1
|
Zhou X, Guo Z, Pan Y, Yang S, Yan R. PRSS23-eIF4E-c-Myc axis promotes gastric tumorigenesis and progression. Oncogene 2025; 44:1167-1181. [PMID: 39920289 DOI: 10.1038/s41388-025-03294-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 01/09/2025] [Accepted: 01/30/2025] [Indexed: 02/09/2025]
Abstract
Gastric cancer is one of the most common malignant tumors. Our previous study showed that PRSS23 expression is increased in human gastric cancer tissues and PRSS23 knockdown inhibits gastric cancer cell growth. This current study aims to uncover the mechanism underlying PRSS23's involvement in gastric tumorigenesis and progression. We established that PRSS23 influences gastric cancer growth both in vitro and in vivo by modulating the eIF4E-c-Myc axis (eIF4E, p-eIF4E, 4EBP1, p-4EBP1, and c-Myc). Our investigation revealed that PRSS23 interacts with eIF4E via its trypsin domain, while eIF4E binds to PRSS23 through the amino acid residue S209, as confirmed by co-IP and immunofluorescence assays. Multiplexed immunofluorescence assay demonstrated a significant elevation of PRSS23 and p-4EBP1 levels in 232 paired gastric cancer tissues. Moreover, in 49 patients exhibiting relatively high PRSS23 expression, the levels of eIF4E-c-Myc axis-related proteins were increased. Importantly, higher PRSS23 expression correlated significantly with increased lymph node metastasis and advanced clinical staging, leading to poorer patient prognosis. These results highlight the role of upregulated PRSS23 in promoting gastric tumorigenesis and progression by activating the eIF4E-c-Myc axis, underscoring the PRSS23-eIF4E-c-Myc axis as a promising therapeutic target for gastric cancer.
Collapse
Affiliation(s)
- Xiaodong Zhou
- Department of Gastroenterology, Jiangxi Provincial Key Laboratory of Digestive Diseases, Jiangxi Clinical Research Center for Gastroenterology, Digestive Disease Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Zixiang Guo
- Department of Gastroenterology, Jiangxi Provincial Key Laboratory of Digestive Diseases, Jiangxi Clinical Research Center for Gastroenterology, Digestive Disease Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Yating Pan
- Department of Gastroenterology, Jiangxi Provincial Key Laboratory of Digestive Diseases, Jiangxi Clinical Research Center for Gastroenterology, Digestive Disease Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Sicheng Yang
- Department of Gastroenterology, Jiangxi Provincial Key Laboratory of Digestive Diseases, Jiangxi Clinical Research Center for Gastroenterology, Digestive Disease Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Runwei Yan
- Department of Gastroenterology, Jiangxi Provincial Key Laboratory of Digestive Diseases, Jiangxi Clinical Research Center for Gastroenterology, Digestive Disease Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China.
| |
Collapse
|
2
|
Phelps PE, Ha SM, Khankan RR, Mekonnen MA, Juarez G, Ingraham Dixie KL, Chen YW, Yang X. Olfactory ensheathing cells from adult female rats are hybrid glia that promote neural repair. eLife 2025; 13:RP95629. [PMID: 40297980 PMCID: PMC12040321 DOI: 10.7554/elife.95629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/30/2025] Open
Abstract
Olfactory ensheathing cells (OECs) are unique glial cells found in both central and peripheral nervous systems where they support continuous axonal outgrowth of olfactory sensory neurons to their targets. Previously, we reported that following severe spinal cord injury, OECs transplanted near the injury site modify the inhibitory glial scar and facilitate axon regeneration past the scar border and into the lesion. To better understand the mechanisms underlying the reparative properties of OECs, we used single-cell RNA-sequencing of OECs from adult rats to study their gene expression programs. Our analyses revealed five diverse OEC subtypes, each expressing novel marker genes and pathways indicative of progenitor, axonal regeneration, secreted molecules, or microglia-like functions. We found substantial overlap of OEC genes with those of Schwann cells, but also with microglia, astrocytes, and oligodendrocytes. We confirmed established markers on cultured OECs, and localized select top genes of OEC subtypes in olfactory bulb tissue. We also show that OECs secrete Reelin and Connective tissue growth factor, extracellular matrix molecules which are important for neural repair and axonal outgrowth. Our results support that OECs are a unique hybrid glia, some with progenitor characteristics, and that their gene expression patterns indicate functions related to wound healing, injury repair, and axonal regeneration.
Collapse
Affiliation(s)
- Patricia E Phelps
- Department of Integrative Biology and Physiology, UCLALos AngelesUnited States
| | - Sung Min Ha
- Department of Integrative Biology and Physiology, UCLALos AngelesUnited States
| | - Rana R Khankan
- Department of Integrative Biology and Physiology, UCLALos AngelesUnited States
| | - Mahlet A Mekonnen
- Department of Integrative Biology and Physiology, UCLALos AngelesUnited States
| | - Giovanni Juarez
- Department of Integrative Biology and Physiology, UCLALos AngelesUnited States
| | | | - Yen-Wei Chen
- Department of Integrative Biology and Physiology, UCLALos AngelesUnited States
| | - Xia Yang
- Department of Integrative Biology and Physiology, UCLALos AngelesUnited States
| |
Collapse
|
3
|
Yang J, Wang Z, Zhou Y, Jiang S, Qin X, Xu Z, Wang Y, Zuo M, Meng Z, Chen S, Wang Q, Wang J, Sun K. Manic Fringe promotes endothelial-to-mesenchymal transition mediated by the Notch signalling pathway during heart valve development. J Mol Med (Berl) 2025; 103:51-71. [PMID: 39528804 PMCID: PMC11739230 DOI: 10.1007/s00109-024-02492-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 09/15/2024] [Accepted: 09/23/2024] [Indexed: 11/16/2024]
Abstract
A fundamental event in the formation of heart valves involves the transformation of endocardial cells within the outflow tract (OFT) and atrioventricular canal (AVC) cushions through a process known as endothelial-to-mesenchymal transition (EndMT). Aberrant EndMT is a primary cause of congenital valvular malformations. Manic Fringe (MFNG) has been previously associated with cardiovascular development, although its role in heart valve development remains underexplored. In this study, we seek to enhance our understanding of MFNG's involvement in valve formation and its association with EndMT. Staining results of histological section revealed the expression of MFNG in the AVC and OFT from embryonic day 9.5 to 10.5 (E9.5-E10.5), when EndMT takes place. Cellular data demonstrated that MFNG exerts a positive regulatory influence on the EndMT process, promoting endothelial cell (EC) migration by enhancing the activity of the Notch signalling pathway. MFNG knockdown mediated by antisense morpholino oligonucleotides (MO) injection caused abnormal development of the heart and valves in zebrafish. Furthermore, through whole-exome sequencing (WES), we identified a heterozygous MFNG mutation in patients diagnosed with tetralogy of Fallot-pulmonary valve stenosis (TOF-PS). Cellular and molecular assays confirmed that this deleterious mutation reduced MFNG expression and hindered the EndMT process. In summary, our study verifies that MFNG plays a role in promoting EndMT mediated by the Notch signalling pathway during the heart and valve development. The MFNG deleterious variant induces MFNG loss of function, potentially elucidating the underlying molecular mechanisms of MFNG's involvement in the pathogenesis of congenital heart valve defects. These observations contribute to our current genetic understanding of congenital heart valve disease and may provide a potential target for prenatal diagnosis and treatment. KEY MESSAGES: Our examination revealed, for the first time, that MFNG exhibited high expression levels during EndMT of heart valve development in mice. Our findings provide compelling evidence that MFNG plays a role in promoting EndMT mediated by the Notch signalling pathway. Our results identified, for the first time, a deleterious MFNG p. T77M variant that inhibited the EndMT process by downregulating the activity of the Notch signalling pathway, thereby preventing the normal valve formation. MFNG may serve as an early diagnostic marker and an effective therapeutic target for the clinical treatment of congenital heart valve defects.
Collapse
Affiliation(s)
- Junjie Yang
- Department of Pediatric Cardiology, The Second Affiliated Hospital, Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
- Department of Pediatric Cardiology, School of Medicine, Xinhua Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Zhi Wang
- Department of Pediatric Cardiology, School of Medicine, Xinhua Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Yue Zhou
- Department of Pediatric Cardiology, School of Medicine, Xinhua Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Shiwei Jiang
- Department of Pediatric Cardiology, School of Medicine, Xinhua Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Xiji Qin
- Department of Pediatric Cardiology, School of Medicine, Xinhua Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Zhikang Xu
- Department of Pediatric Cardiology, School of Medicine, Xinhua Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Yu Wang
- Department of Pediatric Cardiology, The Second Affiliated Hospital, Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
- Department of Pediatric Cardiology, School of Medicine, Xinhua Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Mengying Zuo
- Department of Pediatric Cardiology, School of Medicine, Xinhua Hospital, Shanghai Jiao Tong University, Shanghai, China
- Department of Cardiology, Children's Hospital of Soochow University, Suzhou, China
| | - Zhuo Meng
- Department of Pediatric Cardiology, School of Medicine, Xinhua Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Sun Chen
- Department of Pediatric Cardiology, School of Medicine, Xinhua Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Qingjie Wang
- Department of Pediatric Cardiology, School of Medicine, Xinhua Hospital, Shanghai Jiao Tong University, Shanghai, China.
| | - Jian Wang
- Department of Pediatric Cardiology, School of Medicine, Xinhua Hospital, Shanghai Jiao Tong University, Shanghai, China.
| | - Kun Sun
- Department of Pediatric Cardiology, The Second Affiliated Hospital, Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China.
- Department of Pediatric Cardiology, School of Medicine, Xinhua Hospital, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
4
|
Tietze E, Barbosa AR, Araujo B, Euclydes V, Spiegelberg B, Cho HJ, Lee YK, Wang Y, McCord A, Lorenzetti A, Feltrin A, van de Leemput J, Di Carlo P, Ursini G, Benjamin KJ, Brentani H, Kleinman JE, Hyde TM, Weinberger DR, McKay R, Shin JH, Sawada T, Paquola ACM, Erwin JA. Human archetypal pluripotent stem cells differentiate into trophoblast stem cells via endogenous BMP5/7 induction without transitioning through naive state. Sci Rep 2024; 14:3291. [PMID: 38332235 PMCID: PMC10853519 DOI: 10.1038/s41598-024-53381-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Accepted: 01/31/2024] [Indexed: 02/10/2024] Open
Abstract
Primary human trophoblast stem cells (TSCs) and TSCs derived from human pluripotent stem cells (hPSCs) can potentially model placental processes in vitro. Yet, the pluripotent states and factors involved in the differentiation of hPSCs to TSCs remain poorly understood. In this study, we demonstrate that the primed pluripotent state can generate TSCs by activating pathways such as Epidermal Growth Factor (EGF) and Wingless-related integration site (WNT), and by suppressing tumor growth factor beta (TGFβ), histone deacetylases (HDAC), and Rho-associated protein kinase (ROCK) signaling pathways, all without the addition of exogenous Bone morphogenetic protein 4 (BMP4)-a condition we refer to as the TS condition. We characterized this process using temporal single-cell RNA sequencing to compare TS conditions with differentiation protocols involving BMP4 activation alone or BMP4 activation in conjunction with WNT inhibition. The TS condition consistently produced a stable, proliferative cell type that closely mimics first-trimester placental cytotrophoblasts, marked by the activation of endogenous retroviral genes and the absence of amnion expression. This was observed across multiple cell lines, including various primed induced pluripotent stem cell (iPSC) and embryonic stem cell (ESC) lines. Primed-derived TSCs can proliferate for over 30 passages and further specify into multinucleated syncytiotrophoblasts and extravillous trophoblast cells. Our research establishes that the differentiation of primed hPSCs to TSC under TS conditions triggers the induction of TMSB4X, BMP5/7, GATA3, and TFAP2A without progressing through a naive state. These findings propose that the primed hPSC state is part of a continuum of potency with the capacity to differentiate into TSCs through multiple routes.
