251
|
Garcia-Segura ME, Durainayagam BR, Liggi S, Graça G, Jimenez B, Dehghan A, Tzoulaki I, Karaman I, Elliott P, Griffin JL. Pathway-based integration of multi-omics data reveals lipidomics alterations validated in an Alzheimer's disease mouse model and risk loci carriers. J Neurochem 2023. [PMID: 36326588 DOI: 10.1101/2021.05.10.21255052v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/06/2023]
Abstract
Alzheimer's disease (AD) is a highly prevalent neurodegenerative disorder. Despite increasing evidence of the importance of metabolic dysregulation in AD, the underlying metabolic changes that may impact amyloid plaque formation are not understood, particularly for late-onset AD. This study analyzed genome-wide association studies (GWAS), transcriptomics, and proteomics data obtained from several data repositories to obtain differentially expressed (DE) multi-omics elements in mouse models of AD. We characterized the metabolic modulation in these data sets using gene ontology, transcription factor, pathway, and cell-type enrichment analyses. A predicted lipid signature was extracted from genome-scale metabolic networks (GSMN) and subsequently validated in a lipidomic data set derived from cortical tissue of ABCA-7 null mice, a mouse model of one of the genes associated with late-onset AD. Moreover, a metabolome-wide association study (MWAS) was performed to further characterize the association between dysregulated lipid metabolism in human blood serum and genes associated with AD risk. We found 203 DE transcripts, 164 DE proteins, and 58 DE GWAS-derived mouse orthologs associated with significantly enriched metabolic biological processes. Lipid and bioenergetic metabolic pathways were significantly over-represented across the AD multi-omics data sets. Microglia and astrocytes were significantly enriched in the lipid-predominant AD-metabolic transcriptome. We also extracted a predicted lipid signature that was validated and robustly modeled class separation in the ABCA7 mice cortical lipidome, with 11 of these lipid species exhibiting statistically significant modulations. MWAS revealed 298 AD single nucleotide polymorphisms-metabolite associations, of which 70% corresponded to lipid classes. These results support the importance of lipid metabolism dysregulation in AD and highlight the suitability of mapping AD multi-omics data into GSMNs to identify metabolic alterations.
Collapse
Affiliation(s)
- Monica Emili Garcia-Segura
- Department of Brain Sciences, Imperial College London, London, UK
- Section of Biomolecular Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Brenan R Durainayagam
- Section of Biomolecular Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
- UK-Dementia Research Institute (UK-DRI) at Imperial College London, London, UK
| | - Sonia Liggi
- Section of Biomolecular Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Gonçalo Graça
- Section of Bioinformatics, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Beatriz Jimenez
- Section of Bioanalytical Chemistry and the National Phenome Centre, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Abbas Dehghan
- UK-Dementia Research Institute (UK-DRI) at Imperial College London, London, UK
- Department of Epidemiology and Biostatistics, Imperial College London, London, UK
- MRC Centre for Environment and Health, Imperial College London, London, UK
| | - Ioanna Tzoulaki
- UK-Dementia Research Institute (UK-DRI) at Imperial College London, London, UK
- Department of Epidemiology and Biostatistics, Imperial College London, London, UK
- National Institute for Health Research Imperial Biomedical Research Centre, Imperial College London, UK
- Department of Hygiene and Epidemiology, University of Ioannina Medical School, Ioannina, Greece
| | - Ibrahim Karaman
- Section of Bioinformatics, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
- Department of Epidemiology and Biostatistics, Imperial College London, London, UK
| | - Paul Elliott
- UK-Dementia Research Institute (UK-DRI) at Imperial College London, London, UK
- Department of Epidemiology and Biostatistics, Imperial College London, London, UK
- MRC Centre for Environment and Health, Imperial College London, London, UK
- National Institute for Health Research Imperial Biomedical Research Centre, Imperial College London, UK
| | - Julian L Griffin
- Section of Biomolecular Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
- UK-Dementia Research Institute (UK-DRI) at Imperial College London, London, UK
- Department of Biochemistry and Cambridge Systems Biology Centre, University of Cambridge, Cambridge, UK
- The Rowett Institute, University of Aberdeen, Aberdeen, Scotland
| |
Collapse
|
252
|
Cascalho M, Platt JL. TNFRSF13B in B cell responses to organ transplantation. Hum Immunol 2023; 84:27-33. [PMID: 36333165 PMCID: PMC10429825 DOI: 10.1016/j.humimm.2022.09.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 09/14/2022] [Accepted: 09/27/2022] [Indexed: 11/07/2022]
Abstract
Antibodies directed against organ transplants are thought to pose the most vexing hurdle to enduring function and survival of the transplants, particularly organ xenotransplants, and accordingly basic and clinical investigation has focused on elucidating the specificity and pathogenicity of graft-specific antibodies. While much has been learned about these matters, far less is known about the B cells producing graft-specific antibodies and why these antibodies appear to injure some grafts but not others. With the goal of addressing those questions, we have investigated the properties of tumor necrosis factor receptor super family-13B (TNFRSF13B), which regulates various aspects of B cell responses. A full understanding of the functions of TNFRSF13B however is hindered by extreme polymorphism and by diversity of interactions of the protein. Nevertheless, TNFRSF13B variants have been found to exert distinct impact on natural and elicited antibody responses and host defense and mutations of TNFRSF13B have been found to influence the propensity for development of antibody-mediated rejection of organ transplants. Because B cell responses potentially limit application of xenotransplantation, understanding how TNFRSF13B diversity and TNFRSF13B variants govern immunity in xenotransplantation could inspire development of novel therapeutics that could in turn accelerate clinical implementation of xenotransplantation.
Collapse
Affiliation(s)
- Marilia Cascalho
- Department of Surgery and Department of Microbiology & Immunology, University of Michigan, Ann Arbor, MI, United States.
| | - Jeffrey L Platt
- Department of Surgery and Department of Microbiology & Immunology, University of Michigan, Ann Arbor, MI, United States.
| |
Collapse
|
253
|
Naaz S, Sakib N, Houserova D, Badve R, Crucello A, Borchert GM. Characterization of a novel sRNA contributing to biofilm formation in Salmonella enterica serovar Typhimurium. MICROPUBLICATION BIOLOGY 2023; 2023:10.17912/micropub.biology.000796. [PMID: 37151214 PMCID: PMC10160853 DOI: 10.17912/micropub.biology.000796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Figures] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Revised: 04/20/2023] [Accepted: 01/01/1970] [Indexed: 05/09/2023]
Abstract
Small RNAs (sRNAs) are short noncoding RNAs of ~50-200 nucleotides believed to primarily function in regulating crucial activities in bacteria during periods of cellular stress. This study examined the relevance of specific sRNAs on biofilm formation in nutrient starved Salmonella enterica serovar Typhimurium. Eight unique sRNAs were selected for deletion primarily based on their genomic location and/or putative targets. Quantitative and qualitative analyses confirm one of these, sRNA1186573, is required for efficient biofilm formation in S. enterica further highlighting the significance of sRNAs during Salmonella stress response.
Collapse
Affiliation(s)
- Sayema Naaz
- Department of Pharmacology, College of Medicine, University of South Alabama, Mobile, AL
| | - Najmuj Sakib
- Department of Pharmacology, College of Medicine, University of South Alabama, Mobile, AL
| | - Dominika Houserova
- Department of Pharmacology, College of Medicine, University of South Alabama, Mobile, AL
| | - Rani Badve
- Department of Pharmacology, College of Medicine, University of South Alabama, Mobile, AL
| | - Aline Crucello
- Department of Pharmacology, College of Medicine, University of South Alabama, Mobile, AL
| | - Glen M Borchert
- Department of Pharmacology, College of Medicine, University of South Alabama, Mobile, AL
- Correspondence to: Glen M Borchert (
)
| |
Collapse
|
254
|
Sghaier N, Ayed RB, Rebai A. Application of data fusion modeling for the prediction of auxin response elements in Zea mays for food security purposes. Genomics Inform 2022; 20:e45. [PMID: 36617652 PMCID: PMC9847374 DOI: 10.5808/gi.22056] [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: 08/30/2022] [Accepted: 12/12/2022] [Indexed: 12/31/2022] Open
Abstract
Food security will be affected by climate change worldwide, particularly in the developingworld, where the most important food products originate from plants. Plants are often exposed to environmental stresses that may affect their growth, development, yield, and foodquality. Auxin is a hormone that plays a critical role in improving plants' tolerance of environmental conditions. Auxin controls the expression of many stress-responsive genes inplants by interacting with specific cis-regulatory elements called auxin-responsive elements (AuxREs). In this work, we performed an in silico prediction of AuxREs in promotersof five auxin-responsive genes in Zea mays. We applied a data fusion approach based onthe combined use of Dempster-Shafer evidence theory and fuzzy sets. Auxin has a directimpact on cell membrane proteins. The short-term auxin response may be represented bythe regulation of transmembrane gene expression. The detection of an AuxRE in the promoter of prolyl oligopeptidase (POP) in Z. mays and the 3-fold overexpression of this geneunder auxin treatment for 30 min indicated the role of POP in maize auxin response. POP isregulated by auxin to perform stress adaptation. In addition, the detection of two AuxRETGTCTC motifs in the upstream sequence of the bx1 gene suggests that bx1 can be regulated by auxin. Auxin may also be involved in the regulation of dehydration-responsive element-binding and some members of the protein kinase superfamily.
Collapse
Affiliation(s)
- Nesrine Sghaier
- Laboratory of Advanced Technology and Intelligent Systems, National Engineering School of Sousse, Sousse 4023, Tunisia,Corresponding author E-mail:
| | - Rayda Ben Ayed
- Department of Agronomy and Plant Biotechnology, National Institute of Agronomy of Tunisia (INAT), 43 Avenue Charles Nicolle, 1082 El Mahrajène, University of Carthage-Tunis, Tunisia,Laboratory of Extremophile Plants, Centre of Biotechnology of Borj-Cédria, B.P. 901, Hammam Lif 2050, TunisiaTunisia
| | - Ahmed Rebai
- Laboratory of Molecular and Cellular Screening Processes, Sfax Biotechnology Center, B.P 1177, Sfax 3018,Tunisia
| |
Collapse
|
255
|
Morel B, Williams TA, Stamatakis A. Asteroid: a new algorithm to infer species trees from gene trees under high proportions of missing data. Bioinformatics 2022; 39:6964379. [PMID: 36576010 PMCID: PMC9838317 DOI: 10.1093/bioinformatics/btac832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 12/12/2022] [Accepted: 12/26/2022] [Indexed: 12/29/2022] Open
Abstract
MOTIVATION Missing data and incomplete lineage sorting (ILS) are two major obstacles to accurate species tree inference. Gene tree summary methods such as ASTRAL and ASTRID have been developed to account for ILS. However, they can be severely affected by high levels of missing data. RESULTS We present Asteroid, a novel algorithm that infers an unrooted species tree from a set of unrooted gene trees. We show on both empirical and simulated datasets that Asteroid is substantially more accurate than ASTRAL and ASTRID for very high proportions (>80%) of missing data. Asteroid is several orders of magnitude faster than ASTRAL for datasets that contain thousands of genes. It offers advanced features such as parallelization, support value computation and support for multi-copy and multifurcating gene trees. AVAILABILITY AND IMPLEMENTATION Asteroid is freely available at https://github.com/BenoitMorel/Asteroid. SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
| | - Tom A Williams
- School of Biological Sciences, University of Bristol, Bristol BS8, UK
| | - Alexandros Stamatakis
- Computational Molecular Evolution Group, Heidelberg Institute for Theoretical Studies, Heidelberg 69118, Germany,Institute for Theoretical Informatics, Karlsruhe Institute of Technology, Karlsruhe 76131, Germany
| |
Collapse
|
256
|
Avramouli A, Krokidis MG, Exarchos TP, Vlamos P. In Silico Structural Analysis Predicting the Pathogenicity of PLP1 Mutations in Multiple Sclerosis. Brain Sci 2022; 13:42. [PMID: 36672024 PMCID: PMC9856082 DOI: 10.3390/brainsci13010042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 12/19/2022] [Accepted: 12/20/2022] [Indexed: 12/28/2022] Open
Abstract
The X chromosome gene PLP1 encodes myelin proteolipid protein (PLP), the most prevalent protein in the myelin sheath surrounding the central nervous system. X-linked dysmyelinating disorders such as Pelizaeus-Merzbacher disease (PMD) or spastic paraplegia type 2 (SPG2) are typically caused by point mutations in PLP1. Nevertheless, numerous case reports have shown individuals with PLP1 missense point mutations which also presented clinical symptoms and indications that were consistent with the diagnostic criteria of multiple sclerosis (MS), a disabling disease of the brain and spinal cord with no current cure. Computational structural biology methods were used to assess the impact of these mutations on the stability and flexibility of PLP structure in order to determine the role of PLP1 mutations in MS pathogenicity. The analysis showed that most of the variants can alter the functionality of the protein structure such as R137W variants which results in loss of helix and H140Y which alters the ordered protein interface. In silico genomic methods were also performed to predict the significance of these mutations associated with impairments in protein functionality and could suggest a better definition for therapeutic strategies and clinical application in MS patients.
Collapse
Affiliation(s)
| | - Marios G. Krokidis
- Bioinformatics and Human Electrophysiology Laboratory, Department of Informatics, Ionian University, 491 00 Corfu, Greece
| | | | | |
Collapse
|
257
|
Muscle Regeneration in Holothurians without the Upregulation of Muscle Genes. Int J Mol Sci 2022; 23:ijms232416037. [PMID: 36555677 PMCID: PMC9785333 DOI: 10.3390/ijms232416037] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 12/10/2022] [Accepted: 12/14/2022] [Indexed: 12/23/2022] Open
Abstract
The holothurian Eupentacta fraudatrix is capable of fully restoring its muscles after transverse dissection. Although the regeneration of these structures is well studied at the cellular level, the molecular basis of the process remains poorly understood. To identify genes that may be involved in the regulation of muscle regeneration, the transcriptome of the longitudinal muscle band of E. fraudatrix has been sequenced at different time periods post-injury. An analysis of the map of biological processes and pathways has shown that most genes associated with myogenesis decrease their expression during the regeneration. The only exception is the genes united by the GO term "heart valve development". This may indicate the antiquity of mechanisms of mesodermal structure transformation, which was co-opted into various morphogeneses in deuterostomes. Two groups of genes that play a key role in the regeneration have been analyzed: transcription factors and matrix metalloproteinases. A total of six transcription factor genes (Ef-HOX5, Ef-ZEB2, Ef-RARB, Ef-RUNX1, Ef-SOX17, and Ef-ZNF318) and seven matrix metalloproteinase genes (Ef-MMP11, Ef-MMP13, Ef-MMP13-1, Ef-MMP16-2, Ef-MMP16-3, Ef-MMP24, and Ef-MMP24-1) showing differential expression during myogenesis have been revealed. The identified genes are assumed to be involved in the muscle regeneration in holothurians.
Collapse
|
258
|
Haque OI, Chandrasekaran A, Nabi F, Ahmad O, Marques JP, Ahmad T. A novel compound heterozygous BEST1 gene mutation in two siblings causing autosomal recessive bestrophinopathy. BMC Ophthalmol 2022; 22:493. [PMID: 36527004 PMCID: PMC9756692 DOI: 10.1186/s12886-022-02703-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 11/23/2022] [Indexed: 12/23/2022] Open
Abstract
PURPOSE To describe the clinical features, imaging characteristics, and genetic test results associated with a novel compound heterozygous mutation of the BEST1 gene in two siblings with autosomal recessive bestrophinopathy. METHODS Two siblings underwent a complete ophthalmic examination, including dilated fundus examination, fundus photography, fundus autofluorescence imaging, spectral-domain optical coherence tomography, fluorescein angiography, electroretinography, and electrooculography. A clinical diagnosis of autosomal recessive bestrophinopathy was established based on ocular examination and multimodal retinal imaging. Subsequently, clinical exome sequencing consisting of a panel of 6670 genes was carried out to confirm the diagnosis and assess genetic alterations in the protein-coding region of the genome of the patients. The identified mutations were tested in the two affected siblings and one of their parents. RESULTS Two siblings (a 17-year-old female and a 15-year-old male) presented with reduced visual acuity and bilaterally symmetrical subretinal deposits of hyperautofluorescent materials in the posterior pole, which showed staining in the late phase of fluorescein angiogram. Spectral-domain optical coherence tomography demonstrated hyperreflective subretinal deposits and subretinal fluid accumulation. Both patients shared two mutations in the protein-coding region of the BEST1 gene, c.103G > A, p.(Glu35Lys) and c.313C > A, p.(Arg105Ser) (a novel disease-causing mutation). Sanger sequencing confirmed that the unaffected mother of the proband was carrying p.(Glu35Lys) variant in a heterozygous state. CONCLUSIONS We have identified and described the phenotype of a novel disease-causing mutation NM_004183.4:c.313C > A, p.(Arg105Ser) in a heterozygous state along with a previously reported mutation NM_004183.4:c.103G > A, p.(Glu35Lys) of the BEST1 gene in two related patients with autosomal recessive bestrophinopathy.
Collapse
Affiliation(s)
| | | | - Faisal Nabi
- Interdisciplinary Biotechnology Unit, Aligarh Muslim University, Aligarh, India
| | - Owais Ahmad
- Department of Microbiology and Cell Biology, Indian Institute of Science, Bangalore, India
| | - João Pedro Marques
- Ophthalmology Unit, Centro Hospitalar E Universitário de Coimbra (CHUC), Coimbra, Portugal
| | | |
Collapse
|
259
|
Joyce W, Ripley DM, Gillis T, Black AC, Shiels HA, Hoffmann FG. A Revised Perspective on the Evolution of Troponin I and Troponin T Gene Families in Vertebrates. Genome Biol Evol 2022; 15:6904147. [PMID: 36518048 PMCID: PMC9825255 DOI: 10.1093/gbe/evac173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 11/29/2022] [Accepted: 12/05/2022] [Indexed: 12/23/2022] Open
Abstract
The troponin (Tn) complex, responsible for the Ca2+ activation of striated muscle, is composed of three interacting protein subunits: TnC, TnI, and TnT, encoded by TNNC, TNNI, and TNNT genes. TNNI and TNNT are sister gene families, and in mammals the three TNNI paralogs (TNNI1, TNNI2, TNNI3), which encode proteins with tissue-specific expression, are each in close genomic proximity with one of the three TNNT paralogs (TNNT2, TNNT3, TNNT1, respectively). It has been widely presumed that all vertebrates broadly possess genes of these same three classes, although earlier work has overlooked jawless fishes (cyclostomes) and cartilaginous fishes (chimeras, rays, and sharks), which are distantly related to other jawed vertebrates. With a new phylogenetic and synteny analysis of a diverse array of vertebrates including these taxonomic groups, we define five distinct TNNI classes (TNNI1-5), with TNNI4 and TNNI5 being only present in non-amniote vertebrates and typically found in tandem, and four classes of TNNT (TNNT1-4). These genes are located in four genomic loci that were generated by the 2R whole-genome duplications. TNNI3, encoding "cardiac TnI" in tetrapods, was independently lost in cartilaginous and ray-finned fishes. Instead, ray-finned fishes predominantly express TNNI1 in the heart. TNNI5 is highly expressed in shark hearts and contains a N-terminal extension similar to that of TNNI3 found in tetrapod hearts. Given that TNNI3 and TNNI5 are distantly related, this supports the hypothesis that the N-terminal extension may be an ancestral feature of vertebrate TNNI and not an innovation unique to TNNI3, as has been commonly believed.
