201
|
Villa CE, Cheroni C, Dotter CP, López-Tóbon A, Oliveira B, Sacco R, Yahya AÇ, Morandell J, Gabriele M, Tavakoli MR, Lyudchik J, Sommer C, Gabitto M, Danzl JG, Testa G, Novarino G. CHD8 haploinsufficiency links autism to transient alterations in excitatory and inhibitory trajectories. Cell Rep 2022; 39:110615. [PMID: 35385734 DOI: 10.1016/j.celrep.2022.110615] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 11/18/2021] [Accepted: 03/13/2022] [Indexed: 12/13/2022] Open
Abstract
Mutations in the chromodomain helicase DNA-binding 8 (CHD8) gene are a frequent cause of autism spectrum disorder (ASD). While its phenotypic spectrum often encompasses macrocephaly, implicating cortical abnormalities, how CHD8 haploinsufficiency affects neurodevelopmental is unclear. Here, employing human cerebral organoids, we find that CHD8 haploinsufficiency disrupted neurodevelopmental trajectories with an accelerated and delayed generation of, respectively, inhibitory and excitatory neurons that yields, at days 60 and 120, symmetrically opposite expansions in their proportions. This imbalance is consistent with an enlargement of cerebral organoids as an in vitro correlate of patients' macrocephaly. Through an isogenic design of patient-specific mutations and mosaic organoids, we define genotype-phenotype relationships and uncover their cell-autonomous nature. Our results define cell-type-specific CHD8-dependent molecular defects related to an abnormal program of proliferation and alternative splicing. By identifying cell-type-specific effects of CHD8 mutations, our study uncovers reproducible developmental alterations that may be employed for neurodevelopmental disease modeling.
Collapse
Affiliation(s)
- Carlo Emanuele Villa
- Department of Experimental Oncology, IEO, European Institute of Oncology, IRCCS, 20139 Milan, Italy; Human Technopole, Viale Rita Levi Montalcini 1, 20157 Milan, Italy
| | - Cristina Cheroni
- Department of Experimental Oncology, IEO, European Institute of Oncology, IRCCS, 20139 Milan, Italy; Human Technopole, Viale Rita Levi Montalcini 1, 20157 Milan, Italy; Department of Oncology and Hemato-oncology, University of Milan, 20122 Milan, Italy
| | - Christoph P Dotter
- Institute of Science and Technology (IST) Austria, 3400 Klosterneuburg, Austria
| | - Alejandro López-Tóbon
- Department of Experimental Oncology, IEO, European Institute of Oncology, IRCCS, 20139 Milan, Italy; Human Technopole, Viale Rita Levi Montalcini 1, 20157 Milan, Italy; Department of Oncology and Hemato-oncology, University of Milan, 20122 Milan, Italy
| | - Bárbara Oliveira
- Institute of Science and Technology (IST) Austria, 3400 Klosterneuburg, Austria
| | - Roberto Sacco
- Institute of Science and Technology (IST) Austria, 3400 Klosterneuburg, Austria
| | - Aysan Çerağ Yahya
- Institute of Science and Technology (IST) Austria, 3400 Klosterneuburg, Austria
| | - Jasmin Morandell
- Institute of Science and Technology (IST) Austria, 3400 Klosterneuburg, Austria
| | - Michele Gabriele
- Department of Experimental Oncology, IEO, European Institute of Oncology, IRCCS, 20139 Milan, Italy
| | - Mojtaba R Tavakoli
- Institute of Science and Technology (IST) Austria, 3400 Klosterneuburg, Austria
| | - Julia Lyudchik
- Institute of Science and Technology (IST) Austria, 3400 Klosterneuburg, Austria
| | - Christoph Sommer
- Institute of Science and Technology (IST) Austria, 3400 Klosterneuburg, Austria
| | | | - Johann G Danzl
- Institute of Science and Technology (IST) Austria, 3400 Klosterneuburg, Austria
| | - Giuseppe Testa
- Department of Experimental Oncology, IEO, European Institute of Oncology, IRCCS, 20139 Milan, Italy; Human Technopole, Viale Rita Levi Montalcini 1, 20157 Milan, Italy; Department of Oncology and Hemato-oncology, University of Milan, 20122 Milan, Italy.
| | - Gaia Novarino
- Institute of Science and Technology (IST) Austria, 3400 Klosterneuburg, Austria.
| |
Collapse
|
202
|
Jin Z, Kang J, Yu T. Feature selection and classification over the network with missing node observations. Stat Med 2022; 41:1242-1262. [PMID: 34816464 PMCID: PMC9773124 DOI: 10.1002/sim.9267] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 09/14/2021] [Accepted: 10/29/2021] [Indexed: 12/25/2022]
Abstract
Jointly analyzing transcriptomic data and the existing biological networks can yield more robust and informative feature selection results, as well as better understanding of the biological mechanisms. Selecting and classifying node features over genome-scale networks has become increasingly important in genomic biology and genomic medicine. Existing methods have some critical drawbacks. The first is they do not allow flexible modeling of different subtypes of selected nodes. The second is they ignore nodes with missing values, very likely to increase bias in estimation. To address these limitations, we propose a general modeling framework for Bayesian node classification (BNC) with missing values. A new prior model is developed for the class indicators incorporating the network structure. For posterior computation, we resort to the Swendsen-Wang algorithm for efficiently updating class indicators. BNC can naturally handle missing values in the Bayesian modeling framework, which improves the node classification accuracy and reduces the bias in estimating gene effects. We demonstrate the advantages of our methods via extensive simulation studies and the analysis of the cutaneous melanoma dataset from The Cancer Genome Atlas.
Collapse
Affiliation(s)
| | - Jian Kang
- Department of Biostatistics, University of Michigan, Ann Arbor, Michigan
| | - Tianwei Yu
- School of Data Science and Warshel Institute, The Chinese University of Hong Kong - Shenzhen, and Shenzhen Research Institute of Big Data, Shenzhen, China
| |
Collapse
|
203
|
Larson A, Codden CJ, Huggins GS, Rastegar H, Chen FY, Maron BJ, Rowin EJ, Maron MS, Chin MT. Altered intercellular communication and extracellular matrix signaling as a potential disease mechanism in human hypertrophic cardiomyopathy. Sci Rep 2022; 12:5211. [PMID: 35338173 PMCID: PMC8956620 DOI: 10.1038/s41598-022-08561-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 03/08/2022] [Indexed: 11/17/2022] Open
Abstract
Hypertrophic cardiomyopathy (HCM) is considered a primary disorder of the sarcomere resulting in unexplained left ventricular hypertrophy but the paradoxical association of nonmyocyte phenotypes such as fibrosis, mitral valve anomalies and microvascular occlusion is unexplained. To understand the interplay between cardiomyocyte and nonmyocyte cell types in human HCM, single nuclei RNA-sequencing was performed on myectomy specimens from HCM patients with left ventricular outflow tract obstruction and control samples from donor hearts free of cardiovascular disease. Clustering analysis based on gene expression patterns identified a total of 34 distinct cell populations, which were classified into 10 different cell types based on marker gene expression. Differential gene expression analysis comparing HCM to Normal datasets revealed differences in sarcomere and extracellular matrix gene expression. Analysis of expressed ligand-receptor pairs across multiple cell types indicated profound alteration in HCM intercellular communication, particularly between cardiomyocytes and fibroblasts, fibroblasts and lymphocytes and involving integrin β1 and its multiple extracellular matrix (ECM) cognate ligands. These findings provide a paradigm for how sarcomere dysfunction is associated with reduced cardiomyocyte secretion of ECM ligands, altered fibroblast ligand-receptor interactions with other cell types and increased fibroblast to lymphocyte signaling, which can further alter the ECM composition and promote nonmyocyte phenotypes.
Collapse
Affiliation(s)
- Amy Larson
- Molecular Cardiology Research Institute, Tufts Medical Center, 800 Washington Street, Box 80, Boston, MA, 02111, USA
| | - Christina J Codden
- Molecular Cardiology Research Institute, Tufts Medical Center, 800 Washington Street, Box 80, Boston, MA, 02111, USA
| | - Gordon S Huggins
- Molecular Cardiology Research Institute, Tufts Medical Center, 800 Washington Street, Box 80, Boston, MA, 02111, USA.,CardioVascular Center, Tufts Medical Center, Boston, MA, USA
| | - Hassan Rastegar
- CardioVascular Center, Tufts Medical Center, Boston, MA, USA.,Hypertrophic Cardiomyopathy Center, Tufts Medical Center, Boston, MA, USA
| | | | - Barry J Maron
- CardioVascular Center, Tufts Medical Center, Boston, MA, USA.,Hypertrophic Cardiomyopathy Center, Tufts Medical Center, Boston, MA, USA
| | - Ethan J Rowin
- CardioVascular Center, Tufts Medical Center, Boston, MA, USA.,Hypertrophic Cardiomyopathy Center, Tufts Medical Center, Boston, MA, USA
| | - Martin S Maron
- CardioVascular Center, Tufts Medical Center, Boston, MA, USA.,Hypertrophic Cardiomyopathy Center, Tufts Medical Center, Boston, MA, USA
| | - Michael T Chin
- Molecular Cardiology Research Institute, Tufts Medical Center, 800 Washington Street, Box 80, Boston, MA, 02111, USA. .,CardioVascular Center, Tufts Medical Center, Boston, MA, USA. .,Hypertrophic Cardiomyopathy Center, Tufts Medical Center, Boston, MA, USA.
| |
Collapse
|
204
|
Sivakumar Harish T, Ramesh Babu P, Shrestha A, Balasubramanian B, Chinnathambi A, Ali Alharbi S. Development of a Model System to Study Expression Profile of RAC2 Gene in Breast Cancer MDA-MB-231 Cell Line. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2022; 2022:2077850. [PMID: 35368753 PMCID: PMC8970810 DOI: 10.1155/2022/2077850] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Accepted: 02/19/2022] [Indexed: 12/24/2022]
Abstract
The RAC2 gene encoding GTPases involve cellular signaling of actin polymerization, cell migration, and formation of the phagocytic NADPH oxidase complex. Oncogenic mutations in the RAC2 gene have been identified in various cancers, and extensive research is in progress to delineate its signaling pathways and identify potential therapeutic targets in breast cancers. This paper explored developing a bioinformatics model system to understand the RAC2 gene expression pattern concerning estrogenic receptor status in breast cancers. We have used the MDA-MB-231 breast cancer cell line to identify RAC2 gene expression. To simplify the development of model system with one dataset, we retrieved the microarray dataset GSE27515 from the Gene Expression Omnibus (GEO) for the differential gene expression analysis. Then, network analysis, pathway enrichment analysis, volcano plot, ORA, and the up/downregulated genes were used to highlight genes involved in signaling network pathways. We observed that the RAC2 gene is upregulated in the GSM679722, GSM676923, and GSM679724 downregulated in the samples GSM676925, GSM676926, and GSM676927 from the GEO dataset. Our observation found that the RAC2 gene is upregulated in the estrogen receptor (ER) negative breast cancers and downregulated in ER-positive breast cancer, involving pathways such as focal adhesion, MAPK signaling, axon guidance, and VEGF signaling pathway.
Collapse
Affiliation(s)
- Thogulva Sivakumar Harish
- Department of Genetic Engineering, Bharath Institute of Higher Education and Research, Selaiyur, Chennai-73, India
| | - Polani Ramesh Babu
- Center for Materials Engineering and Regenerative Medicine, Bharath Institute of Higher Education and Research, Selaiyur, Chennai-73, India
| | - Anupama Shrestha
- Department of Biotechnology, School of Science, Kathmandu University, P.O Box: 6250, Dhulikhel, Nepal
| | | | - Arunachalam Chinnathambi
- Department of Botany and Microbiology, College of Science, King Saud University, PO Box-2455, Riyadh 11451, Saudi Arabia
| | - Sulaiman Ali Alharbi
- Department of Botany and Microbiology, College of Science, King Saud University, PO Box-2455, Riyadh 11451, Saudi Arabia
| |
Collapse
|
205
|
Identification and characterisation of sPEPs in Cryptococcus neoformans. Fungal Genet Biol 2022; 160:103688. [PMID: 35339703 DOI: 10.1016/j.fgb.2022.103688] [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: 01/04/2022] [Revised: 03/02/2022] [Accepted: 03/21/2022] [Indexed: 11/24/2022]
Abstract
Short open reading frame (sORF)-encoded peptides (sPEPs) have been found across a wide range of genomic locations in a variety of species. To date, their identification, validation, and characterisation in the human fungal pathogen Cryptococcus neoformans has been limited due to a lack of standardised protocols. We have developed an enrichment process that enables sPEP detection within a protein sample from this polysaccharide-encapsulated yeast, and implemented proteogenomics to provide insights into the validity of predicted and hypothetical sORFs annotated in the C. neoformans genome. Novel sORFs were discovered within the 5' and 3' UTRs of known transcripts as well as in "non-coding" RNAs. One novel candidate, dubbed NPB1, that resided in an RNA annotated as "non-coding", was chosen for characterisation. Through the creation of both specific point mutations and a full deletion allele, the function of the new sPEP, Npb1, was shown to resemble that of the bacterial trans-translation protein SmpB.
Collapse
|
206
|
Podkalicka P, Mucha O, Kaziród K, Szade K, Stępniewski J, Ivanishchuk L, Hirao H, Pośpiech E, Józkowicz A, Kupiec-Weglinski JW, Dulak J, Łoboda A. miR-378 affects metabolic disturbances in the mdx model of Duchenne muscular dystrophy. Sci Rep 2022; 12:3945. [PMID: 35273230 PMCID: PMC8913680 DOI: 10.1038/s41598-022-07868-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Accepted: 02/22/2022] [Indexed: 02/08/2023] Open
Abstract
Although Duchenne muscular dystrophy (DMD) primarily affects muscle tissues, the alterations to systemic metabolism manifested in DMD patients contribute to the severe phenotype of this fatal disorder. We propose that microRNA-378a (miR-378) alters carbohydrate and lipid metabolism in dystrophic mdx mice. In our study, we utilized double knockout animals which lacked both dystrophin and miR-378 (mdx/miR-378-/-). RNA sequencing of the liver identified 561 and 194 differentially expressed genes that distinguished mdx versus wild-type (WT) and mdx/miR-378-/- versus mdx counterparts, respectively. Bioinformatics analysis predicted, among others, carbohydrate metabolism disorder in dystrophic mice, as functionally proven by impaired glucose tolerance and insulin sensitivity. The lack of miR-378 in mdx animals mitigated those effects with a faster glucose clearance in a glucose tolerance test (GTT) and normalization of liver glycogen levels. The absence of miR-378 also restored the expression of genes regulating lipid homeostasis, such as Acly, Fasn, Gpam, Pnpla3, and Scd1. In conclusion, we report for the first time that miR-378 loss results in increased systemic metabolism of mdx mice. Together with our previous finding, demonstrating alleviation of the muscle-related symptoms of DMD, we propose that the inhibition of miR-378 may represent a new strategy to attenuate the multifaceted symptoms of DMD.
Collapse
Affiliation(s)
- Paulina Podkalicka
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University in Krakow, 30-387, Kraków, Poland
| | - Olga Mucha
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University in Krakow, 30-387, Kraków, Poland
| | - Katarzyna Kaziród
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University in Krakow, 30-387, Kraków, Poland
| | - Krzysztof Szade
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University in Krakow, 30-387, Kraków, Poland
| | - Jacek Stępniewski
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University in Krakow, 30-387, Kraków, Poland
| | - Liudmyla Ivanishchuk
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University in Krakow, 30-387, Kraków, Poland
| | - Hirofumi Hirao
- The Dumont-UCLA Transplantation Center, Department of Surgery, Division of Liver and Pancreas Transplantation, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA
| | - Ewelina Pośpiech
- Malopolska Centre of Biotechnology, Jagiellonian University in Krakow, 30-387, Kraków, Poland
| | - Alicja Józkowicz
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University in Krakow, 30-387, Kraków, Poland
| | - Jerzy W Kupiec-Weglinski
- The Dumont-UCLA Transplantation Center, Department of Surgery, Division of Liver and Pancreas Transplantation, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA
| | - Józef Dulak
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University in Krakow, 30-387, Kraków, Poland
| | - Agnieszka Łoboda
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University in Krakow, 30-387, Kraków, Poland.
| |
Collapse
|
207
|
Schoen MS, Singh RH. Plasma metabolomic profile changes in females with phenylketonuria following a camp intervention. Am J Clin Nutr 2022; 115:811-821. [PMID: 34864852 PMCID: PMC8895208 DOI: 10.1093/ajcn/nqab400] [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: 04/17/2021] [Accepted: 11/29/2021] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND There remains a limited understanding of the metabolic perturbations, beyond phenylalanine (Phe) metabolism, that contribute to phenotypic variability in phenylketonuria (PKU). OBJECTIVES This study aimed to characterize changes in the PKU plasma metabolome following a 5-d metabolic camp intervention and to compare PKU profiles with those of matched healthy controls. METHODS In 28 females (aged 12-57 y), fasting plasma samples were collected on the first (day 1) and final (day 5) days of camp to measure metabolic control and to complete untargeted metabolomic profiling. Three-day dietary records were collected to assess changes in dietary adherence and composition. Univariate (Wilcoxon signed-rank and Mann-Whitney U test) and multivariate (random forest, hierarchical clustering) analyses were performed to identify clinical and metabolic features that were associated with the intervention and disease state. RESULTS Relative to healthy controls, Phe catabolites, ketones, and carnitine- and glycine-conjugated fatty acids were elevated in females with PKU at baseline, whereas fatty acylcholine metabolites were substantially lower. After the camp intervention, plasma Phe concentrations decreased [median change: -173 µmol/L (IQR: -325, -28 µmol/L)] and 70% of PKU participants demonstrated improved dietary adherence by decreasing Phe intake and/or increasing medical food consumption. This was accompanied by a shift in abundance for 223 metabolites (q < 0.05). Compounds associated with the metabolism of Phe, fatty acids, and choline contributed most to profile differences between camp days 1 and 5. CONCLUSIONS In females with PKU, untargeted metabolomics identified prominent perturbations in amino acid and lipid metabolites associated with bioenergetic impairment and oxidative stress. Choline-conjugated lipids could have fundamental roles in these pathways and they have not been previously evaluated in PKU. A short-term camp intervention was effective for improving or fully normalizing the abundance of the identified discriminatory metabolites.