Collapse
Affiliation(s)
- Ethan Tietze
- Lieber Institute for Brain Development, Baltimore, MD, USA
| | - Andre Rocha Barbosa
- Lieber Institute for Brain Development, Baltimore, MD, USA
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Inter-Institutional Graduate Program on Bioinformatics, University of São Paulo, São Paulo, SP, Brazil
| | - Bruno Araujo
- Lieber Institute for Brain Development, Baltimore, MD, USA
| | - Veronica Euclydes
- Lieber Institute for Brain Development, Baltimore, MD, USA
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Department of Psychiatry, University of Sao Paulo, Medical School, São Paulo, Brazil
| | - Bailey Spiegelberg
- Lieber Institute for Brain Development, Baltimore, MD, USA
- Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Hyeon Jin Cho
- Lieber Institute for Brain Development, Baltimore, MD, USA
| | - Yong Kyu Lee
- Lieber Institute for Brain Development, Baltimore, MD, USA
| | - Yanhong Wang
- Lieber Institute for Brain Development, Baltimore, MD, USA
| | | | | | - Arthur Feltrin
- Lieber Institute for Brain Development, Baltimore, MD, USA
- Center for Mathematics, Computation and Cognition, Federal University of ABC, Santo André, SP, Brazil
| | - Joyce van de Leemput
- Lieber Institute for Brain Development, Baltimore, MD, USA
- Center for Precision Disease Modeling and Division of Endocrinology, Diabetes and Nutrition, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Pasquale Di Carlo
- Lieber Institute for Brain Development, Baltimore, MD, USA
- Department of Basic Medical Science, Neuroscience, and Sense Organs, University of Bari Aldo Moro, Bari, Italy
| | - Gianluca Ursini
- Lieber Institute for Brain Development, Baltimore, MD, USA
- Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Kynon J Benjamin
- Lieber Institute for Brain Development, Baltimore, MD, USA
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Department of Psychiatry & Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Helena Brentani
- Inter-Institutional Graduate Program on Bioinformatics, University of São Paulo, São Paulo, SP, Brazil
- Department of Psychiatry, University of Sao Paulo, Medical School, São Paulo, Brazil
| | - Joel E Kleinman
- Lieber Institute for Brain Development, Baltimore, MD, USA
- Department of Psychiatry & Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Thomas M Hyde
- Lieber Institute for Brain Development, Baltimore, MD, USA
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Department of Psychiatry & Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Daniel R Weinberger
- Lieber Institute for Brain Development, Baltimore, MD, USA
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Department of Psychiatry & Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Ronald McKay
- Lieber Institute for Brain Development, Baltimore, MD, USA
- Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Joo Heon Shin
- Lieber Institute for Brain Development, Baltimore, MD, USA
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Tomoyo Sawada
- Lieber Institute for Brain Development, Baltimore, MD, USA.
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD, USA.
| | - Apua C M Paquola
- Lieber Institute for Brain Development, Baltimore, MD, USA
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Jennifer A Erwin
- Lieber Institute for Brain Development, Baltimore, MD, USA.
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD, USA.
- Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
5
|
Yan YM, Jin MZ, Li SH, Wu Y, Wang Q, Hu FF, Shen C, Yin WH. Hub genes, diagnostic model, and predicted drugs in systemic sclerosis by integrated bioinformatics analysis. Front Genet 2023; 14:1202561. [PMID: 37501723 PMCID: PMC10369177 DOI: 10.3389/fgene.2023.1202561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2023] [Accepted: 07/04/2023] [Indexed: 07/29/2023] Open
Abstract
Background: Systemic sclerosis (scleroderma; SSc), a rare and heterogeneous connective tissue disease, remains unclear in terms of its underlying causative genes and effective therapeutic approaches. The purpose of the present study was to identify hub genes, diagnostic markers and explore potential small-molecule drugs of SSc. Methods: The cohorts of data used in this study were downloaded from the Gene Expression Complex (GEO) database. Integrated bioinformatic tools were utilized for exploration, including Weighted Gene Co-Expression Network Analysis (WGCNA), least absolute shrinkage and selection operator (LASSO) regression, gene set enrichment analysis (GSEA), Connectivity Map (CMap) analysis, molecular docking, and pharmacokinetic/toxicity properties exploration. Results: Seven hub genes (THY1, SULF1, PRSS23, COL5A2, NNMT, SLCO2B1, and TIMP1) were obtained in the merged gene expression profiles of GSE45485 and GSE76885. GSEA results have shown that they are associated with autoimmune diseases, microorganism infections, inflammatory related pathways, immune responses, and fibrosis process. Among them, THY1 and SULF1 were identified as diagnostic markers and validated in skin samples from GSE32413, GSE95065, GSE58095 and GSE125362. Finally, ten small-molecule drugs with potential therapeutic effects were identified, mainly including phosphodiesterase (PDE) inhibitors (BRL-50481, dipyridamole), TGF-β receptor inhibitor (SB-525334), and so on. Conclusion: This study provides new sights into a deeper understanding the molecular mechanisms in the pathogenesis of SSc. More importantly, the results may offer promising clues for further experimental studies and novel treatment strategies.
Collapse
Affiliation(s)
- Yue-Mei Yan
- Department of Dermatology, The First Hospital of Jiaxing, The Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Meng-Zhu Jin
- Department of Dermatology, The First Hospital of Jiaxing, The Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Sheng-Hua Li
- Department of Dermatology, The First Hospital of Jiaxing, The Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Yun Wu
- Department of Dermatology, The First Hospital of Jiaxing, The Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Qiang Wang
- Department of Dermatology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Fei-Fei Hu
- Department of Dermatology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Chen Shen
- Department of Dermatology, Shanghai Skin Disease Hospital, Tongji University, Shanghai, China
| | - Wen-Hao Yin
- Department of Dermatology, The First Hospital of Jiaxing, The Affiliated Hospital of Jiaxing University, Jiaxing, China
| |
Collapse
|
6
|
Cheng CK, Lin X, Pu Y, Tse JKY, Wang Y, Zhang CL, Cao X, Lau CW, Huang J, He L, Luo JY, Shih YT, Wan S, Ng CF, Wang L, Ma RCW, Chiu JJ, Chan TF, Yu Tian X, Huang Y. SOX4 is a novel phenotypic regulator of endothelial cells in atherosclerosis revealed by single-cell analysis. J Adv Res 2022; 43:187-203. [PMID: 36585108 PMCID: PMC9811326 DOI: 10.1016/j.jare.2022.02.017] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 02/25/2022] [Accepted: 02/27/2022] [Indexed: 01/07/2023] Open
Abstract
INTRODUCTION Atherosclerotic complications represent the leading cause of cardiovascular mortality globally. Dysfunction of endothelial cells (ECs) often initiates the pathological events in atherosclerosis. OBJECTIVES In this study, we sought to investigate the transcriptional profile of atherosclerotic aortae, identify novel regulator in dysfunctional ECs and hence provide mechanistic insights into atherosclerotic progression. METHODS We applied single-cell RNA sequencing (scRNA-seq) on aortic cells from Western diet-fed apolipoprotein E-deficient (ApoE-/-) mice to explore the transcriptional landscape and heterogeneity of dysfunctional ECs. In vivo validation of SOX4 upregulation in ECs were performed in atherosclerotic tissues, including mouse aortic tissues, human coronary arteries, and human renal arteries. Single-cell analysis on human aortic aneurysmal tissue was also performed. Downstream vascular abnormalities induced by EC-specific SOX4 overexpression, and upstream modulators of SOX4 were revealed by biochemical assays, immunostaining, and wire myography. Effects of shear stress on endothelial SOX4 expression was investigated by in vitro hemodynamic study. RESULTS Among the compendium of aortic cells, mesenchymal markers in ECs were significantly enriched. Two EC subsets were subsequently distinguished, as the 'endothelial-like' and 'mesenchymal-like' subsets. Conventional assays consistently identified SOX4 as a novel atherosclerotic marker in mouse and different human arteries, additional to a cancer marker. EC-specific SOX4 overexpression promoted atherogenesis and endothelial-to-mesenchymal transition (EndoMT). Importantly, hyperlipidemia-associated cytokines and oscillatory blood flow upregulated, whereas the anti-diabetic drug metformin pharmacologically suppressed SOX4 level in ECs. CONCLUSION Our study unravels SOX4 as a novel phenotypic regulator during endothelial dysfunction, which exacerbates atherogenesis. Our study also pinpoints hyperlipidemia-associated cytokines and oscillatory blood flow as endogenous SOX4 inducers, providing more therapeutic insights against atherosclerotic diseases.
Collapse
Affiliation(s)
- Chak Kwong Cheng
- School of Biomedical Sciences and Li Ka Shing Institute of Health Science, The Chinese University of Hong Kong, 999077, Hong Kong Special Administrative Region; Heart and Vascular Institute and Shenzhen Research Institute, The Chinese University of Hong Kong, 999077, Hong Kong Special Administrative Region; Department of Biomedical Sciences, City University of Hong Kong, 999077, Hong Kong Special Administrative Region
| | - Xiao Lin
- School of Life Sciences, The Chinese University of Hong Kong, 999077, Hong Kong Special Administrative Region
| | - Yujie Pu
- School of Biomedical Sciences and Li Ka Shing Institute of Health Science, The Chinese University of Hong Kong, 999077, Hong Kong Special Administrative Region; Heart and Vascular Institute and Shenzhen Research Institute, The Chinese University of Hong Kong, 999077, Hong Kong Special Administrative Region
| | - Joyce Ka Yu Tse
- School of Life Sciences, The Chinese University of Hong Kong, 999077, Hong Kong Special Administrative Region
| | - Yu Wang
- School of Biomedical Sciences and Li Ka Shing Institute of Health Science, The Chinese University of Hong Kong, 999077, Hong Kong Special Administrative Region; Heart and Vascular Institute and Shenzhen Research Institute, The Chinese University of Hong Kong, 999077, Hong Kong Special Administrative Region
| | - Cheng-Lin Zhang
- School of Biomedical Sciences and Li Ka Shing Institute of Health Science, The Chinese University of Hong Kong, 999077, Hong Kong Special Administrative Region; Heart and Vascular Institute and Shenzhen Research Institute, The Chinese University of Hong Kong, 999077, Hong Kong Special Administrative Region
| | - Xiaoyun Cao
- School of Biomedical Sciences and Li Ka Shing Institute of Health Science, The Chinese University of Hong Kong, 999077, Hong Kong Special Administrative Region; Heart and Vascular Institute and Shenzhen Research Institute, The Chinese University of Hong Kong, 999077, Hong Kong Special Administrative Region
| | - Chi Wai Lau
- School of Biomedical Sciences and Li Ka Shing Institute of Health Science, The Chinese University of Hong Kong, 999077, Hong Kong Special Administrative Region; Heart and Vascular Institute and Shenzhen Research Institute, The Chinese University of Hong Kong, 999077, Hong Kong Special Administrative Region
| | - Juan Huang
- School of Biomedical Sciences and Li Ka Shing Institute of Health Science, The Chinese University of Hong Kong, 999077, Hong Kong Special Administrative Region; Heart and Vascular Institute and Shenzhen Research Institute, The Chinese University of Hong Kong, 999077, Hong Kong Special Administrative Region
| | - Lei He
- School of Biomedical Sciences and Li Ka Shing Institute of Health Science, The Chinese University of Hong Kong, 999077, Hong Kong Special Administrative Region; Heart and Vascular Institute and Shenzhen Research Institute, The Chinese University of Hong Kong, 999077, Hong Kong Special Administrative Region
| | - Jiang-Yun Luo
- School of Biomedical Sciences and Li Ka Shing Institute of Health Science, The Chinese University of Hong Kong, 999077, Hong Kong Special Administrative Region; Heart and Vascular Institute and Shenzhen Research Institute, The Chinese University of Hong Kong, 999077, Hong Kong Special Administrative Region
| | - Yu-Tsung Shih
- Institute of Cellular and System Medicine, National Health Research Institutes, Miaoli 35053, Taiwan
| | - Song Wan
- Department of Surgery, The Chinese University of Hong Kong, 999077, Hong Kong Special Administrative Region
| | - Chi Fai Ng
- Department of Surgery, The Chinese University of Hong Kong, 999077, Hong Kong Special Administrative Region
| | - Li Wang
- School of Biomedical Sciences and Li Ka Shing Institute of Health Science, The Chinese University of Hong Kong, 999077, Hong Kong Special Administrative Region; Heart and Vascular Institute and Shenzhen Research Institute, The Chinese University of Hong Kong, 999077, Hong Kong Special Administrative Region
| | - Ronald Ching Wan Ma
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, 999077, Hong Kong Special Administrative Region; Hong Kong Institute of Diabetes and Obesity, The Chinese University of Hong Kong, 999077, Hong Kong Special Administrative Region
| | - Jeng-Jiann Chiu
- Institute of Cellular and System Medicine, National Health Research Institutes, Miaoli 35053, Taiwan; School of Medical Laboratory Science and Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan
| | - Ting Fung Chan
- School of Life Sciences, The Chinese University of Hong Kong, 999077, Hong Kong Special Administrative Region
| | - Xiao Yu Tian
- School of Biomedical Sciences and Li Ka Shing Institute of Health Science, The Chinese University of Hong Kong, 999077, Hong Kong Special Administrative Region; Heart and Vascular Institute and Shenzhen Research Institute, The Chinese University of Hong Kong, 999077, Hong Kong Special Administrative Region.