Collapse
Affiliation(s)
| | - Daniel M Ripley
- Division of Cardiovascular Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9PL, United Kingdom
| | - Todd Gillis
- Department of Integrative Biology, University of Guelph, Guelph, Ontario N1G 2W1, Canada
| | - Amanda Coward Black
- Department of Biochemistry, Molecular Biology, Entomology, and Plant Pathology, Mississippi State University, Starkville, Mississippi 39762, USA
| | - Holly A Shiels
- Division of Cardiovascular Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9PL, United Kingdom
| | | |
Collapse
|
260
|
Liu M, Chen MY, Huang JM, Liu Q, Wang L, Liu R, Yang N, Huang WH, Zhang W. LncRNA weighted gene co-expression network analysis reveals novel biomarkers related to prostate cancer metastasis. BMC Med Genomics 2022; 15:256. [PMID: 36514044 PMCID: PMC9745985 DOI: 10.1186/s12920-022-01410-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Accepted: 12/07/2022] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Most prostate cancer patients die from metastasis and lack accurate efficacious biomarkers to monitor the disease behavior, optimize treatment and assess prognosis. Herein, we aimed to identify meaningful lncRNA biomarkers associated with prostate cancer metastatic progression. METHODS By repurposing microarray probes, 11,624 lncRNAs in prostate cancer were obtained from Gene Expression Omnibus database (GSE46691, N = 545; GSE29079, N = 235; GSE94767, N = 130). Weighted gene co-expression network analysis was applied to determine the co-expression lncRNA network pertinent to metastasis. Hub lncRNAs were screened. RNA-seq and clinical data from the Cancer Genome Atlas prostate cancer (TCGA-PRAD) cohort (N = 531) were analyzed. Transwell assay and bioinformatic analysis were performed for mechanism research. RESULTS The high expression levels of nine hub lncRNAs (FTX, AC005261.1, NORAD, LINC01578, AC004542.2, ZFAS1, EBLN3P, THUMPD3-AS1, GAS5) were significantly associated with Gleason score and increased probability of metastatic progression. Among these lncRNAs, ZFAS1 had the consistent trends of expression in all of the analysis from different cohorts, and the Kaplan-Meier survival analyses showed higher expression of ZFAS1 was associated with shorter relapse free survival. In-vitro studies confirmed that downregulation of ZFAS1 decreased prostate cancer cell migration. CONCLUSION We offered some new insights into discovering lncRNA markers correlated with metastatic progression of prostate cancer using the WGCNA. Some may serve as potential prognostic biomarkers and therapeutic targets for advanced metastatic prostate cancer.
Collapse
Affiliation(s)
- Miao Liu
- grid.216417.70000 0001 0379 7164Department of Clinical Pharmacology, Xiangya Hospital, Central South University, 87 Xiangya Road, 410008 Changsha, People’s Republic of China ,grid.216417.70000 0001 0379 7164Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University, 110 Xiangya Road, 410078 Changsha, People’s Republic of China ,National Clinical Research Center for Geriatric Disorders, 87 Xiangya Road, Hunan 410008 Changsha, People’s Republic of China
| | - Man-Yun Chen
- grid.216417.70000 0001 0379 7164Department of Clinical Pharmacology, Xiangya Hospital, Central South University, 87 Xiangya Road, 410008 Changsha, People’s Republic of China ,grid.216417.70000 0001 0379 7164Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University, 110 Xiangya Road, 410078 Changsha, People’s Republic of China ,National Clinical Research Center for Geriatric Disorders, 87 Xiangya Road, Hunan 410008 Changsha, People’s Republic of China
| | - Jia-Meng Huang
- grid.216417.70000 0001 0379 7164Department of Clinical Pharmacology, Xiangya Hospital, Central South University, 87 Xiangya Road, 410008 Changsha, People’s Republic of China ,grid.216417.70000 0001 0379 7164Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University, 110 Xiangya Road, 410078 Changsha, People’s Republic of China ,National Clinical Research Center for Geriatric Disorders, 87 Xiangya Road, Hunan 410008 Changsha, People’s Republic of China
| | - Qian Liu
- grid.216417.70000 0001 0379 7164Department of Clinical Pharmacology, Xiangya Hospital, Central South University, 87 Xiangya Road, 410008 Changsha, People’s Republic of China ,grid.216417.70000 0001 0379 7164Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University, 110 Xiangya Road, 410078 Changsha, People’s Republic of China ,National Clinical Research Center for Geriatric Disorders, 87 Xiangya Road, Hunan 410008 Changsha, People’s Republic of China
| | - Lin Wang
- grid.216417.70000 0001 0379 7164Department of Clinical Pharmacology, Xiangya Hospital, Central South University, 87 Xiangya Road, 410008 Changsha, People’s Republic of China ,grid.216417.70000 0001 0379 7164Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University, 110 Xiangya Road, 410078 Changsha, People’s Republic of China ,National Clinical Research Center for Geriatric Disorders, 87 Xiangya Road, Hunan 410008 Changsha, People’s Republic of China
| | - Rong Liu
- grid.216417.70000 0001 0379 7164Department of Clinical Pharmacology, Xiangya Hospital, Central South University, 87 Xiangya Road, 410008 Changsha, People’s Republic of China ,grid.216417.70000 0001 0379 7164Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University, 110 Xiangya Road, 410078 Changsha, People’s Republic of China ,National Clinical Research Center for Geriatric Disorders, 87 Xiangya Road, Hunan 410008 Changsha, People’s Republic of China
| | - Nian Yang
- grid.216417.70000 0001 0379 7164Department of Clinical Pharmacology, Xiangya Hospital, Central South University, 87 Xiangya Road, 410008 Changsha, People’s Republic of China ,grid.216417.70000 0001 0379 7164Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University, 110 Xiangya Road, 410078 Changsha, People’s Republic of China ,National Clinical Research Center for Geriatric Disorders, 87 Xiangya Road, Hunan 410008 Changsha, People’s Republic of China
| | - Wei-Hua Huang
- grid.216417.70000 0001 0379 7164Department of Clinical Pharmacology, Xiangya Hospital, Central South University, 87 Xiangya Road, 410008 Changsha, People’s Republic of China ,grid.216417.70000 0001 0379 7164Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University, 110 Xiangya Road, 410078 Changsha, People’s Republic of China ,National Clinical Research Center for Geriatric Disorders, 87 Xiangya Road, Hunan 410008 Changsha, People’s Republic of China
| | - Wei Zhang
- grid.216417.70000 0001 0379 7164Department of Clinical Pharmacology, Xiangya Hospital, Central South University, 87 Xiangya Road, 410008 Changsha, People’s Republic of China ,grid.216417.70000 0001 0379 7164Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University, 110 Xiangya Road, 410078 Changsha, People’s Republic of China ,National Clinical Research Center for Geriatric Disorders, 87 Xiangya Road, Hunan 410008 Changsha, People’s Republic of China
| |
Collapse
|
261
|
Grigoriadis D, Perdikopanis N, Georgakilas GK, Hatzigeorgiou AG. DeepTSS: multi-branch convolutional neural network for transcription start site identification from CAGE data. BMC Bioinformatics 2022; 23:395. [PMID: 36510136 PMCID: PMC9743497 DOI: 10.1186/s12859-022-04945-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Accepted: 09/16/2022] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND The widespread usage of Cap Analysis of Gene Expression (CAGE) has led to numerous breakthroughs in understanding the transcription mechanisms. Recent evidence in the literature, however, suggests that CAGE suffers from transcriptional and technical noise. Regardless of the sample quality, there is a significant number of CAGE peaks that are not associated with transcription initiation events. This type of signal is typically attributed to technical noise and more frequently to random five-prime capping or transcription bioproducts. Thus, the need for computational methods emerges, that can accurately increase the signal-to-noise ratio in CAGE data, resulting in error-free transcription start site (TSS) annotation and quantification of regulatory region usage. In this study, we present DeepTSS, a novel computational method for processing CAGE samples, that combines genomic signal processing (GSP), structural DNA features, evolutionary conservation evidence and raw DNA sequence with Deep Learning (DL) to provide single-nucleotide TSS predictions with unprecedented levels of performance. RESULTS To evaluate DeepTSS, we utilized experimental data, protein-coding gene annotations and computationally-derived genome segmentations by chromatin states. DeepTSS was found to outperform existing algorithms on all benchmarks, achieving 98% precision and 96% sensitivity (accuracy 95.4%) on the protein-coding gene strategy, with 96.66% of its positive predictions overlapping active chromatin, 98.27% and 92.04% co-localized with at least one transcription factor and H3K4me3 peak. CONCLUSIONS CAGE is a key protocol in deciphering the language of transcription, however, as every experimental protocol, it suffers from biological and technical noise that can severely affect downstream analyses. DeepTSS is a novel DL-based method for effectively removing noisy CAGE signal. In contrast to existing software, DeepTSS does not require feature selection since the embedded convolutional layers can readily identify patterns and only utilize the important ones for the classification task. This study highlights the key role that DL can play in Molecular Biology, by removing the inherent flaws of experimental protocols, that form the backbone of contemporary research. Here, we show how DeepTSS can unleash the full potential of an already popular and mature method such as CAGE, and push the boundaries of coding and non-coding gene expression regulator research even further.
Collapse
Affiliation(s)
- Dimitris Grigoriadis
- grid.418497.7Hellenic Pasteur Institute, 11521 Athens, Greece ,grid.410558.d0000 0001 0035 6670Department of Computer Science and Biomedical Informatics, University of Thessaly, 35131 Lamia, Greece
| | - Nikos Perdikopanis
- grid.418497.7Hellenic Pasteur Institute, 11521 Athens, Greece ,grid.5216.00000 0001 2155 0800Department of Informatics and Telecommunications, National and Kapodistrian University of Athens, 15784 Athens, Greece ,grid.410558.d0000 0001 0035 6670Department of Electrical and Computer Engineering, University of Thessaly, 38221 Volos, Greece
| | - Georgios K. Georgakilas
- grid.410558.d0000 0001 0035 6670Department of Electrical and Computer Engineering, University of Thessaly, 38221 Volos, Greece ,ommAI Technologies, Tallinn, Estonia
| | - Artemis G. Hatzigeorgiou
- grid.418497.7Hellenic Pasteur Institute, 11521 Athens, Greece ,grid.410558.d0000 0001 0035 6670Department of Computer Science and Biomedical Informatics, University of Thessaly, 35131 Lamia, Greece
| |
Collapse
|
262
|
Bizet M, Defrance M, Calonne E, Bontempi G, Sotiriou C, Fuks F, Jeschke J. Improving Infinium MethylationEPIC data processing: re-annotation of enhancers and long noncoding RNA genes and benchmarking of normalization methods. Epigenetics 2022; 17:2434-2454. [DOI: 10.1080/15592294.2022.2135201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Affiliation(s)
- Martin Bizet
- Laboratory of Cancer Epigenetics, Faculty of Medicine, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Matthieu Defrance
- Interuniversity Institute of Bioinformatics in Brussels (IB2), Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Emilie Calonne
- Laboratory of Cancer Epigenetics, Faculty of Medicine, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Gianluca Bontempi
- Interuniversity Institute of Bioinformatics in Brussels (IB2), Université Libre de Bruxelles (ULB), Brussels, Belgium
| | | | - François Fuks
- Laboratory of Cancer Epigenetics, Faculty of Medicine, Université Libre de Bruxelles (ULB), Brussels, Belgium
- Institut Jules Bordet, ULB, Brussels, Belgium
| | - Jana Jeschke
- Laboratory of Cancer Epigenetics, Faculty of Medicine, Université Libre de Bruxelles (ULB), Brussels, Belgium
- Institut Jules Bordet, ULB, Brussels, Belgium
| |
Collapse
|
263
|
Orabi B, Xie N, McConeghy B, Dong X, Chauve C, Hach F. Freddie: annotation-independent detection and discovery of transcriptomic alternative splicing isoforms using long-read sequencing. Nucleic Acids Res 2022; 51:e11. [PMID: 36478271 PMCID: PMC9881145 DOI: 10.1093/nar/gkac1112] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 10/26/2022] [Accepted: 11/08/2022] [Indexed: 12/13/2022] Open
Abstract
Alternative splicing (AS) is an important mechanism in the development of many cancers, as novel or aberrant AS patterns play an important role as an independent onco-driver. In addition, cancer-specific AS is potentially an effective target of personalized cancer therapeutics. However, detecting AS events remains a challenging task, especially if these AS events are novel. This is exacerbated by the fact that existing transcriptome annotation databases are far from being comprehensive, especially with regard to cancer-specific AS. Additionally, traditional sequencing technologies are severely limited by the short length of the generated reads, which rarely spans more than a single splice junction site. Given these challenges, transcriptomic long-read (LR) sequencing presents a promising potential for the detection and discovery of AS. We present Freddie, a computational annotation-independent isoform discovery and detection tool. Freddie takes as input transcriptomic LR sequencing of a sample alongside its genomic split alignment and computes a set of isoforms for the given sample. It then partitions the input reads into sets that can be processed independently and in parallel. For each partition, Freddie segments the genomic alignment of the reads into canonical exon segments. The goal of this segmentation is to be able to represent any potential isoform as a subset of these canonical exons. This segmentation is formulated as an optimization problem and is solved with a dynamic programming algorithm. Then, Freddie reconstructs the isoforms by jointly clustering and error-correcting the reads using the canonical segmentation as a succinct representation. The clustering and error-correcting step is formulated as an optimization problem-the Minimum Error Clustering into Isoforms (MErCi) problem-and is solved using integer linear programming (ILP). We compare the performance of Freddie on simulated datasets with other isoform detection tools with varying dependence on annotation databases. We show that Freddie outperforms the other tools in its accuracy, including those given the complete ground truth annotation. We also run Freddie on a transcriptomic LR dataset generated in-house from a prostate cancer cell line with a matched short-read RNA-seq dataset. Freddie results in isoforms with a higher short-read cross-validation rate than the other tested tools. Freddie is open source and available at https://github.com/vpc-ccg/freddie/.
Collapse
Affiliation(s)
- Baraa Orabi
- Department of Computer Science, the University of British Columbia, Vancouver, BC, Canada
| | - Ning Xie
- Vancouver Prostate Centre, Vancouver, BC, Canada
| | | | - Xuesen Dong
- Vancouver Prostate Centre, Vancouver, BC, Canada,Department of Urologic Sciences, the University of British Columbia, Vancouver, BC, Canada
| | - Cedric Chauve
- Department of Mathematics, Simon Fraser University, Burnaby, BC, Canada
| | - Faraz Hach
- To whom correspondence should be addressed.
| |
Collapse
|
264
|
Investigation of MicroRNA Biomarkers in Equine Distal Interphalangeal Joint Osteoarthritis. Int J Mol Sci 2022; 23:ijms232415526. [PMID: 36555166 PMCID: PMC9779011 DOI: 10.3390/ijms232415526] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 11/30/2022] [Accepted: 12/06/2022] [Indexed: 12/13/2022] Open
Abstract
Osteoarthritis of the equine distal interphalangeal joint is a common cause of lameness. MicroRNAs from biofluids are promising biomarkers and therapeutic candidates. Synovial fluid samples from horses with mild and severe equine distal interphalangeal joint osteoarthritis were submitted for small RNA sequencing. The results demonstrated that miR-92a was downregulated in equine synovial fluid from horses with severe osteoarthritis and there was a significant increase in COMP, COL1A2, RUNX2 and SOX9 following miR-92a mimic treatment of equine chondrocytes in monolayer culture. This is the first equine study to evaluate the role of miR-92a in osteoarthritic chondrocytes in vitro.
Collapse
|
265
|
Kumar CPR, Tamhankar PM, Manohar R, Sharda S, Madhavilatha GK, Thenral SG, Nair S, Bojamma AK. Exome sequencing and microarray identified a novel large exonic deletion in SYT2 gene in an ultra-rare case with recessive CMS type 7. J Genet 2022. [DOI: 10.1007/s12041-022-01409-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
266
|
Kaserman JE, Werder RB, Wang F, Matte T, Higgins MI, Dodge M, Lindstrom-Vautrin J, Bawa P, Hinds A, Bullitt E, Caballero IS, Shi X, Gerszten RE, Brunetti-Pierri N, Liesa M, Villacorta-Martin C, Hollenberg AN, Kotton DN, Wilson AA. Human iPSC-hepatocyte modeling of alpha-1 antitrypsin heterozygosity reveals metabolic dysregulation and cellular heterogeneity. Cell Rep 2022; 41:111775. [PMID: 36476855 PMCID: PMC9780780 DOI: 10.1016/j.celrep.2022.111775] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 09/28/2022] [Accepted: 11/14/2022] [Indexed: 12/12/2022] Open
Abstract
Individuals homozygous for the "Z" mutation in alpha-1 antitrypsin deficiency are known to be at increased risk for liver disease. It has also become clear that some degree of risk is similarly conferred by the heterozygous state. A lack of model systems that recapitulate heterozygosity in human hepatocytes has limited the ability to study the impact of a single Z alpha-1 antitrypsin (ZAAT) allele on hepatocyte biology. Here, we describe the derivation of syngeneic induced pluripotent stem cells (iPSCs) engineered to determine the effects of ZAAT heterozygosity in iPSC-hepatocytes (iHeps). We find that heterozygous MZ iHeps exhibit an intermediate disease phenotype and share with ZZ iHeps alterations in AAT protein processing and downstream perturbations including altered endoplasmic reticulum (ER) and mitochondrial morphology, reduced mitochondrial respiration, and branch-specific activation of the unfolded protein response in cell subpopulations. Our model of MZ heterozygosity thus provides evidence that a single Z allele is sufficient to disrupt hepatocyte homeostatic function.