Collapse
Affiliation(s)
- Meriah S Schoen
- Nutrition and Health Sciences, Laney Graduate School, Emory University, Atlanta, GA, USA
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, USA
| | - Rani H Singh
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, USA
| |
Collapse
|
208
|
Qian S, Liu H, Yuan X, Wei W, Chen S, Yan H. Row and Column Structure-Based Biclustering for Gene Expression Data. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2022; 19:1117-1129. [PMID: 32894722 DOI: 10.1109/tcbb.2020.3022085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Due to the development of high-throughput technologies for gene analysis, the biclustering method has attracted much attention. However, existing methods have problems with high time and space complexity. This paper proposes a biclustering method, called Row and Column Structure-based Biclustering (RCSBC), with low time and space complexity to find checkerboard patterns within microarray data. First, the paper describes the structure of bicluster by using the structure of rows and columns. Second, the paper chooses the representative rows and columns with two algorithms. Finally, the gene expression data are biclustered on the space spanned by representative rows and columns. To the best of our knowledge, this paper is the first to exploit the relationship between the row/column structure of a gene expression matrix and the structure of biclusters. Both the synthetic datasets and the real-life gene expression datasets are used to validate the effectiveness of our method. It can be seen from the experiment results that the RCSBC outperforms the state-of-the-art algorithms both on clustering accuracy and time/space complexity. This study offers new insights into biclustering the large-scale gene expression data without loading the whole data into memory.
Collapse
|
209
|
Chen HM, MacDonald JA. Network analysis of TCGA and GTEx gene expression datasets for identification of trait-associated biomarkers in human cancer. STAR Protoc 2022; 3:101168. [PMID: 35199033 PMCID: PMC8841814 DOI: 10.1016/j.xpro.2022.101168] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Advances in high-throughput sequencing technologies now yield unprecedented volumes of OMICs data with opportunities to conduct systematic data analyses and derive novel biological insights. Here, we provide protocols to perform differential-expressed gene analysis of TCGA and GTEx RNA-Seq data from human cancers, complete integrative GO and network analyses with focus on clinical and survival data, and identify differential correlation of trait-associated biomarkers. For complete details on the use and execution of this protocol, please refer to Chen and MacDonald (2021). Protocols for the identification of trait-associated molecular correlates in cancer Differentially-expressed gene (DEG) analysis of TCGA and GTEx transcriptomic data Protocols for integrative network analysis of RNA-seq, clinical, and survival data Differential correlation of trait-associated biomarkers for hypothesis testing
Collapse
|
210
|
Lemay MA, Sibbesen JA, Torkamaneh D, Hamel J, Levesque RC, Belzile F. Combined use of Oxford Nanopore and Illumina sequencing yields insights into soybean structural variation biology. BMC Biol 2022; 20:53. [PMID: 35197050 PMCID: PMC8867729 DOI: 10.1186/s12915-022-01255-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 02/16/2022] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Structural variants (SVs), including deletions, insertions, duplications, and inversions, are relatively long genomic variations implicated in a diverse range of processes from human disease to ecology and evolution. Given their complex signatures, tendency to occur in repeated regions, and large size, discovering SVs based on short reads is challenging compared to single-nucleotide variants. The increasing availability of long-read technologies has greatly facilitated SV discovery; however, these technologies remain too costly to apply routinely to population-level studies. Here, we combined short-read and long-read sequencing technologies to provide a comprehensive population-scale assessment of structural variation in a panel of Canadian soybean cultivars. RESULTS We used Oxford Nanopore long-read sequencing data (~12× mean coverage) for 17 samples to both benchmark SV calls made from Illumina short-read data and predict SVs that were subsequently genotyped in a population of 102 samples using Illumina data. Benchmarking results show that variants discovered using Oxford Nanopore can be accurately genotyped from the Illumina data. We first use the genotyped deletions and insertions for population genetics analyses and show that results are comparable to those based on single-nucleotide variants. We observe that the population frequency and distribution within the genome of deletions and insertions are constrained by the location of genes. Gene Ontology and PFAM domain enrichment analyses also confirm previous reports that genes harboring high-frequency deletions and insertions are enriched for functions in defense response. Finally, we discover polymorphic transposable elements from the deletions and insertions and report evidence of the recent activity of a Stowaway MITE. CONCLUSIONS We show that structural variants discovered using Oxford Nanopore data can be genotyped with high accuracy from Illumina data. Our results demonstrate that long-read and short-read sequencing technologies can be efficiently combined to enhance SV analysis in large populations, providing a reusable framework for their study in a wider range of samples and non-model species.
Collapse
Affiliation(s)
- Marc-André Lemay
- Département de phytologie, Université Laval, Quebec, Canada
- Institut de biologie intégrative et des systèmes, Université Laval, Quebec, Canada
| | | | - Davoud Torkamaneh
- Département de phytologie, Université Laval, Quebec, Canada
- Institut de biologie intégrative et des systèmes, Université Laval, Quebec, Canada
| | - Jérémie Hamel
- Institut de biologie intégrative et des systèmes, Université Laval, Quebec, Canada
- Département de microbiologie-infectiologie et d’immunologie, Université Laval, Quebec, Canada
| | - Roger C. Levesque
- Institut de biologie intégrative et des systèmes, Université Laval, Quebec, Canada
- Département de microbiologie-infectiologie et d’immunologie, Université Laval, Quebec, Canada
| | - François Belzile
- Département de phytologie, Université Laval, Quebec, Canada
- Institut de biologie intégrative et des systèmes, Université Laval, Quebec, Canada
| |
Collapse
|
211
|
Feng Y, Zhao Y, Li Y, Zhou J, Li Y, Shi H. Physiological and transcriptome analysis reveals the differences in nitrate content between lamina and midrib of flue-cured tobacco. Sci Rep 2022; 12:2932. [PMID: 35190651 PMCID: PMC8861034 DOI: 10.1038/s41598-022-07011-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2021] [Accepted: 02/07/2022] [Indexed: 11/09/2022] Open
Abstract
Nitrate is an important precursor of tobacco-specific nitrosamines (TSNAs) and a remarkable difference in nitrate accumulation between lamina and midrib of flue-cured tobacco has long been observed. However, the physiological and molecular mechanisms underpinning this difference remain poorly understood. In this study, physiological and genetic factors impacting nitrate accumulation were identified in pot experiments using flue-cured tobacco K326 with contrasting nitrate content between lamina and midrib. The results showed that three times higher NO3-N content was observed in midrib than that in the lamina, along with lower pigment, NH4-N content, nitrate reductase activity (NRA), sucrose synthetase activity (SSA), and glutamine synthetase activity (GSA) in midrib. Transcriptome analysis revealed that expression of genes involved in porphyrin and chlorophyll metabolism, carotenoid biosynthesis, photosynthesis-antenna proteins, photosynthesis, carbon fixation in photosynthetic organisms, starch and sucrose metabolism, nitrogen metabolism, and biosynthesis of amino acids were significantly lower in midrib than in lamina. qRT-PCR results showed that the expression level of nitrate transporter genes LOC107782967, LOC107806749, LOC107775674, LOC107829632, LOC107799198, LOC107768465 decreased by 2.74, 1.81, 49.5, 3.5, 2.64 and 2.96-folds while LOC107789301 increased by 8.23-folds in midrib but not in lamina. Reduced chlorophyll content might result in low carbohydrate formation which is the source of energy and carbon skeleton supply, then the low capacity of nitrogen reduction, assimilation and transportation, and the poor ability of nitrate reallocation but the high capacity of accumulation might lead to nitrate accumulation in midrib. The results laid the foundation for reducing nitrate content and TSNA formation in tobacco midribs and their products.
Collapse
Affiliation(s)
- Yuqing Feng
- National Tobacco Cultivation & Physiology & Biochemistry Research Center, Tobacco Harm Reduction Research Center of China Tobacco, Henan Agricultural University, Zhengzhou, 450002, China
| | - Yuanyuan Zhao
- National Tobacco Cultivation & Physiology & Biochemistry Research Center, Tobacco Harm Reduction Research Center of China Tobacco, Henan Agricultural University, Zhengzhou, 450002, China
| | - Yafei Li
- National Tobacco Cultivation & Physiology & Biochemistry Research Center, Tobacco Harm Reduction Research Center of China Tobacco, Henan Agricultural University, Zhengzhou, 450002, China
| | - Jun Zhou
- Beijing Cigarette Factory, Shanghai Tobacco Group Co., Ltd., Beijing, 100024, China
| | - Yujing Li
- National Tobacco Cultivation & Physiology & Biochemistry Research Center, Tobacco Harm Reduction Research Center of China Tobacco, Henan Agricultural University, Zhengzhou, 450002, China
| | - Hongzhi Shi
- National Tobacco Cultivation & Physiology & Biochemistry Research Center, Tobacco Harm Reduction Research Center of China Tobacco, Henan Agricultural University, Zhengzhou, 450002, China.
| |
Collapse
|
212
|
The Nephrotoxin Puromycin Aminonucleoside Induces Injury in Kidney Organoids Differentiated from Induced Pluripotent Stem Cells. Cells 2022; 11:cells11040635. [PMID: 35203286 PMCID: PMC8870209 DOI: 10.3390/cells11040635] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 01/29/2022] [Accepted: 02/09/2022] [Indexed: 11/17/2022] Open
Abstract
Kidney diseases, including acute kidney injury (AKI) and chronic kidney disease (CKD), which can progress to end stage renal disease (ESRD), are a worldwide health burden. Organ transplantation or kidney dialysis are the only effective available therapeutic tools. Therefore, in vitro models of kidney diseases and the development of prospective therapeutic options are urgently needed. Within the kidney, the glomeruli are involved in blood filtration and waste excretion and are easily affected by changing cellular conditions. Puromycin aminonucleoside (PAN) is a nephrotoxin, which can be employed to induce acute glomerular damage and to model glomerular disease. For this reason, we generated kidney organoids from three iPSC lines and treated these with PAN in order to induce kidney injury. Morphological observations revealed the disruption of glomerular and tubular structures within the kidney organoids upon PAN treatment, which were confirmed by transcriptome analyses. Subsequent analyses revealed an upregulation of immune response as well as inflammatory and cell-death-related processes. We conclude that the treatment of iPSC-derived kidney organoids with PAN induces kidney injury mediated by an intertwined network of inflammation, cytoskeletal re-arrangement, DNA damage, apoptosis and cell death. Furthermore, urine-stem-cell-derived kidney organoids can be used to model kidney-associated diseases and drug discovery.
Collapse
|
213
|
Merz KE, Tunduguru R, Ahn M, Salunkhe VA, Veluthakal R, Hwang J, Bhattacharya S, McCown EM, Garcia PA, Zhou C, Oh E, Yoder SM, Elmendorf JS, Thurmond DC. Changes in Skeletal Muscle PAK1 Levels Regulate Tissue Crosstalk to Impact Whole Body Glucose Homeostasis. Front Endocrinol (Lausanne) 2022; 13:821849. [PMID: 35222279 PMCID: PMC8881144 DOI: 10.3389/fendo.2022.821849] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Accepted: 01/13/2022] [Indexed: 12/13/2022] Open
Abstract
Skeletal muscle accounts for ~80% of insulin-stimulated glucose uptake. The Group I p21-activated kinase 1 (PAK1) is required for the non-canonical insulin-stimulated GLUT4 vesicle translocation in skeletal muscle cells. We found that the abundances of PAK1 protein and its downstream effector in muscle, ARPC1B, are significantly reduced in the skeletal muscle of humans with type 2 diabetes, compared to the non-diabetic controls, making skeletal muscle PAK1 a candidate regulator of glucose homeostasis. Although whole-body PAK1 knockout mice exhibit glucose intolerance and are insulin resistant, the contribution of skeletal muscle PAK1 in particular was unknown. As such, we developed inducible skeletal muscle-specific PAK1 knockout (skmPAK1-iKO) and overexpression (skmPAK1-iOE) mouse models to evaluate the role of PAK1 in skeletal muscle insulin sensitivity and glucose homeostasis. Using intraperitoneal glucose tolerance and insulin tolerance testing, we found that skeletal muscle PAK1 is required for maintaining whole body glucose homeostasis. Moreover, PAK1 enrichment in GLUT4-myc-L6 myoblasts preserves normal insulin-stimulated GLUT4 translocation under insulin resistance conditions. Unexpectedly, skmPAK1-iKO also showed aberrant plasma insulin levels following a glucose challenge. By applying conditioned media from PAK1-enriched myotubes or myoblasts to β-cells in culture, we established that a muscle-derived circulating factor(s) could enhance β-cell function. Taken together, these data suggest that PAK1 levels in the skeletal muscle can regulate not only skeletal muscle insulin sensitivity, but can also engage in tissue crosstalk with pancreatic β-cells, unveiling a new molecular mechanism by which PAK1 regulates whole-body glucose homeostasis.
Collapse
Affiliation(s)
- Karla E. Merz
- Department of Molecular & Cellular Endocrinology, Arthur Riggs Diabetes and Metabolism Research Institute of City of Hope, Duarte, CA, United States
| | - Ragadeepthi Tunduguru
- Department of Diabetes Complications and Metabolism, Arthur Riggs Diabetes and Metabolism Research Institute of City of Hope, Duarte, CA, United States
| | - Miwon Ahn
- Department of Molecular & Cellular Endocrinology, Arthur Riggs Diabetes and Metabolism Research Institute of City of Hope, Duarte, CA, United States
| | - Vishal A. Salunkhe
- Sahlgrenska Academy, Institute of Neuroscience and Physiology, Metabolism Research Unit, University of Gothenburg, Gothenburg, Sweden
| | - Rajakrishnan Veluthakal
- Department of Molecular & Cellular Endocrinology, Arthur Riggs Diabetes and Metabolism Research Institute of City of Hope, Duarte, CA, United States
| | - Jinhee Hwang
- Department of Molecular & Cellular Endocrinology, Arthur Riggs Diabetes and Metabolism Research Institute of City of Hope, Duarte, CA, United States
| | - Supriyo Bhattacharya
- Division of Translational Bioinformatics, City of Hope, Duarte, CA, United States
| | - Erika M. McCown
- Department of Molecular & Cellular Endocrinology, Arthur Riggs Diabetes and Metabolism Research Institute of City of Hope, Duarte, CA, United States
| | - Pablo A. Garcia
- Department of Molecular & Cellular Endocrinology, Arthur Riggs Diabetes and Metabolism Research Institute of City of Hope, Duarte, CA, United States
| | - Chunxue Zhou
- Department of Molecular & Cellular Endocrinology, Arthur Riggs Diabetes and Metabolism Research Institute of City of Hope, Duarte, CA, United States
| | - Eunjin Oh
- Department of Molecular & Cellular Endocrinology, Arthur Riggs Diabetes and Metabolism Research Institute of City of Hope, Duarte, CA, United States
| | - Stephanie M. Yoder
- Global Scientific Communications, Eli Lilly & Company, Indianapolis, IN, United States
| | - Jeffrey S. Elmendorf
- Department of Anatomy, Cell Biology and Physiology, Center for Diabetes and Metabolic Disease, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Debbie C. Thurmond
- Department of Molecular & Cellular Endocrinology, Arthur Riggs Diabetes and Metabolism Research Institute of City of Hope, Duarte, CA, United States
| |
Collapse
|
214
|
Iwanicki NS, Botelho ABRZ, Klingen I, Júnior ID, Rossmann S, Lysøe E. Genomic signatures and insights into host niche adaptation of the entomopathogenic fungus Metarhizium humberi. G3 (BETHESDA, MD.) 2022; 12:6449448. [PMID: 34865006 PMCID: PMC9210286 DOI: 10.1093/g3journal/jkab416] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 11/22/2021] [Indexed: 12/30/2022]
Abstract
The genus Metarhizium is composed of species used in biological control programs of agricultural pests worldwide. This genus includes common fungal pathogen of many insects and mites and endophytes that can increase plant growth. Metarhizium humberi was recently described as a new species. This species is highly virulent against some insect pests and promotes growth in sugarcane, strawberry, and soybean crops. In this study, we sequenced the genome of M. humberi, isolate ESALQ1638, and performed a functional analysis to determine its genomic signatures and highlight the genes and biological processes associated with its lifestyle. The genome annotation predicted 10633 genes in M. humberi, of which 92.0% are assigned putative functions, and ∼17% of the genome was annotated as repetitive sequences. We found that 18.5% of the M. humberi genome is similar to experimentally validated proteins associated with pathogen-host interaction. Compared to the genomes of eight Metarhizium species, the M. humberi ESALQ1638 genome revealed some unique traits that stood out, e.g., more genes functionally annotated as polyketide synthases (PKSs), overrepresended GO-terms associated to transport of ions, organic and amino acid, a higher percentage of repetitive elements, and higher levels of RIP-induced point mutations. The M. humberi genome will serve as a resource for promoting studies on genome structure and evolution that can contribute to research on biological control and plant biostimulation. Thus, the genomic data supported the broad host range of this species within the generalist PARB clade and suggested that M. humberi ESALQ1638 might be particularly good at producing secondary metabolites and might be more efficient in transporting amino acids and organic compounds.