| | - Yu Huang
- School of Biomedical Sciences and Li Ka Shing Institute of Health Science, The Chinese University of Hong Kong, 999077, Hong Kong Special Administrative Region; Heart and Vascular Institute and Shenzhen Research Institute, The Chinese University of Hong Kong, 999077, Hong Kong Special Administrative Region; Department of Biomedical Sciences, City University of Hong Kong, 999077, Hong Kong Special Administrative Region.
| |
Collapse
|
7
|
Multi-time scale transcriptomic analysis on the dynamic process of tamoxifen resistance development in breast cancer cell lines. Breast Cancer 2022; 29:458-467. [PMID: 35041152 DOI: 10.1007/s12282-021-01325-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Accepted: 12/19/2021] [Indexed: 11/02/2022]
Abstract
BACKGROUND Approximately 30% of breast cancer patients develop endocrine resistance after tamoxifen therapy. There still lacks a comprehensive understanding on the mechanism of tamoxifen resistance. This study aims to explore the dynamic process of ER + breast cancer resistance to tamoxifen through the time course transcriptomic analysis. METHODS The transcriptome profiles of human breast cancer cell line MCF-7 treated with tamoxifen at different time scales were collected from LINCS, SRA and GEO databases. Differentially expressed genes (DEGs) were identified in the short-term tamoxifen treatment and tamoxifen-resistant cell lines. The time course analysis was used to explore the dynamic development of tamoxifen resistance using the transcriptome profiles of tamoxifen-cultured MCF-7 for 1-12 weeks. RESULTS After the short-term treatment of MCF-7 with tamoxifen for 6 h or 24 h, the expression level of gene PRSS23 was significantly reduced. However, its expression recovered in the resistant cell lines. The time course analysis identified 9 clusters of the DEGs based on the temporal trend of their expression levels. Gene PRSS23 belongs to cluster 2 in which the expression levels were significantly down-regulated in the first 4 weeks but gradually recovered afterwards. Functional enrichment analysis of the DEGs in cluster 2 showed that they are significantly enriched in DNA replication, mismatch repair and cell cycle pathways. Their specific role in the resistance development needs to be further explored. The protein-protein interaction network analysis indicates that gene PRSS23 participates in the drug resistance by regulating multiple tamoxifen drug targets. CONCLUSIONS The acquired drug resistance in ER + breast cancer is a complex and dynamic biological process. PRSS23 plays an important role in the development of resistance and is a potential target for overcoming resistance.
Collapse
|
8
|
Cui X, Li X, He Y, Yu J, Dong N, Zhao RC. Slight up-regulation of Kir2.1 channel promotes endothelial progenitor cells to transdifferentiate into a pericyte phenotype by Akt/mTOR/Snail pathway. J Cell Mol Med 2021; 25:10088-10100. [PMID: 34592781 PMCID: PMC8572793 DOI: 10.1111/jcmm.16944] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 08/22/2021] [Accepted: 09/19/2021] [Indexed: 12/27/2022] Open
Abstract
It was shown that endothelial progenitor cells (EPCs) have bidirectional differentiation potential and thus perform different biological functions. The purpose of this study was to investigate the effects of slight up‐regulation of the Kir2.1 channel on EPC transdifferentiation and the potential mechanism on cell function and transformed cell type. First, we found that the slight up‐regulation of Kir2.1 expression promoted the expression of the stem cell stemness factors ZFX and NS and inhibited the expression of senescence‐associated β‐galactosidase. Further studies showed the slightly increased expression of Kir2.1 could also improve the expression of pericyte molecular markers NG2, PDGFRβ and Desmin. Moreover, adenovirus‐mediated Kir2.1 overexpression had an enhanced contractile response to norepinephrine of EPCs. These results suggest that the up‐regulated expression of the Kir2.1 channel promotes EPC transdifferentiation into a pericyte phenotype. Furthermore, the mechanism of EPC transdifferentiation to mesenchymal cells (pericytes) was found to be closely related to the channel functional activity of Kir2.1 and revealed that this channel could promote EPC EndoMT by activating the Akt/mTOR/Snail signalling pathway. Overall, this study suggested that in the early stage of inflammatory response, regulating the Kir2.1 channel expression affects the biological function of EPCs, thereby determining the maturation and stability of neovascularization.
Collapse
Affiliation(s)
- Xiaodong Cui
- Department of Basic Medicine, Institute of Stem Cell and Regenerative Medicine, Qingdao University Medical College, Qingdao University, Qingdao, China.,School of Basic Medicine Sciences, Weifang Medical University, Weifang, China
| | - Xiaoxia Li
- Department of Basic Medicine, Institute of Stem Cell and Regenerative Medicine, Qingdao University Medical College, Qingdao University, Qingdao, China
| | - Yanting He
- School of Basic Medicine Sciences, Weifang Medical University, Weifang, China
| | - Jie Yu
- School of Basic Medicine Sciences, Weifang Medical University, Weifang, China
| | - Naijun Dong
- Department of Basic Medicine, Institute of Stem Cell and Regenerative Medicine, Qingdao University Medical College, Qingdao University, Qingdao, China
| | - Robert Chunhua Zhao
- Department of Basic Medicine, Institute of Stem Cell and Regenerative Medicine, Qingdao University Medical College, Qingdao University, Qingdao, China
| |
Collapse
|
9
|
Abu Nahia K, Migdał M, Quinn TA, Poon KL, Łapiński M, Sulej A, Liu J, Mondal SS, Pawlak M, Bugajski Ł, Piwocka K, Brand T, Kohl P, Korzh V, Winata C. Genomic and physiological analyses of the zebrafish atrioventricular canal reveal molecular building blocks of the secondary pacemaker region. Cell Mol Life Sci 2021; 78:6669-6687. [PMID: 34557935 PMCID: PMC8558220 DOI: 10.1007/s00018-021-03939-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 08/06/2021] [Accepted: 09/10/2021] [Indexed: 01/06/2023]
Abstract
The atrioventricular canal (AVC) is the site where key structures responsible for functional division between heart regions are established, most importantly, the atrioventricular (AV) conduction system and cardiac valves. To elucidate the mechanism underlying AVC development and function, we utilized transgenic zebrafish line sqet31Et expressing EGFP in the AVC to isolate this cell population and profile its transcriptome at 48 and 72 hpf. The zebrafish AVC transcriptome exhibits hallmarks of mammalian AV node, including the expression of genes implicated in its development and those encoding connexins forming low conductance gap junctions. Transcriptome analysis uncovered protein-coding and noncoding transcripts enriched in AVC, which have not been previously associated with this structure, as well as dynamic expression of epithelial-to-mesenchymal transition markers and components of TGF-β, Notch, and Wnt signaling pathways likely reflecting ongoing AVC and valve development. Using transgenic line Tg(myl7:mermaid) encoding voltage-sensitive fluorescent protein, we show that abolishing the pacemaker-containing sinoatrial ring (SAR) through Isl1 loss of function resulted in spontaneous activation in the AVC region, suggesting that it possesses inherent automaticity although insufficient to replace the SAR. The SAR and AVC transcriptomes express partially overlapping species of ion channels and gap junction proteins, reflecting their distinct roles. Besides identifying conserved aspects between zebrafish and mammalian conduction systems, our results established molecular hallmarks of the developing AVC which underlies its role in structural and electrophysiological separation between heart chambers. This data constitutes a valuable resource for studying AVC development and function, and identification of novel candidate genes implicated in these processes.
Collapse
Affiliation(s)
- Karim Abu Nahia
- International Institute of Molecular and Cell Biology in Warsaw, Warsaw, Poland
| | - Maciej Migdał
- International Institute of Molecular and Cell Biology in Warsaw, Warsaw, Poland
| | - T Alexander Quinn
- Department of Physiology and Biophysics, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Kar-Lai Poon
- Institute of Molecular and Cell Biology, 61 Biopolis Dr, Singapore , Singapore.,Developmental Dynamics, National Heart and Lung Institute, Imperial College London, London, UK
| | - Maciej Łapiński
- International Institute of Molecular and Cell Biology in Warsaw, Warsaw, Poland
| | - Agata Sulej
- International Institute of Molecular and Cell Biology in Warsaw, Warsaw, Poland
| | - Jiandong Liu
- McAllister Heart Institute, University of North Carolina, Chapel Hill, USA
| | - Shamba S Mondal
- International Institute of Molecular and Cell Biology in Warsaw, Warsaw, Poland
| | - Michał Pawlak
- International Institute of Molecular and Cell Biology in Warsaw, Warsaw, Poland
| | | | | | - Thomas Brand
- Developmental Dynamics, National Heart and Lung Institute, Imperial College London, London, UK
| | - Peter Kohl
- Institute for Experimental Cardiovascular Medicine, University Heart Centre, Faculty of Medicine, and Faculty of Engineering, University of Freiburg, Freiburg im Breisgau, Germany
| | - Vladimir Korzh
- International Institute of Molecular and Cell Biology in Warsaw, Warsaw, Poland.
| | - Cecilia Winata
- International Institute of Molecular and Cell Biology in Warsaw, Warsaw, Poland.
| |
Collapse
|
10
|
Einstein JM, Perelis M, Chaim IA, Meena JK, Nussbacher JK, Tankka AT, Yee BA, Li H, Madrigal AA, Neill NJ, Shankar A, Tyagi S, Westbrook TF, Yeo GW. Inhibition of YTHDF2 triggers proteotoxic cell death in MYC-driven breast cancer. Mol Cell 2021; 81:3048-3064.e9. [PMID: 34216543 PMCID: PMC8359670 DOI: 10.1016/j.molcel.2021.06.014] [Citation(s) in RCA: 102] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 03/11/2021] [Accepted: 06/10/2021] [Indexed: 01/18/2023]
Abstract
RNA-binding proteins (RBPs) are critical regulators of post-transcriptional gene expression, and aberrant RBP-RNA interactions can promote cancer progression. Here, we interrogate the function of RBPs in cancer using pooled CRISPR-Cas9 screening and identify 57 RBP candidates with distinct roles in supporting MYC-driven oncogenic pathways. We find that disrupting YTHDF2-dependent mRNA degradation triggers apoptosis in triple-negative breast cancer (TNBC) cells and tumors. eCLIP and m6A sequencing reveal that YTHDF2 interacts with mRNAs encoding proteins in the MAPK pathway that, when stabilized, induce epithelial-to-mesenchymal transition and increase global translation rates. scRibo-STAMP profiling of translating mRNAs reveals unique alterations in the translatome of single cells within YTHDF2-depleted solid tumors, which selectively contribute to endoplasmic reticulum stress-induced apoptosis in TNBC cells. Thus, our work highlights the therapeutic potential of RBPs by uncovering a critical role for YTHDF2 in counteracting the global increase of mRNA synthesis in MYC-driven breast cancers.