Collapse
Affiliation(s)
- Joseph E. Kaserman
- Center for Regenerative Medicine (CReM) of Boston University and Boston Medical Center, Boston, MA 02118, USA,The Pulmonary Center and Department of Medicine, Boston University School of Medicine, Boston, MA 02118, USA
| | - Rhiannon B. Werder
- Center for Regenerative Medicine (CReM) of Boston University and Boston Medical Center, Boston, MA 02118, USA
| | - Feiya Wang
- Center for Regenerative Medicine (CReM) of Boston University and Boston Medical Center, Boston, MA 02118, USA
| | - Taylor Matte
- Center for Regenerative Medicine (CReM) of Boston University and Boston Medical Center, Boston, MA 02118, USA
| | - Michelle I. Higgins
- Center for Regenerative Medicine (CReM) of Boston University and Boston Medical Center, Boston, MA 02118, USA
| | - Mark Dodge
- Center for Regenerative Medicine (CReM) of Boston University and Boston Medical Center, Boston, MA 02118, USA
| | - Jonathan Lindstrom-Vautrin
- Center for Regenerative Medicine (CReM) of Boston University and Boston Medical Center, Boston, MA 02118, USA
| | - Pushpinder Bawa
- Center for Regenerative Medicine (CReM) of Boston University and Boston Medical Center, Boston, MA 02118, USA
| | - Anne Hinds
- The Pulmonary Center and Department of Medicine, Boston University School of Medicine, Boston, MA 02118, USA
| | - Esther Bullitt
- Department of Physiology and Biophysics, Boston University, Boston, MA 02118, USA
| | - Ignacio S. Caballero
- Center for Regenerative Medicine (CReM) of Boston University and Boston Medical Center, Boston, MA 02118, USA
| | - Xu Shi
- Division of Cardiovascular Medicine, Beth Israel Deaconess Hospital, Boston, MA 02118, USA
| | - Robert E. Gerszten
- Division of Cardiovascular Medicine, Beth Israel Deaconess Hospital, Boston, MA 02118, USA
| | - Nicola Brunetti-Pierri
- Telethon Institute of Genetics and Medicine, 80078 Pozzuoli, Naples, Italy,Department of Translational Medicine, Federico II University, 80131 Naples, Italy
| | - Marc Liesa
- Departments of Medicine, Endocrinology, and Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA,Institut de Biologia Molecular de Barcelona (IBMB-CSIC), 08028 Barcelona, Catalonia, Spain
| | - Carlos Villacorta-Martin
- Center for Regenerative Medicine (CReM) of Boston University and Boston Medical Center, Boston, MA 02118, USA
| | - Anthony N. Hollenberg
- Joan and Sanford I. Weill Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10065, USA
| | - Darrell N. Kotton
- Center for Regenerative Medicine (CReM) of Boston University and Boston Medical Center, Boston, MA 02118, USA,The Pulmonary Center and Department of Medicine, Boston University School of Medicine, Boston, MA 02118, USA
| | - Andrew A. Wilson
- Center for Regenerative Medicine (CReM) of Boston University and Boston Medical Center, Boston, MA 02118, USA,The Pulmonary Center and Department of Medicine, Boston University School of Medicine, Boston, MA 02118, USA,Lead contact,Correspondence:
| |
Collapse
|
267
|
Leman R, Parfait B, Vidaud D, Girodon E, Pacot L, Le Gac G, Ka C, Ferec C, Fichou Y, Quesnelle C, Aucouturier C, Muller E, Vaur D, Castera L, Boulouard F, Ricou A, Tubeuf H, Soukarieh O, Gaildrat P, Riant F, Guillaud‐Bataille M, Caputo SM, Caux‐Moncoutier V, Boutry‐Kryza N, Bonnet‐Dorion F, Schultz I, Rossing M, Quenez O, Goldenberg L, Harter V, Parsons MT, Spurdle AB, Frébourg T, Martins A, Houdayer C, Krieger S. SPiP: Splicing Prediction Pipeline, a machine learning tool for massive detection of exonic and intronic variant effects on mRNA splicing. Hum Mutat 2022; 43:2308-2323. [PMID: 36273432 PMCID: PMC10946553 DOI: 10.1002/humu.24491] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 10/06/2022] [Accepted: 10/18/2022] [Indexed: 01/25/2023]
Abstract
Modeling splicing is essential for tackling the challenge of variant interpretation as each nucleotide variation can be pathogenic by affecting pre-mRNA splicing via disruption/creation of splicing motifs such as 5'/3' splice sites, branch sites, or splicing regulatory elements. Unfortunately, most in silico tools focus on a specific type of splicing motif, which is why we developed the Splicing Prediction Pipeline (SPiP) to perform, in one single bioinformatic analysis based on a machine learning approach, a comprehensive assessment of the variant effect on different splicing motifs. We gathered a curated set of 4616 variants scattered all along the sequence of 227 genes, with their corresponding splicing studies. The Bayesian analysis provided us with the number of control variants, that is, variants without impact on splicing, to mimic the deluge of variants from high-throughput sequencing data. Results show that SPiP can deal with the diversity of splicing alterations, with 83.13% sensitivity and 99% specificity to detect spliceogenic variants. Overall performance as measured by area under the receiving operator curve was 0.986, better than SpliceAI and SQUIRLS (0.965 and 0.766) for the same data set. SPiP lends itself to a unique suite for comprehensive prediction of spliceogenicity in the genomic medicine era. SPiP is available at: https://sourceforge.net/projects/splicing-prediction-pipeline/.
Collapse
Affiliation(s)
- Raphaël Leman
- Laboratoire de Biologie et Génétique du CancerCentre François BaclesseCaenFrance
- Inserm U1245, UNIROUEN, FHU‐G4 génomiqueNormandie UniversitéRouenFrance
- UNICAENNormandie UniversitéCaenFrance
| | - Béatrice Parfait
- Service de Génétique et Biologie Moléculaires, APHP, HUPCHôpital CochinParisFrance
| | - Dominique Vidaud
- Service de Génétique et Biologie Moléculaires, APHP, HUPCHôpital CochinParisFrance
| | - Emmanuelle Girodon
- Service de Génétique et Biologie Moléculaires, APHP, HUPCHôpital CochinParisFrance
| | - Laurence Pacot
- Service de Génétique et Biologie Moléculaires, APHP, HUPCHôpital CochinParisFrance
| | - Gérald Le Gac
- Inserm UMR1078, Genetics, Functional Genomics and BiotechnologyUniversité de Bretagne OccidentaleBrestFrance
| | - Chandran Ka
- Inserm UMR1078, Genetics, Functional Genomics and BiotechnologyUniversité de Bretagne OccidentaleBrestFrance
| | - Claude Ferec
- Inserm UMR1078, Genetics, Functional Genomics and BiotechnologyUniversité de Bretagne OccidentaleBrestFrance
| | - Yann Fichou
- Inserm UMR1078, Genetics, Functional Genomics and BiotechnologyUniversité de Bretagne OccidentaleBrestFrance
| | - Céline Quesnelle
- Laboratoire de Biologie et Génétique du CancerCentre François BaclesseCaenFrance
| | - Camille Aucouturier
- Laboratoire de Biologie et Génétique du CancerCentre François BaclesseCaenFrance
- Inserm U1245, UNIROUEN, FHU‐G4 génomiqueNormandie UniversitéRouenFrance
| | - Etienne Muller
- Laboratoire de Biologie et Génétique du CancerCentre François BaclesseCaenFrance
| | - Dominique Vaur
- Laboratoire de Biologie et Génétique du CancerCentre François BaclesseCaenFrance
- Inserm U1245, UNIROUEN, FHU‐G4 génomiqueNormandie UniversitéRouenFrance
| | - Laurent Castera
- Laboratoire de Biologie et Génétique du CancerCentre François BaclesseCaenFrance
- Inserm U1245, UNIROUEN, FHU‐G4 génomiqueNormandie UniversitéRouenFrance
| | - Flavie Boulouard
- Laboratoire de Biologie et Génétique du CancerCentre François BaclesseCaenFrance
- Inserm U1245, UNIROUEN, FHU‐G4 génomiqueNormandie UniversitéRouenFrance
| | - Agathe Ricou
- Laboratoire de Biologie et Génétique du CancerCentre François BaclesseCaenFrance
- Inserm U1245, UNIROUEN, FHU‐G4 génomiqueNormandie UniversitéRouenFrance
| | - Hélène Tubeuf
- Inserm U1245, UNIROUEN, FHU‐G4 génomiqueNormandie UniversitéRouenFrance
- Integrative BiosoftwareRouenFrance
| | - Omar Soukarieh
- Inserm U1245, UNIROUEN, FHU‐G4 génomiqueNormandie UniversitéRouenFrance
| | | | - Florence Riant
- Laboratoire de Génétique, AP‐HPGH Saint‐Louis‐Lariboisière‐Fernand WidalParisFrance
| | | | - Sandrine M. Caputo
- Department of Genetics, Institut CurieParis Sciences Lettres Research UniversityParisFrance
| | | | - Nadia Boutry‐Kryza
- Unité Mixte de Génétique Constitutionnelle des Cancers FréquentsHospices Civils de LyonLyonFrance
| | - Françoise Bonnet‐Dorion
- Departement de Biopathologie Unité de Génétique ConstitutionnelleInstitut Bergonie—INSERM U1218BordeauxFrance
| | - Ines Schultz
- Laboratoire d'OncogénétiqueCentre Paul StraussStrasbourgFrance
| | - Maria Rossing
- Centre for Genomic Medicine, RigshospitaletUniversity of CopenhagenCopenhagenDenmark
| | - Olivier Quenez
- Inserm U1245, UNIROUEN, FHU‐G4 génomiqueNormandie UniversitéRouenFrance
| | - Louis Goldenberg
- Inserm U1245, UNIROUEN, FHU‐G4 génomiqueNormandie UniversitéRouenFrance
| | - Valentin Harter
- Department of BiostatisticsBaclesse Unicancer CenterCaenFrance
| | - Michael T. Parsons
- Department of Genetics and Computational BiologyQIMR Berghofer Medical Research InstituteHerstonQueenslandAustralia
| | - Amanda B. Spurdle
- Department of Genetics and Computational BiologyQIMR Berghofer Medical Research InstituteHerstonQueenslandAustralia
| | - Thierry Frébourg
- Inserm U1245, UNIROUEN, FHU‐G4 génomiqueNormandie UniversitéRouenFrance
- Department of geneticsRouen University HospitalRouenFrance
| | - Alexandra Martins
- Inserm U1245, UNIROUEN, FHU‐G4 génomiqueNormandie UniversitéRouenFrance
| | - Claude Houdayer
- Inserm U1245, UNIROUEN, FHU‐G4 génomiqueNormandie UniversitéRouenFrance
- Department of geneticsRouen University HospitalRouenFrance
| | - Sophie Krieger
- Laboratoire de Biologie et Génétique du CancerCentre François BaclesseCaenFrance
- Inserm U1245, UNIROUEN, FHU‐G4 génomiqueNormandie UniversitéRouenFrance
- UNICAENNormandie UniversitéCaenFrance
| |
Collapse
|
268
|
Carracedo S, Lirussi L, Alsøe L, Segers F, Wang C, Bartosova Z, Bohov P, Tekin NB, Kong XY, Esbensen QY, Chen L, Wennerström A, Kroustallaki P, Ceolotto D, Tönjes A, Berge RK, Bruheim P, Wong G, Böttcher Y, Halvorsen B, Nilsen H. SMUG1 regulates fat homeostasis leading to a fatty liver phenotype in mice. DNA Repair (Amst) 2022; 120:103410. [PMID: 36244177 DOI: 10.1016/j.dnarep.2022.103410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 08/08/2022] [Accepted: 10/01/2022] [Indexed: 11/25/2022]
|
269
|
Al-Akkad W, Acedo P, Vilia MG, Frenguelli L, Ney A, Rodriguez-Hernandez I, Labib PL, Tamburrino D, Spoletini G, Hall AR, Canestrari S, Osnato A, Garcia-Bernardo J, Sejour L, Vassileva V, Vlachos IS, Fusai G, Luong TV, Whittaker SR, Pereira SP, Vallier L, Pinzani M, Rombouts K, Mazza G. Tissue-Specific Human Extracellular Matrix Scaffolds Promote Pancreatic Tumour Progression and Chemotherapy Resistance. Cells 2022; 11:3652. [PMID: 36429078 PMCID: PMC9688243 DOI: 10.3390/cells11223652] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 11/01/2022] [Accepted: 11/08/2022] [Indexed: 11/19/2022] Open
Abstract
Over 80% of patients with pancreatic ductal adenocarcinoma (PDAC) are diagnosed at a late stage and are locally advanced or with concurrent metastases. The aggressive phenotype and relative chemo- and radiotherapeutic resistance of PDAC is thought to be mediated largely by its prominent stroma, which is supported by an extracellular matrix (ECM). Therefore, we investigated the impact of tissue-matched human ECM in driving PDAC and the role of the ECM in promoting chemotherapy resistance. Decellularized human pancreata and livers were recellularized with PANC-1 and MIA PaCa-2 (PDAC cell lines), as well as PK-1 cells (liver-derived metastatic PDAC cell line). PANC-1 cells migrated into the pancreatic scaffolds, MIA PaCa-2 cells were able to migrate into both scaffolds, whereas PK-1 cells were able to migrate into the liver scaffolds only. These differences were supported by significant deregulations in gene and protein expression between the pancreas scaffolds, liver scaffolds, and 2D culture. Moreover, these cell lines were significantly more resistant to gemcitabine and doxorubicin chemotherapy treatments in the 3D models compared to 2D cultures, even after confirmed uptake by confocal microscopy. These results suggest that tissue-specific ECM provides the preserved native cues for primary and metastatic PDAC cells necessary for a more reliable in vitro cell culture.
Collapse
Affiliation(s)
- Walid Al-Akkad
- UCL Institute for Liver and Digestive Health, Royal Free Hospital, University College London, London NW3 2PF, UK
- Engitix Therapeutics, The Westworks, 195 Wood Lane, Shepherd’s Bush, London W12 7FQ, UK
| | - Pilar Acedo
- UCL Institute for Liver and Digestive Health, Royal Free Hospital, University College London, London NW3 2PF, UK
| | - Maria-Giovanna Vilia
- UCL Institute for Liver and Digestive Health, Royal Free Hospital, University College London, London NW3 2PF, UK
- Engitix Therapeutics, The Westworks, 195 Wood Lane, Shepherd’s Bush, London W12 7FQ, UK
| | - Luca Frenguelli
- UCL Institute for Liver and Digestive Health, Royal Free Hospital, University College London, London NW3 2PF, UK
- Engitix Therapeutics, The Westworks, 195 Wood Lane, Shepherd’s Bush, London W12 7FQ, UK
| | - Alexander Ney
- UCL Institute for Liver and Digestive Health, Royal Free Hospital, University College London, London NW3 2PF, UK
| | | | - Peter L. Labib
- UCL Institute for Liver and Digestive Health, Royal Free Hospital, University College London, London NW3 2PF, UK
| | - Domenico Tamburrino
- Division of Surgery, Royal Free London NHS Foundation Trust, University College London, London NW3 2QG, UK
| | - Gabriele Spoletini
- Division of Surgery, Royal Free London NHS Foundation Trust, University College London, London NW3 2QG, UK
| | - Andrew R. Hall
- UCL Institute for Liver and Digestive Health, Royal Free Hospital, University College London, London NW3 2PF, UK
- Sheila Sherlock Liver Centre, Royal Free London NHS Foundation Trust, London NW3 2PF, UK
| | - Simone Canestrari
- UCL Institute for Liver and Digestive Health, Royal Free Hospital, University College London, London NW3 2PF, UK
| | - Anna Osnato
- Wellcome Trust-MRC Cambridge Stem Cell Institute, Anne McLaren Laboratory, University of Cambridge, Cambridge CB2 0AW, UK
- Wellcome Trust Sanger Institute, Hinxton, Cambridgeshire CB10 1RQ, UK
| | | | - Leinal Sejour
- Cancer Research Institute, HMS Initiative for RNA Medicine, Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Vessela Vassileva
- Department of Surgery and Cancer, Imperial College London, London SW7 2AZ, UK
| | - Ioannis S. Vlachos
- Cancer Research Institute, HMS Initiative for RNA Medicine, Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Giuseppe Fusai
- Division of Surgery, Royal Free London NHS Foundation Trust, University College London, London NW3 2QG, UK
| | - Tu Vinh Luong
- UCL Institute for Liver and Digestive Health, Royal Free Hospital, University College London, London NW3 2PF, UK
- Sheila Sherlock Liver Centre, Royal Free London NHS Foundation Trust, London NW3 2PF, UK
| | - Steven R. Whittaker
- Engitix Therapeutics, The Westworks, 195 Wood Lane, Shepherd’s Bush, London W12 7FQ, UK
| | - Stephen P. Pereira
- UCL Institute for Liver and Digestive Health, Royal Free Hospital, University College London, London NW3 2PF, UK
| | - Ludovic Vallier
- Wellcome Trust-MRC Cambridge Stem Cell Institute, Anne McLaren Laboratory, University of Cambridge, Cambridge CB2 0AW, UK
- Wellcome Trust Sanger Institute, Hinxton, Cambridgeshire CB10 1RQ, UK
| | - Massimo Pinzani
- UCL Institute for Liver and Digestive Health, Royal Free Hospital, University College London, London NW3 2PF, UK
| | - Krista Rombouts
- UCL Institute for Liver and Digestive Health, Royal Free Hospital, University College London, London NW3 2PF, UK
| | - Giuseppe Mazza
- UCL Institute for Liver and Digestive Health, Royal Free Hospital, University College London, London NW3 2PF, UK
- Engitix Therapeutics, The Westworks, 195 Wood Lane, Shepherd’s Bush, London W12 7FQ, UK
| |
Collapse
|
270
|
Gupta N, Magatha LS, Jayaraman D, Scott JX, Antony SB, Koshy T. Mercaptopurine induced myelosuppression in a child with a NUDT15 rs116855232 homozygous variant. J Oncol Pharm Pract 2022:10781552221137709. [DOI: 10.1177/10781552221137709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Introduction Mercaptopurine (6-MP) is the backbone of the consolidation and maintenance therapy for paediatric acute lymphoblastic leukaemia (ALL). Nevertheless, it can cause critical myelosuppression. Predicting adverse reactions to 6-MP often involves the investigation of pharmacogenetic variants; in particular thiopurine S-methyltransferase ( TPMT) and nudix hydrolase 15 ( NUDT15). Lately, NUDT15 variants have been shown to play a significant pharmacogenetic role in predicting 6-MP intolerance in children of Asian descent. Case Report We present a six-year-old male child of Indian origin with persistent cytopenia after treatment. This prompted targeted sequencing of the genes TPMT and NUD15. The results revealed two copies of the variant of NUD15 rs116855232, that is, NUDT15*2 genotype. Management and Outcome Since the NUDT15*2 allele classified the patient as a poor metabolizer, he was restarted on a low dose of 6-MP, which he tolerated. Discussion Individuals with the NUDT15*2allele (*2/*2 genotype) are poor metabolizers of thiopurines which results in an adverse reaction to 6-MP. About 3.5% of Indians show variations in the TPMT gene as compared to 19.4% variations observed in NUDT15, which makes the latter a more reliable disease marker.