Collapse
Affiliation(s)
- Natasha Sant′Anna Iwanicki
- Department of Entomology and Acarology, “Luiz de Queiroz” College of Agriculture (ESALQ/USP), Piracicaba 13418-900, Brazil
- Corresponding author: (N.S.I.); (E.L.)
| | | | - Ingeborg Klingen
- Division of Biotechnology and Plant Health, Norwegian Institute of Bioeconomy Research (NIBIO), Ås 1431, Norway
| | - Italo Delalibera Júnior
- Department of Entomology and Acarology, “Luiz de Queiroz” College of Agriculture (ESALQ/USP), Piracicaba 13418-900, Brazil
| | - Simeon Rossmann
- Division of Biotechnology and Plant Health, Norwegian Institute of Bioeconomy Research (NIBIO), Ås 1431, Norway
| | - Erik Lysøe
- Division of Biotechnology and Plant Health, Norwegian Institute of Bioeconomy Research (NIBIO), Ås 1431, Norway
- Corresponding author: (N.S.I.); (E.L.)
| |
Collapse
|
215
|
Zhu W, Yang C, Chen X, Liu Q, Li Q, Peng M, Wang H, Chen X, Yang Q, Liao Z, Li M, Pan C, Feng P, Zeng D, Zhao Y. Single-Cell Ribonucleic Acid Sequencing Clarifies Cold Tolerance Mechanisms in the Pacific White Shrimp ( Litopenaeus Vannamei). Front Genet 2022; 12:792172. [PMID: 35096009 PMCID: PMC8790290 DOI: 10.3389/fgene.2021.792172] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Accepted: 11/25/2021] [Indexed: 12/12/2022] Open
Abstract
To characterize the cold tolerance mechanism of the Pacific white shrimp (Litopenaeus vannamei), we performed single-cell RNA sequencing (scRNA-seq) of ∼5185 hepatopancreas cells from cold-tolerant (Lv-T) and common (Lv-C) L. vannamei at preferred and low temperatures (28°C and 10°C, respectively). The cells fell into 10 clusters and 4 cell types: embryonic, resorptive, blister-like, and fibrillar. We identified differentially expressed genes between Lv-T and Lv-C, which were mainly associated with the terms “immune system,” “cytoskeleton,” “antioxidant system,” “digestive enzyme,” and “detoxification,” as well as the pathways “metabolic pathways of oxidative phosphorylation,” “metabolism of xenobiotics by cytochrome P450,” “chemical carcinogenesis,” “drug metabolism-cytochrome P450,” and “fatty acid metabolism.” Reconstruction of fibrillar cell trajectories showed that, under low temperature stress, hepatopancreas cells had two distinct fates, cell fate 1 and cell fate 2. Cell fate 1 was mainly involved in signal transduction and sensory organ development. Cell fate 2 was mainly involved in metabolic processes. This study preliminarily clarifies the molecular mechanisms underlying cold tolerance in L. vannamei, which will be useful for the breeding of shrimp with greater cold tolerance.
Collapse
Affiliation(s)
- Weilin Zhu
- Guangxi Key Laboratory of Aquatic Genetic Breeding and Healthy Aquaculture, Guangxi Academy of Fishery Sciences, Nanning, China
| | - Chunling Yang
- Guangxi Key Laboratory of Aquatic Genetic Breeding and Healthy Aquaculture, Guangxi Academy of Fishery Sciences, Nanning, China
| | - Xiuli Chen
- Guangxi Key Laboratory of Aquatic Genetic Breeding and Healthy Aquaculture, Guangxi Academy of Fishery Sciences, Nanning, China
| | - Qingyun Liu
- Guangxi Key Laboratory of Aquatic Genetic Breeding and Healthy Aquaculture, Guangxi Academy of Fishery Sciences, Nanning, China.,Guangxi Shrimp and Crab Breeding Engineering Technology Research Center, Guangxi Academy of Fishery Sciences, Nanning, China
| | - Qiangyong Li
- Guangxi Key Laboratory of Aquatic Genetic Breeding and Healthy Aquaculture, Guangxi Academy of Fishery Sciences, Nanning, China.,Guangxi Shrimp and Crab Breeding Engineering Technology Research Center, Guangxi Academy of Fishery Sciences, Nanning, China
| | - Min Peng
- Guangxi Key Laboratory of Aquatic Genetic Breeding and Healthy Aquaculture, Guangxi Academy of Fishery Sciences, Nanning, China
| | - Huanling Wang
- Key Lab of Freshwater Animal Breeding, Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Ministry of Education, College of Fishery, Huazhong Agriculture University, Wuhan, China
| | - Xiaohan Chen
- Guangxi Key Laboratory of Aquatic Genetic Breeding and Healthy Aquaculture, Guangxi Academy of Fishery Sciences, Nanning, China
| | - Qiong Yang
- Guangxi Key Laboratory of Aquatic Genetic Breeding and Healthy Aquaculture, Guangxi Academy of Fishery Sciences, Nanning, China
| | - Zhenping Liao
- Guangxi Key Laboratory of Aquatic Genetic Breeding and Healthy Aquaculture, Guangxi Academy of Fishery Sciences, Nanning, China
| | - Min Li
- Guangxi Key Laboratory of Aquatic Genetic Breeding and Healthy Aquaculture, Guangxi Academy of Fishery Sciences, Nanning, China
| | - Chuanyan Pan
- Guangxi Key Laboratory of Aquatic Genetic Breeding and Healthy Aquaculture, Guangxi Academy of Fishery Sciences, Nanning, China
| | - Pengfei Feng
- Guangxi Key Laboratory of Aquatic Genetic Breeding and Healthy Aquaculture, Guangxi Academy of Fishery Sciences, Nanning, China
| | - Digang Zeng
- Guangxi Key Laboratory of Aquatic Genetic Breeding and Healthy Aquaculture, Guangxi Academy of Fishery Sciences, Nanning, China
| | - Yongzhen Zhao
- Guangxi Key Laboratory of Aquatic Genetic Breeding and Healthy Aquaculture, Guangxi Academy of Fishery Sciences, Nanning, China.,Guangxi Shrimp and Crab Breeding Engineering Technology Research Center, Guangxi Academy of Fishery Sciences, Nanning, China
| |
Collapse
|
216
|
Riederer I, Mendes-da-Cruz DA, da Fonseca GC, González MN, Brustolini O, Rocha C, Loss G, de Carvalho JB, Menezes MT, Raphael LMS, Gerber A, Bonaldo MC, Butler-Browne G, Mouly V, Cotta-de-Almeida V, Savino W, Ribeiro de Vasconcelos AT. Zika virus disrupts gene expression in human myoblasts and myotubes: Relationship with susceptibility to infection. PLoS Negl Trop Dis 2022; 16:e0010166. [PMID: 35171909 PMCID: PMC8923442 DOI: 10.1371/journal.pntd.0010166] [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: 06/25/2021] [Revised: 03/15/2022] [Accepted: 01/12/2022] [Indexed: 11/30/2022] Open
Abstract
The tropism of Zika virus (ZIKV) has been described in the nervous system, blood, placenta, thymus, and skeletal muscle. We investigated the mechanisms of skeletal muscle susceptibility to ZIKV using an in vitro model of human skeletal muscle myogenesis, in which myoblasts differentiate into myotubes. Myoblasts were permissive to ZIKV infection, generating productive viral particles, while myotubes controlled ZIKV replication. To investigate the underlying mechanisms, we used gene expression profiling. First, we assessed gene changes in myotubes compared with myoblasts in the model without infection. As expected, we observed an increase in genes and pathways related to the contractile muscle system in the myotubes, a reduction in processes linked to proliferation, migration and cytokine production, among others, confirming the myogenic capacity of our system in vitro. A comparison between non-infected and infected myoblasts revealed more than 500 differentially expressed genes (DEGs). In contrast, infected myotubes showed almost 2,000 DEGs, among which we detected genes and pathways highly or exclusively expressed in myotubes, including those related to antiviral and innate immune responses. Such gene modulation could explain our findings showing that ZIKV also invades myotubes but does not replicate in these differentiated cells. In conclusion, we showed that ZIKV largely (but differentially) disrupts gene expression in human myoblasts and myotubes. Identifying genes involved in myotube resistance can shed light on potential antiviral mechanisms against ZIKV infection.
Collapse
Affiliation(s)
- Ingo Riederer
- Laboratory on Thymus Research, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
- National Institute of Science and Technology on Neuroimmunomodulation (INCT-NIM); Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
- Rio de Janeiro Research Network on Neuroinflammation (RENEURIN), Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
- School of Pharmacy and Biomedical Sciences, University of Central Lancashire, Preston, England, United Kingdom
| | - Daniella Arêas Mendes-da-Cruz
- Laboratory on Thymus Research, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
- National Institute of Science and Technology on Neuroimmunomodulation (INCT-NIM); Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
- Rio de Janeiro Research Network on Neuroinflammation (RENEURIN), Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
- School of Pharmacy and Biomedical Sciences, University of Central Lancashire, Preston, England, United Kingdom
| | | | - Mariela Natacha González
- Laboratory on Thymus Research, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
- National Institute of Science and Technology on Neuroimmunomodulation (INCT-NIM); Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Otavio Brustolini
- Bioinformatics Laboratory, National Laboratory for Scientific Computing, Petropolis, Rio de Janeiro, Brazil
| | - Cássia Rocha
- Laboratory on Thymus Research, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
- National Institute of Science and Technology on Neuroimmunomodulation (INCT-NIM); Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Guilherme Loss
- Bioinformatics Laboratory, National Laboratory for Scientific Computing, Petropolis, Rio de Janeiro, Brazil
| | - Joseane Biso de Carvalho
- Bioinformatics Laboratory, National Laboratory for Scientific Computing, Petropolis, Rio de Janeiro, Brazil
| | - Mariane Talon Menezes
- Department of Genetics, Institute of Biology, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Lidiane Menezes Souza Raphael
- Laboratory of Molecular Biology of Flavivirus, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Alexandra Gerber
- Bioinformatics Laboratory, National Laboratory for Scientific Computing, Petropolis, Rio de Janeiro, Brazil
| | - Myrna Cristina Bonaldo
- Laboratory of Molecular Biology of Flavivirus, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Gillian Butler-Browne
- Sorbonne Université, Inserm, Institut de Myologie, Centre de Recherche en Myologie, Paris, France
| | - Vincent Mouly
- Sorbonne Université, Inserm, Institut de Myologie, Centre de Recherche en Myologie, Paris, France
| | - Vinicius Cotta-de-Almeida
- Laboratory on Thymus Research, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
- National Institute of Science and Technology on Neuroimmunomodulation (INCT-NIM); Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
- Rio de Janeiro Research Network on Neuroinflammation (RENEURIN), Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Wilson Savino
- Laboratory on Thymus Research, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
- National Institute of Science and Technology on Neuroimmunomodulation (INCT-NIM); Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
- Rio de Janeiro Research Network on Neuroinflammation (RENEURIN), Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | | |
Collapse
|
217
|
Spatiotemporal analysis identifies ABF2 and ABF3 as key hubs of endodermal response to nitrate. Proc Natl Acad Sci U S A 2022; 119:2107879119. [PMID: 35046022 PMCID: PMC8794810 DOI: 10.1073/pnas.2107879119] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/14/2021] [Indexed: 12/24/2022] Open
Abstract
Nitrate is a nutrient and a potent signal that impacts global gene expression in plants. However, the regulatory factors controlling temporal and cell type-specific nitrate responses remain largely unknown. We assayed nitrate-responsive transcriptome changes in five major root cell types of the Arabidopsis thaliana root as a function of time. We found that gene-expression response to nitrate is dynamic and highly localized and predicted cell type-specific transcription factor (TF)-target interactions. Among cell types, the endodermis stands out as having the largest and most connected nitrate-regulatory gene network. ABF2 and ABF3 are major hubs for transcriptional responses in the endodermis cell layer. We experimentally validated TF-target interactions for ABF2 and ABF3 by chromatin immunoprecipitation followed by sequencing and a cell-based system to detect TF regulation genome-wide. Validated targets of ABF2 and ABF3 account for more than 50% of the nitrate-responsive transcriptome in the endodermis. Moreover, ABF2 and ABF3 are involved in nitrate-induced lateral root growth. Our approach offers an unprecedented spatiotemporal resolution of the root response to nitrate and identifies important components of cell-specific gene regulatory networks.
Collapse
|
218
|
Tian Z, Fang H, Ye Y, Zhu Z. A novel gene functional similarity calculation model by utilizing the specificity of terms and relationships in gene ontology. BMC Bioinformatics 2022; 23:47. [PMID: 35057740 PMCID: PMC8772239 DOI: 10.1186/s12859-022-04557-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 01/03/2022] [Indexed: 11/18/2022] Open
Abstract
Background Recently, with the foundation and development of gene ontology (GO) resources, numerous works have been proposed to compute functional similarity of genes and achieved series of successes in some research fields. Focusing on the calculation of the information content (IC) of terms is the main idea of these methods, which is essential for measuring functional similarity of genes. However, most approaches have some deficiencies, especially when measuring the IC of both GO terms and their corresponding annotated term sets. To this end, measuring functional similarity of genes accurately is still challenging. Results In this article, we proposed a novel gene functional similarity calculation method, which especially encapsulates the specificity of terms and edges (STE). The proposed method mainly contains three steps. Firstly, a novel computing model is put forward to compute the IC of terms. This model has the ability to exploit the specific structural information of GO terms. Secondly, the IC of term sets are computed by capturing the genetic structure between the terms contained in the set. Lastly, we measure the gene functional similarity according to the IC overlap ratio of the corresponding annotated genes sets. The proposed method accurately measures the IC of not only GO terms but also the annotated term sets by leveraging the specificity of edges in the GO graph. Conclusions We conduct experiments on gene functional classification in biological pathways, gene expression datasets, and protein-protein interaction datasets. Extensive experimental results show the better performances of our proposed STE against several baseline methods.
Collapse
|
219
|
Iouranova A, Grun D, Rossy T, Duc J, Coudray A, Imbeault M, de Tribolet-Hardy J, Turelli P, Persat A, Trono D. KRAB zinc finger protein ZNF676 controls the transcriptional influence of LTR12-related endogenous retrovirus sequences. Mob DNA 2022; 13:4. [PMID: 35042549 PMCID: PMC8767690 DOI: 10.1186/s13100-021-00260-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Accepted: 12/23/2021] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Transposable element-embedded regulatory sequences (TEeRS) and their KRAB-containing zinc finger protein (KZFP) controllers are increasingly recognized as modulators of gene expression. We aim to characterize the contribution of this system to gene regulation in early human development and germ cells. RESULTS Here, after studying genes driven by the long terminal repeat (LTR) of endogenous retroviruses, we identify the ape-restricted ZNF676 as the sequence-specific repressor of a subset of contemporary LTR12 integrants responsible for a large fraction of transpochimeric gene transcripts (TcGTs) generated during human early embryogenesis. We go on to reveal that the binding of this KZFP correlates with the epigenetic marking of these TEeRS in the germline, and is crucial to the control of genes involved in ciliogenesis/flagellogenesis, a biological process that dates back to the last common ancestor of eukaryotes. CONCLUSION These results illustrate how KZFPs and their TE targets contribute to the evolutionary turnover of transcription networks and participate in the transgenerational inheritance of epigenetic traits.
Collapse
Affiliation(s)
| | - Delphine Grun
- School of Life Sciences, EPFL, Lausanne, Switzerland
| | - Tamara Rossy
- School of Life Sciences, EPFL, Lausanne, Switzerland
| | - Julien Duc
- School of Life Sciences, EPFL, Lausanne, Switzerland
| | | | - Michael Imbeault
- School of Life Sciences, EPFL, Lausanne, Switzerland
- Department of Genetics, University of Cambridge, Cambridge, UK
| | | | | | | | - Didier Trono
- School of Life Sciences, EPFL, Lausanne, Switzerland.
| |
Collapse
|
220
|
Hwang T, Kim S, Chowdhury T, Yu HJ, Kim KM, Kang H, Won JK, Park SH, Shin JH, Park CK. Genome-wide perturbations of Alu expression and Alu-associated post-transcriptional regulations distinguish oligodendroglioma from other gliomas. Commun Biol 2022; 5:62. [PMID: 35042936 PMCID: PMC8766575 DOI: 10.1038/s42003-022-03011-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Accepted: 12/27/2021] [Indexed: 01/09/2023] Open
Abstract
AbstractAlu is a primate-specific repeat element in the human genome and has been increasingly appreciated as a regulatory element in many biological processes. But the appreciation of Alu has been limited in tumorigenesis, especially for brain tumor. To investigate the relevance of Alu to the gliomagenesis, we studied Alu element-associated post-transcriptional processes and the RNA expression of the element by performing RNA-seq for a total of 41 pairs of neurotypical and diverse glioma brain tissues. We find that A-to-I editing and circular RNA levels, as well as Alu RNA expression, are decreased overall in gliomas, compared to normal tissue. Interestingly, grade 2 oligodendrogliomas are least affected in A-to-I editing and circular RNA levels among gliomas, whereas they have a higher proportion of down-regulated Alu subfamilies, compared to the other gliomas. These findings collectively imply a unique pattern of Alu-associated transcriptomes in grade 2 oligodendroglioma, providing an insight to gliomagenesis from the perspective of an evolutionary genetic element.
Collapse
|
221
|
Codden CJ, Chin MT. Common and Distinctive Intercellular Communication Patterns in Human Obstructive and Nonobstructive Hypertrophic Cardiomyopathy. Int J Mol Sci 2022; 23:946. [PMID: 35055131 PMCID: PMC8780670 DOI: 10.3390/ijms23020946] [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: 12/29/2021] [Revised: 01/12/2022] [Accepted: 01/13/2022] [Indexed: 01/27/2023] Open
Abstract
Hypertrophic Cardiomyopathy (HCM) is a common inherited disorder characterized by unexplained left ventricular hypertrophy with or without left ventricular outflow tract (LVOT) obstruction. Single-nuclei RNA-sequencing (snRNA-seq) of both obstructive and nonobstructive HCM patient samples has revealed alterations in communication between various cell types, but no direct and integrated comparison between the two HCM phenotypes has been reported. We performed a bioinformatic analysis of HCM snRNA-seq datasets from obstructive and nonobstructive patient samples to identify differentially expressed genes and distinctive patterns of intercellular communication. Differential gene expression analysis revealed 37 differentially expressed genes, predominantly in cardiomyocytes but also in other cell types, relevant to aging, muscle contraction, cell motility, and the extracellular matrix. Intercellular communication was generally reduced in HCM, affecting the extracellular matrix, growth factor binding, integrin binding, PDGF binding, and SMAD binding, but with increases in adenylate cyclase binding, calcium channel inhibitor activity, and serine-threonine kinase activity in nonobstructive HCM. Increases in neuron to leukocyte and dendritic cell communication, in fibroblast to leukocyte and dendritic cell communication, and in endothelial cell communication to other cell types, largely through changes in the expression of integrin-β1 and its cognate ligands, were also noted. These findings indicate both common and distinct physiological mechanisms affecting the pathogenesis of obstructive and nonobstructive HCM and provide opportunities for the personalized management of different HCM phenotypes.