Collapse
Affiliation(s)
- Jaclyn M Einstein
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA, USA; Stem Cell Program, University of California, San Diego, La Jolla, CA, USA; Institute for Genomic Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Mark Perelis
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA, USA; Stem Cell Program, University of California, San Diego, La Jolla, CA, USA; Institute for Genomic Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Isaac A Chaim
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA, USA; Stem Cell Program, University of California, San Diego, La Jolla, CA, USA; Institute for Genomic Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Jitendra K Meena
- Verna & Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Julia K Nussbacher
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA, USA; Stem Cell Program, University of California, San Diego, La Jolla, CA, USA; Institute for Genomic Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Alexandra T Tankka
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA, USA; Stem Cell Program, University of California, San Diego, La Jolla, CA, USA; Institute for Genomic Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Brian A Yee
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA, USA; Stem Cell Program, University of California, San Diego, La Jolla, CA, USA; Institute for Genomic Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Heyuan Li
- Verna & Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Assael A Madrigal
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA, USA; Stem Cell Program, University of California, San Diego, La Jolla, CA, USA; Institute for Genomic Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Nicholas J Neill
- Verna & Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Archana Shankar
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA, USA; Stem Cell Program, University of California, San Diego, La Jolla, CA, USA; Institute for Genomic Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Siddhartha Tyagi
- Verna & Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Thomas F Westbrook
- Verna & Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA; Therapeutic Innovation Center (THINC), Baylor College of Medicine, Houston, TX 77030, USA
| | - Gene W Yeo
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA, USA; Stem Cell Program, University of California, San Diego, La Jolla, CA, USA; Institute for Genomic Medicine, University of California, San Diego, La Jolla, CA, USA.
| |
Collapse
|
11
|
Wang Z, Gao D, Wang S, Lin H, Wang Y, Xu W. Exosomal microRNA-1246 from human umbilical cord mesenchymal stem cells potentiates myocardial angiogenesis in chronic heart failure. Cell Biol Int 2021; 45:2211-2225. [PMID: 34270841 DOI: 10.1002/cbin.11664] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 06/08/2021] [Accepted: 07/03/2021] [Indexed: 12/19/2022]
Abstract
microRNAs (miRNAs) are of importance to chronic heart failure (CHF). However, the relevance of the exosomal miRNAs produced during CHF remains unknown. Our purpose here was to examine the relevance of exosomal microRNA-1246 (miR-1246) released from human umbilical cord mesenchymal stem cell (hucMSC) during CHF and the mechanism of action. Cardiac function, myocardial infarction area, apoptosis, and angiogenesis were all evaluated in a CHF rat model following treatment with hucMSC-derived exosomes (hucMSC-Exos). H9C2 and human umbilical vascular endothelial cells (HUVECs) were subjected to oxygen and glucose deprivation and exosome treatment to quantify the cell proliferation and apoptosis in H9C2 cells and the tube formation capacity of the HUVECs. A dual-luciferase activity reporter assay was conducted to validate the interaction between miR-1246 and serine protease 23 (PRSS23). HucMSCs treatment led to a reduction in H9C2 apoptosis and an increase in HUVEC angiogenesis, which were mitigated when hucMSCs were treated with a miR-1246 inhibitor. We also confirmed that PRSS23 is a putative target of miR-1246 and that miR-1246 attenuated hypoxia-induced myocardial tissue damage by targeting PRSS23 and inhibiting the activation of the Snail/alpha-smooth muscle actin signaling. Our findings suggest that exosomal miR-1246 from hucMSCs protects the heart from failure by targeting PRSS23.
Collapse
Affiliation(s)
- Zicheng Wang
- Department of Cardiovascular Medicine, Ningbo Medical Center Lihuili Hospital, Ningbo, Zhejiang, China
| | - Da Gao
- Department of Cardiovascular Medicine, Ningbo Medical Center Lihuili Hospital, Ningbo, Zhejiang, China
| | - Shengjie Wang
- Department of Cardiovascular Medicine, Ningbo Medical Center Lihuili Hospital, Ningbo, Zhejiang, China
| | - Haiyan Lin
- Department of Cardiovascular Medicine, Ningbo Medical Center Lihuili Hospital, Ningbo, Zhejiang, China
| | - Yanwei Wang
- Department of Cardiovascular Medicine, Ningbo Medical Center Lihuili Hospital, Ningbo, Zhejiang, China
| | - Weifeng Xu
- Department of Cardiovascular Medicine, Ningbo Medical Center Lihuili Hospital, Ningbo, Zhejiang, China
| |
Collapse
|
12
|
Qiu X, Liu Y, Shen H, Wang Z, Gong Y, Yang J, Li X, Zhang H, Chen Y, Zhou C, Lv W, Cheng L, Hu Y, Li B, Shen W, Zhu X, Tan LJ, Xiao HM, Deng HW. Single-cell RNA sequencing of human femoral head in vivo. Aging (Albany NY) 2021; 13:15595-15619. [PMID: 34111027 PMCID: PMC8221309 DOI: 10.18632/aging.203124] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 05/13/2021] [Indexed: 11/25/2022]
Abstract
The homeostasis of bone metabolism depends on the coupling and precise regulation of various types of cells in bone tissue. However, the communication and interaction between bone tissue cells at the single-cell level remains poorly understood. Thus, we performed single-cell RNA sequencing (scRNA-seq) on the primary human femoral head tissue cells (FHTCs). Nine cell types were identified in 26,574 primary human FHTCs, including granulocytes, T cells, monocytes, B cells, red blood cells, osteoblastic lineage cells, endothelial cells, endothelial progenitor cells (EPCs) and plasmacytoid dendritic cells. We identified serine protease 23 (PRSS23) and matrix remodeling associated protein 8 (MXRA8) as novel bone metabolism-related genes. Additionally, we found that several subtypes of monocytes, T cells and B cells were related to bone metabolism. Cell-cell communication analysis showed that collagen, chemokine, transforming growth factor and their ligands have significant roles in the crosstalks between FHTCs. In particular, EPCs communicated with osteoblastic lineage cells closely via the "COL2A1-ITGB1" interaction pair. Collectively, this study provided an initial characterization of the cellular composition of the human FHTCs and the complex crosstalks between them at the single-cell level. It is a unique starting resource for in-depth insights into bone metabolism.
Collapse
Affiliation(s)
- Xiang Qiu
- Center for System Biology, Data Sciences, and Reproductive Health, School of Basic Medical Science, Central South University, Yuelu, Changsha 410013, China
| | - Ying Liu
- Laboratory of Molecular and Statistical Genetics, College of Life Sciences, Human Normal University, Changsha 410081, China
| | - Hui Shen
- Tulane Center of Biomedical Informatics and Genomics, Deming Department of Medicine, School of Medicine, Tulane University, New Orleans, LA 70112, USA
| | - Zun Wang
- Xiangya Nursing School, Central South University, Changsha 410013, China
| | - Yun Gong
- Tulane Center of Biomedical Informatics and Genomics, Deming Department of Medicine, School of Medicine, Tulane University, New Orleans, LA 70112, USA
| | - Junxiao Yang
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Xiaohua Li
- Laboratory of Molecular and Statistical Genetics, College of Life Sciences, Human Normal University, Changsha 410081, China
| | - Huixi Zhang
- Laboratory of Molecular and Statistical Genetics, College of Life Sciences, Human Normal University, Changsha 410081, China
| | - Yu Chen
- Laboratory of Molecular and Statistical Genetics, College of Life Sciences, Human Normal University, Changsha 410081, China
| | - Cui Zhou
- Laboratory of Molecular and Statistical Genetics, College of Life Sciences, Human Normal University, Changsha 410081, China
| | - Wanqiang Lv
- Center for System Biology, Data Sciences, and Reproductive Health, School of Basic Medical Science, Central South University, Yuelu, Changsha 410013, China
| | - Liang Cheng
- Department of Orthopedics and National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Yihe Hu
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Boyang Li
- Center for System Biology, Data Sciences, and Reproductive Health, School of Basic Medical Science, Central South University, Yuelu, Changsha 410013, China
| | - Wendi Shen
- Center for System Biology, Data Sciences, and Reproductive Health, School of Basic Medical Science, Central South University, Yuelu, Changsha 410013, China
| | - Xuezhen Zhu
- Center for System Biology, Data Sciences, and Reproductive Health, School of Basic Medical Science, Central South University, Yuelu, Changsha 410013, China
| | - Li-Jun Tan
- Laboratory of Molecular and Statistical Genetics, College of Life Sciences, Human Normal University, Changsha 410081, China
| | - Hong-Mei Xiao
- Center for System Biology, Data Sciences, and Reproductive Health, School of Basic Medical Science, Central South University, Yuelu, Changsha 410013, China
| | - Hong-Wen Deng
- Center for System Biology, Data Sciences, and Reproductive Health, School of Basic Medical Science, Central South University, Yuelu, Changsha 410013, China
- Tulane Center of Biomedical Informatics and Genomics, Deming Department of Medicine, School of Medicine, Tulane University, New Orleans, LA 70112, USA
| |
Collapse
|
13
|
Podvin S, Jones A, Liu Q, Aulston B, Mosier C, Ames J, Winston C, Lietz CB, Jiang Z, O’Donoghue AJ, Ikezu T, Rissman RA, Yuan SH, Hook V. Mutant Presenilin 1 Dysregulates Exosomal Proteome Cargo Produced by Human-Induced Pluripotent Stem Cell Neurons. ACS OMEGA 2021; 6:13033-13056. [PMID: 34056454 PMCID: PMC8158845 DOI: 10.1021/acsomega.1c00660] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Accepted: 04/16/2021] [Indexed: 05/28/2023]
Abstract
The accumulation and propagation of hyperphosphorylated tau (p-Tau) is a neuropathological hallmark occurring with neurodegeneration of Alzheimer's disease (AD). Extracellular vesicles, exosomes, have been shown to initiate tau propagation in the brain. Notably, exosomes from human-induced pluripotent stem cell (iPSC) neurons expressing the AD familial A246E mutant form of presenilin 1 (mPS1) are capable of inducing tau deposits in the mouse brain after in vivo injection. To gain insights into the exosome proteome cargo that participates in propagating tau pathology, this study conducted proteomic analysis of exosomes produced by human iPSC neurons expressing A246E mPS1. Significantly, mPS1 altered the profile of exosome cargo proteins to result in (1) proteins present only in mPS1 exosomes and not in controls, (2) the absence of proteins in the mPS1 exosomes which were present only in controls, and (3) shared proteins which were upregulated or downregulated in the mPS1 exosomes compared to controls. These results show that mPS1 dysregulates the proteome cargo of exosomes to result in the acquisition of proteins involved in the extracellular matrix and protease functions, deletion of proteins involved in RNA and protein translation systems along with proteasome and related functions, combined with the upregulation and downregulation of shared proteins, including the upregulation of amyloid precursor protein. Notably, mPS1 neuron-derived exosomes displayed altered profiles of protein phosphatases and kinases involved in regulating the status of p-tau. The dysregulation of exosome cargo proteins by mPS1 may be associated with the ability of mPS1 neuron-derived exosomes to propagate tau pathology.
Collapse
Affiliation(s)
- Sonia Podvin
- Skaggs
School of Pharmacy and Pharmaceutical Sciences, University of California San Diego,
La Jolla, San Diego 92093, California, United States
| | - Alexander Jones
- Biomedical
Sciences Graduate Program, University of
California, San Diego, La Jolla, San Diego 92093, California, United States
| | - Qing Liu
- Department
of Neurosciences, School of Medicine, University
of California, San Diego, La Jolla, San Diego 92093, California, United States
| | - Brent Aulston
- Department
of Neurosciences, School of Medicine, University
of California, San Diego, La Jolla, San Diego 92093, California, United States
| | - Charles Mosier
- Skaggs
School of Pharmacy and Pharmaceutical Sciences, University of California San Diego,
La Jolla, San Diego 92093, California, United States
| | - Janneca Ames
- Skaggs
School of Pharmacy and Pharmaceutical Sciences, University of California San Diego,
La Jolla, San Diego 92093, California, United States
| | - Charisse Winston
- Department
of Neurosciences, School of Medicine, University
of California, San Diego, La Jolla, San Diego 92093, California, United States
| | - Christopher B. Lietz
- Skaggs
School of Pharmacy and Pharmaceutical Sciences, University of California San Diego,
La Jolla, San Diego 92093, California, United States
| | - Zhenze Jiang
- Skaggs
School of Pharmacy and Pharmaceutical Sciences, University of California San Diego,
La Jolla, San Diego 92093, California, United States
| | - Anthony J. O’Donoghue
- Skaggs
School of Pharmacy and Pharmaceutical Sciences, University of California San Diego,
La Jolla, San Diego 92093, California, United States
| | - Tsuneya Ikezu
- Department
of Pharmacology and Experimental Therapeutics, Department of Neurology,
Alzheimer’s Disease Research Center, Boston University, School of Medicine, Boston 02118, Massachusetts, United States
| | - Robert A. Rissman
- Department
of Neurosciences, School of Medicine, University
of California, San Diego, La Jolla, San Diego 92093, California, United States
- Veterans
Affairs San Diego Healthcare System,
La Jolla, San Diego 92161, California, United States
| | - Shauna H. Yuan
- Department
of Neurosciences, School of Medicine, University
of California, San Diego, La Jolla, San Diego 92093, California, United States
| | - Vivian Hook
- Skaggs
School of Pharmacy and Pharmaceutical Sciences, University of California San Diego,
La Jolla, San Diego 92093, California, United States
- Biomedical
Sciences Graduate Program, University of
California, San Diego, La Jolla, San Diego 92093, California, United States
- Department
of Neurosciences, School of Medicine, University
of California, San Diego, La Jolla, San Diego 92093, California, United States
| |
Collapse
|
14
|
Queckbörner S, von Grothusen C, Boggavarapu NR, Francis RM, Davies LC, Gemzell-Danielsson K. Stromal Heterogeneity in the Human Proliferative Endometrium-A Single-Cell RNA Sequencing Study. J Pers Med 2021; 11:jpm11060448. [PMID: 34067358 PMCID: PMC8224746 DOI: 10.3390/jpm11060448] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 05/15/2021] [Accepted: 05/17/2021] [Indexed: 12/11/2022] Open
Abstract
The endometrium undergoes regular regeneration and stromal proliferation as part of the normal menstrual cycle. To better understand cellular interactions driving the mechanisms in endometrial regeneration we employed single-cell RNA sequencing. Endometrial biopsies were obtained during the proliferative phase of the menstrual cycle from healthy fertile women and processed to single-cell suspensions which were submitted for sequencing. In addition to known endometrial cell types, bioinformatic analysis revealed multiple stromal populations suggestive of specific stromal niches with the ability to control inflammation and extracellular matrix composition. Ten different stromal cells and two pericyte subsets were identified. Applying different R packages (Seurat, SingleR, Velocyto) we established cell cluster diversity and cell lineage/trajectory, while using external data to validate our findings. By understanding healthy regeneration in the described stromal compartments, we aim to identify points of further investigation and possible targets for novel therapy development for benign gynecological disorders affecting endometrial regeneration and proliferation such as endometriosis and Asherman’s syndrome.
