosis Unit, Department of Medicine, Reina Sofia University Hospital, University of Córdoba, Córdoba, Spain
| | - Raul M Luque
- Maimonides Institute for Biomedical Research of Cordoba (IMIBIC), Córdoba, Spain.; Universidad de Córdoba, Córdoba, Spain.; Reina Sofia University Hospital, Córdoba, Spain.; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Córdoba, Spain..
| |
Collapse
|
33
|
|
34
|
Abstract
Alternative splicing is a well-studied gene regulatory mechanism that produces biological diversity by allowing the production of multiple protein isoforms from a single gene. An involvement of alternative splicing in the key biological signalling Hippo pathway is emerging and offers new therapeutic avenues. This review discusses examples of alternative splicing in the Hippo pathway, how deregulation of these processes may contribute to disease and whether these processes offer new potential therapeutic targets.
Collapse
|
35
|
Yu F, Wei R, Yang J, Liu J, Yang K, Wang H, Mu Y, Hong T. FoxO1 inhibition promotes differentiation of human embryonic stem cells into insulin producing cells. Exp Cell Res 2017; 362:227-234. [PMID: 29157981 DOI: 10.1016/j.yexcr.2017.11.022] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Revised: 11/13/2017] [Accepted: 11/15/2017] [Indexed: 12/14/2022]
Abstract
Insulin-producing cells (IPCs) derived from human embryonic stem cells (hESCs) hold great potential for cell transplantation therapy in diabetes. Tremendous progress has been made in inducing differentiation of hESCs into IPCs in vitro, of which definitive endoderm (DE) protocol mimicking foetal pancreatic development has been widely used. However, immaturity of the obtained IPCs limits their further applications in treating diabetes. Forkhead box O1 (FoxO1) is involved in the differentiation and functional maintenance of murine pancreatic β cells, but its role in human β cell differentiation is under elucidation. Here, we showed that although FoxO1 expression level remained consistent, cytoplasmic phosphorylated FoxO1 protein level increased during IPC differentiation of hESCs induced by DE protocol. Lentiviral silencing of FoxO1 in pancreatic progenitors upregulated the levels of pancreatic islet differentiation-related genes and improved glucose-stimulated insulin secretion response in their progeny IPCs, whereas overexpression of FoxO1 showed the opposite effects. Notably, treatment with the FoxO1 inhibitor AS1842856 displayed similar effects with FoxO1 knockdown in pancreatic progenitors. These effects were closely associated with the mutually exclusive nucleocytoplasmic shuttling of FoxO1 and Pdx1 in the AS1842856-treated pancreatic progenitors. Our data demonstrated a promising effect of FoxO1 inhibition by the small molecule on gene expression profile during the differentiation, and in turn, on determining IPC maturation via modulating subcellular location of FoxO1 and Pdx1. Therefore, we identify a novel role of FoxO1 inhibition in promoting IPC differentiation of hESCs, which may provide clues for induction of mature β cells from hESCs and clinical applications in regenerative medicine.
Collapse
Affiliation(s)
- Fei Yu
- Department of Endocrinology and Metabolism, Peking University Third Hospital, 49 North Garden Road, Haidian District, Beijing 100191, China; Clinical Stem Cell Research Center, Peking University Third Hospital, Beijing 100191, China
| | - Rui Wei
- Department of Endocrinology and Metabolism, Peking University Third Hospital, 49 North Garden Road, Haidian District, Beijing 100191, China; Clinical Stem Cell Research Center, Peking University Third Hospital, Beijing 100191, China.
| | - Jin Yang
- Department of Endocrinology and Metabolism, Peking University Third Hospital, 49 North Garden Road, Haidian District, Beijing 100191, China; Clinical Stem Cell Research Center, Peking University Third Hospital, Beijing 100191, China
| | - Junling Liu
- Department of Endocrinology and Metabolism, Peking University Third Hospital, 49 North Garden Road, Haidian District, Beijing 100191, China; Clinical Stem Cell Research Center, Peking University Third Hospital, Beijing 100191, China
| | - Kun Yang
- Department of Endocrinology and Metabolism, Peking University Third Hospital, 49 North Garden Road, Haidian District, Beijing 100191, China; Clinical Stem Cell Research Center, Peking University Third Hospital, Beijing 100191, China
| | - Haining Wang
- Department of Endocrinology and Metabolism, Peking University Third Hospital, 49 North Garden Road, Haidian District, Beijing 100191, China; Clinical Stem Cell Research Center, Peking University Third Hospital, Beijing 100191, China
| | - Yiming Mu
- Department of Endocrinology, Chinese PLA General Hospital, Beijing 100853, China
| | - Tianpei Hong
- Department of Endocrinology and Metabolism, Peking University Third Hospital, 49 North Garden Road, Haidian District, Beijing 100191, China; Clinical Stem Cell Research Center, Peking University Third Hospital, Beijing 100191, China.
| |
Collapse
|
36
|
Zhu HQ, Gao FH. The Molecular Mechanisms of Regulation on USP2's Alternative Splicing and the Significance of Its Products. Int J Biol Sci 2017; 13:1489-1496. [PMID: 29230097 PMCID: PMC5723915 DOI: 10.7150/ijbs.21637] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Accepted: 09/10/2017] [Indexed: 01/06/2023] Open
Abstract
Ubiquitin-specific protease 2 (USP2) has a regulatory function in cell growth or death and is involved in the pathogenesis of various diseases. USP2 gene can generate 7 splicing variants through alternative splicing, and 5 variants respectively as USP2-201, USP2-202, USP2-204, USP2-205, USP2-206 can encode proteins. The influence of circadian rhythm, nutrition and androgen on specific signaling molecules or cytokines can regulate the alternative splicing of USP2. Specifically, PKC activator, IL-1β, TNF-α, PDGF-BB, TGF-β1 are all regulatory factors for USP2's alternative splicing. USP2-201 plays a crucial role in cell cycle progression, and is also of great significance in EGFR recycling. USP2-202 can activate apoptosis signaling pathway to participate in cell apoptosis, and USP2-204 can induce cell anti-virus reaction to decrease. In general, we collect and summarize the factors involved in the alternative splicing of USP2 in this review to further understand the mechanism behind the USP2's alternative splicing.
Collapse
Affiliation(s)
| | - Feng-Hou Gao
- Department of Oncology, Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine, 639 Zhi Zao Ju Rd, Shanghai, China
| |
Collapse
|