Collapse
Affiliation(s)
- Christina J. Codden
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA 02111, USA;
| | - Michael T. Chin
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA 02111, USA;
- Tufts Hypertrophic Cardiomyopathy Center and Research Institute, Boston, MA 02111, USA
| |
Collapse
|
222
|
Erber L, Liu S, Gong Y, Tran P, Chen Y. Quantitative Proteome and Transcriptome Dynamics Analysis Reveals Iron Deficiency Response Networks and Signature in Neuronal Cells. Molecules 2022; 27:484. [PMID: 35056799 PMCID: PMC8779535 DOI: 10.3390/molecules27020484] [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: 12/15/2021] [Revised: 01/05/2022] [Accepted: 01/10/2022] [Indexed: 01/17/2023] Open
Abstract
Iron and oxygen deficiencies are common features in pathophysiological conditions, such as ischemia, neurological diseases, and cancer. Cellular adaptive responses to such deficiencies include repression of mitochondrial respiration, promotion of angiogenesis, and cell cycle control. We applied a systematic proteomics analysis to determine the global proteomic changes caused by acute hypoxia and chronic and acute iron deficiency (ID) in hippocampal neuronal cells. Our analysis identified over 8600 proteins, revealing similar and differential effects of each treatment on activation and inhibition of pathways regulating neuronal development. In addition, comparative analysis of ID-induced proteomics changes in cultured cells and transcriptomic changes in the rat hippocampus identified common altered pathways, indicating specific neuronal effects. Transcription factor enrichment and correlation analysis identified key transcription factors that were activated in both cultured cells and tissue by iron deficiency, including those implicated in iron regulation, such as HIF1, NFY, and NRF1. We further identified MEF2 as a novel transcription factor whose activity was induced by ID in both HT22 proteome and rat hippocampal transcriptome, thus linking iron deficiency to MEF2-dependent cellular signaling pathways in neuronal development. Taken together, our study results identified diverse signaling networks that were differentially regulated by hypoxia and ID in neuronal cells.
Collapse
Affiliation(s)
- Luke Erber
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota at Twin Cities, Minneapolis, MN 55455, USA; (L.E.); (Y.G.)
| | - Shirelle Liu
- Department of Pediatrics, University of Minnesota at Twin Cities, Minneapolis, MN 55455, USA;
| | - Yao Gong
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota at Twin Cities, Minneapolis, MN 55455, USA; (L.E.); (Y.G.)
| | - Phu Tran
- Department of Pediatrics, University of Minnesota at Twin Cities, Minneapolis, MN 55455, USA;
| | - Yue Chen
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota at Twin Cities, Minneapolis, MN 55455, USA; (L.E.); (Y.G.)
| |
Collapse
|
223
|
Thuy-Boun PS, Wang AY, Crissien-Martinez A, Xu JH, Chatterjee S, Stupp GS, Su AI, Coyle WJ, Wolan DW. Quantitative metaproteomics and activity-based protein profiling of patient fecal microbiome identifies host and microbial serine-type endopeptidase activity associated with ulcerative colitis. Mol Cell Proteomics 2022; 21:100197. [PMID: 35033677 PMCID: PMC8941213 DOI: 10.1016/j.mcpro.2022.100197] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 01/10/2022] [Accepted: 01/11/2022] [Indexed: 12/12/2022] Open
Abstract
The gut microbiota plays an important yet incompletely understood role in the induction and propagation of ulcerative colitis (UC). Organism-level efforts to identify UC-associated microbes have revealed the importance of community structure, but less is known about the molecular effectors of disease. We performed 16S rRNA gene sequencing in parallel with label-free data-dependent LC-MS/MS proteomics to characterize the stool microbiomes of healthy (n = 8) and UC (n = 10) patients. Comparisons of taxonomic composition between techniques revealed major differences in community structure partially attributable to the additional detection of host, fungal, viral, and food peptides by metaproteomics. Differential expression analysis of metaproteomic data identified 176 significantly enriched protein groups between healthy and UC patients. Gene ontology analysis revealed several enriched functions with serine-type endopeptidase activity overrepresented in UC patients. Using a biotinylated fluorophosphonate probe and streptavidin-based enrichment, we show that serine endopeptidases are active in patient fecal samples and that additional putative serine hydrolases are detectable by this approach compared with unenriched profiling. Finally, as metaproteomic databases expand, they are expected to asymptotically approach completeness. Using ComPIL and de novo peptide sequencing, we estimate the size of the probable peptide space unidentified (“dark peptidome”) by our large database approach to establish a rough benchmark for database sufficiency. Despite high variability inherent in patient samples, our analysis yielded a catalog of differentially enriched proteins between healthy and UC fecal proteomes. This catalog provides a clinically relevant jumping-off point for further molecular-level studies aimed at identifying the microbial underpinnings of UC. Identified 176 significantly altered protein groups between healthy and UC patients. Serine-type endopeptidase activity is overrepresented in UC patients. Fluorophosphonate ABPP shows that endopeptidases are active in fecal samples. ABPP enrichment helps identify additional putative serine hydrolases in samples. De novo sequencing used to estimate number of MS2 spectra unidentified by ComPIL.
Collapse
Affiliation(s)
- Peter S Thuy-Boun
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037
| | - Ana Y Wang
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037
| | | | - Janice H Xu
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037
| | - Sandip Chatterjee
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037
| | - Gregory S Stupp
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037
| | - Andrew I Su
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037
| | - Walter J Coyle
- Scripps Clinic Gastroenterology Division, La Jolla, CA 92037
| | - Dennis W Wolan
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037; Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037.
| |
Collapse
|
224
|
Czapiewski R, Batrakou DG, de Las Heras JI, Carter RN, Sivakumar A, Sliwinska M, Dixon CR, Webb S, Lattanzi G, Morton NM, Schirmer EC. Genomic loci mispositioning in Tmem120a knockout mice yields latent lipodystrophy. Nat Commun 2022; 13:321. [PMID: 35027552 PMCID: PMC8758788 DOI: 10.1038/s41467-021-27869-2] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 12/19/2021] [Indexed: 12/13/2022] Open
Abstract
Little is known about how the observed fat-specific pattern of 3D-spatial genome organisation is established. Here we report that adipocyte-specific knockout of the gene encoding nuclear envelope transmembrane protein Tmem120a disrupts fat genome organisation, thus causing a lipodystrophy syndrome. Tmem120a deficiency broadly suppresses lipid metabolism pathway gene expression and induces myogenic gene expression by repositioning genes, enhancers and miRNA-encoding loci between the nuclear periphery and interior. Tmem120a-/- mice, particularly females, exhibit a lipodystrophy syndrome similar to human familial partial lipodystrophy FPLD2, with profound insulin resistance and metabolic defects that manifest upon exposure to an obesogenic diet. Interestingly, similar genome organisation defects occurred in cells from FPLD2 patients that harbour nuclear envelope protein encoding LMNA mutations. Our data indicate TMEM120A genome organisation functions affect many adipose functions and its loss may yield adiposity spectrum disorders, including a miRNA-based mechanism that could explain muscle hypertrophy in human lipodystrophy.
Collapse
Affiliation(s)
- Rafal Czapiewski
- Institute of Cell Biology, University of Edinburgh, Edinburgh, EH9 3BF, UK
| | - Dzmitry G Batrakou
- Institute of Cell Biology, University of Edinburgh, Edinburgh, EH9 3BF, UK
| | | | - Roderick N Carter
- Molecular Metabolism Group, University/BHF Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, EH16 4TJ, UK
| | | | | | - Charles R Dixon
- Institute of Cell Biology, University of Edinburgh, Edinburgh, EH9 3BF, UK
- Wellcome Centre for Cell Biology, University of Edinburgh, Edinburgh, EH9 3BF, UK
| | - Shaun Webb
- Wellcome Centre for Cell Biology, University of Edinburgh, Edinburgh, EH9 3BF, UK
| | - Giovanna Lattanzi
- CNR - National Research Council of Italy, Institute of Molecular Genetics "Luigi Luca Cavalli-Sforza", Unit of Bologna, Bologna, 40136, Italy
- IRCCS, Istituto Ortopedico Rizzoli, Bologna, 40136, Italy
| | - Nicholas M Morton
- Molecular Metabolism Group, University/BHF Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, EH16 4TJ, UK
| | - Eric C Schirmer
- Institute of Cell Biology, University of Edinburgh, Edinburgh, EH9 3BF, UK.
| |
Collapse
|
225
|
Bhatti G, Romero R, Gomez-Lopez N, Chaiworapongsa T, Jung E, Gotsch F, Pique-Regi R, Pacora P, Hsu CD, Kavdia M, Tarca AL. The amniotic fluid proteome changes with gestational age in normal pregnancy: a cross-sectional study. Sci Rep 2022; 12:601. [PMID: 35022423 PMCID: PMC8755742 DOI: 10.1038/s41598-021-04050-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 12/02/2021] [Indexed: 11/28/2022] Open
Abstract
The cell-free transcriptome in amniotic fluid (AF) has been shown to be informative of physiologic and pathologic processes in pregnancy; however, the change in AF proteome with gestational age has mostly been studied by targeted approaches. The objective of this study was to describe the gestational age-dependent changes in the AF proteome during normal pregnancy by using an omics platform. The abundance of 1310 proteins was measured on a high-throughput aptamer-based proteomics platform in AF samples collected from women during midtrimester (16-24 weeks of gestation, n = 15) and at term without labor (37-42 weeks of gestation, n = 13). Only pregnancies without obstetrical complications were included in the study. Almost 25% (320) of AF proteins significantly changed in abundance between the midtrimester and term gestation. Of these, 154 (48.1%) proteins increased, and 166 (51.9%) decreased in abundance at term compared to midtrimester. Tissue-specific signatures of the trachea, salivary glands, brain regions, and immune system were increased while those of the gestational tissues (uterus, placenta, and ovary), cardiac myocytes, and fetal liver were decreased at term compared to midtrimester. The changes in AF protein abundance were correlated with those previously reported in the cell-free AF transcriptome. Intersecting gestational age-modulated AF proteins and their corresponding mRNAs previously reported in the maternal blood identified neutrophil-related protein/mRNA pairs that were modulated in the same direction. The first study to utilize an aptamer-based assay to profile the AF proteome modulation with gestational age, it reveals that almost one-quarter of the proteins are modulated as gestation advances, which is more than twice the fraction of altered plasma proteins (~ 10%). The results reported herein have implications for future studies focused on discovering biomarkers to predict, monitor, and diagnose obstetrical diseases.
Collapse
Affiliation(s)
- Gaurav Bhatti
- Perinatology Research Branch, US Department of Health and Human Services, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
- Department of Biomedical Engineering, Wayne State University College of Engineering, Detroit, MI, USA
| | - Roberto Romero
- Perinatology Research Branch, US Department of Health and Human Services, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Detroit, MI, USA.
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI, USA.
- Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, MI, USA.
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI, USA.
- Detroit Medical Center, Detroit, MI, USA.
| | - Nardhy Gomez-Lopez
- Perinatology Research Branch, US Department of Health and Human Services, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
- Department of Biochemistry, Microbiology and Immunology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Tinnakorn Chaiworapongsa
- Perinatology Research Branch, US Department of Health and Human Services, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Eunjung Jung
- Perinatology Research Branch, US Department of Health and Human Services, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Francesca Gotsch
- Perinatology Research Branch, US Department of Health and Human Services, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
- Office of Women's Health, Integrative Biosciences Center, Wayne State University, Detroit, MI, USA
| | - Roger Pique-Regi
- Perinatology Research Branch, US Department of Health and Human Services, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI, USA
| | - Percy Pacora
- Perinatology Research Branch, US Department of Health and Human Services, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
- Department of Obstetrics, Gynecology & Reproductive Sciences, The University of Texas Health Sciences Center at Houston, Houston, TX, USA
| | - Chaur-Dong Hsu
- Perinatology Research Branch, US Department of Health and Human Services, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
- Department of Physiology, Wayne State University School of Medicine, Detroit, MI, USA
- Department of Obstetrics & Gynecology, University of Arizona College of Medicine -Tucson, Tucson, AZ, USA
| | - Mahendra Kavdia
- Department of Biomedical Engineering, Wayne State University College of Engineering, Detroit, MI, USA
| | - Adi L Tarca
- Perinatology Research Branch, US Department of Health and Human Services, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Detroit, MI, USA.
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA.
- Department of Computer Science, Wayne State University College of Engineering, Detroit, MI, USA.
| |
Collapse
|
226
|
Manakanatas C, Ghadge SK, Agic A, Sarigol F, Fichtinger P, Fischer I, Foisner R, Osmanagic-Myers S. Endothelial and systemic upregulation of miR-34a-5p fine-tunes senescence in progeria. Aging (Albany NY) 2022; 14:195-224. [PMID: 35020601 PMCID: PMC8791216 DOI: 10.18632/aging.203820] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2021] [Accepted: 12/25/2021] [Indexed: 11/25/2022]
Abstract
Endothelial defects significantly contribute to cardiovascular pathology in the premature aging disease Hutchinson-Gilford progeria syndrome (HGPS). Using an endothelium-specific progeria mouse model, we identify a novel, endothelium-specific microRNA (miR) signature linked to the p53-senescence pathway and a senescence-associated secretory phenotype (SASP). Progerin-expressing endothelial cells exert profound cell-non-autonomous effects initiating senescence in non-endothelial cell populations and causing immune cell infiltrates around blood vessels. Comparative miR expression analyses revealed unique upregulation of senescence-associated miR34a-5p in endothelial cells with strong accumulation at atheroprone aortic arch regions but also, in whole cardiac- and lung tissues as well as in the circulation of progeria mice. Mechanistically, miR34a-5p knockdown reduced not only p53 levels but also late-stage senescence regulator p16 with no effect on p21 levels, while p53 knockdown reduced miR34a-5p and partially rescued p21-mediated cell cycle inhibition with a moderate effect on SASP. These data demonstrate that miR34a-5p reinforces two separate senescence regulating branches in progerin-expressing endothelial cells, the p53- and p16-associated pathways, which synergistically maintain a senescence phenotype that contributes to cardiovascular pathology. Thus, the key function of circulatory miR34a-5p in endothelial dysfunction-linked cardiovascular pathology offers novel routes for diagnosis, prognosis and treatment for cardiovascular aging in HGPS and potentially geriatric patients.
Collapse
Affiliation(s)
- Christina Manakanatas
- Max Perutz Labs, Center for Medical Biochemistry, Medical University of Vienna, Vienna Biocenter Campus (VBC), Vienna A-1030, Austria
| | - Santhosh Kumar Ghadge
- Max Perutz Labs, Center for Medical Biochemistry, Medical University of Vienna, Vienna Biocenter Campus (VBC), Vienna A-1030, Austria
| | - Azra Agic
- Max Perutz Labs, Center for Medical Biochemistry, Medical University of Vienna, Vienna Biocenter Campus (VBC), Vienna A-1030, Austria
| | - Fatih Sarigol
- Max Perutz Labs, Center for Medical Biochemistry, Medical University of Vienna, Vienna Biocenter Campus (VBC), Vienna A-1030, Austria
| | - Petra Fichtinger
- Max Perutz Labs, Center for Medical Biochemistry, Medical University of Vienna, Vienna Biocenter Campus (VBC), Vienna A-1030, Austria
| | - Irmgard Fischer
- Max Perutz Labs, Center for Medical Biochemistry, Medical University of Vienna, Vienna Biocenter Campus (VBC), Vienna A-1030, Austria
| | - Roland Foisner
- Max Perutz Labs, Center for Medical Biochemistry, Medical University of Vienna, Vienna Biocenter Campus (VBC), Vienna A-1030, Austria
| | - Selma Osmanagic-Myers
- Max Perutz Labs, Center for Medical Biochemistry, Medical University of Vienna, Vienna Biocenter Campus (VBC), Vienna A-1030, Austria
- Institute of Medical Chemistry, Center for Pathobiochemistry and Genetics, Medical University of Vienna, Vienna A-1090, Austria
| |
Collapse
|
227
|
Sadovska L, Auders J, Keiša L, Romanchikova N, Silamiķele L, Kreišmane M, Zayakin P, Takahashi S, Kalniņa Z, Linē A. Exercise-Induced Extracellular Vesicles Delay the Progression of Prostate Cancer. Front Mol Biosci 2022; 8:784080. [PMID: 35087866 PMCID: PMC8787363 DOI: 10.3389/fmolb.2021.784080] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 12/17/2021] [Indexed: 11/23/2022] Open
Abstract
Increasing evidence suggests that regular physical exercise not only reduces the risk of cancer but also improves functional capacity, treatment efficacy and disease outcome in cancer patients. At least partially, these effects are mediated by the secretome of the tissues responding to exercise. The secreted molecules can be released in a carrier-free form or enclosed into extracellular vesicles (EVs). Several recent studies have shown that EVs are actively released into circulation during physical exercise. Here, we for the first time investigated the effects of exercise-induced EVs on the progression of cancer in an F344 rat model of metastatic prostate cancer. Although we did not observe a consistent increase in the circulating EV levels, RNA sequencing analysis demonstrated substantial changes in the RNA content of EVs collected before and immediately after forced wheel running exercise as well as differences between EVs from runners at resting state and sedentary rats. The major RNA biotype in EVs was mRNA, followed by miRNA and rRNA. Molecular functions of differentially expressed RNAs reflected various physiological processes including protein folding, metabolism and regulation of immune responses triggered by the exercise in the parental cells. Intravenous administration of exercise-induced EVs into F344 rats with orthotopically injected syngeneic prostate cancer cells PLS10, demonstrated reduction of the primary tumor volume by 35% and possibly—attenuation of lung metastases. Hence, our data provide the first evidence that exercise-induced EVs may modulate tumor physiology and delay the progression of cancer.