Collapse
Affiliation(s)
- Suzanna Queckbörner
- Department of Women’s and Children’s Health, Division of Obstetrics and Gynecology, Karolinska Institutet, and Karolinska University Hospital, S-171 64 Solna, Sweden; (S.Q.); (N.R.B.); (K.G.-D.)
| | - Carolina von Grothusen
- Department of Women’s and Children’s Health, Division of Obstetrics and Gynecology, Karolinska Institutet, and Karolinska University Hospital, S-171 64 Solna, Sweden; (S.Q.); (N.R.B.); (K.G.-D.)
- Correspondence:
| | - Nageswara Rao Boggavarapu
- Department of Women’s and Children’s Health, Division of Obstetrics and Gynecology, Karolinska Institutet, and Karolinska University Hospital, S-171 64 Solna, Sweden; (S.Q.); (N.R.B.); (K.G.-D.)
| | - Roy Mathew Francis
- Department of Medical Biochemistry and Microbiology (IMBIM), Uppsala University, BMC, Husargatan 3, 752 37 Uppsala, Sweden;
- National Bioinformatics Infrastructure Sweden (NBIS), Department of Cell and Molecular Biology (ICM), Uppsala University, SciLifeLab, 751 24 Uppsala, Sweden
| | - Lindsay C. Davies
- Department of Laboratory Medicine, Karolinska Institutet, S-141 52 Huddinge, Sweden;
| | - Kristina Gemzell-Danielsson
- Department of Women’s and Children’s Health, Division of Obstetrics and Gynecology, Karolinska Institutet, and Karolinska University Hospital, S-171 64 Solna, Sweden; (S.Q.); (N.R.B.); (K.G.-D.)
| |
Collapse
|
15
|
Greenspan LJ, Weinstein BM. To be or not to be: endothelial cell plasticity in development, repair, and disease. Angiogenesis 2021; 24:251-269. [PMID: 33449300 PMCID: PMC8205957 DOI: 10.1007/s10456-020-09761-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Accepted: 12/14/2020] [Indexed: 02/08/2023]
Abstract
Endothelial cells display an extraordinary plasticity both during development and throughout adult life. During early development, endothelial cells assume arterial, venous, or lymphatic identity, while selected endothelial cells undergo additional fate changes to become hematopoietic progenitor, cardiac valve, and other cell types. Adult endothelial cells are some of the longest-lived cells in the body and their participation as stable components of the vascular wall is critical for the proper function of both the circulatory and lymphatic systems, yet these cells also display a remarkable capacity to undergo changes in their differentiated identity during injury, disease, and even normal physiological changes in the vasculature. Here, we discuss how endothelial cells become specified during development as arterial, venous, or lymphatic endothelial cells or convert into hematopoietic stem and progenitor cells or cardiac valve cells. We compare findings from in vitro and in vivo studies with a focus on the zebrafish as a valuable model for exploring the signaling pathways and environmental cues that drive these transitions. We also discuss how endothelial plasticity can aid in revascularization and repair of tissue after damage- but may have detrimental consequences under disease conditions. By better understanding endothelial plasticity and the mechanisms underlying endothelial fate transitions, we can begin to explore new therapeutic avenues.
Collapse
Affiliation(s)
- Leah J Greenspan
- Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Brant M Weinstein
- Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, 20892, USA.
| |
Collapse
|
16
|
Qin S, Li B, Li R, Cai Y, Zheng K, Huang H, Xiao F, Zeng M, Xu X. Proteomic characteristics and identification of PM 2.5-induced differentially expressed proteins in hepatocytes and c-Myc silenced hepatocytes. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2021; 209:111838. [PMID: 33387776 DOI: 10.1016/j.ecoenv.2020.111838] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 12/16/2020] [Accepted: 12/19/2020] [Indexed: 06/12/2023]
Abstract
Proteomics and bioinformatics were applied to explore PM2.5-induced differentially expressed proteins (DEPs) in hepatocytes (L02 cells) and c-Myc-silenced hepatocytes. L02 cells and c-Myc-silenced hepatocytes were treated with PM2.5 for 24 h. Fifty-two DEPs were screened in L02 hepatocytes, of which 28 were upregulated and 24 were downregulated. Forty-one DEPs were screened in the c-Myc-silenced hepatocytes, of which 31 were upregulated and 10 were downregulated. GO analysis showed that DEPs in L02 cells were mainly concentrated in the cytosol and were involved in biological processes such as the response to metal ions. DEPs in c-Myc-silenced cells were mainly enriched in the extracellular space and were involved in lipoprotein metabolism. KEGG analysis showed that DEPs in L02 cells were mainly involved in arachidonic acid metabolism and mineral absorption. DEPs in c-Myc-silenced cells were mainly enriched in pathways involving nerve absorption, complement and coagulation cascades, and other pathways. Twenty key proteins, including Metallothionein-2A (MT2A), Metallothionein-1X (MT1X), zinc transporter ZIP10 (SLC39A10) and Serine protease 23 (PRSS23) were screened in two groups through analysis of protein-protein interactions. Based on the identification of the selected DEPs, PRSS23 and SLC39A10 might be the potential biomarker of PM2.5-induced carcinogenesis, which provide the scientific basis for further research into the carcinogenic mechanisms of PM2.5.
Collapse
Affiliation(s)
- Shuangjian Qin
- Xiangya School of Public Health, Central South University, Changsha, Hunan 410078, China; Institute of environment and health, Shenzhen center for disease control and prevention, Shenzhen, Guangdong 518055, China
| | - Boru Li
- Xiangya School of Public Health, Central South University, Changsha, Hunan 410078, China; Institute of environment and health, Shenzhen center for disease control and prevention, Shenzhen, Guangdong 518055, China
| | - Runbing Li
- Institute of environment and health, Shenzhen center for disease control and prevention, Shenzhen, Guangdong 518055, China; School of public health, University of South China, Hengyang, Hunan 421001, China
| | - Ying Cai
- Institute of environment and health, Shenzhen center for disease control and prevention, Shenzhen, Guangdong 518055, China; School of public health, University of South China, Hengyang, Hunan 421001, China
| | - Kai Zheng
- Institute of environment and health, Shenzhen center for disease control and prevention, Shenzhen, Guangdong 518055, China; School of public health, University of South China, Hengyang, Hunan 421001, China
| | - Haiyan Huang
- Institute of environment and health, Shenzhen center for disease control and prevention, Shenzhen, Guangdong 518055, China
| | - Fang Xiao
- Xiangya School of Public Health, Central South University, Changsha, Hunan 410078, China.
| | - Ming Zeng
- Xiangya School of Public Health, Central South University, Changsha, Hunan 410078, China.
| | - Xinyun Xu
- Institute of environment and health, Shenzhen center for disease control and prevention, Shenzhen, Guangdong 518055, China.
| |
Collapse
|
17
|
Apoptotic exosome-like vesicles regulate endothelial gene expression, inflammatory signaling, and function through the NF-κB signaling pathway. Sci Rep 2020; 10:12562. [PMID: 32724121 PMCID: PMC7387353 DOI: 10.1038/s41598-020-69548-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 07/06/2020] [Indexed: 12/19/2022] Open
Abstract
Persistent endothelial injury promotes maladaptive responses by favoring the release of factors leading to perturbation in vascular homeostasis and tissue architecture. Caspase-3 dependent death of microvascular endothelial cells leads to the release of unique apoptotic exosome-like vesicles (ApoExo). Here, we evaluate the impact of ApoExo on endothelial gene expression and function in the context of a pro-apoptotic stimulus. Endothelial cells exposed to ApoExo differentially express genes involved in cell death, inflammation, differentiation, and cell movement. Endothelial cells exposed to ApoExo showed inhibition of apoptosis, improved wound closure along with reduced angiogenic activity and reduced expression of endothelial markers consistent with the first phase of endothelial-to-mesenchymal transition (endoMT). ApoExo interaction with endothelial cells also led to NF-κB activation. NF-κB is known to participate in endothelial dysfunction in numerous diseases. Silencing NF-κB reversed the anti-apoptotic effect and the pro-migratory state and prevented angiostatic properties and CD31 downregulation in endothelial cells exposed to ApoExo. This study identifies vascular injury-derived extracellular vesicles (ApoExo) as novel drivers of NF-κB activation in endothelial cells and demonstrates the pivotal role of this signaling pathway in coordinating ApoExo-induced functional changes in endothelial cells. Hence, targeting ApoExo-mediated NF-κB activation in endothelial cells opens new avenues to prevent endothelial dysfunction.
Collapse
|
18
|
Larsson C, Cordeddu L, Siggens L, Pandzic T, Kundu S, He L, Ali MA, Pristovšek N, Hartman K, Ekwall K, Sjöblom T. Restoration of KMT2C/MLL3 in human colorectal cancer cells reinforces genome-wide H3K4me1 profiles and influences cell growth and gene expression. Clin Epigenetics 2020; 12:74. [PMID: 32471474 PMCID: PMC7257146 DOI: 10.1186/s13148-020-00863-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Accepted: 05/14/2020] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND The histone 3 lysine 4 (H3K4) monomethylase KMT2C is mutated across several cancer types; however, the effects of mutations on epigenome organization, gene expression, and cell growth are not clear. A frequently recurring mutation in colorectal cancer (CRC) with microsatellite instability is a single nucleotide deletion within the exon 38 poly-A(9) repeat (c.8390delA) which results in frameshift preceding the functional carboxy-terminal SET domain. To study effects of KMT2C expression in CRC cells, we restored one allele to wild type KMT2C in the two CRC cell lines RKO and HCT116, which both are homozygous c.8390delA mutant. RESULTS Gene editing resulted in increased KMT2C expression, increased H3K4me1 levels, altered gene expression profiles, and subtle negative effects on cell growth, where higher dependence and stronger effects of KMT2C expression were observed in RKO compared to HCT116 cells. Surprisingly, we found that the two RKO and HCT116 CRC cell lines have distinct baseline H3K4me1 epigenomic profiles. In RKO cells, a flatter genome-wide H3K4me1 profile was associated with more increased H3K4me1 deposition at enhancers, reduced cell growth, and more differential gene expression relative to HCT116 cells when KMT2C was restored. Profiling of H3K4me1 did not indicate a highly specific regulation of gene expression as KMT2C-induced H3K4me1 deposition was found globally and not at a specific enhancer sub-set in the engineered cells. Although we observed variation in differentially regulated gene sets between cell lines and individual clones, differentially expressed genes in both cell lines included genes linked to known cancer signaling pathways, estrogen response, hypoxia response, and aspects of immune system regulation. CONCLUSIONS Here, KMT2C restoration reduced CRC cell growth and reinforced genome-wide H3K4me1 deposition at enhancers; however, the effects varied depending upon the H3K4me1 status of KMT2C deficient cells. Results indicate that KMT2C inactivation may promote colorectal cancer development through transcriptional dysregulation in several pathways with known cancer relevance.