Collapse
Affiliation(s)
- Navya Gupta
- Pediatic Hemato-oncology Unit, Department of Pediatrics, Sri Ramachandra Medical College and Research Institute, Porur, Chennai, India
| | - Latha Sneha Magatha
- Pediatic Hemato-oncology Unit, Department of Pediatrics, Sri Ramachandra Medical College and Research Institute, Porur, Chennai, India
| | - Dhaarani Jayaraman
- Pediatic Hemato-oncology Unit, Department of Pediatrics, Sri Ramachandra Medical College and Research Institute, Porur, Chennai, India
| | - Julius Xavier Scott
- Pediatic Hemato-oncology Unit, Department of Pediatrics, Sri Ramachandra Medical College and Research Institute, Porur, Chennai, India
| | - Sharon Benita Antony
- Department of Human Genetics, Sri Ramachandra Faculty of Biomedical Science and Technology, Sri Ramachandra Institute of Higher Education and Research, Porur, Chennai, India
| | - Teena Koshy
- Department of Human Genetics, Sri Ramachandra Faculty of Biomedical Science and Technology, Sri Ramachandra Institute of Higher Education and Research, Porur, Chennai, India
| |
Collapse
|
271
|
Vega Thurber R, Schmeltzer ER, Grottoli AG, van Woesik R, Toonen RJ, Warner M, Dobson KL, McLachlan RH, Barott K, Barshis DJ, Baumann J, Chapron L, Combosch DJ, Correa AMS, DeCarlo TM, Hagedorn M, Hédouin L, Hoadley K, Felis T, Ferrier-Pagès C, Kenkel C, Kuffner IB, Matthews J, Medina M, Meyer C, Oster C, Price J, Putnam HM, Sawall Y. Unified methods in collecting, preserving, and archiving coral bleaching and restoration specimens to increase sample utility and interdisciplinary collaboration. PeerJ 2022; 10:e14176. [PMID: 36345483 PMCID: PMC9636870 DOI: 10.7717/peerj.14176] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Accepted: 09/13/2022] [Indexed: 12/15/2022] Open
Abstract
Coral reefs are declining worldwide primarily because of bleaching and subsequent mortality resulting from thermal stress. Currently, extensive efforts to engage in more holistic research and restoration endeavors have considerably expanded the techniques applied to examine coral samples. Despite such advances, coral bleaching and restoration studies are often conducted within a specific disciplinary focus, where specimens are collected, preserved, and archived in ways that are not always conducive to further downstream analyses by specialists in other disciplines. This approach may prevent the full utilization of unexpended specimens, leading to siloed research, duplicative efforts, unnecessary loss of additional corals to research endeavors, and overall increased costs. A recent US National Science Foundation-sponsored workshop set out to consolidate our collective knowledge across the disciplines of Omics, Physiology, and Microscopy and Imaging regarding the methods used for coral sample collection, preservation, and archiving. Here, we highlight knowledge gaps and propose some simple steps for collecting, preserving, and archiving coral-bleaching specimens that can increase the impact of individual coral bleaching and restoration studies, as well as foster additional analyses and future discoveries through collaboration. Rapid freezing of samples in liquid nitrogen or placing at -80 °C to -20 °C is optimal for most Omics and Physiology studies with a few exceptions; however, freezing samples removes the potential for many Microscopy and Imaging-based analyses due to the alteration of tissue integrity during freezing. For Microscopy and Imaging, samples are best stored in aldehydes. The use of sterile gloves and receptacles during collection supports the downstream analysis of host-associated bacterial and viral communities which are particularly germane to disease and restoration efforts. Across all disciplines, the use of aseptic techniques during collection, preservation, and archiving maximizes the research potential of coral specimens and allows for the greatest number of possible downstream analyses.
Collapse
Affiliation(s)
- Rebecca Vega Thurber
- Department of Microbiology, Oregon State University, Corvallis, OR, United States
| | - Emily R. Schmeltzer
- Department of Microbiology, Oregon State University, Corvallis, OR, United States
| | - Andréa G. Grottoli
- School of Earth Sciences, Ohio State University, Columbus, OH, United States
| | - Robert van Woesik
- Institute for Global Ecology, Florida Institute of Technology, Melbourne, Fl, United States
| | - Robert J. Toonen
- Hawai’i Institute of Marine Biology, University of Hawai’i at Mānoa, Kāne’ohe, HI, United States
| | - Mark Warner
- School of Marine Science and Policy, University of Delaware, Lewes, DE, United States
| | - Kerri L. Dobson
- School of Earth Sciences, Ohio State University, Columbus, OH, United States
| | - Rowan H. McLachlan
- Department of Microbiology, Oregon State University, Corvallis, OR, United States,School of Earth Sciences, Ohio State University, Columbus, OH, United States
| | - Katie Barott
- Department of Biology, University of Pennsylvania, Philadelphia, PA, United States
| | - Daniel J. Barshis
- Department of Biological Sciences, Old Dominion University, Norfolk, VA, United States
| | - Justin Baumann
- Biology Department, Bowdoin College, Brunswick, ME, United States
| | - Leila Chapron
- School of Earth Sciences, Ohio State University, Columbus, OH, United States
| | | | | | - Thomas M. DeCarlo
- College of Natural and Computational Sciences, Hawai’i Pacific University, Honolulu, HI, United States
| | - Mary Hagedorn
- Hawai’i Institute of Marine Biology, University of Hawai’i at Mānoa, Kāne’ohe, HI, United States,Conservation Biology Institute, Smithsonian, Kāne’ohe, HI, United States
| | - Laetitia Hédouin
- Centre de Recherches Insulaires et Observatoire de l’Environnement, Chargée de Recherches CNRS, Papetō’ai, Moorea, French Polynesia
| | - Kenneth Hoadley
- Department of Biological Sciences, University of Alabama – Tuscaloosa, Tuscaloosa, AL, United States
| | - Thomas Felis
- MARUM – Center for Marine Environmental Sciences, University of Bremen, Bremen, Germany
| | | | - Carly Kenkel
- Department of Biological Sciences, University of Southern California, Los Angeles, CA, United States
| | | | - Jennifer Matthews
- Climate Change Cluster, University of Technology Sydney, Sydney, Australia
| | - Mónica Medina
- Department of Biology, Pennsylvania State University, University Park, PA, United States
| | - Christopher Meyer
- Department of Invertebrate Zoology, National Museum of Natural History, Smithsonian, Washington DC, United States
| | - Corinna Oster
- MARUM – Center for Marine Environmental Sciences, University of Bremen, Bremen, Germany
| | - James Price
- School of Earth Sciences, Ohio State University, Columbus, OH, United States
| | - Hollie M. Putnam
- Department of Biological Sciences, University of Rhode Island, Kingston, RI, United States
| | - Yvonne Sawall
- Bermuda Institute of Ocean Sciences, St. George’s, St. George’s, Bermuda
| |
Collapse
|
272
|
Falcone M, Uribe AH, Papalazarou V, Newman AC, Athineos D, Stevenson K, Sauvé CEG, Gao Y, Kim JK, Del Latto M, Kierstead M, Wu C, Smith JJ, Romesser PB, Chalmers AJ, Blyth K, Maddocks ODK. Sensitisation of cancer cells to radiotherapy by serine and glycine starvation. Br J Cancer 2022; 127:1773-1786. [PMID: 36115879 PMCID: PMC9643498 DOI: 10.1038/s41416-022-01965-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 08/10/2022] [Accepted: 08/19/2022] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Cellular metabolism is an integral component of cellular adaptation to stress, playing a pivotal role in the resistance of cancer cells to various treatment modalities, including radiotherapy. In response to radiotherapy, cancer cells engage antioxidant and DNA repair mechanisms which mitigate and remove DNA damage, facilitating cancer cell survival. Given the reliance of these resistance mechanisms on amino acid metabolism, we hypothesised that controlling the exogenous availability of the non-essential amino acids serine and glycine would radiosensitise cancer cells. METHODS We exposed colorectal, breast and pancreatic cancer cell lines/organoids to radiation in vitro and in vivo in the presence and absence of exogenous serine and glycine. We performed phenotypic assays for DNA damage, cell cycle, ROS levels and cell death, combined with a high-resolution untargeted LCMS metabolomics and RNA-Seq. RESULTS Serine and glycine restriction sensitised a range of cancer cell lines, patient-derived organoids and syngeneic mouse tumour models to radiotherapy. Comprehensive metabolomic and transcriptomic analysis of central carbon metabolism revealed that amino acid restriction impacted not only antioxidant response and nucleotide synthesis but had a marked inhibitory effect on the TCA cycle. CONCLUSION Dietary restriction of serine and glycine is a viable radio-sensitisation strategy in cancer.
Collapse
Affiliation(s)
- Mattia Falcone
- School of Cancer Sciences, Wolfson Wohl Cancer Research Centre, University of Glasgow, Glasgow, UK
- Division of Oncogenomics, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands
| | - Alejandro Huerta Uribe
- School of Cancer Sciences, Wolfson Wohl Cancer Research Centre, University of Glasgow, Glasgow, UK
| | - Vasileios Papalazarou
- School of Cancer Sciences, Wolfson Wohl Cancer Research Centre, University of Glasgow, Glasgow, UK
| | - Alice C Newman
- School of Cancer Sciences, Wolfson Wohl Cancer Research Centre, University of Glasgow, Glasgow, UK
| | | | - Katrina Stevenson
- School of Cancer Sciences, Wolfson Wohl Cancer Research Centre, University of Glasgow, Glasgow, UK
| | | | - Yajing Gao
- Colorectal Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Jin K Kim
- Colorectal Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Michael Del Latto
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Maria Kierstead
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Chao Wu
- Colorectal Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - J Joshua Smith
- Colorectal Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Paul B Romesser
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
- Early Drug Development Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Anthony J Chalmers
- School of Cancer Sciences, Wolfson Wohl Cancer Research Centre, University of Glasgow, Glasgow, UK
| | - Karen Blyth
- School of Cancer Sciences, Wolfson Wohl Cancer Research Centre, University of Glasgow, Glasgow, UK
- Cancer Research UK Beatson Institute, Glasgow, UK
| | - Oliver D K Maddocks
- School of Cancer Sciences, Wolfson Wohl Cancer Research Centre, University of Glasgow, Glasgow, UK.
| |
Collapse
|
273
|
Meena JP, Pathak N, Gupta AK, Bakhshi S, Gupta R, Makkar H, Seth R. Molecular evaluation of gene mutation profiles and copy number variations in pediatric acute myeloid leukemia. Leuk Res 2022; 122:106954. [PMID: 36162216 DOI: 10.1016/j.leukres.2022.106954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 08/19/2022] [Accepted: 09/17/2022] [Indexed: 11/24/2022]
Abstract
BACKGROUND The objectives of this study were to investigate the mutation profiles of targeted genes and copy number variations (CNVs) in normal cytogenetics (CN) pediatric acute myeloid leukemia (AML). METHODS This prospective study was conducted from October 2018 to December 2020. The next-generation sequencing (NGS) and chromosomal microarray analyses (CMA) were performed in pediatric CN-AML patients. RESULTS Out of 94 children (aged ≤18 years), 70 patients with AML (24 excluded) underwent conventional karyotyping/cytogenetic analyses. Forty-five (64.3%) of patients had abnormal/ recurrent cytogenetic abnormalities and 25 (35.7%) had normal cytogenetics. Twenty-three out of 25 CN-AML were further processed for gene mutation profile and CNVs using NGS and CMA, respectively. Twenty-two out of 23 (95.7%) patients were detected to have mutations in various genes. The common mutations were: NRAS, NPM1, CEBPA, KRAS, KIT, RUNX1, NOTCH1, WT1, GATA1, GATA2, FLT3, KMT2D, FLT3-TKD, and PHF6. Copy number variations (CNVs) were detected in nine patients (39%), and eight (34.8%) had a long contiguous stretch of homozygosity (LCSH) /loss of heterozygosity (LOH). An LCSH was detected on chromosomes 5, 7, 11, and 19. The gains were more common than losses (8 vs 2). The gains were observed on chromosomes 8, 9, 14, 19, 21, and 22, and the losses were detected on chromosomes 7 and 10. Monosomy was observed in three patients. Three patients (monosomy7, n = 2, and FLT-ITD, n = 1) were reclassified into the high-risk category. Post-induction, complete remission was achieved in all evaluable patients. CONCLUSION CN-AML patients have genetic abnormalities that can be detected by more advanced techniques like NGS and CMA. These genetic abnormalities play a role in risk stratification that may remain hidden in otherwise CN-AML.
Collapse
Affiliation(s)
- Jagdish Prasad Meena
- Division of Pediatric Oncology, Department of Pediatrics, All India Institute of Medical Sciences, New Delhi 110029, India.
| | - Nivedita Pathak
- Division of Pediatric Oncology, Department of Pediatrics, All India Institute of Medical Sciences, New Delhi 110029, India.
| | - Aditya Kumar Gupta
- Division of Pediatric Oncology, Department of Pediatrics, All India Institute of Medical Sciences, New Delhi 110029, India.
| | - Sameer Bakhshi
- Department of Medical Oncology, Dr. B.R.A. IRCH, All India Institute of Medical Sciences, New Delhi 110029, India.
| | - Ritu Gupta
- Laboratory Oncology Unit, Dr. B.R.A. IRCH, All India Institute of Medical Sciences, New Delhi 110029, India.
| | - Harshita Makkar
- Division of Pediatric Oncology, Department of Pediatrics, All India Institute of Medical Sciences, New Delhi 110029, India.
| | - Rachna Seth
- Division of Pediatric Oncology, Department of Pediatrics, All India Institute of Medical Sciences, New Delhi 110029, India.
| |
Collapse
|
274
|
Wyss AB, Hoang TT, Vindenes HK, White JD, Sikdar S, Richards M, Beane-Freeman LE, Parks CG, Lee M, Umbach DM, London SJ. Early-life farm exposures and eczema among adults in the Agricultural Lung Health Study. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY. GLOBAL 2022; 1:248-256. [PMID: 36569583 PMCID: PMC9784317 DOI: 10.1016/j.jacig.2022.06.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Background Several studies conducted in Europe have suggested a protective association between early-life farming exposures and childhood eczema or atopic dermatitis; few studies have examined associations in adults. Objectives To investigate associations between early-life exposures and eczema among 3217 adult farmers and farm spouses (mean age 62.8 years) in a case-control study nested within an US agricultural cohort. Methods We used sampling-weighted logistic regression to estimate odds ratios (ORs) and 95% confidence intervals (95%CIs) for associations between early-life exposures and self-reported doctor-diagnosed eczema (273 cases) and polytomous logistic regression to estimate ORs (95%CIs) for a 4-level outcome combining information on eczema and atopy (specific IgE≥0.35). Additionally, we explored genetic and gene-environment associations with eczema. Results Although early-life farming exposures were not associated with eczema overall, several early-life exposures were associated with a reduced risk of having both eczema and atopy. Notably, results suggest stronger protective associations among individuals with both eczema and atopy than among those with either atopy alone or eczema alone. For example, ORs (95%CIs) for having a mother who did farm work while pregnant were 1.01 (0.60-1.69) for eczema alone and 0.80 (0.65-0.99) for atopy alone, but 0.54 (0.33-0.80) for having both eczema and atopy. A genetic risk score based on previously identified atopic dermatitis variants was strongly positively associated with eczema, and interaction testing suggested protective effects of several early-life farming exposures only in individuals at lower genetic risk. Conclusions In utero and childhood farming exposures are associated with decreased odds of having eczema with atopy in adults.
Collapse
Affiliation(s)
- Annah B Wyss
- Epidemiology Branch, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, NC
| | - Thanh T Hoang
- Epidemiology Branch, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, NC
| | - Hilde K Vindenes
- Epidemiology Branch, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, NC
- Department of Occupational Medicine, Haukeland University Hospital, Bergen, Norway
- Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Julie D White
- Epidemiology Branch, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, NC
| | - Sinjini Sikdar
- Epidemiology Branch, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, NC
- Department of Mathematics and Statistics, Old Dominion University, Norfolk, VA
| | | | - Laura E Beane-Freeman
- Occupational and Environmental Epidemiology Branch, National Cancer Institute, National Institutes of Health, Department of Health and Human Services, Bethesda, MD
| | - Christine G Parks
- Epidemiology Branch, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, NC
| | - Mikyeong Lee
- Epidemiology Branch, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, NC
| | - David M Umbach
- Biostatistics and Computation Biology Branch, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, NC
| | - Stephanie J London
- Epidemiology Branch, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, NC
- Immunity, Inflammation and Disease Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, NC
| |
Collapse
|
275
|
Li Z, Yao F, Yu P, Li D, Zhang M, Mao L, Shen X, Ren Z, Wang L, Zhou B. Postnatal state transition of cardiomyocyte as a primary step in heart maturation. Protein Cell 2022; 13:842-862. [PMID: 35394262 PMCID: PMC9237199 DOI: 10.1007/s13238-022-00908-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Accepted: 01/17/2022] [Indexed: 11/26/2022] Open
Abstract
Postnatal heart maturation is the basis of normal cardiac function and provides critical insights into heart repair and regenerative medicine. While static snapshots of the maturing heart have provided much insight into its molecular signatures, few key events during postnatal cardiomyocyte maturation have been uncovered. Here, we report that cardiomyocytes (CMs) experience epigenetic and transcriptional decline of cardiac gene expression immediately after birth, leading to a transition state of CMs at postnatal day 7 (P7) that was essential for CM subtype specification during heart maturation. Large-scale single-cell analysis and genetic lineage tracing confirm the presence of transition state CMs at P7 bridging immature state and mature states. Silencing of key transcription factor JUN in P1-hearts significantly repressed CM transition, resulting in perturbed CM subtype proportions and reduced cardiac function in mature hearts. In addition, transplantation of P7-CMs into infarcted hearts exhibited cardiac repair potential superior to P1-CMs. Collectively, our data uncover CM state transition as a key event in postnatal heart maturation, which not only provides insights into molecular foundations of heart maturation, but also opens an avenue for manipulation of cardiomyocyte fate in disease and regenerative medicine.