Collapse
Affiliation(s)
- Lilite Sadovska
- Cancer Biomarker Group, Latvian Biomedical Research and Study Centre, Riga, Latvia
| | - Jānis Auders
- Cancer Biomarker Group, Latvian Biomedical Research and Study Centre, Riga, Latvia
- Faculty of Medicine, University of Latvia, Riga, Latvia
| | - Laura Keiša
- Cancer Biomarker Group, Latvian Biomedical Research and Study Centre, Riga, Latvia
- Faculty of Medicine, University of Latvia, Riga, Latvia
| | | | - Laila Silamiķele
- Laboratory Animal Core Facility, Latvian Biomedical Research and Study Centre, Riga, Latvia
| | - Madara Kreišmane
- Laboratory Animal Core Facility, Latvian Biomedical Research and Study Centre, Riga, Latvia
| | - Pawel Zayakin
- Cancer Biomarker Group, Latvian Biomedical Research and Study Centre, Riga, Latvia
| | - Satoru Takahashi
- Department of Experimental Pathology and Tumor Biology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Zane Kalniņa
- Laboratory Animal Core Facility, Latvian Biomedical Research and Study Centre, Riga, Latvia
| | - Aija Linē
- Cancer Biomarker Group, Latvian Biomedical Research and Study Centre, Riga, Latvia
- Faculty of Biology, University of Latvia, Riga, Latvia
- *Correspondence: Aija Linē,
| |
Collapse
|
228
|
Grigolon G, Araldi E, Erni R, Wu JY, Thomas C, La Fortezza M, Laube B, Pöhlmann D, Stoffel M, Zarse K, Carreira EM, Ristow M, Fischer F. Grainyhead 1 acts as a drug-inducible conserved transcriptional regulator linked to insulin signaling and lifespan. Nat Commun 2022; 13:107. [PMID: 35013237 PMCID: PMC8748497 DOI: 10.1038/s41467-021-27732-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Accepted: 12/06/2021] [Indexed: 12/13/2022] Open
Abstract
Aging is impacted by interventions across species, often converging on metabolic pathways. Transcription factors regulate longevity yet approaches for their pharmacological modulation to exert geroprotection remain sparse. We show that increased expression of the transcription factor Grainyhead 1 (GRH-1) promotes lifespan and pathogen resistance in Caenorhabditis elegans. A compound screen identifies FDA-approved drugs able to activate human GRHL1 and promote nematodal GRH-1-dependent longevity. GRHL1 activity is regulated by post-translational lysine methylation and the phosphoinositide (PI) 3-kinase C2A. Consistently, nematodal longevity following impairment of the PI 3-kinase or insulin/IGF-1 receptor requires grh-1. In BXD mice, Grhl1 expression is positively correlated with lifespan and insulin sensitivity. In humans, GRHL1 expression positively correlates with insulin receptor signaling and also with lifespan. Fasting blood glucose levels, including in individuals with type 2 diabetes, are negatively correlated with GRHL1 expression. Thereby, GRH-1/GRHL1 is identified as a pharmacologically malleable transcription factor impacting insulin signaling and lifespan. Life- and healthspan of organisms can be modulated by dietary, genetic, or pharmacological interventions, which often affect metabolic pathways. Here the authors report that Grainyhead 1 is an evolutionarily conserved, drug-inducible transcription factor that promotes longevity in C. elegans, and thus a potential target for the development of geroprotective drugs.
Collapse
Affiliation(s)
- Giovanna Grigolon
- Energy Metabolism Laboratory, Institute of Translational Medicine, Department of Health Sciences and Technology, Swiss Federal Institute of Technology (ETH) Zurich, Schwerzenbach, CH-8603, Switzerland
| | - Elisa Araldi
- Energy Metabolism Laboratory, Institute of Translational Medicine, Department of Health Sciences and Technology, Swiss Federal Institute of Technology (ETH) Zurich, Schwerzenbach, CH-8603, Switzerland.,Metabolism and Metabolic Disease Laboratory, Institute for Molecular Health Sciences, Department of Biology, Swiss Federal Institute of Technology (ETH) Zurich, Zurich, CH-8093, Switzerland
| | - Reto Erni
- Laboratory of Organic Chemistry, Department of Chemistry and Applied Biosciences, Swiss Federal Institute of Technology (ETH) Zurich, Zurich, CH-8093, Switzerland
| | - Jia Yee Wu
- Energy Metabolism Laboratory, Institute of Translational Medicine, Department of Health Sciences and Technology, Swiss Federal Institute of Technology (ETH) Zurich, Schwerzenbach, CH-8603, Switzerland
| | - Carolin Thomas
- Energy Metabolism Laboratory, Institute of Translational Medicine, Department of Health Sciences and Technology, Swiss Federal Institute of Technology (ETH) Zurich, Schwerzenbach, CH-8603, Switzerland
| | - Marco La Fortezza
- Evolutionary Biology Laboratory, Department of Environmental Systems Science, Swiss Federal Institute of Technology (ETH) Zurich, Zurich, CH-8092, Switzerland
| | - Beate Laube
- Energy Metabolism Laboratory, Institute of Translational Medicine, Department of Health Sciences and Technology, Swiss Federal Institute of Technology (ETH) Zurich, Schwerzenbach, CH-8603, Switzerland
| | - Doris Pöhlmann
- Energy Metabolism Laboratory, Institute of Translational Medicine, Department of Health Sciences and Technology, Swiss Federal Institute of Technology (ETH) Zurich, Schwerzenbach, CH-8603, Switzerland
| | - Markus Stoffel
- Metabolism and Metabolic Disease Laboratory, Institute for Molecular Health Sciences, Department of Biology, Swiss Federal Institute of Technology (ETH) Zurich, Zurich, CH-8093, Switzerland
| | - Kim Zarse
- Energy Metabolism Laboratory, Institute of Translational Medicine, Department of Health Sciences and Technology, Swiss Federal Institute of Technology (ETH) Zurich, Schwerzenbach, CH-8603, Switzerland
| | - Erick M Carreira
- Laboratory of Organic Chemistry, Department of Chemistry and Applied Biosciences, Swiss Federal Institute of Technology (ETH) Zurich, Zurich, CH-8093, Switzerland
| | - Michael Ristow
- Energy Metabolism Laboratory, Institute of Translational Medicine, Department of Health Sciences and Technology, Swiss Federal Institute of Technology (ETH) Zurich, Schwerzenbach, CH-8603, Switzerland.
| | - Fabian Fischer
- Energy Metabolism Laboratory, Institute of Translational Medicine, Department of Health Sciences and Technology, Swiss Federal Institute of Technology (ETH) Zurich, Schwerzenbach, CH-8603, Switzerland
| |
Collapse
|
229
|
McInturf SA, Khan MA, Gokul A, Castro-Guerrero NA, Höhner R, Li J, Marjault HB, Fichman Y, Kunz HH, Goggin FL, Keyster M, Nechushtai R, Mittler R, Mendoza-Cózatl DG. Cadmium interference with iron sensing reveals transcriptional programs sensitive and insensitive to reactive oxygen species. JOURNAL OF EXPERIMENTAL BOTANY 2022; 73:324-338. [PMID: 34499172 DOI: 10.1093/jxb/erab393] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Accepted: 08/25/2021] [Indexed: 06/13/2023]
Abstract
Iron (Fe) is an essential micronutrient whose uptake is tightly regulated to prevent either deficiency or toxicity. Cadmium (Cd) is a non-essential element that induces both Fe deficiency and toxicity; however, the mechanisms behind these Fe/Cd-induced responses are still elusive. Here we explored Cd- and Fe-associated responses in wild-type Arabidopsis and in a mutant that overaccumulates Fe (opt3-2). Gene expression profiling revealed a large overlap between transcripts induced by Fe deficiency and Cd exposure. Interestingly, the use of opt3-2 allowed us to identify additional gene clusters originally induced by Cd in the wild type but repressed in the opt3-2 background. Based on the high levels of H2O2 found in opt3-2, we propose a model where reactive oxygen species prevent the induction of genes that are induced in the wild type by either Fe deficiency or Cd. Interestingly, a defined cluster of Fe-responsive genes was found to be insensitive to this negative feedback, suggesting that their induction by Cd is more likely to be the result of an impaired Fe sensing. Overall, our data suggest that Fe deficiency responses are governed by multiple inputs and that a hierarchical regulation of Fe homeostasis prevents the induction of specific networks when Fe and H2O2 levels are elevated.
Collapse
Affiliation(s)
- Samuel A McInturf
- Division of Plant Sciences, C.S. Bond Life Sciences Center, University of Missouri, Columbia, MO 65211, USA
| | - Mather A Khan
- Division of Plant Sciences, C.S. Bond Life Sciences Center, University of Missouri, Columbia, MO 65211, USA
| | - Arun Gokul
- Environmental Biotechnology Laboratory, Department of Biotechnology, University of the Western Cape, Bellville, Cape Town, 7535, South Africa
| | - Norma A Castro-Guerrero
- Division of Plant Sciences, C.S. Bond Life Sciences Center, University of Missouri, Columbia, MO 65211, USA
| | - Ricarda Höhner
- School of Biological Sciences, Washington State University, PO Box 644236, Pullman, WA 99164-4236, USA
| | - Jiamei Li
- Department of Entomology and Plant Pathology, 217 Plant Sciences Building, University of Arkansas System Division of Agriculture, Fayetteville, AR 72701, USA
| | | | - Yosef Fichman
- Division of Plant Sciences, C.S. Bond Life Sciences Center, University of Missouri, Columbia, MO 65211, USA
| | - Hans-Henning Kunz
- School of Biological Sciences, Washington State University, PO Box 644236, Pullman, WA 99164-4236, USA
- Biozentrum der LMU München, Germany
| | - Fiona L Goggin
- Department of Entomology and Plant Pathology, 217 Plant Sciences Building, University of Arkansas System Division of Agriculture, Fayetteville, AR 72701, USA
| | - Marshall Keyster
- Environmental Biotechnology Laboratory, Department of Biotechnology, University of the Western Cape, Bellville, Cape Town, 7535, South Africa
| | - Rachel Nechushtai
- Institute of Life Science, The Hebrew University of Jerusalem, Jerusalem, 91904Israel
| | - Ron Mittler
- Division of Plant Sciences, C.S. Bond Life Sciences Center, University of Missouri, Columbia, MO 65211, USA
| | - David G Mendoza-Cózatl
- Division of Plant Sciences, C.S. Bond Life Sciences Center, University of Missouri, Columbia, MO 65211, USA
- Environmental Biotechnology Laboratory, Department of Biotechnology, University of the Western Cape, Bellville, Cape Town, 7535, South Africa
| |
Collapse
|
230
|
Deneweth J, Van de Peer Y, Vermeirssen V. Nearby transposable elements impact plant stress gene regulatory networks: a meta-analysis in A. thaliana and S. lycopersicum. BMC Genomics 2022; 23:18. [PMID: 34983397 PMCID: PMC8725346 DOI: 10.1186/s12864-021-08215-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 11/09/2021] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Transposable elements (TE) make up a large portion of many plant genomes and are playing innovative roles in genome evolution. Several TEs can contribute to gene regulation by influencing expression of nearby genes as stress-responsive regulatory motifs. To delineate TE-mediated plant stress regulatory networks, we took a 2-step computational approach consisting of identifying TEs in the proximity of stress-responsive genes, followed by searching for cis-regulatory motifs in these TE sequences and linking them to known regulatory factors. Through a systematic meta-analysis of RNA-seq expression profiles and genome annotations, we investigated the relation between the presence of TE superfamilies upstream, downstream or within introns of nearby genes and the differential expression of these genes in various stress conditions in the TE-poor Arabidopsis thaliana and the TE-rich Solanum lycopersicum. RESULTS We found that stress conditions frequently expressed genes having members of various TE superfamilies in their genomic proximity, such as SINE upon proteotoxic stress and Copia and Gypsy upon heat stress in A. thaliana, and EPRV and hAT upon infection, and Harbinger, LINE and Retrotransposon upon light stress in S. lycopersicum. These stress-specific gene-proximal TEs were mostly located within introns and more detected near upregulated than downregulated genes. Similar stress conditions were often related to the same TE superfamily. Additionally, we detected both novel and known motifs in the sequences of those TEs pointing to regulatory cooption of these TEs upon stress. Next, we constructed the regulatory network of TFs that act through binding these TEs to their target genes upon stress and discovered TE-mediated regulons targeted by TFs such as BRB/BPC, HD, HSF, GATA, NAC, DREB/CBF and MYB factors in Arabidopsis and AP2/ERF/B3, NAC, NF-Y, MYB, CXC and HD factors in tomato. CONCLUSIONS Overall, we map TE-mediated plant stress regulatory networks using numerous stress expression profile studies for two contrasting plant species to study the regulatory role TEs play in the response to stress. As TE-mediated gene regulation allows plants to adapt more rapidly to new environmental conditions, this study contributes to the future development of climate-resilient plants.
Collapse
Affiliation(s)
- Jan Deneweth
- Department of Plant Biotechnology and Bioinformatics, Ghent University, Ghent, Belgium
| | - Yves Van de Peer
- Department of Plant Biotechnology and Bioinformatics, Ghent University, Ghent, Belgium.,VIB Center for Plant Systems Biology, Ghent, Belgium.,Department of Biochemistry, Genetics and Microbiology, University of Pretoria, Pretoria, South Africa
| | - Vanessa Vermeirssen
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium. .,Department of Biomolecular Medicine, Ghent University, Ghent, Belgium. .,Lab for Computational Biology, Integromics and Gene Regulation (CBIGR), Cancer Research Institute Ghent (CRIG), Ghent, Belgium.
| |
Collapse
|
231
|
Pesaranghader A, Matwin S, Sokolova M, Grenier JC, Beiko RG, Hussin J. OUP accepted manuscript. Bioinformatics 2022; 38:3051-3061. [PMID: 35536192 PMCID: PMC9154256 DOI: 10.1093/bioinformatics/btac304] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 02/12/2022] [Indexed: 11/24/2022] Open
Abstract
Motivation There is a plethora of measures to evaluate functional similarity (FS) of genes based on their co-expression, protein–protein interactions and sequence similarity. These measures are typically derived from hand-engineered and application-specific metrics to quantify the degree of shared information between two genes using their Gene Ontology (GO) annotations. Results We introduce deepSimDEF, a deep learning method to automatically learn FS estimation of gene pairs given a set of genes and their GO annotations. deepSimDEF’s key novelty is its ability to learn low-dimensional embedding vector representations of GO terms and gene products and then calculate FS using these learned vectors. We show that deepSimDEF can predict the FS of new genes using their annotations: it outperformed all other FS measures by >5–10% on yeast and human reference datasets on protein–protein interactions, gene co-expression and sequence homology tasks. Thus, deepSimDEF offers a powerful and adaptable deep neural architecture that can benefit a wide range of problems in genomics and proteomics, and its architecture is flexible enough to support its extension to any organism. Availability and implementation Source code and data are available at https://github.com/ahmadpgh/deepSimDEF Supplementary information Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
| | - Stan Matwin
- Faculty of Computer Science, Dalhousie University, Halifax B3H 4R2, Canada
- Institute for Big Data Analytics, Dalhousie University, Halifax B3H 4R2, Canada
- Institute of Computer Science, Polish Academy of Sciences, Warsaw, Poland
| | - Marina Sokolova
- Institute for Big Data Analytics, Dalhousie University, Halifax B3H 4R2, Canada
- Faculty of Medicine and Faculty of Engineering, University of Ottawa, Ottawa K1H 8M5, Canada
| | | | - Robert G Beiko
- Faculty of Computer Science, Dalhousie University, Halifax B3H 4R2, Canada
- Institute for Big Data Analytics, Dalhousie University, Halifax B3H 4R2, Canada
| | | |
Collapse
|
232
|
Wang IY, Chung CF, Babayeva S, Sogomonian T, Torban E. Loss of Planar Cell Polarity Effector Fuzzy Causes Renal Hypoplasia by Disrupting Several Signaling Pathways. J Dev Biol 2021; 10:jdb10010001. [PMID: 35076510 PMCID: PMC8788523 DOI: 10.3390/jdb10010001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 12/09/2021] [Accepted: 12/19/2021] [Indexed: 12/20/2022] Open
Abstract
In vertebrates, the planar cell polarity (PCP) pathway regulates tissue morphogenesis during organogenesis, including the kidney. Mutations in human PCP effector proteins have been associated with severe syndromic ciliopathies. Importantly, renal hypoplasia has been reported in some patients. However, the developmental disturbance that causes renal hypoplasia is unknown. Here, we describe the early onset of profound renal hypoplasia in mice homozygous for null mutation of the PCP effector gene, Fuzzy. We found that this phenotype is caused by defective branching morphogenesis of the ureteric bud (UB) in the absence of defects in nephron progenitor specification or in early steps of nephrogenesis. By using various experimental approaches, we show that the loss of Fuzzy affects multiple signaling pathways. Specifically, we found mild involvement of GDNF/c-Ret pathway that drives UB branching. We noted the deficient expression of molecules belonging to the Bmp, Fgf and Shh pathways. Analysis of the primary cilia in the UB structures revealed a significant decrease in ciliary length. We conclude that renal hypoplasia in the mouse Fuzzy mutants is caused by defective UB branching associated with dysregulation of ciliary and non-ciliary signaling pathways. Our work suggests a PCP effector-dependent pathogenetic mechanism that contributes to renal hypoplasia in mice and humans.
Collapse
Affiliation(s)
- Irene-Yanran Wang
- Department of Medicine, McGill University, Montreal, QC H4A 3J1, Canada; (I.-Y.W.); (C.-F.C.); (S.B.); (T.S.)
- McGill University Health Center Research Institute, Montreal, QC H4A 3J1, Canada
| | - Chen-Fang Chung
- Department of Medicine, McGill University, Montreal, QC H4A 3J1, Canada; (I.-Y.W.); (C.-F.C.); (S.B.); (T.S.)
- McGill University Health Center Research Institute, Montreal, QC H4A 3J1, Canada
| | - Sima Babayeva
- Department of Medicine, McGill University, Montreal, QC H4A 3J1, Canada; (I.-Y.W.); (C.-F.C.); (S.B.); (T.S.)
- McGill University Health Center Research Institute, Montreal, QC H4A 3J1, Canada
| | - Tamara Sogomonian
- Department of Medicine, McGill University, Montreal, QC H4A 3J1, Canada; (I.-Y.W.); (C.-F.C.); (S.B.); (T.S.)
- McGill University Health Center Research Institute, Montreal, QC H4A 3J1, Canada
| | - Elena Torban
- Department of Medicine, McGill University, Montreal, QC H4A 3J1, Canada; (I.-Y.W.); (C.-F.C.); (S.B.); (T.S.)