Collapse
Affiliation(s)
- Chatarina Larsson
- Department of Immunology, Genetics and Pathology, Uppsala University, Rudbeck Laboratory, SE-751 85, Uppsala, Sweden
| | - Lina Cordeddu
- Department of Biosciences and Nutrition, NEO, Karolinska Institute, SE-141 83, Huddinge, Sweden
| | - Lee Siggens
- Department of Biosciences and Nutrition, NEO, Karolinska Institute, SE-141 83, Huddinge, Sweden
| | - Tatjana Pandzic
- Department of Immunology, Genetics and Pathology, Uppsala University, Rudbeck Laboratory, SE-751 85, Uppsala, Sweden
| | - Snehangshu Kundu
- Department of Immunology, Genetics and Pathology, Uppsala University, Rudbeck Laboratory, SE-751 85, Uppsala, Sweden
| | - Liqun He
- Department of Immunology, Genetics and Pathology, Uppsala University, Rudbeck Laboratory, SE-751 85, Uppsala, Sweden
| | - Muhammad Akhtar Ali
- Department of Immunology, Genetics and Pathology, Uppsala University, Rudbeck Laboratory, SE-751 85, Uppsala, Sweden.,Present address: School of Biological Sciences, University of the Punjab, Lahore, Pakistan
| | - Nuša Pristovšek
- Department of Immunology, Genetics and Pathology, Uppsala University, Rudbeck Laboratory, SE-751 85, Uppsala, Sweden.,Present address: The Novo Nordisk Foundation for Biosustainability, Technical University of Denmark, Kemitorvet, Building 220, 2800, Kgs. Lyngby, Denmark
| | - Karin Hartman
- Department of Immunology, Genetics and Pathology, Uppsala University, Rudbeck Laboratory, SE-751 85, Uppsala, Sweden
| | - Karl Ekwall
- Department of Biosciences and Nutrition, NEO, Karolinska Institute, SE-141 83, Huddinge, Sweden
| | - Tobias Sjöblom
- Department of Immunology, Genetics and Pathology, Uppsala University, Rudbeck Laboratory, SE-751 85, Uppsala, Sweden.
| |
Collapse
|
19
|
Jiang L, Xu J, Sang M, Zhang Y, Ye M, Zhang H, Wu B, Zhu Y, Xu P, Tai R, Zhao Z, Jiang Y, Dong C, Sun L, Griffin CH, Gragnoli C, Wu R. A Drive to Driven Model of Mapping Intraspecific Interaction Networks. iScience 2019; 22:109-122. [PMID: 31765992 PMCID: PMC6883333 DOI: 10.1016/j.isci.2019.11.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Revised: 09/23/2019] [Accepted: 11/01/2019] [Indexed: 12/14/2022] Open
Abstract
Community ecology theory suggests that an individual's phenotype is determined by the phenotypes of its coexisting members to the extent at which this process can shape community evolution. Here, we develop a mapping theory to identify interaction quantitative trait loci (QTL) governing inter-individual dependence. We mathematically formulate the decision-making strategy of interacting individuals. We integrate these mathematical descriptors into a statistical procedure, enabling the joint characterization of how QTL drive the strengths of ecological interactions and how the genetic architecture of QTL is driven by ecological networks. In three fish full-sib mapping experiments, we identify a set of genome-wide QTL that control a range of societal behaviors, including mutualism, altruism, aggression, and antagonism, and find that these intraspecific interactions increase the genetic variation of body mass by about 50%. We showcase how the interaction QTL can be used as editors to reconstruct and engineer new social networks for ecological communities. We develop a new theory for complex-trait mapping by integrating behavioral ecology This theory can characterize how QTL drive cooperation or competition in populations It can also illustrate how the activation of QTL is driven by ecological interactions The new theory leverages interdisciplinary studies of genetics, ecology, and evolution
Collapse
Affiliation(s)
- Libo Jiang
- Beijing Advanced Innovation Center for Tree Breeding by Molecular Design, Center for Computational Biology, College of Biological Sciences and Technology, Beijing Forestry University, Beijing 100083, China
| | - Jian Xu
- Key Laboratory of Aquatic Genomics, Ministry of Agriculture, CAFS Key Laboratory of Aquatic Genomics and Beijing Key Laboratory of Fishery Biotechnology, Chinese Academy of Fishery Sciences, Beijing 100141, China
| | - Mengmeng Sang
- Beijing Advanced Innovation Center for Tree Breeding by Molecular Design, Center for Computational Biology, College of Biological Sciences and Technology, Beijing Forestry University, Beijing 100083, China
| | - Yan Zhang
- Key Laboratory of Aquatic Genomics, Ministry of Agriculture, CAFS Key Laboratory of Aquatic Genomics and Beijing Key Laboratory of Fishery Biotechnology, Chinese Academy of Fishery Sciences, Beijing 100141, China
| | - Meixia Ye
- Beijing Advanced Innovation Center for Tree Breeding by Molecular Design, Center for Computational Biology, College of Biological Sciences and Technology, Beijing Forestry University, Beijing 100083, China
| | - Hanyuan Zhang
- Key Laboratory of Aquatic Genomics, Ministry of Agriculture, CAFS Key Laboratory of Aquatic Genomics and Beijing Key Laboratory of Fishery Biotechnology, Chinese Academy of Fishery Sciences, Beijing 100141, China
| | - Biyin Wu
- Key Laboratory of Aquatic Genomics, Ministry of Agriculture, CAFS Key Laboratory of Aquatic Genomics and Beijing Key Laboratory of Fishery Biotechnology, Chinese Academy of Fishery Sciences, Beijing 100141, China
| | - Youxiu Zhu
- Key Laboratory of Aquatic Genomics, Ministry of Agriculture, CAFS Key Laboratory of Aquatic Genomics and Beijing Key Laboratory of Fishery Biotechnology, Chinese Academy of Fishery Sciences, Beijing 100141, China
| | - Peng Xu
- Key Laboratory of Aquatic Genomics, Ministry of Agriculture, CAFS Key Laboratory of Aquatic Genomics and Beijing Key Laboratory of Fishery Biotechnology, Chinese Academy of Fishery Sciences, Beijing 100141, China; Fujian Collaborative Innovation Center for Exploitation and Utilization of Marine Biological Resources, Xiamen University, Xiamen, Fujian 361102, China
| | - Ruyu Tai
- Key Laboratory of Aquatic Genomics, Ministry of Agriculture, CAFS Key Laboratory of Aquatic Genomics and Beijing Key Laboratory of Fishery Biotechnology, Chinese Academy of Fishery Sciences, Beijing 100141, China
| | - Zixia Zhao
- Key Laboratory of Aquatic Genomics, Ministry of Agriculture, CAFS Key Laboratory of Aquatic Genomics and Beijing Key Laboratory of Fishery Biotechnology, Chinese Academy of Fishery Sciences, Beijing 100141, China
| | - Yanliang Jiang
- Key Laboratory of Aquatic Genomics, Ministry of Agriculture, CAFS Key Laboratory of Aquatic Genomics and Beijing Key Laboratory of Fishery Biotechnology, Chinese Academy of Fishery Sciences, Beijing 100141, China
| | - Chuanju Dong
- Key Laboratory of Aquatic Genomics, Ministry of Agriculture, CAFS Key Laboratory of Aquatic Genomics and Beijing Key Laboratory of Fishery Biotechnology, Chinese Academy of Fishery Sciences, Beijing 100141, China; College of Fishery, Henan Normal University, Xinxiang, Henan 453007, China
| | - Lidan Sun
- Beijing Advanced Innovation Center for Tree Breeding by Molecular Design, Center for Computational Biology, College of Biological Sciences and Technology, Beijing Forestry University, Beijing 100083, China
| | - Christopher H Griffin
- Applied Research Laboratory, The Pennsylvania State University, University Park, PA 16802, USA
| | - Claudia Gragnoli
- Division of Endocrinology, Diabetes, and Metabolic Disease, Translational Medicine, Department of Medicine, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19106, USA; Molecular Biology Laboratory, Bios Biotech Multi Diagnostic Health Center, Rome 00197, Italy
| | - Rongling Wu
- Beijing Advanced Innovation Center for Tree Breeding by Molecular Design, Center for Computational Biology, College of Biological Sciences and Technology, Beijing Forestry University, Beijing 100083, China; Center for Statistical Genetics, The Pennsylvania State University, Hershey, PA 17033, USA.
| |
Collapse
|
20
|
A bigenic mouse model of FSGS reveals perturbed pathways in podocytes, mesangial cells and endothelial cells. PLoS One 2019; 14:e0216261. [PMID: 31461442 PMCID: PMC6713350 DOI: 10.1371/journal.pone.0216261] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Accepted: 08/12/2019] [Indexed: 11/24/2022] Open
Abstract
Focal segmental glomerulosclerosis is a major cause of end stage renal disease. Many patients prove unresponsive to available therapies. An improved understanding of the molecular basis of the disease process could provide insights leading to novel therapeutic approaches. In this study we carried out an RNA-seq analysis of the altered gene expression patterns of podocytes, mesangial cells and glomerular endothelial cells of the bigenic Cd2ap+/-, Fyn-/- mutant mouse model of FSGS. In the podocytes we observed upregulation of many genes related to the Tgfβ family/pathway, including Gdnf, Tgfβ1, Tgfβ2, Snai2, Vegfb, Bmp4, and Tnc. The mutant podocytes also showed upregulation of Acta2, a marker of smooth muscle and associated with myofibroblasts, which are implicated in driving fibrosis. GO analysis of the podocyte upregulated genes identified elevated protein kinase activity, increased expression of growth factors, and negative regulation of cell adhesion, perhaps related to the observed podocyte loss. Both podocytes and mesangial cells showed strong upregulation of aldehyde dehydrogenase genes involved in the synthesis of retinoic acid. Similarly, the Cd2ap+/-, Fyn-/- mesangial cells, as well as podocytes in other genetic models, and the glomeruli of human FSGS patients, all show upregulation of the serine protease Prss23, with the common thread suggesting important functionality. Another gene with strong upregulation in the Cd2ap+/-, Fyn-/- mutant mesangial cells as well as multiple other mutant mouse models of FSGS was thrombospondin, which activates the secreted inactive form of Tgfβ. The Cd2ap+/-, Fyn-/- mutant endothelial cells showed elevated expression of genes involved in cell proliferation, angioblast migration, angiogenesis, and neovasculature, all consistent with the formation of new blood vessels in the diseased glomerulus. The resulting global definition of the perturbed molecular pathways in the three major cell types of the mutant glomerulus provide deeper understanding of the molecular pathogenic pathways.
Collapse
|
21
|
Nicolosi PA, Tombetti E, Giovenzana A, Donè E, Pulcinelli E, Meneveri R, Tirone M, Maugeri N, Rovere-Querini P, Manfredi AA, Brunelli S. Macrophages Guard Endothelial Lineage by Hindering Endothelial-to-Mesenchymal Transition: Implications for the Pathogenesis of Systemic Sclerosis. THE JOURNAL OF IMMUNOLOGY 2019; 203:247-258. [PMID: 31127033 DOI: 10.4049/jimmunol.1800883] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Accepted: 04/23/2019] [Indexed: 11/19/2022]
Abstract
The signals that control endothelial plasticity in inflamed tissues have only been partially characterized. For example, it has been shown that inadequate vasculogenesis in systemic sclerosis (SSc) has been associated with an endothelial defect. We used a genetic lineage tracing model to investigate whether endothelial cells die or change phenotypically after fibrosis induction and whether signals released by cells of the innate immune system and in the blood of patients influence their commitment. We observed that in the lineage-tracing transgenic mice Cdh5-CreERT2::R26R-EYFP, endothelial-derived cells (EdCs) underwent fibrosis after treatment with bleomycin, and EdCs retrieved from the lung showed expression of endothelial-to-mesenchymal transition (EndoMT) markers. Liposome-encapsulated clodronate was used to assess macrophage impact on EdCs. Clodronate treatment affected the number of alternatively activated macrophages in the lung, with upregulated expression of EndoMT markers in lung EdCs. Endothelial fate and function were investigated in vitro upon challenge with serum signals from SSc patients or released by activated macrophages. Sera of SSc patients with anti-Scl70 Abs, at higher risk of visceral organ fibrosis, induced EndoMT and jeopardized endothelial function. In conclusion, EdCs in SSc might be defective because of commitment to a mesenchymal fate, which is sustained by soluble signals in the patient's blood. Macrophages contribute to preserve the endothelial identity of precursor cells. Altered macrophage-dependent plasticity of EdCs could contribute to link vasculopathy with fibrosis.