Collapse
Affiliation(s)
- Zheng Li
- Shenzhen Key Laboratory of Cardiovascular Disease, Fuwai Hospital Chinese Academy of Medical Sciences, Shenzhen, 518057, China
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
- Key Laboratory of Pluripotent Stem Cells in Cardiac Repair and Regeneration, Chinese Academy of Medical Sciences, Beijing, 100037, China
| | - Fang Yao
- Shenzhen Key Laboratory of Cardiovascular Disease, Fuwai Hospital Chinese Academy of Medical Sciences, Shenzhen, 518057, China
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
- Key Laboratory of Pluripotent Stem Cells in Cardiac Repair and Regeneration, Chinese Academy of Medical Sciences, Beijing, 100037, China
| | - Peng Yu
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
| | - Dandan Li
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
| | - Mingzhi Zhang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
| | - Lin Mao
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
| | - Xiaomeng Shen
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
| | - Zongna Ren
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
| | - Li Wang
- Shenzhen Key Laboratory of Cardiovascular Disease, Fuwai Hospital Chinese Academy of Medical Sciences, Shenzhen, 518057, China.
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China.
- Key Laboratory of Pluripotent Stem Cells in Cardiac Repair and Regeneration, Chinese Academy of Medical Sciences, Beijing, 100037, China.
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, People's Republic of China.
| | - Bingying Zhou
- Shenzhen Key Laboratory of Cardiovascular Disease, Fuwai Hospital Chinese Academy of Medical Sciences, Shenzhen, 518057, China.
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China.
- Key Laboratory of Pluripotent Stem Cells in Cardiac Repair and Regeneration, Chinese Academy of Medical Sciences, Beijing, 100037, China.
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, People's Republic of China.
| |
Collapse
|
276
|
Monoclonal Antibody Sequence Variants Disguised as Fragments: Identification, Characterization, and Their Removal by Purification Process Optimization. J Pharm Sci 2022; 111:3009-3016. [PMID: 35940243 DOI: 10.1016/j.xphs.2022.08.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 08/02/2022] [Accepted: 08/02/2022] [Indexed: 12/14/2022]
Abstract
During early stage development of a therapeutic IgG1 monoclonal antibody, high levels of low molecular weight (LMW) peaks were observed by high performance size-exclusion chromatography and capillary electrophoresis. Further characterization of the LMW peak enriched HPSEC fractions using reversed phase liquid chromatography coupled to mass spectrometry showed these LMW species were 47 kDa and 50 kDa in size. However, the measured masses could not be matched to any fragments resulting from peptide bond hydrolysis. To identify these unknown LMW species, molecular characterization methods were employed, including high-throughput sequencing of RNA. Transcriptomic analysis revealed the LMW species were generated by mis-splicing events in the heavy chain transcript, which produced truncated heavy chain products that assembled with the light chain to mimic the appearance of fragments identified by routine purity assays. In an effort to improve product quality, an optimized purification process was developed. Characterization of the process intermediates confirmed removal of both LMW species by the optimized process. Our study demonstrates that deep-dive analytical characterization of biotherapeutics is critical to ensure product quality and inform process development. Transcriptomic analysis tools can help identify the cause of unknown species, and plays a key role in product and process characterization.
Collapse
|
277
|
Horak M, Fairweather D, Kokkonen P, Bednar D, Bienertova-Vasku J. Follistatin-like 1 and its paralogs in heart development and cardiovascular disease. Heart Fail Rev 2022; 27:2251-2265. [PMID: 35867287 PMCID: PMC11140762 DOI: 10.1007/s10741-022-10262-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/10/2022] [Indexed: 11/29/2022]
Abstract
Cardiovascular diseases (CVDs) are a group of disorders affecting the heart and blood vessels and a leading cause of death worldwide. Thus, there is a need to identify new cardiokines that may protect the heart from damage as reported in GBD 2017 Causes of Death Collaborators (2018) (The Lancet 392:1736-1788). Follistatin-like 1 (FSTL1) is a cardiokine that is highly expressed in the heart and released to the serum after cardiac injury where it is associated with CVD and predicts poor outcome. The action of FSTL1 likely depends not only on the tissue source but also post-translation modifications that are target tissue- and cell-specific. Animal studies examining the effect of FSTL1 in various models of heart disease have exploded over the past 15 years and primarily report a protective effect spanning from inhibiting inflammation via transforming growth factor, preventing remodeling and fibrosis to promoting angiogenesis and hypertrophy. A better understanding of FSTL1 and its homologs is needed to determine whether this protein could be a useful novel biomarker to predict poor outcome and death and whether it has therapeutic potential. The aim of this review is to provide a comprehensive description of the literature for this family of proteins in order to better understand their role in normal physiology and CVD.
Collapse
Affiliation(s)
- Martin Horak
- Department of Pathological Physiology, Faculty of Medicine, Masaryk University, Kamenice 5, Brno, 625 00, Czech Republic
- Research Centre for Toxic Compounds in the Environment, Faculty of Science, Masaryk University, Kamenice 5, Brno, 625 00, Czech Republic
| | - DeLisa Fairweather
- Department of Cardiovascular Medicine, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL, 32224, USA
| | - Piia Kokkonen
- Research Centre for Toxic Compounds in the Environment, Faculty of Science, Masaryk University, Kamenice 5, Brno, 625 00, Czech Republic
| | - David Bednar
- Research Centre for Toxic Compounds in the Environment, Faculty of Science, Masaryk University, Kamenice 5, Brno, 625 00, Czech Republic
| | - Julie Bienertova-Vasku
- Department of Pathological Physiology, Faculty of Medicine, Masaryk University, Kamenice 5, Brno, 625 00, Czech Republic.
- Research Centre for Toxic Compounds in the Environment, Faculty of Science, Masaryk University, Kamenice 5, Brno, 625 00, Czech Republic.
| |
Collapse
|
278
|
Ganther M, Lippold E, Bienert MD, Bouffaud ML, Bauer M, Baumann L, Bienert GP, Vetterlein D, Heintz-Buschart A, Tarkka MT. Plant Age and Soil Texture Rather Than the Presence of Root Hairs Cause Differences in Maize Resource Allocation and Root Gene Expression in the Field. PLANTS (BASEL, SWITZERLAND) 2022; 11:2883. [PMID: 36365336 PMCID: PMC9657941 DOI: 10.3390/plants11212883] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 10/20/2022] [Accepted: 10/25/2022] [Indexed: 06/16/2023]
Abstract
Understanding the biological roles of root hairs is key to projecting their contributions to plant growth and to assess their relevance for plant breeding. The objective of this study was to assess the importance of root hairs for maize nutrition, carbon allocation and root gene expression in a field experiment. Applying wild type and root hairless rth3 maize grown on loam and sand, we examined the period of growth including 4-leaf, 9-leaf and tassel emergence stages, accompanied with a low precipitation rate. rth3 maize had lower shoot growth and lower total amounts of mineral nutrients than wild type, but the concentrations of mineral elements, root gene expression, or carbon allocation were largely unchanged. For these parameters, growth stage accounted for the main differences, followed by substrate. Substrate-related changes were pronounced during tassel emergence, where the concentrations of several elements in leaves as well as cell wall formation-related root gene expression and C allocation decreased. In conclusion, the presence of root hairs stimulated maize shoot growth and total nutrient uptake, but other parameters were more impacted by growth stage and soil texture. Further research should relate root hair functioning to the observed losses in maize productivity and growth efficiency.
Collapse
Affiliation(s)
- Minh Ganther
- Helmholtz Centre for Environmental Research, Theodor-Lieser-Str. 4, 06120 Halle, Germany
| | - Eva Lippold
- Helmholtz Centre for Environmental Research, Theodor-Lieser-Str. 4, 06120 Halle, Germany
| | - Manuela Désirée Bienert
- TUM School of Life Sciences, Technical University of Munich, Alte Akademie 12, 85354 Freising, Germany
| | - Marie-Lara Bouffaud
- Helmholtz Centre for Environmental Research, Theodor-Lieser-Str. 4, 06120 Halle, Germany
| | - Mario Bauer
- Helmholtz Centre for Environmental Research, Permoserstr. 15, 04318 Leipzig, Germany
| | - Louis Baumann
- Helmholtz Centre for Environmental Research, Theodor-Lieser-Str. 4, 06120 Halle, Germany
| | - Gerd Patrick Bienert
- TUM School of Life Sciences, Technical University of Munich, Alte Akademie 12, 85354 Freising, Germany
| | - Doris Vetterlein
- Helmholtz Centre for Environmental Research, Theodor-Lieser-Str. 4, 06120 Halle, Germany
- Institute of Agricultural and Nutritional Sciences, Martin Luther University Halle-Wittenberg, Von-Seckendorff-Platz 3, 06120 Halle/Saale, Germany
| | - Anna Heintz-Buschart
- Helmholtz Centre for Environmental Research, Theodor-Lieser-Str. 4, 06120 Halle, Germany
- Swammerdam Institute for Life Sciences, University of Amsterdam, Science Park 904, 1098 XH Amsterdam, The Netherlands
| | - Mika Tapio Tarkka
- Helmholtz Centre for Environmental Research, Theodor-Lieser-Str. 4, 06120 Halle, Germany
- German Centre for Integrative Biodiversity Research (iDiv) Halle-Jena-Leipzig, Deutscher Platz 5e, 04103 Leipzig, Germany
| |
Collapse
|
279
|
Friedl MS, Djakovic L, Kluge M, Hennig T, Whisnant AW, Backes S, Dölken L, Friedel CC. HSV-1 and influenza infection induce linear and circular splicing of the long NEAT1 isoform. PLoS One 2022; 17:e0276467. [PMID: 36279270 PMCID: PMC9591066 DOI: 10.1371/journal.pone.0276467] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Accepted: 10/07/2022] [Indexed: 11/18/2022] Open
Abstract
The herpes simplex virus 1 (HSV-1) virion host shut-off (vhs) protein cleaves both cellular and viral mRNAs by a translation-initiation-dependent mechanism, which should spare circular RNAs (circRNAs). Here, we show that vhs-mediated degradation of linear mRNAs leads to an enrichment of circRNAs relative to linear mRNAs during HSV-1 infection. This was also observed in influenza A virus (IAV) infection, likely due to degradation of linear host mRNAs mediated by the IAV PA-X protein and cap-snatching RNA-dependent RNA polymerase. For most circRNAs, enrichment was not due to increased circRNA synthesis but due to a general loss of linear RNAs. In contrast, biogenesis of a circRNA originating from the long isoform (NEAT1_2) of the nuclear paraspeckle assembly transcript 1 (NEAT1) was induced both in HSV-1 infection-in a vhs-independent manner-and in IAV infection. This was associated with induction of novel linear splicing of NEAT1_2 both within and downstream of the circRNA. NEAT1_2 forms a scaffold for paraspeckles, nuclear bodies located in the interchromatin space, must likely remain unspliced for paraspeckle assembly and is up-regulated in HSV-1 and IAV infection. We show that NEAT1_2 splicing and up-regulation can be induced by ectopic co-expression of the HSV-1 immediate-early proteins ICP22 and ICP27, potentially linking increased expression and splicing of NEAT1_2. To identify other conditions with NEAT1_2 splicing, we performed a large-scale screen of published RNA-seq data. This uncovered both induction of NEAT1_2 splicing and poly(A) read-through similar to HSV-1 and IAV infection in cancer cells upon inhibition or knockdown of CDK7 or the MED1 subunit of the Mediator complex phosphorylated by CDK7. In summary, our study reveals induction of novel circular and linear NEAT1_2 splicing isoforms as a common characteristic of HSV-1 and IAV infection and highlights a potential role of CDK7 in HSV-1 or IAV infection.
Collapse
Affiliation(s)
- Marie-Sophie Friedl
- Institute of Informatics, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Lara Djakovic
- Institute for Virology and Immunobiology, Julius-Maximilians-University Würzburg, Würzburg, Germany
| | - Michael Kluge
- Institute of Informatics, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Thomas Hennig
- Institute for Virology and Immunobiology, Julius-Maximilians-University Würzburg, Würzburg, Germany
| | - Adam W. Whisnant
- Institute for Virology and Immunobiology, Julius-Maximilians-University Würzburg, Würzburg, Germany
| | - Simone Backes
- Institute for Virology and Immunobiology, Julius-Maximilians-University Würzburg, Würzburg, Germany
| | - Lars Dölken
- Institute for Virology and Immunobiology, Julius-Maximilians-University Würzburg, Würzburg, Germany
| | - Caroline C. Friedel
- Institute of Informatics, Ludwig-Maximilians-Universität München, Munich, Germany
- * E-mail:
| |
Collapse
|
280
|
An automatic hypothesis generation for plausible linkage between xanthium and diabetes. Sci Rep 2022; 12:17547. [PMID: 36266295 PMCID: PMC9585073 DOI: 10.1038/s41598-022-20752-0] [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: 12/07/2021] [Accepted: 09/19/2022] [Indexed: 01/13/2023] Open
Abstract
There has been a significant increase in text mining implementation for biomedical literature in recent years. Previous studies introduced the implementation of text mining and literature-based discovery to generate hypotheses of potential candidates for drug development. By conducting a hypothesis-generation step and using evidence from published journal articles or proceedings, previous studies have managed to reduce experimental time and costs. First, we applied the closed discovery approach from Swanson's ABC model to collect publications related to 36 Xanthium compounds or diabetes. Second, we extracted biomedical entities and relations using a knowledge extraction engine, the Public Knowledge Discovery Engine for Java or PKDE4J. Third, we built a knowledge graph using the obtained bio entities and relations and then generated paths with Xanthium compounds as source nodes and diabetes as the target node. Lastly, we employed graph embeddings to rank each path and evaluated the results based on domain experts' opinions and literature. Among 36 Xanthium compounds, 35 had direct paths to five diabetes-related nodes. We ranked 2,740,314 paths in total between 35 Xanthium compounds and three diabetes-related phrases: type 1 diabetes, type 2 diabetes, and diabetes mellitus. Based on the top five percentile paths, we concluded that adenosine, choline, beta-sitosterol, rhamnose, and scopoletin were potential candidates for diabetes drug development using natural products. Our framework for hypothesis generation employs a closed discovery from Swanson's ABC model that has proven very helpful in discovering biological linkages between bio entities. The PKDE4J tools we used to capture bio entities from our document collection could label entities into five categories: genes, compounds, phenotypes, biological processes, and molecular functions. Using the BioPREP model, we managed to interpret the semantic relatedness between two nodes and provided paths containing valuable hypotheses. Lastly, using a graph-embedding algorithm in our path-ranking analysis, we exploited the semantic relatedness while preserving the graph structure properties.
Collapse
|
281
|
Nicolet BP, Wolkers MC. The relationship of mRNA with protein expression in CD8+ T cells associates with gene class and gene characteristics. PLoS One 2022; 17:e0276294. [PMID: 36260607 PMCID: PMC9581405 DOI: 10.1371/journal.pone.0276294] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Accepted: 10/04/2022] [Indexed: 11/19/2022] Open
Abstract
T cells are key players in our defence against infections and malignancies. When T cells differentiate or become activated, they undergo substantial alterations in gene expression. Even though RNA expression levels are now well documented throughout different stages of T cells, it is not well understood how mRNA expression translates into the protein landscape. By combining paired RNA sequencing and mass spectrometry data of primary human CD8+ T cells, we report that mRNA expression is a poor proxy for the overall protein output, irrespective of the differentiation or activation status. Yet, gene class stratification revealed a function-specific correlation of mRNA with protein expression. This gene class-specific expression pattern associated with differences in gene characteristics such as sequence conservation and untranslated region (UTR) lengths. In addition, the presence of AU-rich elements in the 3'UTR associated with alterations in mRNA and protein abundance T cell activation dependent, gene class-specific manner. In conclusion, our study highlights the role of gene characteristics as a determinant for gene expression in T cells.
Collapse
Affiliation(s)
- Benoît P. Nicolet
- Department of Hematopoiesis, Sanquin Research, Amsterdam, The Netherlands
- Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Oncode Institute, Utrecht, The Netherlands
| | - Monika C. Wolkers
- Department of Hematopoiesis, Sanquin Research, Amsterdam, The Netherlands
- Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Oncode Institute, Utrecht, The Netherlands
- * E-mail:
| |
Collapse
|
282
|
Yuan L, Roy B, Ratna P, Uhler C, Shivashankar GV. Lateral confined growth of cells activates Lef1 dependent pathways to regulate cell-state transitions. Sci Rep 2022; 12:17318. [PMID: 36243826 PMCID: PMC9569372 DOI: 10.1038/s41598-022-21596-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 09/29/2022] [Indexed: 01/10/2023] Open
Abstract
Long-term sustained mechano-chemical signals in tissue microenvironment regulate cell-state transitions. In recent work, we showed that laterally confined growth of fibroblasts induce dedifferentiation programs. However, the molecular mechanisms underlying such mechanically induced cell-state transitions are poorly understood. In this paper, we identify Lef1 as a critical somatic transcription factor for the mechanical regulation of de-differentiation pathways. Network optimization methods applied to time-lapse RNA-seq data identify Lef1 dependent signaling as potential regulators of such cell-state transitions. We show that Lef1 knockdown results in the down-regulation of fibroblast de-differentiation and that Lef1 directly interacts with the promoter regions of downstream reprogramming factors. We also evaluate the potential upstream activation pathways of Lef1, including the Smad4, Atf2, NFkB and Beta-catenin pathways, thereby identifying that Smad4 and Atf2 may be critical for Lef1 activation. Collectively, we describe an important mechanotransduction pathway, including Lef1, which upon activation, through progressive lateral cell confinement, results in fibroblast de-differentiation.
Collapse
Affiliation(s)
- Luezhen Yuan
- Division of Biology and Chemistry, Paul Scherrer Institut, 5232, Villigen, Switzerland
- Department of Health Sciences and Technology, ETH Zurich, 8092, Zurich, Switzerland
- Mechanobiology Institute, National University of Singapore, Singapore, 117411, Singapore
| | - Bibhas Roy
- Division of Biology and Chemistry, Paul Scherrer Institut, 5232, Villigen, Switzerland
- Mechanobiology Institute, National University of Singapore, Singapore, 117411, Singapore
- Institute of Molecular Oncology, Italian Foundation for Cancer Research, 20139, Milan, Italy
| | - Prasuna Ratna
- Mechanobiology Institute, National University of Singapore, Singapore, 117411, Singapore
| | - Caroline Uhler
- Massachusetts Institute of Technology, Cambridge, MA, USA
| | - G V Shivashankar
- Division of Biology and Chemistry, Paul Scherrer Institut, 5232, Villigen, Switzerland.