- McGill University Health Center Research Institute, Montreal, QC H4A 3J1, Canada
- Correspondence:
| |
Collapse
|
233
|
Honey Bee Larval and Adult Microbiome Life Stages Are Effectively Decoupled with Vertical Transmission Overcoming Early Life Perturbations. mBio 2021; 12:e0296621. [PMID: 34933445 PMCID: PMC8689520 DOI: 10.1128/mbio.02966-21] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Microbiomes provide a range of benefits to their hosts which can lead to the coevolution of a joint ecological niche. However, holometabolous insects, some of the most successful organisms on Earth, occupy different niches throughout development, with larvae and adults being physiologically and morphologically highly distinct. Furthermore, transition between the stages usually involves the loss of the gut microbiome since the gut is remodeled during pupation. Most eusocial organisms appear to have evolved a workaround to this problem by sharing their communal microbiome across generations. However, whether this vertical microbiome transmission can overcome perturbations of the larval microbiome remains untested. Honey bees have a relatively simple, conserved, coevolved adult microbiome which is socially transmitted and affects many aspects of their biology. In contrast, larval microbiomes are more variable, with less clear roles. Here, we manipulated the gut microbiome of in vitro-reared larvae, and after pupation of the larvae, we inoculated the emerged bees with adult microbiome to test whether adult and larval microbiome stages may be coupled (e.g., through immune priming). Larval treatments differed in bacterial composition and abundance, depending on diet, which also drove larval gene expression. Nonetheless, adults converged on the typical core taxa and showed limited gene expression variation. This work demonstrates that honey bee adult and larval stages are effectively microbiologically decoupled, and the core adult microbiome is remarkably stable to early developmental perturbations. Combined with the transmission of the microbiome in early adulthood, this allows the formation of long-term host-microbiome associations. IMPORTANCE This work investigated host-microbiome interactions during a crucial developmental stage-the transition from larvae to adults, which is a challenge to both, the insect host and its microbiome. Using the honey bee as a tractable model system, we showed that microbiome transfer after emergence overrides any variation in the larvae, indicating that larval and adult microbiome stages are effectively decoupled. Together with the reliable vertical transfer in the eusocial system, this decoupling ensures that the adults are colonized with a consistent and derived microbiome after eclosion. Taken all together, our data provide additional support that the evolution of sociality, at least in the honey bee system tested here, is linked with host-microbiome relationships.
Collapse
|
234
|
Bojanowski K, Ibeji CU, Singh P, Swindell WR, Chaudhuri RK. A Sensitization-Free Dimethyl Fumarate Prodrug, Isosorbide Di-(Methyl Fumarate), Provides a Topical Treatment Candidate for Psoriasis. JID INNOVATIONS 2021; 1:100040. [PMID: 34909741 PMCID: PMC8659395 DOI: 10.1016/j.xjidi.2021.100040] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 05/28/2021] [Accepted: 06/07/2021] [Indexed: 12/27/2022] Open
Abstract
Dimethyl fumarate (DMF) is an effective oral treatment for psoriasis administered in Europe for nearly 60 years. However, its potential has been limited by contact dermatitis that prohibits topical application. This paper characterizes a DMF derivative, isosorbide DMF (IDMF), which was designed to have antipsoriatic effects without skin-sensitizing properties. We show that IDMF exhibits neither genotoxicity nor radiation sensitivity in skin fibroblasts and is nonirritating and nonsensitizing in animal models (rat, rabbit, guinea pig). Microarray analysis of cytokine-stimulated keratinocytes showed that IDMF represses the expression of genes specifically upregulated in psoriatic skin lesions but not those of other skin diseases. IDMF also downregulated genes induced by IL-17A and TNF in keratinocytes as well as predicted targets of NF-κB and the antidifferentiation noncoding RNA (i.e., ANCR). IDMF further stimulated the transcription of oxidative stress response genes (NQO1, GPX2, GSR) with stronger NRF2/ARE activation compared to DMF. Finally, IDMF reduced erythema and scaling while repressing the expression of immune response genes in psoriasiform lesions elicited by topical application of imiquimod in mice. These data show that IDMF exhibits antipsoriatic activity that is similar or improved compared with that exhibited by DMF, without the harsh skin-sensitizing effects that have prevented topical delivery of the parent molecule.
Collapse
Key Words
- ARE, antioxidant response element
- CES2, carboxylesterase 2
- CPD, cyclobutane pyrimidine dimer
- CTRL, control
- DEG, differentially expressed gene
- DMF, dimethyl fumarate
- FC, fold change
- FDR, false discovery rate
- GSH, glutathione
- IDMF, isosorbide di-(methyl fumarate)
- IMQ, imiquimod
- KC, keratinocyte
- MMF, monomethyl fumarate
- PN, uninvolved skin from psoriasis patient
- PP, lesional skin from psoriasis patient
- RNA-seq, RNA sequencing
- VEH, vehicle
Collapse
Affiliation(s)
- Krzysztof Bojanowski
- Sunny BioDiscovery, Inc, Santa Paula, California, USA.,Symbionyx Pharmaceuticals Inc, Boonton, New Jersey, USA
| | - Collins U Ibeji
- Department of Pure and Industrial Chemistry, Faculty of Physical Sciences, University of Nigeria, Nsukka, Nigeria
| | - Parvesh Singh
- School of Chemistry & Physics, University of KwaZulu-Natal, Durban, South Africa
| | - William R Swindell
- Department of Internal Medicine, The Jewish Hospital, Cincinnati, Ohio, USA
| | - Ratan K Chaudhuri
- Symbionyx Pharmaceuticals Inc, Boonton, New Jersey, USA.,Sytheon Ltd, Boonton, New Jersey, USA
| |
Collapse
|
235
|
Gamez S, Chaverra-Rodriguez D, Buchman A, Kandul NP, Mendez-Sanchez SC, Bennett JB, Sánchez C HM, Yang T, Antoshechkin I, Duque JE, Papathanos PA, Marshall JM, Akbari OS. Exploiting a Y chromosome-linked Cas9 for sex selection and gene drive. Nat Commun 2021; 12:7202. [PMID: 34893590 PMCID: PMC8664916 DOI: 10.1038/s41467-021-27333-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Accepted: 11/03/2021] [Indexed: 02/06/2023] Open
Abstract
CRISPR-based genetic engineering tools aimed to bias sex ratios, or drive effector genes into animal populations, often integrate the transgenes into autosomal chromosomes. However, in species with heterogametic sex chromsomes (e.g. XY, ZW), sex linkage of endonucleases could be beneficial to drive the expression in a sex-specific manner to produce genetic sexing systems, sex ratio distorters, or even sex-specific gene drives, for example. To explore this possibility, here we develop a transgenic line of Drosophila melanogaster expressing Cas9 from the Y chromosome. We functionally characterize the utility of this strain for both sex selection and gene drive finding it to be quite effective. To explore its utility for population control, we built mathematical models illustrating its dynamics as compared to other state-of-the-art systems designed for both population modification and suppression. Taken together, our results contribute to the development of current CRISPR genetic control tools and demonstrate the utility of using sex-linked Cas9 strains for genetic control of animals.
Collapse
Affiliation(s)
- Stephanie Gamez
- Division of Biological Sciences, Section of Cell and Developmental Biology, University of California, San Diego, La Jolla, CA, 92093, USA
- Agragene Inc., San Diego, CA, 92121, USA
| | - Duverney Chaverra-Rodriguez
- Division of Biological Sciences, Section of Cell and Developmental Biology, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Anna Buchman
- Division of Biological Sciences, Section of Cell and Developmental Biology, University of California, San Diego, La Jolla, CA, 92093, USA
- Verily Life Sciences, South San Francisco, CA, 94080, USA
| | - Nikolay P Kandul
- Division of Biological Sciences, Section of Cell and Developmental Biology, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Stelia C Mendez-Sanchez
- Division of Biological Sciences, Section of Cell and Developmental Biology, University of California, San Diego, La Jolla, CA, 92093, USA
- Group for Research in Biochemistry and Microbiology (Grupo de Investigación en Bioquímica Y Microbiología-GIBIM), School of Chemistry, Universidad Industrial de Santander, Bucaramanga, Colombia
| | - Jared B Bennett
- Biophysics Graduate Group, University of California, Berkeley, CA, 94720, USA
- Divisions of Epidemiology & Biostatistics, School of Public Health, University of California, Berkeley, CA, 94720, USA
| | - Héctor M Sánchez C
- Divisions of Epidemiology & Biostatistics, School of Public Health, University of California, Berkeley, CA, 94720, USA
| | - Ting Yang
- Division of Biological Sciences, Section of Cell and Developmental Biology, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Igor Antoshechkin
- Division of Biology and Biological Engineering (BBE), California Institute of Technology, Pasadena, CA, 91125, USA
| | - Jonny E Duque
- Division of Biological Sciences, Section of Cell and Developmental Biology, University of California, San Diego, La Jolla, CA, 92093, USA
- Centro de Investigaciones en Enfermedades Tropicales - CINTROP, Facultad de Salud, Escuela de Medicina, Departamento de Ciencias Básicas, Universidad Industrial de Santander, Piedecuesta, Santander, Colombia
| | - Philippos A Papathanos
- Department of Entomology, Robert H. Smith Faculty of Agriculture, Food and Environment, Hebrew University of Jerusalem, Rehovot, 7610001, Israel
| | - John M Marshall
- Divisions of Epidemiology & Biostatistics, School of Public Health, University of California, Berkeley, CA, 94720, USA
- Innovative Genomics Institute, University of California, Berkeley, CA, 94720, USA
| | - Omar S Akbari
- Division of Biological Sciences, Section of Cell and Developmental Biology, University of California, San Diego, La Jolla, CA, 92093, USA.
| |
Collapse
|
236
|
Astarita JL, Keerthivasan S, Husain B, Şenbabaoğlu Y, Verschueren E, Gierke S, Pham VC, Peterson SM, Chalouni C, Pierce AA, Lill JR, Gonzalez LC, Martinez-Martin N, Turley SJ. The neutrophil protein CD177 is a novel PDPN receptor that regulates human cancer-associated fibroblast physiology. PLoS One 2021; 16:e0260800. [PMID: 34879110 PMCID: PMC8654239 DOI: 10.1371/journal.pone.0260800] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 11/18/2021] [Indexed: 01/12/2023] Open
Abstract
The cancer-associated fibroblast (CAF) marker podoplanin (PDPN) is generally correlated with poor clinical outcomes in cancer patients and thus represents a promising therapeutic target. Despite its biomedical relevance, basic aspects of PDPN biology such as its cellular functions and cell surface ligands remain poorly uncharacterized, thus challenging drug development. Here, we utilize a high throughput platform to elucidate the PDPN cell surface interactome, and uncover the neutrophil protein CD177 as a new binding partner. Quantitative proteomics analysis of the CAF phosphoproteome reveals a role for PDPN in cell signaling, growth and actomyosin contractility, among other processes. Moreover, cellular assays demonstrate that CD177 is a functional antagonist, recapitulating the phenotype observed in PDPN-deficient CAFs. In sum, starting from the unbiased elucidation of the PDPN co-receptome, our work provides insights into PDPN functions and reveals the PDPN/CD177 axis as a possible modulator of fibroblast physiology in the tumor microenvironment.
Collapse
Affiliation(s)
- Jillian L. Astarita
- Department of Cancer Immunology, Genentech, South San Francisco, California, United States of America
| | - Shilpa Keerthivasan
- Department of Cancer Immunology, Genentech, South San Francisco, California, United States of America
| | - Bushra Husain
- Department of Microchemistry, Proteomics and Lipidomics, Genentech, South San Francisco, California, United States of America
| | - Yasin Şenbabaoğlu
- Department of Bioinformatics and Computational Biology, Genentech, South San Francisco, California, United States of America
| | - Erik Verschueren
- Department of Microchemistry, Proteomics and Lipidomics, Genentech, South San Francisco, California, United States of America
| | - Sarah Gierke
- Center for Advanced Light Microscopy, Genentech, South San Francisco, California, United States of America
| | - Victoria C. Pham
- Department of Microchemistry, Proteomics and Lipidomics, Genentech, South San Francisco, California, United States of America
| | - Sean M. Peterson
- Department of Microchemistry, Proteomics and Lipidomics, Genentech, South San Francisco, California, United States of America
| | - Cecile Chalouni
- Center for Advanced Light Microscopy, Genentech, South San Francisco, California, United States of America
| | - Andrew A. Pierce
- Department of Research Pathology, Genentech, South San Francisco, California, United States of America
| | - Jennie R. Lill
- Department of Microchemistry, Proteomics and Lipidomics, Genentech, South San Francisco, California, United States of America
| | - Lino C. Gonzalez
- Department of Microchemistry, Proteomics and Lipidomics, Genentech, South San Francisco, California, United States of America
| | - Nadia Martinez-Martin
- Department of Microchemistry, Proteomics and Lipidomics, Genentech, South San Francisco, California, United States of America
- * E-mail: (SJT); (NMM)
| | - Shannon J. Turley
- Department of Cancer Immunology, Genentech, South San Francisco, California, United States of America
- * E-mail: (SJT); (NMM)
| |
Collapse
|
237
|
Gao R, Kalathur RKR, Coto‐Llerena M, Ercan C, Buechel D, Shuang S, Piscuoglio S, Dill MT, Camargo FD, Christofori G, Tang F. YAP/TAZ and ATF4 drive resistance to Sorafenib in hepatocellular carcinoma by preventing ferroptosis. EMBO Mol Med 2021; 13:e14351. [PMID: 34664408 PMCID: PMC8649869 DOI: 10.15252/emmm.202114351] [Citation(s) in RCA: 315] [Impact Index Per Article: 78.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 09/29/2021] [Accepted: 10/04/2021] [Indexed: 12/20/2022] Open
Abstract
Understanding the mechanisms underlying evasive resistance in cancer is an unmet medical need to improve the efficacy of current therapies. In this study, a combination of shRNA-mediated synthetic lethality screening and transcriptomic analysis revealed the transcription factors YAP/TAZ as key drivers of Sorafenib resistance in hepatocellular carcinoma (HCC) by repressing Sorafenib-induced ferroptosis. Mechanistically, in a TEAD-dependent manner, YAP/TAZ induce the expression of SLC7A11, a key transporter maintaining intracellular glutathione homeostasis, thus enabling HCC cells to overcome Sorafenib-induced ferroptosis. At the same time, YAP/TAZ sustain the protein stability, nuclear localization, and transcriptional activity of ATF4 which in turn cooperates to induce SLC7A11 expression. Our study uncovers a critical role of YAP/TAZ in the repression of ferroptosis and thus in the establishment of Sorafenib resistance in HCC, highlighting YAP/TAZ-based rewiring strategies as potential approaches to overcome HCC therapy resistance.
Collapse
Affiliation(s)
- Ruize Gao
- Department of BiomedicineUniversity of BaselBaselSwitzerland
| | | | | | - Caner Ercan
- Institute of PathologyUniversity Hospital BaselBaselSwitzerland
| | - David Buechel
- Department of BiomedicineUniversity of BaselBaselSwitzerland
| | - Song Shuang
- Friedrich Miescher Institute for Biomedical ResearchBaselSwitzerland
| | | | - Michael T Dill
- Stem Cell ProgramBoston Children's HospitalBostonMAUSA
- Department of Stem Cell and Regenerative BiologyHarvard UniversityCambridgeMAUSA
| | - Fernando D Camargo
- Stem Cell ProgramBoston Children's HospitalBostonMAUSA
- Department of Stem Cell and Regenerative BiologyHarvard UniversityCambridgeMAUSA
| | | | - Fengyuan Tang
- Department of BiomedicineUniversity of BaselBaselSwitzerland
| |
Collapse
|
238
|
Sun H, Lu Z, Singh A, Zhou Y, Zheng E, Zhou M, Wang J, Wu X, Hu Z, Gu Z, Campbell JL, Zheng L, Shen B. Error-prone, stress-induced 3' flap-based Okazaki fragment maturation supports cell survival. Science 2021; 374:1252-1258. [PMID: 34855483 PMCID: PMC8852821 DOI: 10.1126/science.abj1013] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
How cells with DNA replication defects acquire mutations that allow them to escape apoptosis under environmental stress is a long-standing question. Here, we report that an error-prone Okazaki fragment maturation (OFM) pathway is activated at restrictive temperatures in rad27Δ yeast cells. Restrictive temperature stress activated Dun1, facilitating transformation of unprocessed 5′ flaps into 3′ flaps, which were removed by 3′ nucleases, including DNA polymerase δ (Polδ). However, at certain regions, 3′ flaps formed secondary structures that facilitated 3′ end extension rather than degradation, producing alternative duplications with short spacer sequences, such as pol3 internal tandem duplications. Consequently, little 5′ flap was formed, suppressing rad27Δ-induced lethality at restrictive temperatures. We define a stress-induced, error-prone OFM pathway that generates mutations that counteract replication defects and drive cellular evolution and survival.
Collapse
Affiliation(s)
- Haitao Sun
- Department of Cancer Genetics and Epigenetics, Beckman Research Institute, City of Hope, 1500 East Duarte Road, Duarte, CA 91010
| | - Zhaoning Lu
- Department of Cancer Genetics and Epigenetics, Beckman Research Institute, City of Hope, 1500 East Duarte Road, Duarte, CA 91010
| | - Amanpreet Singh
- Department of Cancer Genetics and Epigenetics, Beckman Research Institute, City of Hope, 1500 East Duarte Road, Duarte, CA 91010
| | - Yajing Zhou
- Department of Cancer Genetics and Epigenetics, Beckman Research Institute, City of Hope, 1500 East Duarte Road, Duarte, CA 91010
| | - Eric Zheng
- Department of Cancer Genetics and Epigenetics, Beckman Research Institute, City of Hope, 1500 East Duarte Road, Duarte, CA 91010
- Department of Molecular, Cellular, and Developmental Biology, University of California at Santa Barbara, Santa Barbara, CA 93106
| | - Mian Zhou
- Department of Cancer Genetics and Epigenetics, Beckman Research Institute, City of Hope, 1500 East Duarte Road, Duarte, CA 91010
| | - Jinhui Wang
- Department of Molecular and Cellular Biology, Beckman Research Institute, City of Hope, 1500 East Duarte Road, Duarte, CA 91010
| | - Xiwei Wu
- Department of Molecular and Cellular Biology, Beckman Research Institute, City of Hope, 1500 East Duarte Road, Duarte, CA 91010
| | - Zunsong Hu
- Department of Computational and Quantitative Medicine, Beckman Research Institute, City of Hope, 1500 East Duarte Road, Duarte, CA 91010
| | - Zhaohui Gu
- Department of Computational and Quantitative Medicine, Beckman Research Institute, City of Hope, 1500 East Duarte Road, Duarte, CA 91010
| | - Judith L. Campbell
- Divisions of Chemistry and Chemical Engineering and Biology and Biological Engineering California Institute of Technology, Pasadena, CA 91125, USA
| | - Li Zheng
- Department of Cancer Genetics and Epigenetics, Beckman Research Institute, City of Hope, 1500 East Duarte Road, Duarte, CA 91010
| | - Binghui Shen
- Department of Cancer Genetics and Epigenetics, Beckman Research Institute, City of Hope, 1500 East Duarte Road, Duarte, CA 91010
| |
Collapse
|
239
|
Song TY, Long M, Zhao HX, Zou MW, Fan HJ, Liu Y, Geng CL, Song MF, Liu YF, Chen JY, Yang YL, Zhou WR, Huang DW, Peng B, Peng ZG, Cang Y. Tumor evolution selectively inactivates the core microRNA machinery for immune evasion. Nat Commun 2021; 12:7003. [PMID: 34853298 PMCID: PMC8636623 DOI: 10.1038/s41467-021-27331-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Accepted: 11/15/2021] [Indexed: 12/13/2022] Open
Abstract
Cancer cells acquire genetic heterogeneity to escape from immune surveillance during tumor evolution, but a systematic approach to distinguish driver from passenger mutations is lacking. Here we investigate the impact of different immune pressure on tumor clonal dynamics and immune evasion mechanism, by combining massive parallel sequencing of immune edited tumors and CRISPR library screens in syngeneic mouse tumor model and co-culture system. We find that the core microRNA (miRNA) biogenesis and targeting machinery maintains the sensitivity of cancer cells to PD-1-independent T cell-mediated cytotoxicity. Genetic inactivation of the machinery or re-introduction of ANKRD52 frequent patient mutations dampens the JAK-STAT-interferon-γ signaling and antigen presentation in cancer cells, largely by abolishing miR-155-targeted silencing of suppressor of cytokine signaling 1 (SOCS1). Expression of each miRNA machinery component strongly correlates with intratumoral T cell infiltration in nearly all human cancer types. Our data indicate that the evolutionarily conserved miRNA pathway can be exploited by cancer cells to escape from T cell-mediated elimination and immunotherapy.