Collapse
Affiliation(s)
- Pier Andrea Nicolosi
- School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy
| | - Enrico Tombetti
- Division of Immunology, Transplantation and Infectious Disease, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Anna Giovenzana
- School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy
| | - Eleonora Donè
- School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy
| | - Eleonora Pulcinelli
- School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy
| | - Raffaella Meneveri
- School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy
| | - Mario Tirone
- School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy.,Division of Genetics and Cell Biology, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy; and
| | - Norma Maugeri
- Division of Immunology, Transplantation and Infectious Disease, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Patrizia Rovere-Querini
- Division of Immunology, Transplantation and Infectious Disease, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy.,Università Vita-Salute San Raffaele, 20132 Milan, Italy
| | - Angelo A Manfredi
- Division of Immunology, Transplantation and Infectious Disease, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy; .,Università Vita-Salute San Raffaele, 20132 Milan, Italy
| | - Silvia Brunelli
- School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy;
| |
Collapse
|
22
|
Han B, Yang Y, Chen J, He X, Lv N, Yan R. PRSS23 knockdown inhibits gastric tumorigenesis through EIF2 signaling. Pharmacol Res 2019; 142:50-57. [PMID: 30769097 DOI: 10.1016/j.phrs.2019.02.008] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Revised: 01/29/2019] [Accepted: 02/05/2019] [Indexed: 02/06/2023]
Abstract
PRSS23 is a newly discovered serine protease that has been associated with tumor progression in various types of cancers. Our previous study showed PRSS23 is down-regulated obviously in Hedgehog pathway blocked gastric cancer cells. However, the correlation between PRSS23 and tumor progression of gastric cancer remains unclear. Here, the role and mechanism of PRSS23 in tumor progression of gastric cancer were determined. PRSS23 protein levels were significantly increased in gastric cancer tissues compared with the paired adjacent normal gastric mucosa tissues. The high expression of PRSS23 correlated strongly with both poor differentiated histology and cancer region of sinus ventriculi. Gastric cancer patients with low PRSS23 expression displayed a better prognosis. In gastric cancer cells, PRSS23 knockdown inhibited cell proliferation and induced apoptosis. In xenograft tumor model, PRSS23 knockdown led to dramatic decreases of the average tumor volume and the average tumor weight. In addition, PRSS23 knockdown suppressed gastric cancer growth through inhibiting EIF2 signaling using gene expression microarray analysis. Taken together, our results suggest PRSS23 is highly associated with human gastric tumorigenesis and progression. PRSS23 knockdown could suppress tumor growth of gastric cancer in vitro and in vivo through inhibiting EIF2 signaling, and EIF4E maybe a potential target of PRSS23. PRSS23 could serve as a potential target for gastric cancer therapy, and also a biomarker for the prediction of prognosis of gastric cancer.
Collapse
Affiliation(s)
- Bing Han
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Key Laboratory of Digestive Diseases of Jiangxi Province, Nanchang, China; Institute of Tropical Fruit Trees, Hainan Academy of Agricultural Sciences, Haikou, China
| | - Yang Yang
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Key Laboratory of Digestive Diseases of Jiangxi Province, Nanchang, China; Guilin Medical University, Guilin, China
| | - Jiang Chen
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Key Laboratory of Digestive Diseases of Jiangxi Province, Nanchang, China
| | - Xingxing He
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Key Laboratory of Digestive Diseases of Jiangxi Province, Nanchang, China
| | - Nonghua Lv
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Key Laboratory of Digestive Diseases of Jiangxi Province, Nanchang, China.
| | - Runwei Yan
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Key Laboratory of Digestive Diseases of Jiangxi Province, Nanchang, China.
| |
Collapse
|
23
|
Harder JL, Menon R, Otto EA, Zhou J, Eddy S, Wys NL, O'Connor C, Luo J, Nair V, Cebrian C, Spence JR, Bitzer M, Troyanskaya OG, Hodgin JB, Wiggins RC, Freedman BS, Kretzler M. Organoid single cell profiling identifies a transcriptional signature of glomerular disease. JCI Insight 2019; 4:122697. [PMID: 30626756 PMCID: PMC6485369 DOI: 10.1172/jci.insight.122697] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Accepted: 11/27/2018] [Indexed: 12/13/2022] Open
Abstract
Podocyte injury is central to many forms of kidney disease, but transcriptional signatures reflecting podocyte injury and compensation mechanisms are challenging to analyze in vivo. Human kidney organoids derived from pluripotent stem cells (PSCs), a potentially new model for disease and regeneration, present an opportunity to explore the transcriptional plasticity of podocytes. Here, transcriptional profiling of more than 12,000 single cells from human PSC-derived kidney organoid cultures was used to identify robust and reproducible cell lineage gene expression signatures shared with developing human kidneys based on trajectory analysis. Surprisingly, the gene expression signature characteristic of developing glomerular epithelial cells was also observed in glomerular tissue from a kidney disease cohort. This signature correlated with proteinuria and inverse eGFR, and it was confirmed in an independent podocytopathy cohort. Three genes in particular were further characterized as potentially novel components of the glomerular disease signature. We conclude that cells in human PSC-derived kidney organoids reliably recapitulate the developmental transcriptional program of podocytes and other cell lineages in the human kidney and that transcriptional profiles seen in developing podocytes are reactivated in glomerular disease. Our findings demonstrate an approach to identifying potentially novel molecular programs involved in the pathogenesis of glomerulopathies.
Collapse
Affiliation(s)
| | - Rajasree Menon
- Department of Computational Medicine and Bioinformatics, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Edgar A Otto
- Department of Internal Medicine, Division of Nephrology, and
| | - Jian Zhou
- Flatiron Institute, Simons Foundation, New York, New York, USA
| | - Sean Eddy
- Department of Internal Medicine, Division of Nephrology, and
| | - Noel L Wys
- Department of Internal Medicine, Division of Nephrology, and
| | | | | | - Viji Nair
- Department of Internal Medicine, Division of Nephrology, and
| | - Cristina Cebrian
- Department of Internal Medicine, Division of Gastroenterology, University of Michigan, Ann Arbor, Michigan, USA
| | - Jason R Spence
- Department of Internal Medicine, Division of Gastroenterology, University of Michigan, Ann Arbor, Michigan, USA
| | - Markus Bitzer
- Department of Internal Medicine, Division of Nephrology, and
| | - Olga G Troyanskaya
- Flatiron Institute, Simons Foundation, New York, New York, USA.,Lewis-Sigler Institute for Integrative Genomics and.,Department of Computer Science, Princeton University, Princeton, New Jersey, USA
| | | | - Roger C Wiggins
- Department of Internal Medicine, Division of Nephrology, and
| | - Benjamin S Freedman
- Department of Medicine, Division of Nephrology.,Kidney Research Institute.,Institute for Stem Cell and Regenerative Medicine, and.,Department of Pathology, University of Washington School of Medicine, Seattle, Washington, USA
| | - Matthias Kretzler
- Department of Internal Medicine, Division of Nephrology, and.,Department of Computational Medicine and Bioinformatics, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | | | | |
Collapse
|
24
|
Brown AR, Green JM, Moreman J, Gunnarsson LM, Mourabit S, Ball J, Winter MJ, Trznadel M, Correia A, Hacker C, Perry A, Wood ME, Hetheridge MJ, Currie RA, Tyler CR. Cardiovascular Effects and Molecular Mechanisms of Bisphenol A and Its Metabolite MBP in Zebrafish. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2019; 53:463-474. [PMID: 30520632 PMCID: PMC6333396 DOI: 10.1021/acs.est.8b04281] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Revised: 11/12/2018] [Accepted: 12/06/2018] [Indexed: 05/03/2023]
Abstract
The plastic monomer bisphenol A (BPA) is one of the highest production volume chemicals in the world and is frequently detected in wildlife and humans, particularly children. BPA has been associated with numerous adverse health outcomes relating to its estrogenic and other hormonal properties, but direct causal links are unclear in humans and animal models. Here we simulated measured (1×) and predicted worst-case (10× ) maximum fetal exposures for BPA, or equivalent concentrations of its metabolite MBP, using fluorescent reporter embryo-larval zebrafish, capable of quantifying Estrogen Response Element (ERE) activation throughout the body. Heart valves were primary sites for ERE activation by BPA and MBP, and transcriptomic analysis of microdissected heart tissues showed that both chemicals targeted several molecular pathways constituting biomarkers for calcific aortic valve disease (CAVD), including extra-cellular matrix (ECM) alteration. ECM collagen deficiency and impact on heart valve structural integrity were confirmed by histopathology for high-level MBP exposure, and structural defects (abnormal curvature) of the atrio-ventricular valves corresponded with impaired cardiovascular function (reduced ventricular beat rate and blood flow). Our results are the first to demonstrate plausible mechanistic links between ERE activation in the heart valves by BPA's reactive metabolite MBP and the development of valvular-cardiovascular disease states.
Collapse
Affiliation(s)
- A. Ross Brown
- Biosciences,
College of Life and Environmental Sciences, University of Exeter, Geoffrey Pope, Stocker Road, Exeter, Devon EX4 4QD, U.K.
| | - Jon M. Green
- Biosciences,
College of Life and Environmental Sciences, University of Exeter, Geoffrey Pope, Stocker Road, Exeter, Devon EX4 4QD, U.K.
| | - John Moreman
- Biosciences,
College of Life and Environmental Sciences, University of Exeter, Geoffrey Pope, Stocker Road, Exeter, Devon EX4 4QD, U.K.
| | - Lina M. Gunnarsson
- Biosciences,
College of Life and Environmental Sciences, University of Exeter, Geoffrey Pope, Stocker Road, Exeter, Devon EX4 4QD, U.K.
| | - Sulayman Mourabit
- Biosciences,
College of Life and Environmental Sciences, University of Exeter, Geoffrey Pope, Stocker Road, Exeter, Devon EX4 4QD, U.K.
| | - Jonathan Ball
- Biosciences,
College of Life and Environmental Sciences, University of Exeter, Geoffrey Pope, Stocker Road, Exeter, Devon EX4 4QD, U.K.
| | - Matthew J. Winter
- Biosciences,
College of Life and Environmental Sciences, University of Exeter, Geoffrey Pope, Stocker Road, Exeter, Devon EX4 4QD, U.K.
| | - Maciej Trznadel
- Biosciences,
College of Life and Environmental Sciences, University of Exeter, Geoffrey Pope, Stocker Road, Exeter, Devon EX4 4QD, U.K.
| | - Ana Correia
- Biosciences,
College of Life and Environmental Sciences, University of Exeter, Geoffrey Pope, Stocker Road, Exeter, Devon EX4 4QD, U.K.
| | - Christian Hacker
- Biosciences,
College of Life and Environmental Sciences, University of Exeter, Geoffrey Pope, Stocker Road, Exeter, Devon EX4 4QD, U.K.
| | - Alexis Perry
- Biosciences,
College of Life and Environmental Sciences, University of Exeter, Geoffrey Pope, Stocker Road, Exeter, Devon EX4 4QD, U.K.
| | - Mark E. Wood
- Biosciences,
College of Life and Environmental Sciences, University of Exeter, Geoffrey Pope, Stocker Road, Exeter, Devon EX4 4QD, U.K.
| | - Malcolm J. Hetheridge
- Biosciences,
College of Life and Environmental Sciences, University of Exeter, Geoffrey Pope, Stocker Road, Exeter, Devon EX4 4QD, U.K.
| | - Richard A. Currie
- Jealott’s
Hill International Research Centre, Syngenta, Bracknell, Berkshire RG42
6EY, U.K.
| | - Charles R. Tyler
- Biosciences,
College of Life and Environmental Sciences, University of Exeter, Geoffrey Pope, Stocker Road, Exeter, Devon EX4 4QD, U.K.
| |
Collapse
|
25
|
Hong L, Du X, Li W, Mao Y, Sun L, Li X. EndMT: A promising and controversial field. Eur J Cell Biol 2018; 97:493-500. [PMID: 30082099 DOI: 10.1016/j.ejcb.2018.07.005] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Revised: 07/03/2018] [Accepted: 07/27/2018] [Indexed: 12/17/2022] Open
Abstract
The endothelial to mesenchymal transition (EndMT) is the process by which endothelial cells lose a portion of their cellular features and obtain certain characteristics of mesenchymal cells, including loss of tight junctions, increased motility, and increased secretion of extracellular matrix proteins. EndMT is involved in cardiac development and a variety of diseases processes, such as vascular or tissue fibrosis and tumor. However, its role in specific diseases remains under debate. This review summarizes EndMT-related diseases, existing controversies, different types of EndMT, and molecules and signaling pathways associated with the process.