- Department of Health Sciences and Technology, ETH Zurich, 8092, Zurich, Switzerland.
- Mechanobiology Institute, National University of Singapore, Singapore, 117411, Singapore.
- Institute of Molecular Oncology, Italian Foundation for Cancer Research, 20139, Milan, Italy.
| |
Collapse
|
283
|
Sedivy-Haley K, Blimkie T, Falsafi R, Lee AHY, Hancock REW. A transcriptomic analysis of the effects of macrophage polarization and endotoxin tolerance on the response to Salmonella. PLoS One 2022; 17:e0276010. [PMID: 36240188 PMCID: PMC9565388 DOI: 10.1371/journal.pone.0276010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Accepted: 09/27/2022] [Indexed: 11/19/2022] Open
Abstract
Salmonella is an intracellular pathogen causing significant morbidity and mortality. Its ability to grow inside macrophages is important to virulence, and is dependent on the activation state of the macrophages. Classically activated M1 macrophages are non-permissive for Salmonella growth, while alternatively activated M2 macrophages are permissive for Salmonella growth. Here we showed that endotoxin-primed macrophages (MEP), such as those associated with sepsis, showed similar levels of Salmonella resistance to M1 macrophages after 2 hr of intracellular infection, but at the 4 hr and 24 hr time points were susceptible like M2 macrophages. To understand this mechanistically, transcriptomic sequencing, RNA-Seq, was performed. This showed that M1 and MEP macrophages that had not been exposed to Salmonella, demonstrated a process termed here as primed activation, in expressing relatively higher levels of particular anti-infective genes and pathways, including the JAK-STAT (Janus kinase-signal transducer and activator of transcription) pathway. In contrast, in M2 macrophages these genes and pathways were largely expressed only in response to infection. Conversely, in response to infection, M1 macrophages, but not MEP macrophages, modulated additional genes known to be associated with susceptibility to Salmonella infection, possibly contributing to the differences in resistance at later time points. Application of the JAK inhibitor Ruxolitinib before infection reduced resistance in M1 macrophages, supporting the importance of early JAK-STAT signalling in M1 resistance to Salmonella.
Collapse
Affiliation(s)
- Katharine Sedivy-Haley
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Travis Blimkie
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Reza Falsafi
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Amy Huei-Yi Lee
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Robert E W Hancock
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
284
|
Akpokiro V, Martin T, Oluwadare O. EnsembleSplice: ensemble deep learning model for splice site prediction. BMC Bioinformatics 2022; 23:413. [PMID: 36203144 PMCID: PMC9535948 DOI: 10.1186/s12859-022-04971-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Accepted: 09/29/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Identifying splice site regions is an important step in the genomic DNA sequencing pipelines of biomedical and pharmaceutical research. Within this research purview, efficient and accurate splice site detection is highly desirable, and a variety of computational models have been developed toward this end. Neural network architectures have recently been shown to outperform classical machine learning approaches for the task of splice site prediction. Despite these advances, there is still considerable potential for improvement, especially regarding model prediction accuracy, and error rate. RESULTS Given these deficits, we propose EnsembleSplice, an ensemble learning architecture made up of four (4) distinct convolutional neural networks (CNN) model architecture combination that outperform existing splice site detection methods in the experimental evaluation metrics considered including the accuracies and error rates. We trained and tested a variety of ensembles made up of CNNs and DNNs using the five-fold cross-validation method to identify the model that performed the best across the evaluation and diversity metrics. As a result, we developed our diverse and highly effective splice site (SS) detection model, which we evaluated using two (2) genomic Homo sapiens datasets and the Arabidopsis thaliana dataset. The results showed that for of the Homo sapiens EnsembleSplice achieved accuracies of 94.16% for one of the acceptor splice sites and 95.97% for donor splice sites, with an error rate for the same Homo sapiens dataset, 4.03% for the donor splice sites and 5.84% for the acceptor splice sites datasets. CONCLUSIONS Our five-fold cross validation ensured the prediction accuracy of our models are consistent. For reproducibility, all the datasets used, models generated, and results in our work are publicly available in our GitHub repository here: https://github.com/OluwadareLab/EnsembleSplice.
Collapse
Affiliation(s)
- Victor Akpokiro
- Department of Computer Science, University of Colorado, Colorado Springs, CO, 80918, USA
| | - Trevor Martin
- Department of Mathematics, Oberlin College, Oberlin, OH, 44074, USA
| | - Oluwatosin Oluwadare
- Department of Computer Science, University of Colorado, Colorado Springs, CO, 80918, USA.
| |
Collapse
|
285
|
Ghosh R, León-Ruiz M, Singh Sardar S, Naga D, Roy D, Ghosh T, Dubey S, Benito-León J. A novel heterozygous mutation in the hydroxymethylbilane synthase gene in a case with acute intermittent porphyria. Qatar Med J 2022; 2022:46. [PMID: 36304064 PMCID: PMC9577391 DOI: 10.5339/qmj.2022.46] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Accepted: 05/25/2022] [Indexed: 11/03/2022] Open
Abstract
Porphyrias are rare metabolic disorders caused by inherited or acquired enzymatic defects in the heme biosynthetic pathway. They are grouped into acute hepatic porphyrias and photocutaneous porphyrias. Acute intermittent porphyria, the most prevalent subtype of acute hepatic porphyrias, is caused by a mutation in the hydroxymethylbilane synthase gene. In this work, a case of a 13 year-old Indian female presenting with multi-organ involvement (Neurological: episodic seizures, behavioral abnormalities, acute onset progressive flaccid-motor quadriparesis, multiple cranial nerve palsies, respiratory paralysis, dysautonomia, and posterior reversible encephalopathy syndrome; Gastrointestinal: recurrent attacks of abdominal pain, nausea/vomiting, isolated transaminitis, and acute pancreatitis; and Renal: metabolic alkalosis and refractory dyselectrolytemia) which resulted in significant diagnostic dilemmas. She was eventually diagnosed as a case of acute intermittent porphyria harboring a novel hydroxymethylbilane synthase gene mutation (p.Arg173Trp).
Collapse
Affiliation(s)
- Ritwik Ghosh
- Department of General Medicine, Burdwan Medical College & Hospital, Burdwan, West Bengal, India E-mail:
| | - Moisés León-Ruiz
- Section of Clinical Neurophysiology, Department of Neurology, University Hospital “La Paz,” Madrid, Spain
| | - Sona Singh Sardar
- Department of General Medicine, Burdwan Medical College & Hospital, Burdwan, West Bengal, India E-mail:
| | - Dinobandhu Naga
- Department of General Medicine, Burdwan Medical College & Hospital, Burdwan, West Bengal, India E-mail:
| | | | - Tapas Ghosh
- Department of Anatomy, Burdwan Medical College & Hospital, Burdwan, West Bengal, India
| | - Souvik Dubey
- Department of Neuromedicine, Bangur Institute of Neurosciences (BIN), Kolkata, West Bengal, India
| | | |
Collapse
|
286
|
Srikanth K, von Pfeil DJF, Stanley BJ, Griffitts C, Huson HJ. Genome Wide Association Study with Imputed Whole Genome Sequence Data Identifies a 431 kb Risk Haplotype on CFA18 for Congenital Laryngeal Paralysis in Alaskan Sled Dogs. Genes (Basel) 2022; 13:genes13101808. [PMID: 36292693 PMCID: PMC9602090 DOI: 10.3390/genes13101808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 10/03/2022] [Accepted: 10/04/2022] [Indexed: 11/16/2022] Open
Abstract
Congenital laryngeal paralysis (CLP) is an inherited disorder that affects the ability of the dog to exercise and precludes it from functioning as a working sled dog. Though CLP is known to occur in Alaskan sled dogs (ASDs) since 1986, the genetic mutation underlying the disease has not been reported. Using a genome-wide association study (GWAS), we identified a 708 kb region on CFA 18 harboring 226 SNPs to be significantly associated with CLP. The significant SNPs explained 47.06% of the heritability of CLP. We narrowed the region to 431 kb through autozygosity mapping and found 18 of the 20 cases to be homozygous for the risk haplotype. Whole genome sequencing of two cases and a control ASD, and comparison with the genome of 657 dogs from various breeds, confirmed the homozygous status of the risk haplotype to be unique to the CLP cases. Most of the dogs that were homozygous for the risk allele had blue eyes. Gene annotation and a gene-based association study showed that the risk haplotype encompasses genes implicated in developmental and neurodegenerative disorders. Pathway analysis showed enrichment of glycoproteins and glycosaminoglycans biosynthesis, which play a key role in repairing damaged nerves. In conclusion, our results suggest an important role for the identified candidate region in CLP.
Collapse
Affiliation(s)
- Krishnamoorthy Srikanth
- Department of Animal Science, College of Agriculture and Life Science, Cornell University, Ithaca, NY 14850, USA
| | | | - Bryden J. Stanley
- Department of Small Animal Clinical Sciences, Michigan State University, East Lansing, MI 48824, USA
| | | | - Heather J. Huson
- Department of Animal Science, College of Agriculture and Life Science, Cornell University, Ithaca, NY 14850, USA
- Correspondence:
| |
Collapse
|
287
|
Hernandez-Moran BA, Papanastasiou AS, Parry D, Meynert A, Gautier P, Grimes G, Adams IR, Trejo-Reveles V, Bengani H, Keighren M, Jackson IJ, Adams DJ, FitzPatrick DR, Rainger J. Robust Genetic Analysis of the X-Linked Anophthalmic ( Ie) Mouse. Genes (Basel) 2022; 13:1797. [PMID: 36292683 PMCID: PMC9601528 DOI: 10.3390/genes13101797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 09/25/2022] [Accepted: 09/29/2022] [Indexed: 11/24/2022] Open
Abstract
Anophthalmia (missing eye) describes a failure of early embryonic ocular development. Mutations in a relatively small set of genes account for 75% of bilateral anophthalmia cases, yet 25% of families currently are left without a molecular diagnosis. Here, we report our experimental work that aimed to uncover the developmental and genetic basis of the anophthalmia characterising the X-linked Ie (eye-ear reduction) X-ray-induced allele in mouse that was first identified in 1947. Histological analysis of the embryonic phenotype showed failure of normal eye development after the optic vesicle stage with particularly severe malformation of the ventral retina. Linkage analysis mapped this mutation to a ~6 Mb region on the X chromosome. Short- and long-read whole-genome sequencing (WGS) of affected and unaffected male littermates confirmed the Ie linkage but identified no plausible causative variants or structural rearrangements. These analyses did reduce the critical candidate interval and revealed evidence of multiple variants within the ancestral DNA, although none were found that altered coding sequences or that were unique to Ie. To investigate early embryonic events at a genetic level, we then generated mouse ES cells derived from male Ie embryos and wild type littermates. RNA-seq and accessible chromatin sequencing (ATAC-seq) data generated from cultured optic vesicle organoids did not reveal any large differences in gene expression or accessibility of putative cis-regulatory elements between Ie and wild type. However, an unbiased TF-footprinting analysis of accessible chromatin regions did provide evidence of a genome-wide reduction in binding of transcription factors associated with ventral eye development in Ie, and evidence of an increase in binding of the Zic-family of transcription factors, including Zic3, which is located within the Ie-refined critical interval. We conclude that the refined Ie critical region at chrX: 56,145,000-58,385,000 contains multiple genetic variants that may be linked to altered cis regulation but does not contain a convincing causative mutation. Changes in the binding of key transcription factors to chromatin causing altered gene expression during development, possibly through a subtle mis-regulation of Zic3, presents a plausible cause for the anophthalmia phenotype observed in Ie, but further work is required to determine the precise causative allele and its genetic mechanism.
Collapse
Affiliation(s)
- Brianda A. Hernandez-Moran
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Crewe Rd South, Edinburgh EH4 2XU, UK
| | - Andrew S. Papanastasiou
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Crewe Rd South, Edinburgh EH4 2XU, UK
| | - David Parry
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Crewe Rd South, Edinburgh EH4 2XU, UK
| | - Alison Meynert
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Crewe Rd South, Edinburgh EH4 2XU, UK
| | - Philippe Gautier
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Crewe Rd South, Edinburgh EH4 2XU, UK
| | - Graeme Grimes
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Crewe Rd South, Edinburgh EH4 2XU, UK
| | - Ian R. Adams
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Crewe Rd South, Edinburgh EH4 2XU, UK
| | - Violeta Trejo-Reveles
- The Division of Functional Genetics and Development, The Roslin Institute, Midlothian EH25 9RG, UK
| | - Hemant Bengani
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Crewe Rd South, Edinburgh EH4 2XU, UK
| | - Margaret Keighren
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Crewe Rd South, Edinburgh EH4 2XU, UK
| | - Ian J. Jackson
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Crewe Rd South, Edinburgh EH4 2XU, UK
| | - David J. Adams
- Wellcome Sanger Institute, Hinxton, Cambridgeshire CB10 1SA, UK
| | - David R. FitzPatrick
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Crewe Rd South, Edinburgh EH4 2XU, UK
| | - Joe Rainger
- The Division of Functional Genetics and Development, The Roslin Institute, Midlothian EH25 9RG, UK
| |
Collapse
|
288
|
Peitz T, Möhlendick B, Eisenberger U, Siffert W, Heinemann FM, Kribben A, Friebus-Kardash J. CC Genotype of GNAS c.393C>T (rs7121) Polymorphism Has a Protective Effect against Development of BK Viremia and BKV-Associated Nephropathy after Renal Transplant. Pathogens 2022; 11:pathogens11101138. [PMID: 36297195 PMCID: PMC9609707 DOI: 10.3390/pathogens11101138] [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: 08/24/2022] [Revised: 09/23/2022] [Accepted: 09/28/2022] [Indexed: 11/07/2022] Open
Abstract
The GNAS gene encodes the alpha-subunit of the stimulatory G-protein (Gαs) in humans and mice. The single-nucleotide polymorphism of GNAS, c.393C>T, is associated with an elevated production of Gαs and an increased formation of cyclic adenosine monophosphate (cAMP). In the present study, we analyzed the effect of this GNAS polymorphism on a renal allograft outcome. We screened a cohort of 436 renal allograft recipients, who were retrospectively followed up for up to 5 years after transplant. GNAS genotypes were determined with polymerase chain reaction restriction fragment length polymorphism (PCR-RFLP) assays. The 393T allele was detected in 319 (73%) recipients (113 recipients with TT and 206 with CT genotype) and the CC genotype in 117 (27%). The CC genotype was associated with a significantly lower frequency of BK viremia (CC, 17 recipients (15%); T 84 (26%)); p = 0.01; TT, 27 vs. CC, 17, p = 0.07; TT, 27 vs. CT, 57, p = 0. 46; CT, 57 vs. CC, 17, p = 0.01) and BKV-associated nephropathy (CC, 3 recipients (3%); T, 27 (8%); p = 0.03; TT,10 vs. CC, 3, p = 0.04; TT, 10 vs. CT,17, p = 0.85; CT, 17 vs. CC,3, p = 0.04) after transplant. BKV-associated nephropathy-free survival was significantly better among CC genotype carriers than among T allele carriers (p = 0.043; TT vs. CC, p = 0.03; CT vs. CC, p = 0.04; TT vs. CT, p = 0.83). Multivariate analysis indicated an independent protective effect of the CC genotype against the development of both BK viremia (relative risk. 0.54; p = 0.04) and BKV-associated nephropathy after renal transplant (relative risk. 0.27; p = 0.036). The GNAS 393 CC genotype seems to protect renal allograft recipients against the development of BK viremia and BKV-associated nephropathy.
Collapse
Affiliation(s)
- Tobias Peitz
- Department of Nephrology, University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany
| | - Birte Möhlendick
- Institute of Pharmacogenetics, University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany
| | - Ute Eisenberger
- Department of Nephrology, University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany
| | - Winfried Siffert
- Institute of Pharmacogenetics, University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany
| | - Falko Markus Heinemann
- Institute for Transfusion Medicine, Transplantation Diagnostics, University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany
| | - Andreas Kribben
- Department of Nephrology, University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany
| | - Justa Friebus-Kardash
- Department of Nephrology, University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany
- Correspondence: ; Tel.: +49-(0)-201-7236559; Fax: +49-(0)-201-7236907
| |
Collapse
|
289
|
Hashimoto M, Araki K, Cardenas MA, Li P, Jadhav RR, Kissick HT, Hudson WH, McGuire DJ, Obeng RC, Wieland A, Lee J, McManus DT, Ross JL, Im SJ, Lee J, Lin JX, Hu B, West EE, Scharer CD, Freeman GJ, Sharpe AH, Ramalingam SS, Pellerin A, Teichgräber V, Greenleaf WJ, Klein C, Goronzy JJ, Umaña P, Leonard WJ, Smith KA, Ahmed R. PD-1 combination therapy with IL-2 modifies CD8 + T cell exhaustion program. Nature 2022; 610:173-181. [PMID: 36171288 PMCID: PMC9793890 DOI: 10.1038/s41586-022-05257-0] [Citation(s) in RCA: 186] [Impact Index Per Article: 62.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Accepted: 08/22/2022] [Indexed: 12/30/2022]
Abstract
Combination therapy with PD-1 blockade and IL-2 is highly effective during chronic lymphocytic choriomeningitis virus infection1. Here we examine the underlying basis for this synergy. We show that PD-1 + IL-2 combination therapy, in contrast to PD-1 monotherapy, substantially changes the differentiation program of the PD-1+TCF1+ stem-like CD8+ T cells and results in the generation of transcriptionally and epigenetically distinct effector CD8+ T cells that resemble highly functional effector CD8+ T cells seen after an acute viral infection. The generation of these qualitatively superior CD8+ T cells that mediate viral control underlies the synergy between PD-1 and IL-2. Our results show that the PD-1+TCF1+ stem-like CD8+ T cells, also referred to as precursors of exhausted CD8+ T cells, are not fate-locked into the exhaustion program and their differentiation trajectory can be changed by IL-2 signals. These virus-specific effector CD8+ T cells emerging from the stem-like CD8+ T cells after combination therapy expressed increased levels of the high-affinity IL-2 trimeric (CD25-CD122-CD132) receptor. This was not seen after PD-1 blockade alone. Finally, we show that CD25 engagement with IL-2 has an important role in the observed synergy between IL-2 cytokine and PD-1 blockade. Either blocking CD25 with an antibody or using a mutated version of IL-2 that does not bind to CD25 but still binds to CD122 and CD132 almost completely abrogated the synergistic effects observed after PD-1 + IL-2 combination therapy. There is considerable interest in PD-1 + IL-2 combination therapy for patients with cancer2,3, and our fundamental studies defining the underlying mechanisms of how IL-2 synergizes with PD-1 blockade should inform these human translational studies.