Collapse
Affiliation(s)
- Tian-Yu Song
- Gene Editing Center, School of Life Science and Technology, ShanghaiTech University, Shanghai, China
- Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
- Life Sciences Institute, Zhejiang University, Hangzhou, Zhejiang, China
| | - Min Long
- Gene Editing Center, School of Life Science and Technology, ShanghaiTech University, Shanghai, China
- Life Sciences Institute, Zhejiang University, Hangzhou, Zhejiang, China
| | - Hai-Xin Zhao
- Oncology and Immunology Unit, WuXi Biology, WuXi AppTec (Shanghai) Co, Ltd, Shanghai, China
| | - Miao-Wen Zou
- Gene Editing Center, School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Hong-Jie Fan
- Gene Editing Center, School of Life Science and Technology, ShanghaiTech University, Shanghai, China
- University of Chinese Academy of Sciences, 100049, Beijing, China
| | - Yang Liu
- Gene Editing Center, School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Chen-Lu Geng
- Gene Editing Center, School of Life Science and Technology, ShanghaiTech University, Shanghai, China
- Life Sciences Institute, Zhejiang University, Hangzhou, Zhejiang, China
| | - Min-Fang Song
- Gene Editing Center, School of Life Science and Technology, ShanghaiTech University, Shanghai, China
- Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, 100049, Beijing, China
| | - Yu-Feng Liu
- Gene Editing Center, School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Jun-Yi Chen
- Gene Editing Center, School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Yu-Lin Yang
- Oncology and Immunology Unit, WuXi Biology, WuXi AppTec (Shanghai) Co, Ltd, Shanghai, China
| | - Wen-Rong Zhou
- Oncology and Immunology Unit, WuXi Biology, WuXi AppTec (Shanghai) Co, Ltd, Shanghai, China
| | - Da-Wei Huang
- Oncology and Immunology Unit, WuXi Biology, WuXi AppTec (Shanghai) Co, Ltd, Shanghai, China
| | - Bo Peng
- Gene Editing Center, School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Zhen-Gang Peng
- Oncology and Immunology Unit, WuXi Biology, WuXi AppTec (Shanghai) Co, Ltd, Shanghai, China
| | - Yong Cang
- Gene Editing Center, School of Life Science and Technology, ShanghaiTech University, Shanghai, China.
| |
Collapse
|
240
|
Wallach T, Mossmann ZJ, Szczepek M, Wetzel M, Machado R, Raden M, Miladi M, Kleinau G, Krüger C, Dembny P, Adler D, Zhai Y, Kumbol V, Dzaye O, Schüler J, Futschik M, Backofen R, Scheerer P, Lehnardt S. MicroRNA-100-5p and microRNA-298-5p released from apoptotic cortical neurons are endogenous Toll-like receptor 7/8 ligands that contribute to neurodegeneration. Mol Neurodegener 2021; 16:80. [PMID: 34838071 PMCID: PMC8626928 DOI: 10.1186/s13024-021-00498-5] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Accepted: 11/03/2021] [Indexed: 12/30/2022] Open
Abstract
Background MicroRNA (miRNA) expression in the brain is altered in neurodegenerative diseases. Recent studies demonstrated that selected miRNAs conventionally regulating gene expression at the post-transcriptional level can act extracellularly as signaling molecules. The identity of miRNA species serving as membrane receptor ligands involved in neuronal apoptosis in the central nervous system (CNS), as well as the miRNAs’ sequence and structure required for this mode of action remained largely unresolved. Methods Using a microarray-based screening approach we analyzed apoptotic cortical neurons of C56BL/6 mice and their supernatant with respect to alterations in miRNA expression/presence. HEK-Blue Toll-like receptor (TLR) 7/8 reporter cells, primary microglia and macrophages derived from human and mouse were employed to test the potential of the identified miRNAs released from apoptotic neurons to serve as signaling molecules for the RNA-sensing receptors. Biophysical and bioinformatical approaches, as well as immunoassays and sequential microscopy were used to analyze the interaction between candidate miRNA and TLR. Immunocytochemical and -histochemical analyses of murine CNS cultures and adult mice intrathecally injected with miRNAs, respectively, were performed to evaluate the impact of miRNA-induced TLR activation on neuronal survival and microglial activation. Results We identified a specific pattern of miRNAs released from apoptotic cortical neurons that activate TLR7 and/or TLR8, depending on sequence and species. Exposure of microglia and macrophages to certain miRNA classes released from apoptotic neurons resulted in the sequence-specific production of distinct cytokines/chemokines and increased phagocytic activity. Out of those miRNAs miR-100-5p and miR-298-5p, which have consistently been linked to neurodegenerative diseases, entered microglia, located to their endosomes, and directly bound to human TLR8. The miRNA-TLR interaction required novel sequence features, but no specific structure formation of mature miRNA. As a consequence of miR-100-5p- and miR-298-5p-induced TLR activation, cortical neurons underwent cell-autonomous apoptosis. Presence of miR-100-5p and miR-298-5p in cerebrospinal fluid led to neurodegeneration and microglial accumulation in the murine cerebral cortex through TLR7 signaling. Conclusion Our data demonstrate that specific miRNAs are released from apoptotic cortical neurons, serve as endogenous TLR7/8 ligands, and thereby trigger further neuronal apoptosis in the CNS. Our findings underline the recently discovered role of miRNAs as extracellular signaling molecules, particularly in the context of neurodegeneration. Supplementary Information The online version contains supplementary material available at 10.1186/s13024-021-00498-5.
Collapse
Affiliation(s)
- Thomas Wallach
- Institute of Cell Biology and Neurobiology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117, Berlin, Germany.
| | - Zoé J Mossmann
- Institute of Cell Biology and Neurobiology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117, Berlin, Germany
| | - Michal Szczepek
- Institute for Medical Physics and Biophysics, Group Protein X-ray Crystallography & Signal Transduction, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117, Berlin, Germany
| | - Max Wetzel
- Institute of Cell Biology and Neurobiology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117, Berlin, Germany
| | - Rui Machado
- Department of Biomedical Sciences and Medicine, University of Algarve, 8005-139, Faro, Portugal
| | - Martin Raden
- Bioinformatics, Department of Computer Science, Albert-Ludwigs-University Freiburg, Freiburg, Germany
| | - Milad Miladi
- Bioinformatics, Department of Computer Science, Albert-Ludwigs-University Freiburg, Freiburg, Germany
| | - Gunnar Kleinau
- Institute for Medical Physics and Biophysics, Group Protein X-ray Crystallography & Signal Transduction, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117, Berlin, Germany
| | - Christina Krüger
- Institute of Cell Biology and Neurobiology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117, Berlin, Germany
| | - Paul Dembny
- Institute of Cell Biology and Neurobiology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117, Berlin, Germany
| | - Drew Adler
- Institute of Cell Biology and Neurobiology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117, Berlin, Germany
| | - Yuanyuan Zhai
- Institute of Cell Biology and Neurobiology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117, Berlin, Germany
| | - Victor Kumbol
- Institute of Cell Biology and Neurobiology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117, Berlin, Germany
| | - Omar Dzaye
- Department of Radiology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117, Berlin, Germany
| | - Jutta Schüler
- Institute of Cell Biology and Neurobiology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117, Berlin, Germany
| | - Matthias Futschik
- Department of Biomedical Sciences and Medicine, University of Algarve, 8005-139, Faro, Portugal.,School of Biomedical Sciences, Faculty of Health, University of Plymouth, Plymouth, PL6 8BU, UK.,MRC London Institute of Medical Sciences (LMS), Faculty of Medicine, Imperial College London, London, W12 0NN, UK
| | - Rolf Backofen
- Bioinformatics, Department of Computer Science, Albert-Ludwigs-University Freiburg, Freiburg, Germany
| | - Patrick Scheerer
- Institute for Medical Physics and Biophysics, Group Protein X-ray Crystallography & Signal Transduction, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117, Berlin, Germany.,German Centre for Cardiovascular Research, partner site Berlin, Berlin, Germany
| | - Seija Lehnardt
- Institute of Cell Biology and Neurobiology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117, Berlin, Germany. .,Department of Neurology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117, Berlin, Germany.
| |
Collapse
|
241
|
Aghaieabiane N, Koutis I. A Novel Calibration Step in Gene Co-Expression Network Construction. FRONTIERS IN BIOINFORMATICS 2021; 1:704817. [PMID: 36303738 PMCID: PMC9581019 DOI: 10.3389/fbinf.2021.704817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Accepted: 10/22/2021] [Indexed: 12/02/2022] Open
Abstract
High-throughput technologies such as DNA microarrays and RNA-sequencing are used to measure the expression levels of large numbers of genes simultaneously. To support the extraction of biological knowledge, individual gene expression levels are transformed to Gene Co-expression Networks (GCNs). In a GCN, nodes correspond to genes, and the weight of the connection between two nodes is a measure of similarity in the expression behavior of the two genes. In general, GCN construction and analysis includes three steps; 1) calculating a similarity value for each pair of genes 2) using these similarity values to construct a fully connected weighted network 3) finding clusters of genes in the network, commonly called modules. The specific implementation of these three steps can significantly impact the final output and the downstream biological analysis. GCN construction is a well-studied topic. Existing algorithms rely on relatively simple statistical and mathematical tools to implement these steps. Currently, software package WGCNA appears to be the most widely accepted standard. We hypothesize that the raw features provided by sequencing data can be leveraged to extract modules of higher quality. A novel preprocessing step of the gene expression data set is introduced that in effect calibrates the expression levels of individual genes, before computing pairwise similarities. Further, the similarity is computed as an inner-product of positive vectors. In experiments, this provides a significant improvement over WGCNA, as measured by aggregate p-values of the gene ontology term enrichment of the computed modules.
Collapse
|
242
|
Saha B, Chhatriya B, Pramanick S, Goswami S. Bioinformatic Analysis and Integration of Transcriptome and Proteome Results Identify Key Coding and Noncoding Genes Predicting Malignancy in Intraductal Papillary Mucinous Neoplasms of the Pancreas. BIOMED RESEARCH INTERNATIONAL 2021; 2021:1056622. [PMID: 34790815 PMCID: PMC8592698 DOI: 10.1155/2021/1056622] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/10/2021] [Revised: 10/07/2021] [Accepted: 10/21/2021] [Indexed: 12/15/2022]
Abstract
BACKGROUND Intraductal papillary mucinous neoplasms (IPMNs) are precursor lesions of pancreatic ductal adenocarcinoma (PDAC). IPMNs are generally associated with high risk of developing malignancy and therefore need to be diagnosed and assessed accurately, once detected. Existing diagnostic methods are inadequate, and identification of efficient biomarker capable of detecting high-risk IPMNs is necessitated. Moreover, the mechanism of development of malignancy in IPMNs is also elusive. METHODS Gene expression meta-analysis conducted using 12 low-risk IPMN and 23 high-risk IPMN tissue samples. We have also listed all the altered miRNAs and long noncoding RNAs (lncRNAs), identified their target genes, and performed pathway analysis. We further enlisted cyst fluid proteins detected to be altered in high-risk or malignant IPMNs and compared them with fraction of differentially expressed genes secreted into cyst fluid. RESULTS Our meta-analysis identified 270 upregulated and 161 downregulated genes characteristically altered in high-risk IPMNs. We further identified 61 miRNAs and 14 lncRNAs and their target genes and key pathways contributing towards understanding of the gene regulation during the progression of the disease. Most importantly, we have detected 12 genes altered significantly both in cystic lesions and cyst fluid. CONCLUSION Our study reports, for the first time, a meta-analysis identifying key changes in gene expression between low-risk and high-risk IPMNs and also explains the regulatory aspect through construction of a miRNA-lncRNA-mRNA interaction network. The 12-gene-signature could function as potential biomarker in cyst fluid for detection of IPMN with a high risk of developing malignancy.
Collapse
Affiliation(s)
- Barsha Saha
- National Institute of Biomedical Genomics, Kalyani, West Bengal, India
| | | | | | - Srikanta Goswami
- National Institute of Biomedical Genomics, Kalyani, West Bengal, India
| |
Collapse
|
243
|
Gene Expression Signature Associated with Clinical Outcome in ALK-Positive Anaplastic Large Cell Lymphoma. Cancers (Basel) 2021; 13:cancers13215523. [PMID: 34771686 PMCID: PMC8582782 DOI: 10.3390/cancers13215523] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 10/15/2021] [Accepted: 10/28/2021] [Indexed: 11/17/2022] Open
Abstract
Simple Summary Anaplastic large cell lymphomas associated with ALK translocation have a good outcome after CHOP treatment; however, the 2-year relapse rate remains at 30%. Microarray gene-expression profiling, high throughput RT-qPCR, and RNA sequencing of 48 ALK-positive anaplastic large cell lymphoma (ALK+ ALCL) samples obtained at diagnosis enable the identification of genes associated with clinical outcome. More particularly, our molecular signatures indicate that the FN1 gene, a matrix key regulator, might also be involved in the prognosis and the therapeutic response in anaplastic lymphomas. Abstract Anaplastic large cell lymphomas associated with ALK translocation have a good outcome after CHOP treatment; however, the 2-year relapse rate remains at 30%. Microarray gene-expression profiling of 48 samples obtained at diagnosis was used to identify 47 genes that were differentially expressed between patients with early relapse/progression and no relapse. In the relapsing group, the most significant overrepresented genes were related to the regulation of the immune response and T-cell activation while those in the non-relapsing group were involved in the extracellular matrix. Fluidigm technology gave concordant results for 29 genes, of which FN1, FAM179A, and SLC40A1 had the strongest predictive power after logistic regression and two classification algorithms. In parallel with 39 samples, we used a Kallisto/Sleuth pipeline to analyze RNA sequencing data and identified 20 genes common to the 28 genes validated by Fluidigm technology—notably, the FAM179A and FN1 genes. Interestingly, FN1 also belongs to the gene signature predicting longer survival in diffuse large B-cell lymphomas treated with CHOP. Thus, our molecular signatures indicate that the FN1 gene, a matrix key regulator, might also be involved in the prognosis and the therapeutic response in anaplastic lymphomas.
Collapse
|
244
|
Han Z, Yang T, Guo Y, Cui WH, Yao LJ, Li G, Wu AM, Li JH, Liu LJ. The transcription factor PagLBD3 contributes to the regulation of secondary growth in Populus. JOURNAL OF EXPERIMENTAL BOTANY 2021; 72:7092-7106. [PMID: 34313722 DOI: 10.1093/jxb/erab351] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Accepted: 07/24/2021] [Indexed: 06/13/2023]
Abstract
LATERAL ORGAN BOUNDARIES DOMAIN (LBD) genes encode plant-specific transcription factors that participate in regulating various developmental processes. In this study, we genetically characterized PagLBD3 encoding an important regulator of secondary growth in poplar (Populus alba × Populus glandulosa). Overexpression of PagLBD3 increased stem secondary growth in Populus with a significantly higher rate of cambial cell differentiation into phloem, while dominant repression of PagLBD3 significantly decreased the rate of cambial cell differentiation into phloem. Furthermore, we identified 1756 PagLBD3 genome-wide putative direct target genes (DTGs) through RNA sequencing (RNA-seq)-coupled DNA affinity purification followed by sequencing (DAP-seq) assays. Gene Ontology analysis revealed that genes regulated by PagLBD3 were enriched in biological pathways regulating meristem development, xylem development, and auxin transport. Several central regulator genes for vascular development, including PHLOEM INTERCALATED WITH XYLEM (PXY), WUSCHEL RELATED HOMEOBOX4 (WOX4), Secondary Wall-Associated NAC Domain 1s (SND1-B2), and Vascular-Related NAC-Domain 6s (VND6-B1), were identified as PagLBD3 DTGs. Together, our results indicate that PagLBD3 and its DTGs form a complex transcriptional network to modulate cambium activity and phloem/xylem differentiation.