Collapse
Affiliation(s)
- Masao Hashimoto
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, USA
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA, USA
| | - Koichi Araki
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, USA
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA, USA
- Division of Infectious Diseases, Center for Inflammation and Tolerance, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Maria A Cardenas
- Department of Urology, Emory University School of Medicine, Atlanta, GA, USA
| | - Peng Li
- Laboratory of Molecular Immunology and the Immunology Center, National Heart, Lung, and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Rohit R Jadhav
- Department of Immunology, Mayo Clinic School of Medicine and Sciences, Rochester, MN, USA
- Department of Medicine, Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, CA, USA
| | - Haydn T Kissick
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, USA
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA, USA
- Department of Urology, Emory University School of Medicine, Atlanta, GA, USA
- Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - William H Hudson
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, USA
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA, USA
| | - Donald J McGuire
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, USA
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA, USA
| | - Rebecca C Obeng
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, USA
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA, USA
- Department of Pathology, Emory University School of Medicine, Atlanta, GA, USA
- Department of Pathology and Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Andreas Wieland
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, USA
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA, USA
- Department of Otolaryngology, The Ohio State University College of Medicine, Columbus, OH, USA
- The Pelotonia Institute for Immuno-Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| | - Judong Lee
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, USA
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA, USA
| | - Daniel T McManus
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, USA
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA, USA
| | - James L Ross
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, USA
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA, USA
| | - Se Jin Im
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, USA
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA, USA
- Department of Immunology, Sungkyunkwan University School of Medicine, Suwon, Republic of Korea
| | - Junghwa Lee
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, USA
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA, USA
- Department of Precision Medicine, Sungkyunkwan University School of Medicine, Suwon, Republic of Korea
| | - Jian-Xin Lin
- Laboratory of Molecular Immunology and the Immunology Center, National Heart, Lung, and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Bin Hu
- Department of Medicine, Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, CA, USA
| | - Erin E West
- Laboratory of Molecular Immunology and the Immunology Center, National Heart, Lung, and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda, MD, USA
- Complement and Inflammation Research Section (CIRS), National Heart, Lung, and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Christopher D Scharer
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA, USA
| | - Gordon J Freeman
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Arlene H Sharpe
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
- Evergrande Center for Immunological Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA, USA
| | - Suresh S Ramalingam
- Winship Cancer Institute, Emory University, Atlanta, GA, USA
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, USA
| | | | | | - William J Greenleaf
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | | | - Jorg J Goronzy
- Department of Immunology, Mayo Clinic School of Medicine and Sciences, Rochester, MN, USA
- Department of Medicine, Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, CA, USA
| | - Pablo Umaña
- Roche Innovation Center Zurich, Schlieren, Switzerland
| | - Warren J Leonard
- Laboratory of Molecular Immunology and the Immunology Center, National Heart, Lung, and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Kendall A Smith
- Department of Medicine, Division of Immunology, Weill Medical College of Cornell University, New York, NY, USA
| | - Rafi Ahmed
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, USA.
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA, USA.
- Winship Cancer Institute, Emory University, Atlanta, GA, USA.
| |
Collapse
|
290
|
Alfonsi T, Bernasconi A, Canakoglu A, Masseroli M. Genomic data integration and user-defined sample-set extraction for population variant analysis. BMC Bioinformatics 2022; 23:401. [PMID: 36175857 PMCID: PMC9520931 DOI: 10.1186/s12859-022-04927-0] [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: 06/17/2022] [Accepted: 09/13/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Population variant analysis is of great importance for gathering insights into the links between human genotype and phenotype. The 1000 Genomes Project established a valuable reference for human genetic variation; however, the integrative use of the corresponding data with other datasets within existing repositories and pipelines is not fully supported. Particularly, there is a pressing need for flexible and fast selection of population partitions based on their variant and metadata-related characteristics. RESULTS Here, we target general germline or somatic mutation data sources for their seamless inclusion within an interoperable-format repository, supporting integration among them and with other genomic data, as well as their integrated use within bioinformatic workflows. In addition, we provide VarSum, a data summarization service working on sub-populations of interest selected using filters on population metadata and/or variant characteristics. The service is developed as an optimized computational framework with an Application Programming Interface (API) that can be called from within any existing computing pipeline or programming script. Provided example use cases of biological interest show the relevance, power and ease of use of the API functionalities. CONCLUSIONS The proposed data integration pipeline and data set extraction and summarization API pave the way for solid computational infrastructures that quickly process cumbersome variation data, and allow biologists and bioinformaticians to easily perform scalable analysis on user-defined partitions of large cohorts from increasingly available genetic variation studies. With the current tendency to large (cross)nation-wide sequencing and variation initiatives, we expect an ever growing need for the kind of computational support hereby proposed.
Collapse
Affiliation(s)
- Tommaso Alfonsi
- Department of Electronics, Information and Bioengineering, Politecnico di Milano, Via Ponzio 34/5, 20133, Milan, Italy.
| | - Anna Bernasconi
- Department of Electronics, Information and Bioengineering, Politecnico di Milano, Via Ponzio 34/5, 20133, Milan, Italy
| | - Arif Canakoglu
- Department of Electronics, Information and Bioengineering, Politecnico di Milano, Via Ponzio 34/5, 20133, Milan, Italy.,Dipartimento di Anestesia, Rianimazione ed Emergenza-Urgenza, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Policlinico di Milano, Via Francesco Sforza, 35, 20122, Milan, Italy
| | - Marco Masseroli
- Department of Electronics, Information and Bioengineering, Politecnico di Milano, Via Ponzio 34/5, 20133, Milan, Italy
| |
Collapse
|
291
|
de Hoon M, Bonetti A, Plessy C, Ando Y, Hon CC, Ishizu Y, Itoh M, Kato S, Lin D, Maekawa S, Murata M, Nishiyori H, Shin JW, Stolte J, Suzuki AM, Tagami M, Takahashi H, Thongjuea S, Forrest ARR, Hayashizaki Y, Kere J, Carninci P. Deep sequencing of short capped RNAs reveals novel families of noncoding RNAs. Genome Res 2022; 32:1727-1735. [PMID: 35961773 PMCID: PMC9528987 DOI: 10.1101/gr.276647.122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 08/09/2022] [Indexed: 12/03/2022]
Abstract
In eukaryotes, capped RNAs include long transcripts such as messenger RNAs and long noncoding RNAs, as well as shorter transcripts such as spliceosomal RNAs, small nucleolar RNAs, and enhancer RNAs. Long capped transcripts can be profiled using cap analysis gene expression (CAGE) sequencing and other methods. Here, we describe a sequencing library preparation protocol for short capped RNAs, apply it to a differentiation time course of the human cell line THP-1, and systematically compare the landscape of short capped RNAs to that of long capped RNAs. Transcription initiation peaks associated with genes in the sense direction have a strong preference to produce either long or short capped RNAs, with one out of six peaks detected in the short capped RNA libraries only. Gene-associated short capped RNAs have highly specific 3' ends, typically overlapping splice sites. Enhancers also preferentially generate either short or long capped RNAs, with 10% of enhancers observed in the short capped RNA libraries only. Enhancers producing either short or long capped RNAs show enrichment for GWAS-associated disease SNPs. We conclude that deep sequencing of short capped RNAs reveals new families of noncoding RNAs and elucidates the diversity of transcripts generated at known and novel promoters and enhancers.
Collapse
Affiliation(s)
- Michiel de Hoon
- RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa 230-0045, Japan
| | - Alessandro Bonetti
- RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa 230-0045, Japan
| | - Charles Plessy
- RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa 230-0045, Japan
| | - Yoshinari Ando
- RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa 230-0045, Japan
| | - Chung-Chau Hon
- RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa 230-0045, Japan
| | - Yuri Ishizu
- RIKEN Center for Life Science Technologies, Division of Genomic Technologies, Yokohama, Kanagawa 230-0045, Japan
| | - Masayoshi Itoh
- RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa 230-0045, Japan
- RIKEN Preventive Medicine and Diagnosis Innovation Program, Wako, Saitama 351-0198, Japan
| | - Sachi Kato
- RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa 230-0045, Japan
| | - Dongyan Lin
- RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa 230-0045, Japan
- Integrated Program in Neuroscience, McGill University, Montreal, Quebec H3A 1A1, Canada
- Mila, Montreal, Quebec H2S 3H1, Canada
| | - Sho Maekawa
- RIKEN Omics Science Center (OSC), Yokohama, Kanagawa 230-0045, Japan
| | - Mitsuyoshi Murata
- RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa 230-0045, Japan
| | - Hiromi Nishiyori
- RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa 230-0045, Japan
| | - Jay W Shin
- RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa 230-0045, Japan
- Genome Institute of Singapore, Agency for Science, Technology and Research (A*STAR), Singapore, 138632, Singapore
| | - Jens Stolte
- RIKEN Center for Life Science Technologies, Division of Genomic Technologies, Yokohama, Kanagawa 230-0045, Japan
| | - Ana Maria Suzuki
- RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa 230-0045, Japan
| | - Michihira Tagami
- RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa 230-0045, Japan
| | - Hazuki Takahashi
- RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa 230-0045, Japan
| | - Supat Thongjuea
- RIKEN Center for Life Science Technologies, Division of Genomic Technologies, Yokohama, Kanagawa 230-0045, Japan
| | - Alistair R R Forrest
- RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa 230-0045, Japan
- Harry Perkins Institute of Medical Research, QEII Medical Centre and Centre for Medical Research, The University of Western Australia, Nedlands, Perth, Western Australia 6009, Australia
| | - Yoshihide Hayashizaki
- RIKEN Preventive Medicine and Diagnosis Innovation Program, Wako, Saitama 351-0198, Japan
- RIKEN Omics Science Center (OSC), Yokohama, Kanagawa 230-0045, Japan
| | - Juha Kere
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge 14157, Sweden
- Stem Cells and Metabolism Research Program, University of Helsinki and Folkhälsan Research Center, Helsinki 00290, Finland
| | - Piero Carninci
- RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa 230-0045, Japan
- Human Technopole, Milan 20157, Italy
| |
Collapse
|
292
|
Jaeger B, Schupp JC, Plappert L, Terwolbeck O, Artysh N, Kayser G, Engelhard P, Adams TS, Zweigerdt R, Kempf H, Lienenklaus S, Garrels W, Nazarenko I, Jonigk D, Wygrecka M, Klatt D, Schambach A, Kaminski N, Prasse A. Airway basal cells show a dedifferentiated KRT17 highPhenotype and promote fibrosis in idiopathic pulmonary fibrosis. Nat Commun 2022; 13:5637. [PMID: 36163190 PMCID: PMC9513076 DOI: 10.1038/s41467-022-33193-0] [Citation(s) in RCA: 63] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Accepted: 09/07/2022] [Indexed: 11/10/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a fatal disease with limited treatment options. In this study, we focus on the properties of airway basal cells (ABC) obtained from patients with IPF (IPF-ABC). Single cell RNA sequencing (scRNAseq) of bronchial brushes revealed extensive reprogramming of IPF-ABC towards a KRT17high PTENlow dedifferentiated cell type. In the 3D organoid model, compared to ABC obtained from healthy volunteers, IPF-ABC give rise to more bronchospheres, de novo bronchial structures resembling lung developmental processes, induce fibroblast proliferation and extracellular matrix deposition in co-culture. Intratracheal application of IPF-ABC into minimally injured lungs of Rag2-/- or NRG mice causes severe fibrosis, remodeling of the alveolar compartment, and formation of honeycomb cyst-like structures. Connectivity MAP analysis of scRNAseq of bronchial brushings suggested that gene expression changes in IPF-ABC can be reversed by SRC inhibition. After demonstrating enhanced SRC expression and activity in these cells, and in IPF lungs, we tested the effects of saracatinib, a potent SRC inhibitor previously studied in humans. We demonstrate that saracatinib modified in-vitro and in-vivo the profibrotic changes observed in our 3D culture system and novel mouse xenograft model.
Collapse
Affiliation(s)
- Benedikt Jaeger
- Fraunhofer Institute for Toxicology and Experimental Medicine, Hannover, Germany
- German Center for Lung Research, BREATH, Hannover, Germany
| | - Jonas Christian Schupp
- German Center for Lung Research, BREATH, Hannover, Germany
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine, New Haven, CT, USA
- Department of Pulmonology, Hannover Medical School, Hannover, Germany
| | - Linda Plappert
- Fraunhofer Institute for Toxicology and Experimental Medicine, Hannover, Germany
- German Center for Lung Research, BREATH, Hannover, Germany
| | - Oliver Terwolbeck
- Fraunhofer Institute for Toxicology and Experimental Medicine, Hannover, Germany
- German Center for Lung Research, BREATH, Hannover, Germany
| | - Nataliia Artysh
- Fraunhofer Institute for Toxicology and Experimental Medicine, Hannover, Germany
- German Center for Lung Research, BREATH, Hannover, Germany
- Department of Pulmonology, Hannover Medical School, Hannover, Germany
| | - Gian Kayser
- Institute of Surgical Pathology, University Medical Center, Freiburg, Germany
| | - Peggy Engelhard
- Department of Pneumology, University Medical Center, Freiburg, Germany
| | - Taylor Sterling Adams
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Robert Zweigerdt
- Leibniz Research Laboratories for Biotechnology and Artificial Organs, Hannover Medical School, Hannover, Germany
| | - Henning Kempf
- Leibniz Research Laboratories for Biotechnology and Artificial Organs, Hannover Medical School, Hannover, Germany
| | - Stefan Lienenklaus
- Institute for Laboratory Animal Science, Hannover Medical School, Hannover, Germany
| | - Wiebke Garrels
- Institute for Laboratory Animal Science, Hannover Medical School, Hannover, Germany
| | - Irina Nazarenko
- Institute for Infection Prevention and Hospital Epidemiology, Medical Center - University of Freiburg, Freiburg, Germany
- German Cancer Consortium (DKTK), Partner Site Freiburg and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Danny Jonigk
- German Center for Lung Research, BREATH, Hannover, Germany
- Institute of Pathology, Hannover Medical School, Hannover, Germany
| | - Malgorzata Wygrecka
- Department of Biochemistry, Faculty of Medicine, Justus Liebig University, Gießen, Germany
| | - Denise Klatt
- Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany
| | - Axel Schambach
- Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany
- Division of Hematology/Oncology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Naftali Kaminski
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Antje Prasse
- Fraunhofer Institute for Toxicology and Experimental Medicine, Hannover, Germany.
- German Center for Lung Research, BREATH, Hannover, Germany.
- Department of Pulmonology, Hannover Medical School, Hannover, Germany.
| |
Collapse
|
293
|
Wijesinghe SN, Anderson J, Brown TJ, Nanus DE, Housmans B, Green JA, Hackl M, Choi KK, Arkill KP, Welting T, James V, Jones SW, Peffers MJ. The role of extracellular vesicle miRNAs and tRNAs in synovial fibroblast senescence. Front Mol Biosci 2022; 9:971621. [PMID: 36213127 PMCID: PMC9537453 DOI: 10.3389/fmolb.2022.971621] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Accepted: 09/06/2022] [Indexed: 11/24/2022] Open
Abstract
Extracellular vesicles are mediators of intercellular communication with critical roles in cellular senescence and ageing. In arthritis, senescence is linked to the activation of a pro-inflammatory phenotype contributing to chronic arthritis pathogenesis. We hypothesised that senescent osteoarthritic synovial fibroblasts induce senescence and a pro-inflammatory phenotype in non-senescent osteoarthritic fibroblasts, mediated through extracellular vesicle cargo. Small RNA-sequencing and mass spectrometry proteomics were performed on extracellular vesicles isolated from the secretome of non-senescent and irradiation-induced senescent synovial fibroblasts. β-galactosidase staining confirmed senescence in SFs. RNA sequencing identified 17 differentially expressed miRNAs, 11 lncRNAs, 14 tRNAs and one snoRNA and, 21 differentially abundant proteins were identified by mass spectrometry. Bioinformatics analysis of miRNAs identified fibrosis, cell proliferation, autophagy, and cell cycle as significant pathways, tRNA analysis was enriched for signaling pathways including FGF, PI3K/AKT and MAPK, whilst protein analysis identified PAX3-FOXO1, MYC and TFGB1 as enriched upstream regulators involved in senescence and cell cycle arrest. Finally, treatment of non-senescent synovial fibroblasts with senescent extracellular vesicles confirmed the bystander effect, inducing senescence in non-senescent cells potentially through down regulation of NF-κβ and cAMP response element signaling pathways thus supporting our hypothesis. Understanding the exact composition of EV-derived small RNAs of senescent cells in this way will inform our understanding of their roles in inflammation, intercellular communication, and as active molecules in the senescence bystander effect.