Collapse
Affiliation(s)
- Zhen Han
- College of Forestry, State Forestry and Grassland Administration Key Laboratory of Silviculture in downstream areas of the Yellow River, Shandong Agriculture University, Taian, Shandong 271018, China
| | - Tong Yang
- College of Forestry, State Forestry and Grassland Administration Key Laboratory of Silviculture in downstream areas of the Yellow River, Shandong Agriculture University, Taian, Shandong 271018, China
| | - Ying Guo
- College of Forestry, State Forestry and Grassland Administration Key Laboratory of Silviculture in downstream areas of the Yellow River, Shandong Agriculture University, Taian, Shandong 271018, China
| | - Wen-Hui Cui
- College of Forestry, State Forestry and Grassland Administration Key Laboratory of Silviculture in downstream areas of the Yellow River, Shandong Agriculture University, Taian, Shandong 271018, China
| | - Li-Juan Yao
- College of Forestry, State Forestry and Grassland Administration Key Laboratory of Silviculture in downstream areas of the Yellow River, Shandong Agriculture University, Taian, Shandong 271018, China
| | - Gang Li
- College of Life Science, State Key Laboratory of Crop Biology, Shandong Agriculture University, Taian, Shandong 271018, China
| | - Ai-Min Wu
- Guangdong Key Laboratory for Innovative Development and Utilization of Forest Plant Germplasm, College of Forestry and Landscape Architecture, South China Agricultural University, Guangzhou 510642, China
| | - Ji-Hong Li
- College of Forestry, State Forestry and Grassland Administration Key Laboratory of Silviculture in downstream areas of the Yellow River, Shandong Agriculture University, Taian, Shandong 271018, China
| | - Li-Jun Liu
- College of Forestry, State Forestry and Grassland Administration Key Laboratory of Silviculture in downstream areas of the Yellow River, Shandong Agriculture University, Taian, Shandong 271018, China
| |
Collapse
|
245
|
Gomez-Lopez N, Romero R, Galaz J, Bhatti G, Done B, Miller D, Ghita C, Motomura K, Farias-Jofre M, Jung E, Pique-Regi R, Hassan SS, Chaiworapongsa T, Tarca AL. Transcriptome changes in maternal peripheral blood during term parturition mimic perturbations preceding spontaneous Preterm birth†. Biol Reprod 2021; 106:185-199. [PMID: 34686873 DOI: 10.1093/biolre/ioab197] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 09/30/2021] [Accepted: 10/20/2021] [Indexed: 11/14/2022] Open
Abstract
The complex physiologic process of parturition includes the onset of labor, which requires the orchestrated stimulation of a common pathway involving uterine contractility, cervical ripening, and chorioamniotic membrane activation. However, the labor-specific processes taking place in these tissues have limited use as predictive biomarkers unless they can be probed in non-invasive samples, such as the peripheral blood. Herein, we utilized a transcriptomic dataset to assess labor-specific changes in the peripheral blood of women who delivered at term. We identified a set of genes that were differentially expressed with labor and enriched for immunological processes, and these gene expression changes were strongly correlated with results from prior studies, providing in silico validation of our findings. We then identified significant correlations between labor-specific transcriptomic changes in the maternal circulation and those reported in the chorioamniotic membranes, myometrium, and cervix of women at term, demonstrating that tissue-specific labor signatures are partly mirrored in the peripheral blood. Last, we demonstrated a significant overlap between the peripheral blood transcriptomic changes in term parturition and those observed in asymptomatic women prior to the diagnosis of preterm prelabor rupture of membranes who delivered preterm. Collectively, we provide evidence that the normal process of labor at term is characterized by a unique immunological expression signature, which may serve as a useful tool for assessing labor status and potentially identifying women at risk for preterm birth.
Collapse
Affiliation(s)
- Nardhy Gomez-Lopez
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS); Bethesda, MD, and Detroit, MI, USA.,Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA.,Department of Biochemistry, Microbiology and Immunology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Roberto Romero
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS); Bethesda, MD, and Detroit, MI, USA.,Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA.,Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI, USA.,Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, MI, USA.,Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI, USA.,Detroit Medical Center, Detroit, MI, USA.,Department of Computer Science, Wayne State University College of Engineering, Detroit, MI, USA
| | - Jose Galaz
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS); Bethesda, MD, and Detroit, MI, USA.,Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Gaurav Bhatti
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS); Bethesda, MD, and Detroit, MI, USA.,Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Bogdan Done
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS); Bethesda, MD, and Detroit, MI, USA.,Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Derek Miller
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS); Bethesda, MD, and Detroit, MI, USA.,Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Corina Ghita
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS); Bethesda, MD, and Detroit, MI, USA
| | - Kenichiro Motomura
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS); Bethesda, MD, and Detroit, MI, USA.,Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Marcelo Farias-Jofre
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS); Bethesda, MD, and Detroit, MI, USA.,Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Eunjung Jung
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS); Bethesda, MD, and Detroit, MI, USA.,Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Roger Pique-Regi
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS); Bethesda, MD, and Detroit, MI, USA.,Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI, USA
| | - Sonia S Hassan
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Tinnakorn Chaiworapongsa
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS); Bethesda, MD, and Detroit, MI, USA.,Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Adi L Tarca
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS); Bethesda, MD, and Detroit, MI, USA.,Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA.,Department of Computer Science, Wayne State University College of Engineering, Detroit, MI, USA
| |
Collapse
|
246
|
Simon S, Breeschoten T, Jansen HJ, Dirks RP, Schranz ME, Ros VID. Genome and transcriptome analysis of the beet armyworm Spodoptera exigua reveals targets for pest control. G3 (BETHESDA, MD.) 2021; 11:jkab311. [PMID: 34557910 PMCID: PMC8527508 DOI: 10.1093/g3journal/jkab311] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Accepted: 08/19/2021] [Indexed: 12/13/2022]
Abstract
The genus Spodoptera (Lepidoptera: Noctuidae) includes some of the most infamous insect pests of cultivated plants including Spodoptera frugiperda, Spodoptera litura, and Spodoptera exigua. To effectively develop targeted pest control strategies for diverse Spodoptera species, genomic resources are highly desired. To this aim, we provide the genome assembly and developmental transcriptome comprising all major life stages of S. exigua, the beet armyworm. Spodoptera exigua is a polyphagous herbivore that can feed on > 130 host plants, including several economically important crops. The 419 Mb beet armyworm genome was sequenced from a female S. exigua pupa. Using a hybrid genome sequencing approach (Nanopore long-read data and Illumina short read), a high-quality genome assembly was achieved (N50 = 1.1 Mb). An official gene set (18,477 transcripts) was generated by automatic annotation and by using transcriptomic RNA-seq datasets of 18 S. exigua samples as supporting evidence. In-depth analyses of developmental stage-specific expression combined with gene tree analyses of identified homologous genes across Lepidoptera genomes revealed four potential genes of interest (three of them Spodoptera-specific) upregulated during first- and third-instar larval stages for targeted pest-outbreak management. The beet armyworm genome sequence and developmental transcriptome covering all major developmental stages provide critical insights into the biology of this devastating polyphagous insect pest species worldwide. In addition, comparative genomic analyses across Lepidoptera significantly advance our knowledge to further control other invasive Spodoptera species and reveals potential lineage-specific target genes for pest control strategies.
Collapse
Affiliation(s)
- Sabrina Simon
- Biosystematics Group, Wageningen University & Research, 6708 PB Wageningen, The Netherlands
| | - Thijmen Breeschoten
- Biosystematics Group, Wageningen University & Research, 6708 PB Wageningen, The Netherlands
| | - Hans J Jansen
- Future Genomics Technologies, Leiden, The Netherlands
| | - Ron P Dirks
- Future Genomics Technologies, Leiden, The Netherlands
| | - M Eric Schranz
- Biosystematics Group, Wageningen University & Research, 6708 PB Wageningen, The Netherlands
| | - Vera I D Ros
- Laboratory of Virology, Wageningen University & Research, 6708 PB Wageningen, The Netherlands
| |
Collapse
|
247
|
Wruck W, Graffmann N, Spitzhorn LS, Adjaye J. Human Induced Pluripotent Stem Cell-Derived Mesenchymal Stem Cells Acquire Rejuvenation and Reduced Heterogeneity. Front Cell Dev Biol 2021; 9:717772. [PMID: 34604216 PMCID: PMC8481886 DOI: 10.3389/fcell.2021.717772] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Accepted: 08/17/2021] [Indexed: 12/20/2022] Open
Abstract
Despite the uniform selection criteria for the isolation of human mesenchymal stem cells (MSCs), considerable heterogeneity exists which reflects the distinct tissue origins and differences between individuals with respect to their genetic background and age. This heterogeneity is manifested by the variabilities seen in the transcriptomes, proteomes, secretomes, and epigenomes of tissue-specific MSCs. Here, we review literature on different aspects of MSC heterogeneity including the role of epigenetics and the impact of MSC heterogeneity on therapies. We then combine this with a meta-analysis of transcriptome data from distinct MSC subpopulations derived from bone marrow, adipose tissue, cruciate, tonsil, kidney, umbilical cord, fetus, and induced pluripotent stem cells derived MSCs (iMSCs). Beyond that, we investigate transcriptome differences between tissue-specific MSCs and pluripotent stem cells. Our meta-analysis of numerous MSC-related data sets revealed markers and associated biological processes characterizing the heterogeneity and the common features of MSCs from various tissues. We found that this heterogeneity is mainly related to the origin of the MSCs and infer that microenvironment and epigenetics are key drivers. The epigenomes of MSCs alter with age and this has a profound impact on their differentiation capabilities. Epigenetic modifications of MSCs are propagated during cell divisions and manifest in differentiated cells, thus contributing to diseased or healthy phenotypes of the respective tissue. An approach used to reduce heterogeneity caused by age- and tissue-related epigenetic and microenvironmental patterns is the iMSC concept: iMSCs are MSCs generated from induced pluripotent stem cells (iPSCs). During iMSC generation epigenetic and chromatin remodeling result in a gene expression pattern associated with rejuvenation thus allowing to overcome age-related shortcomings (e.g., limited differentiation and proliferation capacity). The importance of the iMSC concept is underlined by multiple clinical trials. In conclusion, we propose the use of rejuvenated iMSCs to bypass tissue- and age-related heterogeneity which are associated with native MSCs.
Collapse
Affiliation(s)
- Wasco Wruck
- Medical Faculty, Institute for Stem Cell Research and Regenerative Medicine, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Nina Graffmann
- Medical Faculty, Institute for Stem Cell Research and Regenerative Medicine, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Lucas-Sebastian Spitzhorn
- Medical Faculty, Institute for Stem Cell Research and Regenerative Medicine, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - James Adjaye
- Medical Faculty, Institute for Stem Cell Research and Regenerative Medicine, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| |
Collapse
|
248
|
Randhawa V, Kumar M. An integrated network analysis approach to identify potential key genes, transcription factors, and microRNAs regulating human hematopoietic stem cell aging. Mol Omics 2021; 17:967-984. [PMID: 34605522 DOI: 10.1039/d1mo00199j] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Hematopoietic stem cells (HSCs) undergo functional deterioration with increasing age that causes loss of their self-renewal and regenerative potential. Despite various efforts, significant success in identifying molecular regulators of HSC aging has not been achieved, one prime reason being the non-availability of appropriate human HSC samples. To demonstrate the scope of integrating and re-analyzing the HSC transcriptomics data available, we used existing tools and databases to structure a sequential data analysis pipeline to predict potential candidate genes, transcription factors, and microRNAs simultaneously. This sequential approach comprises (i) collecting matched young and aged mice HSC sample datasets, (ii) identifying differentially expressed genes, (iii) identifying human homologs of differentially expressed genes, (iv) inferring gene co-expression network modules, and (v) inferring the microRNA-transcription factor-gene regulatory network. Systems-level analyses of HSC interaction networks provided various insights based on which several candidates were predicted. For example, 16 HSC aging-related candidate genes were predicted (e.g., CD38, BRCA1, AGTR1, GSTM1, etc.) from GCN analysis. Following this, the shortest path distance-based analyses of the regulatory network predicted several novel candidate miRNAs and TFs. Among these, miR-124-3p was a common regulator in candidate gene modules, while TFs MYC and SP1 were identified to regulate various candidate genes. Based on the regulatory interactions among candidate genes, TFs, and miRNAs, a potential regulation model of biological processes in each of the candidate modules was predicted, which provided systems-level insights into the molecular complexity of each module to regulate HSC aging.
Collapse
Affiliation(s)
- Vinay Randhawa
- Virology Unit and Bioinformatics Centre, Institute of Microbial Technology, Council of Scientific & Industrial Research, Chandigarh-160036, India.
| | - Manoj Kumar
- Virology Unit and Bioinformatics Centre, Institute of Microbial Technology, Council of Scientific & Industrial Research, Chandigarh-160036, India. .,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad-201002, India
| |
Collapse
|
249
|
Guo Q, Liu Z, Zheng J, Zhao H, Li C. Substances for regenerative wound healing during antler renewal stimulated scar-less restoration of rat cutaneous wounds. Cell Tissue Res 2021; 386:99-116. [PMID: 34390408 DOI: 10.1007/s00441-021-03505-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 07/07/2021] [Indexed: 10/20/2022]
Abstract
Scarification is the outcome of cutaneous wound healing under normal conditions. Although considerable effort has been expended in this field, scar-less healing has not been achieved satisfactorily. The lack of a good model of scar-free healing has contributed to this undesirable situation. However, the annual regeneration of deer antlers, which starts from regenerative wound healing over the top of the pedicles (permanent bony protuberances), may provide such a model. Therefore, in this study, we investigated the process of pedicle wound healing at the organ, tissue, cell, and molecular levels. Our results convincingly demonstrate that wounds over the pedicle preceded a regenerative healing process including regeneration of skin appendages, such as hair follicles. Compared to the scar healing in rats, regenerative healing of the pedicle wound exhibited a weaker inflammatory response, lack of myofibroblast induction, and higher ratios of Col III/Col I, TGF-β3/TGF-β1, and MMP/TIMP. Importantly, our periosteal transplantation experiments in vivo revealed that this regenerative healing process was achieved through induction of antler stem cells (ASCs). Further study showed that this effect of ASCs on regenerative healing was not species-specific but more generic and could be applied to other mammalian species, as injection of ASCs stimulated regenerative healing of full-thickness excisional cutaneous wounds in rats. Overall, our findings show that ASCs may have therapeutic potential in enhancing the quality of wound healing and preventing scar formation in clinical settings.
Collapse
Affiliation(s)
- Qianqian Guo
- Institute of Special Animal and Plant Sciences, Chinese Academy of Agricultural Sciences (CAAS), Changchun, Jilin, 130112, China
| | - Zhen Liu
- Institute of Special Animal and Plant Sciences, Chinese Academy of Agricultural Sciences (CAAS), Changchun, Jilin, 130112, China
| | - Junjun Zheng
- Institute of Special Animal and Plant Sciences, Chinese Academy of Agricultural Sciences (CAAS), Changchun, Jilin, 130112, China
| | - Haiping Zhao
- Institute of Special Animal and Plant Sciences, Chinese Academy of Agricultural Sciences (CAAS), Changchun, Jilin, 130112, China.
| | - Chunyi Li
- Institute of Antler Science and Product Technology, Changchun Sci-Tech University, Changchun, Jilin, 130600, China.
| |
Collapse
|
250
|
Akizawa H, Saito S, Kohri N, Furukawa E, Hayashi Y, Bai H, Nagano M, Yanagawa Y, Tsukahara H, Takahashi M, Kagawa S, Kawahara-Miki R, Kobayashi H, Kono T, Kawahara M. Deciphering two rounds of cell lineage segregations during bovine preimplantation development. FASEB J 2021; 35:e21904. [PMID: 34569650 DOI: 10.1096/fj.202002762rr] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Revised: 08/18/2021] [Accepted: 08/23/2021] [Indexed: 12/12/2022]
Abstract
Blastocyst formation gives rise to the inner cell mass (ICM) and trophectoderm (TE) and is followed by the differentiation of the epiblast (Epi) and primitive endoderm (PrE) within the ICM. Although these two-round cell lineage differentiations underpin proper embryogenesis in every mammal, their spatiotemporal dynamics are quite diverse among species. Here, molecular details of the blastocyst stage in cattle were dissected using an optimized in vitro culture method. Blastocyst embryos were placed on agarose gel filled with nutrient-rich media to expose embryos to both gaseous and liquid phases. Embryos derived from this "on-gel" culture were transferred to surrogate mothers on day (D) 10 after fertilization and successfully implanted. Immunofluorescent studies using on-gel-cultured embryos revealed that the proportion of TE cells expressing the pluripotent ICM marker, OCT4, which was beyond 80% on D8, was rapidly reduced after D9 and reached 0% on D9.5. This first lineage segregation process was temporally parallel with the second one, identified by the spatial separation of Epi cells expressing SOX2 and PrE cells expressing SOX17. RNA-seq comparison of TE cells from D8 in vitro fertilized embryos and D14 in vivo embryos revealed that besides drastic reduction of pluripotency-related genes, TE cells highly expressed Wnt, FGF, and VEGF signaling pathways-related genes to facilitate the functional maturation required for feto-maternal interaction. Quantitative PCR analysis of TE cells derived from on-gel culture further confirmed time-dependent increments in the expression of key TE markers. Altogether, the present study provides platforms to understand species-specific strategies for mammalian preimplantation development.
Collapse
Affiliation(s)
- Hiroki Akizawa
- Laboratory of Animal Genetics and Reproduction, Research Faculty of Agriculture, Hokkaido University, Sapporo, Japan
| | - Shun Saito
- Laboratory of Animal Genetics and Reproduction, Research Faculty of Agriculture, Hokkaido University, Sapporo, Japan
| | - Nanami Kohri
- Laboratory of Animal Genetics and Reproduction, Research Faculty of Agriculture, Hokkaido University, Sapporo, Japan
| | - Eri Furukawa
- Laboratory of Theriogenology, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | - Yoshihiro Hayashi
- Laboratory of Animal Genetics and Reproduction, Research Faculty of Agriculture, Hokkaido University, Sapporo, Japan
| | - Hanako Bai
- Laboratory of Animal Genetics and Reproduction, Research Faculty of Agriculture, Hokkaido University, Sapporo, Japan
| | - Masashi Nagano
- Laboratory of Animal Reproduction, Department of Animal Science, School of Veterinary Medicine, Kitasato University, Towada, Japan
| | - Yojiro Yanagawa
- Laboratory of Theriogenology, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | - Hayato Tsukahara
- Laboratory of Animal Genetics and Reproduction, Research Faculty of Agriculture, Hokkaido University, Sapporo, Japan
| | - Masashi Takahashi
- Global Station for Food, Land and Water Resources, Global Institution for Collaborative Research and Education (GI-CoRE), Hokkaido, Japan
| | - Shinjiro Kagawa
- Livestock Research Institute, Aomori Prefectural Industrial Technology Research Center, Aomori, Japan
| | | | - Hisato Kobayashi
- Department of Embryology, Nara Medical University, Kashihara, Japan
| | - Tomohiro Kono
- Department of Bioscience, Tokyo University of Agriculture, Setagaya, Japan
| | - Manabu Kawahara
- Laboratory of Animal Genetics and Reproduction, Research Faculty of Agriculture, Hokkaido University, Sapporo, Japan
| |
Collapse
|