Collapse
Affiliation(s)
- Susanne N. Wijesinghe
- Institute of Inflammation and Ageing, MRC- Versus Arthritis Centre for Musculoskeletal Ageing Research, University of Birmingham, Birmingham, United Kingdom
| | - James Anderson
- Institute of Life Course and Medical Sciences, Faculty of Health and Life Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Thomas J. Brown
- School of Veterinary Medicine and Science, University of Nottingham, Nottingham, United Kingdom
| | - Dominika E. Nanus
- Institute of Inflammation and Ageing, MRC- Versus Arthritis Centre for Musculoskeletal Ageing Research, University of Birmingham, Birmingham, United Kingdom
| | - Bas Housmans
- Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, Maastricht University Medical Center, Maastricht, Netherlands
| | | | | | - Katie K. Choi
- School of Medicine, University of Nottingham, Nottingham, United Kingdom
| | - Kenton P. Arkill
- School of Medicine, University of Nottingham, Nottingham, United Kingdom
| | - Tim Welting
- Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, Maastricht University Medical Center, Maastricht, Netherlands
| | - Victoria James
- School of Veterinary Medicine and Science, University of Nottingham, Nottingham, United Kingdom
| | - Simon W. Jones
- Institute of Inflammation and Ageing, MRC- Versus Arthritis Centre for Musculoskeletal Ageing Research, University of Birmingham, Birmingham, United Kingdom
| | - Mandy J. Peffers
- Institute of Life Course and Medical Sciences, Faculty of Health and Life Sciences, University of Liverpool, Liverpool, United Kingdom
| |
Collapse
|
294
|
Balcerak A, Macech-Klicka E, Wakula M, Tomecki R, Goryca K, Rydzanicz M, Chmielarczyk M, Szostakowska-Rodzos M, Wisniewska M, Lyczek F, Helwak A, Tollervey D, Kudla G, Grzybowska EA. The RNA-Binding Landscape of HAX1 Protein Indicates Its Involvement in Translation and Ribosome Assembly. Cells 2022; 11:cells11192943. [PMID: 36230905 PMCID: PMC9564044 DOI: 10.3390/cells11192943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 09/13/2022] [Accepted: 09/15/2022] [Indexed: 11/18/2022] Open
Abstract
HAX1 is a human protein with no known homologues or structural domains. Mutations in the HAX1 gene cause severe congenital neutropenia through mechanisms that are poorly understood. Previous studies reported the RNA-binding capacity of HAX1, but the role of this binding in physiology and pathology remains unexplained. Here, we report the transcriptome-wide characterization of HAX1 RNA targets using RIP-seq and CRAC, indicating that HAX1 binds transcripts involved in translation, ribosome biogenesis, and rRNA processing. Using CRISPR knockouts, we find that HAX1 RNA targets partially overlap with transcripts downregulated in HAX1 KO, implying a role in mRNA stabilization. Gene ontology analysis demonstrated that genes differentially expressed in HAX1 KO (including genes involved in ribosome biogenesis and translation) are also enriched in a subset of genes whose expression correlates with HAX1 expression in four analyzed neoplasms. The functional connection to ribosome biogenesis was also demonstrated by gradient sedimentation ribosome profiles, which revealed differences in the small subunit:monosome ratio in HAX1 WT/KO. We speculate that changes in HAX1 expression may be important for the etiology of HAX1-linked diseases through dysregulation of translation.
Collapse
Affiliation(s)
- Anna Balcerak
- Molecular and Translational Oncology, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland
| | - Ewelina Macech-Klicka
- Molecular and Translational Oncology, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland
| | - Maciej Wakula
- Molecular and Translational Oncology, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland
| | - Rafal Tomecki
- Laboratory of RNA Processing and Decay, Institute of Biochemistry and Biophysics, Polish Academy of Sciences, 02-106 Warsaw, Poland
- Faculty of Biology, Institute of Genetics and Biotechnology, University of Warsaw, 02-106 Warsaw, Poland
| | - Krzysztof Goryca
- Genomics Core Facility, Centre of New Technologies University of Warsaw, 02-097 Warsaw, Poland
| | - Malgorzata Rydzanicz
- Department of Medical Genetics, Medical University of Warsaw, 02-106 Warsaw, Poland
| | - Mateusz Chmielarczyk
- Molecular and Translational Oncology, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland
| | - Malgorzata Szostakowska-Rodzos
- Molecular and Translational Oncology, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland
| | - Marta Wisniewska
- Laboratory of Biological Chemistry of Metal Ions, Institute of Biochemistry and Biophysics, Polish Academy of Sciences, 02-106 Warsaw, Poland
| | - Filip Lyczek
- Molecular and Translational Oncology, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland
| | - Aleksandra Helwak
- Wellcome Centre for Cell Biology, University of Edinburgh, Edinburgh EH9 3BF, UK
| | - David Tollervey
- Wellcome Centre for Cell Biology, University of Edinburgh, Edinburgh EH9 3BF, UK
| | - Grzegorz Kudla
- MRC Human Genetics Unit, University of Edinburgh, Edinburgh EH4 2XU, UK
| | - Ewa A. Grzybowska
- Molecular and Translational Oncology, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland
- Correspondence:
| |
Collapse
|
295
|
Marklund M, Schultz N, Friedrich S, Berglund E, Tarish F, Tanoglidi A, Liu Y, Bergenstråhle L, Erickson A, Helleday T, Lamb AD, Sonnhammer E, Lundeberg J. Spatio-temporal analysis of prostate tumors in situ suggests pre-existence of treatment-resistant clones. Nat Commun 2022; 13:5475. [PMID: 36115838 PMCID: PMC9482614 DOI: 10.1038/s41467-022-33069-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Accepted: 08/30/2022] [Indexed: 11/25/2022] Open
Abstract
The molecular mechanisms underlying lethal castration-resistant prostate cancer remain poorly understood, with intratumoral heterogeneity a likely contributing factor. To examine the temporal aspects of resistance, we analyze tumor heterogeneity in needle biopsies collected before and after treatment with androgen deprivation therapy. By doing so, we are able to couple clinical responsiveness and morphological information such as Gleason score to transcriptome-wide data. Our data-driven analysis of transcriptomes identifies several distinct intratumoral cell populations, characterized by their unique gene expression profiles. Certain cell populations present before treatment exhibit gene expression profiles that match those of resistant tumor cell clusters, present after treatment. We confirm that these clusters are resistant by the localization of active androgen receptors to the nuclei in cancer cells post-treatment. Our data also demonstrates that most stromal cells adjacent to resistant clusters do not express the androgen receptor, and we identify differentially expressed genes for these cells. Altogether, this study shows the potential to increase the power in predicting resistant tumors.
Collapse
Affiliation(s)
- Maja Marklund
- Department of Gene Technology, KTH Royal Institute of Technology, Science for Life Laboratory, Solna, Sweden
| | - Niklas Schultz
- Division of Translational Medicine & Chemical Biology, Karolinska Institute, Science for Life Laboratory, Solna, Sweden
| | - Stefanie Friedrich
- Department of Biochemistry and Biophysics, Stockholm University, Science for Laboratory, Solna, Sweden
| | - Emelie Berglund
- Department of Gene Technology, KTH Royal Institute of Technology, Science for Life Laboratory, Solna, Sweden
| | - Firas Tarish
- Division of Translational Medicine & Chemical Biology, Karolinska Institute, Science for Life Laboratory, Solna, Sweden
| | - Anna Tanoglidi
- Department of Pathology, Evangelismos General Hospital, 45-47 Ipsilantou str, Athens, Greece
| | - Yao Liu
- Division of Translational Medicine & Chemical Biology, Karolinska Institute, Science for Life Laboratory, Solna, Sweden
| | - Ludvig Bergenstråhle
- Department of Gene Technology, KTH Royal Institute of Technology, Science for Life Laboratory, Solna, Sweden
| | - Andrew Erickson
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK
| | - Thomas Helleday
- Division of Translational Medicine & Chemical Biology, Karolinska Institute, Science for Life Laboratory, Solna, Sweden
| | - Alastair D Lamb
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK
| | - Erik Sonnhammer
- Department of Biochemistry and Biophysics, Stockholm University, Science for Laboratory, Solna, Sweden.
| | - Joakim Lundeberg
- Department of Gene Technology, KTH Royal Institute of Technology, Science for Life Laboratory, Solna, Sweden.
| |
Collapse
|
296
|
Wu AC, Vivori C, Patel H, Sideri T, Moretto F, van Werven FJ. RSC and GRFs confer promoter directionality by restricting divergent noncoding transcription. Life Sci Alliance 2022; 5:e202201394. [PMID: 36114005 PMCID: PMC9481977 DOI: 10.26508/lsa.202201394] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 09/02/2022] [Accepted: 09/02/2022] [Indexed: 11/24/2022] Open
Abstract
The directionality of gene promoters-the ratio of protein-coding over divergent noncoding transcription-is highly variable. How promoter directionality is controlled remains poorly understood. Here, we show that the chromatin remodelling complex RSC and general regulatory factors (GRFs) dictate promoter directionality by attenuating divergent transcription relative to protein-coding transcription. At gene promoters that are highly directional, depletion of RSC leads to a relative increase in divergent noncoding transcription and thus to a decrease in promoter directionality. We find that RSC has a modest effect on nucleosome positioning upstream in promoters at the sites of divergent transcription. These promoters are also enriched for the binding of GRFs such as Reb1 and Abf1. Ectopic targeting of divergent transcription initiation sites with GRFs or the dCas9 DNA-binding protein suppresses divergent transcription. Our data suggest that RSC and GRFs play a pervasive role in limiting divergent transcription relative to coding direction transcription. We propose that any DNA-binding factor, when stably associated with cryptic transcription start sites, forms a barrier which represses divergent transcription, thereby promoting promoter directionality.
Collapse
Affiliation(s)
- Andrew Ck Wu
- Cell Fate and Gene Regulation Laboratory, The Francis Crick Institute, London, UK
| | - Claudia Vivori
- Cell Fate and Gene Regulation Laboratory, The Francis Crick Institute, London, UK
| | - Harshil Patel
- Bioinformatics and Biostatistics, The Francis Crick Institute, London, UK
| | - Theodora Sideri
- Cell Fate and Gene Regulation Laboratory, The Francis Crick Institute, London, UK
| | - Fabien Moretto
- Cell Fate and Gene Regulation Laboratory, The Francis Crick Institute, London, UK
- Institute of Molecular Biology and Biotechnology (IMBB), Foundation for Research and Technology-Hellas (FORTH), Heraklion, Greece
| | - Folkert J van Werven
- Cell Fate and Gene Regulation Laboratory, The Francis Crick Institute, London, UK
| |
Collapse
|
297
|
Stephenson HN, Streeck R, Grüblinger F, Goosmann C, Herzig A. Hemocytes are essential for Drosophila melanogaster post-embryonic development, independent of control of the microbiota. Development 2022; 149:dev200286. [PMID: 36093870 PMCID: PMC9641648 DOI: 10.1242/dev.200286] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 08/19/2022] [Indexed: 09/22/2023]
Abstract
Proven roles for hemocytes (blood cells) have expanded beyond the control of infections in Drosophila. Despite this, the crucial role of hemocytes in post-embryonic development has long thought to be limited to control of microorganisms during metamorphosis. This has previously been shown by rescue of adult development in hemocyte-ablation models under germ-free conditions. Here, we show that hemocytes have an essential role in post-embryonic development beyond their ability to control the microbiota. Using a newly generated strong hemocyte-specific driver line for the GAL4/UAS system, we show that specific ablation of hemocytes is early pupal lethal, even under axenic conditions. Genetic rescue experiments prove that this is a hemocyte-specific phenomenon. RNA-seq data suggests that dysregulation of the midgut is a prominent consequence of hemocyte ablation in larval stages, resulting in reduced gut lengths. Dissection suggests that multiple processes may be affected during metamorphosis. We believe this previously unreported role for hemocytes during metamorphosis is a major finding for the field.
Collapse
Affiliation(s)
- Holly N. Stephenson
- Department of Cellular Microbiology, Max Planck Institute for Infection Biology, Charitéplatz 1, Berlin 10117, Germany
- Peninsula Medical School, Faculty of Health,University of Plymouth, Plymouth, Devon PL4 8AA, UK
| | - Robert Streeck
- Department of Cellular Microbiology, Max Planck Institute for Infection Biology, Charitéplatz 1, Berlin 10117, Germany
| | - Florian Grüblinger
- Department of Cellular Microbiology, Max Planck Institute for Infection Biology, Charitéplatz 1, Berlin 10117, Germany
| | - Christian Goosmann
- Department of Cellular Microbiology, Max Planck Institute for Infection Biology, Charitéplatz 1, Berlin 10117, Germany
| | - Alf Herzig
- Department of Cellular Microbiology, Max Planck Institute for Infection Biology, Charitéplatz 1, Berlin 10117, Germany
| |
Collapse
|
298
|
Laloum D, Robinson-Rechavi M. Rhythmicity is linked to expression cost at the protein level but to expression precision at the mRNA level. PLoS Comput Biol 2022; 18:e1010399. [PMID: 36095022 PMCID: PMC9518874 DOI: 10.1371/journal.pcbi.1010399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 09/28/2022] [Accepted: 07/17/2022] [Indexed: 11/18/2022] Open
Abstract
Many genes have nycthemeral rhythms of expression, i.e. a 24-hours periodic variation, at either mRNA or protein level or both, and most rhythmic genes are tissue-specific. Here, we investigate and discuss the evolutionary origins of rhythms in gene expression. Our results suggest that rhythmicity of protein expression could have been favored by selection to minimize costs. Trends are consistent in bacteria, plants and animals, and are also supported by tissue-specific patterns in mouse. Unlike for protein level, cost cannot explain rhythm at the RNA level. We suggest that instead it allows to periodically reduce expression noise. Noise control had the strongest support in mouse, with limited evidence in other species. We have also found that genes under stronger purifying selection are rhythmically expressed at the mRNA level, and we propose that this is because they are noise sensitive genes. Finally, the adaptive role of rhythmic expression is supported by rhythmic genes being highly expressed yet tissue-specific. This provides a good evolutionary explanation for the observation that nycthemeral rhythms are often tissue-specific. For many genes, their expression, i.e. the production of RNA and proteins, is rhythmic with a 24-hour period. Here, we study and discuss the evolutionary origins of these rhythms. Our analyses of data from different species suggest that the rhythmicity of protein level may have been favored by selection for cost minimization. Furthermore, we have shown that cost cannot explain the rhythmic variations in RNA levels. Instead, we suggest that it periodically reduces the stochasticity of gene expression. We also found that genes under stronger purifying selection are rhythmically expressed at the mRNA level, and propose that this is because they are noise-sensitive genes. Finally, rhythmic expression involves genes that are often highly expressed and tissue-specific. This provides a good evolutionary explanation for the tissue-specificity of these rhythms.
Collapse
Affiliation(s)
- David Laloum
- Department of Ecology and Evolution, Batiment Biophore, Quartier UNIL-Sorge, Université de Lausanne, Lausanne, Switzerland
- Swiss Institute of Bioinformatics, Batiment Génopode, Quartier UNIL-Sorge, Université de Lausanne, Lausanne, Switzerland
| | - Marc Robinson-Rechavi
- Department of Ecology and Evolution, Batiment Biophore, Quartier UNIL-Sorge, Université de Lausanne, Lausanne, Switzerland
- Swiss Institute of Bioinformatics, Batiment Génopode, Quartier UNIL-Sorge, Université de Lausanne, Lausanne, Switzerland
- * E-mail:
| |
Collapse
|
299
|
Gnocis: An integrated system for interactive and reproducible analysis and modelling of cis-regulatory elements in Python 3. PLoS One 2022; 17:e0274338. [PMID: 36084008 PMCID: PMC9462789 DOI: 10.1371/journal.pone.0274338] [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: 08/07/2021] [Accepted: 08/25/2022] [Indexed: 11/23/2022] Open
Abstract
Gene expression is regulated through cis-regulatory elements (CREs), among which are promoters, enhancers, Polycomb/Trithorax Response Elements (PREs), silencers and insulators. Computational prediction of CREs can be achieved using a variety of statistical and machine learning methods combined with different feature space formulations. Although Python packages for DNA sequence feature sets and for machine learning are available, no existing package facilitates the combination of DNA sequence feature sets with machine learning methods for the genome-wide prediction of candidate CREs. We here present Gnocis, a Python package that streamlines the analysis and the modelling of CRE sequences by providing extensible APIs and implementing the glue required for combining feature sets and models for genome-wide prediction. Gnocis implements a variety of base feature sets, including motif pair occurrence frequencies and the k-spectrum mismatch kernel. It integrates with Scikit-learn and TensorFlow for state-of-the-art machine learning. Gnocis additionally implements a broad suite of tools for the handling and preparation of sequence, region and curve data, which can be useful for general DNA bioinformatics in Python. We also present Deep-MOCCA, a neural network architecture inspired by SVM-MOCCA that achieves moderate to high generalization without prior motif knowledge. To demonstrate the use of Gnocis, we applied multiple machine learning methods to the modelling of D. melanogaster PREs, including a Convolutional Neural Network (CNN), making this the first study to model PREs with CNNs. The models are readily adapted to new CRE modelling problems and to other organisms. In order to produce a high-performance, compiled package for Python 3, we implemented Gnocis in Cython. Gnocis can be installed using the PyPI package manager by running ‘pip install gnocis’. The source code is available on GitHub, at https://github.com/bjornbredesen/gnocis.
Collapse
|
300
|
Shibai A, Kotani H, Sakata N, Furusawa C, Tsuru S. Purifying selection enduringly acts on the sequence evolution of highly expressed proteins in Escherichia coli. G3 GENES|GENOMES|GENETICS 2022; 12:6694045. [PMID: 36073932 PMCID: PMC9635659 DOI: 10.1093/g3journal/jkac235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Accepted: 08/27/2022] [Indexed: 11/17/2022]
Abstract
The evolutionary speed of a protein sequence is constrained by its expression level, with highly expressed proteins evolving relatively slowly. This negative correlation between expression levels and evolutionary rates (known as the E–R anticorrelation) has already been widely observed in past macroevolution between species from bacteria to animals. However, it remains unclear whether this seemingly general law also governs recent evolution, including past and de novo, within a species. However, the advent of genomic sequencing and high-throughput phenotyping, particularly for bacteria, has revealed fundamental gaps between the 2 evolutionary processes and has provided empirical data opposing the possible underlying mechanisms which are widely believed. These conflicts raise questions about the generalization of the E–R anticorrelation and the relevance of plausible mechanisms. To explore the ubiquitous impact of expression levels on molecular evolution and test the relevance of the possible underlying mechanisms, we analyzed the genome sequences of 99 strains of Escherichia coli for evolution within species in nature. We also analyzed genomic mutations accumulated under laboratory conditions as a model of de novo evolution within species. Here, we show that E–R anticorrelation is significant in both past and de novo evolution within species in E. coli. Our data also confirmed ongoing purifying selection on highly expressed genes. Ongoing selection included codon-level purifying selection, supporting the relevance of the underlying mechanisms. However, the impact of codon-level purifying selection on the constraints in evolution within species might be smaller than previously expected from evolution between species.
Collapse
Affiliation(s)
- Atsushi Shibai
- Center for Biosystems Dynamics Research (BDR), RIKEN , Osaka 565-0874, Japan
| | - Hazuki Kotani
- Center for Biosystems Dynamics Research (BDR), RIKEN , Osaka 565-0874, Japan
| | - Natsue Sakata
- Center for Biosystems Dynamics Research (BDR), RIKEN , Osaka 565-0874, Japan
| | - Chikara Furusawa
- Center for Biosystems Dynamics Research (BDR), RIKEN , Osaka 565-0874, Japan
- Universal Biology Institute, School of Science, The University of Tokyo , Tokyo 113-0033, Japan
| | - Saburo Tsuru
- Universal Biology Institute, School of Science, The University of Tokyo , Tokyo 113-0033, Japan
| |
Collapse
|