Collapse
Affiliation(s)
- Lei Hong
- Department of Vascular Surgery, The Second Affiliated Hospital of Soochow University, Sanxiang Road, Suzhou 215000, JiangSu, China.
| | - Xiaolong Du
- Department of Vascular Surgery, The Second Affiliated Hospital of Soochow University, Sanxiang Road, Suzhou 215000, JiangSu, China.
| | - Wendong Li
- Department of Vascular Surgery, The Second Affiliated Hospital of Soochow University, Sanxiang Road, Suzhou 215000, JiangSu, China
| | - Youjun Mao
- Department of Vascular Surgery, The Second Affiliated Hospital of Soochow University, Sanxiang Road, Suzhou 215000, JiangSu, China
| | - Lili Sun
- Department of Vascular Surgery, The Second Affiliated Hospital of Soochow University, Sanxiang Road, Suzhou 215000, JiangSu, China
| | - Xiaoqiang Li
- Department of Vascular Surgery, The Second Affiliated Hospital of Soochow University, Sanxiang Road, Suzhou 215000, JiangSu, China.
| |
Collapse
|
26
|
Bayoumi AS, Teoh JP, Aonuma T, Yuan Z, Ruan X, Tang Y, Su H, Weintraub NL, Kim IM. MicroRNA-532 protects the heart in acute myocardial infarction, and represses prss23, a positive regulator of endothelial-to-mesenchymal transition. Cardiovasc Res 2018; 113:1603-1614. [PMID: 29016706 DOI: 10.1093/cvr/cvx132] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Accepted: 07/07/2017] [Indexed: 01/13/2023] Open
Abstract
Aims Acute myocardial infarction (MI) leads to cardiac remodelling and development of heart failure. Insufficient myocardial capillary density after MI is considered a critical determinant of this process. MicroRNAs (miRs), negative regulators of gene expression, have emerged as important players in MI. We previously showed that miR-532-5p (miR-532) is up-regulated by the β-arrestin-biased β-adrenergic receptor antagonist (β-blocker) carvedilol, which activates protective pathways in the heart independent of G protein-mediated second messenger signalling. Here, we hypothesize that β2-adrenergic receptor/β-arrestin-responsive miR-532 confers cardioprotection against MI. Methods and results Using cultured cardiac endothelial cell (CEC) and in vivo approaches, we show that CECs lacking miR-532 exhibit increased transition to a fibroblast-like phenotype via endothelial-to-mesenchymal transition (EndMT), while CECs over-expressing miR-532 display decreased EndMT. We also demonstrate that knockdown of miR-532 in mice causes abnormalities in cardiac structure and function as well as reduces CEC proliferation and cardiac vascularization after MI. Mechanistically, cardioprotection elicited by miR-532 is in part attributed to direct repression of a positive regulator of maladaptive EndMT, prss23 (a protease serine 23) in CECs. Conclusions In conclusion, these findings reveal a pivotal role for miR-532-prss23 axis in regulating CEC function after MI, and this novel axis could be suitable for therapeutic intervention in ischemic heart disease.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Huabo Su
- Vascular Biology Center.,Department of Pharmacology and Toxicology
| | | | - Il-Man Kim
- Vascular Biology Center.,Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta University, CB-3717, 1459 Laney Walker Blvd, Augusta, GA 30912, USA
| |
Collapse
|
27
|
Shinde V, Hoelting L, Srinivasan SP, Meisig J, Meganathan K, Jagtap S, Grinberg M, Liebing J, Bluethgen N, Rahnenführer J, Rempel E, Stoeber R, Schildknecht S, Förster S, Godoy P, van Thriel C, Gaspar JA, Hescheler J, Waldmann T, Hengstler JG, Leist M, Sachinidis A. Definition of transcriptome-based indices for quantitative characterization of chemically disturbed stem cell development: introduction of the STOP-Tox ukn and STOP-Tox ukk tests. Arch Toxicol 2016; 91:839-864. [PMID: 27188386 PMCID: PMC5306084 DOI: 10.1007/s00204-016-1741-8] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Accepted: 05/04/2016] [Indexed: 01/08/2023]
Abstract
Stem cell-based in vitro test systems can recapitulate specific phases of human development. In the UKK test system, human pluripotent stem cells (hPSCs) randomly differentiate into cells of the three germ layers and their derivatives. In the UKN1 test system, hPSCs differentiate into early neural precursor cells. During the normal differentiation period (14 days) of the UKK system, 570 genes [849 probe sets (PSs)] were regulated >fivefold; in the UKN1 system (6 days), 879 genes (1238 PSs) were regulated. We refer to these genes as 'developmental genes'. In the present study, we used genome-wide expression data of 12 test substances in the UKK and UKN1 test systems to understand the basic principles of how chemicals interfere with the spontaneous transcriptional development in both test systems. The set of test compounds included six histone deacetylase inhibitors (HDACis), six mercury-containing compounds ('mercurials') and thalidomide. All compounds were tested at the maximum non-cytotoxic concentration, while valproic acid and thalidomide were additionally tested over a wide range of concentrations. In total, 242 genes (252 PSs) in the UKK test system and 793 genes (1092 PSs) in the UKN1 test system were deregulated by the 12 test compounds. We identified sets of 'diagnostic genes' appropriate for the identification of the influence of HDACis or mercurials. Test compounds that interfered with the expression of developmental genes usually antagonized their spontaneous development, meaning that up-regulated developmental genes were suppressed and developmental genes whose expression normally decreases were induced. The fraction of compromised developmental genes varied widely between the test compounds, and it reached up to 60 %. To quantitatively describe disturbed development on a genome-wide basis, we recommend a concept of two indices, 'developmental potency' (D p) and 'developmental index' (D i), whereby D p is the fraction of all developmental genes that are up- or down-regulated by a test compound, and D i is the ratio of overrepresentation of developmental genes among all genes deregulated by a test compound. The use of D i makes hazard identification more sensitive because some compounds compromise the expression of only a relatively small number of genes but have a high propensity to deregulate developmental genes specifically, resulting in a low D p but a high D i. In conclusion, the concept based on the indices D p and D i offers the possibility to quantitatively express the propensity of test compounds to interfere with normal development.
Collapse
Affiliation(s)
- Vaibhav Shinde
- Institute of Neurophysiology and Centre for Molecular Medicine Cologne (CMMC), University of Cologne (UKK), Robert-Koch-Str. 39, 50931, Cologne, Germany
| | - Lisa Hoelting
- Doerenkamp-Zbinden Chair for In Vitro Toxicology and Biomedicine, University of Konstanz, Box: M657, 78457, Constance, Germany.,Konstanz Graduate School Chemical Biology KORS-CB, University of Konstanz, 78457, Constance, Germany
| | - Sureshkumar Perumal Srinivasan
- Institute of Neurophysiology and Centre for Molecular Medicine Cologne (CMMC), University of Cologne (UKK), Robert-Koch-Str. 39, 50931, Cologne, Germany
| | - Johannes Meisig
- Institute of Pathology, Charité Universitätsmedizin, 10117, Berlin, Germany.,Integrative Research Institute for the Life Sciences, Institute for Theoretical Biology, Humboldt Universität, 10115, Berlin, Germany
| | - Kesavan Meganathan
- Institute of Neurophysiology and Centre for Molecular Medicine Cologne (CMMC), University of Cologne (UKK), Robert-Koch-Str. 39, 50931, Cologne, Germany
| | - Smita Jagtap
- Institute of Neurophysiology and Centre for Molecular Medicine Cologne (CMMC), University of Cologne (UKK), Robert-Koch-Str. 39, 50931, Cologne, Germany
| | | | - Julia Liebing
- Leibniz Research Centre for Working Environment and Human Factors at the Technical, University of Dortmund (IfADo), Ardeystrasse 67, 44139, Dortmund, Germany
| | - Nils Bluethgen
- Institute of Pathology, Charité Universitätsmedizin, 10117, Berlin, Germany.,Integrative Research Institute for the Life Sciences, Institute for Theoretical Biology, Humboldt Universität, 10115, Berlin, Germany
| | | | - Eugen Rempel
- Department of Statistics, TU Dortmund University, Dortmund, Germany.,Centre for Organismal Studies, Heidelberg University, 69120, Heidelberg, Germany
| | - Regina Stoeber
- Leibniz Research Centre for Working Environment and Human Factors at the Technical, University of Dortmund (IfADo), Ardeystrasse 67, 44139, Dortmund, Germany
| | - Stefan Schildknecht
- Doerenkamp-Zbinden Chair for In Vitro Toxicology and Biomedicine, University of Konstanz, Box: M657, 78457, Constance, Germany
| | - Sunniva Förster
- Doerenkamp-Zbinden Chair for In Vitro Toxicology and Biomedicine, University of Konstanz, Box: M657, 78457, Constance, Germany
| | - Patricio Godoy
- Leibniz Research Centre for Working Environment and Human Factors at the Technical, University of Dortmund (IfADo), Ardeystrasse 67, 44139, Dortmund, Germany
| | - Christoph van Thriel
- Leibniz Research Centre for Working Environment and Human Factors at the Technical, University of Dortmund (IfADo), Ardeystrasse 67, 44139, Dortmund, Germany
| | - John Antonydas Gaspar
- Institute of Neurophysiology and Centre for Molecular Medicine Cologne (CMMC), University of Cologne (UKK), Robert-Koch-Str. 39, 50931, Cologne, Germany
| | - Jürgen Hescheler
- Institute of Neurophysiology and Centre for Molecular Medicine Cologne (CMMC), University of Cologne (UKK), Robert-Koch-Str. 39, 50931, Cologne, Germany
| | - Tanja Waldmann
- Doerenkamp-Zbinden Chair for In Vitro Toxicology and Biomedicine, University of Konstanz, Box: M657, 78457, Constance, Germany
| | - Jan G Hengstler
- Leibniz Research Centre for Working Environment and Human Factors at the Technical, University of Dortmund (IfADo), Ardeystrasse 67, 44139, Dortmund, Germany.
| | - Marcel Leist
- Doerenkamp-Zbinden Chair for In Vitro Toxicology and Biomedicine, University of Konstanz, Box: M657, 78457, Constance, Germany.
| | - Agapios Sachinidis
- Institute of Neurophysiology and Centre for Molecular Medicine Cologne (CMMC), University of Cologne (UKK), Robert-Koch-Str. 39, 50931, Cologne, Germany.
| |
Collapse
|
28
|
Advanced glycation end products induce endothelial-to-mesenchymal transition via downregulating Sirt 1 and upregulating TGF-β in human endothelial cells. BIOMED RESEARCH INTERNATIONAL 2015; 2015:684242. [PMID: 25710021 PMCID: PMC4330956 DOI: 10.1155/2015/684242] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/21/2014] [Accepted: 11/25/2014] [Indexed: 01/09/2023]
Abstract
In the present study, we examined the advanced glycation end products- (AGEs-) induced endothelial-to-mesenchymal transition (EndMT) in human umbilical vein endothelial cells (HUVECs). Results demonstrated that AGE-BSAs significantly reduced the cluster of differentiation 31 (CD 31) expression, whereas they promoted the expression of fibroblast-specific protein-1 (FSP-1), α-smooth muscle antibody (α-SMA), and collagen I at both mRNA and protein levels in HUVECs. And the AGE-BSAs also promoted the receptors for AGEs (RAGEs) and receptor I for TGF-β (TGFR I) markedly with a dose dependence, whereas the Sirt 1 was significantly downregulated by the AGE-BSA at both mRNA and protein levels. Moreover, the Sirt 1 activity manipulation with its activator, resveratrol (RSV), or its inhibitor, EX527, markedly inhibited or ameliorated the AGE-mediated TGF-β upregulation. And the manipulated Sirt 1 activity positively regulated the AGE-induced CD31, whereas it negatively regulated the AGE-induced FSP-1. Thus, Sirt 1 was confirmed to regulate the AGE-induced EndMT via TGF-β. In summary, we found that AGE-BSA induced EndMT in HUVECs via upregulating TGF-β and downregulating Sirt 1, which also negatively regulated TGF-β in the cell. This study implied the EndMT probably as an important mechanism of AGE-induced cardiovascular injury.
Collapse
|
29
|
J. D'Souza RC, Knittle AM, Nagaraj N, van Dinther M, Choudhary C, ten Dijke P, Mann M, Sharma K. Time-resolved dissection of early phosphoproteome and ensuing proteome changes in response to TGF-. Sci Signal 2014; 7:rs5. [DOI: 10.1126/scisignal.2004856] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
30
|
Kim KL, Choi C, Suh W. Analysis of disease progression-associated gene expression profile in fibrillin-1 mutant mice: new insight into molecular pathogenesis of marfan syndrome. Biomol Ther (Seoul) 2014; 22:143-8. [PMID: 24753820 PMCID: PMC3975469 DOI: 10.4062/biomolther.2014.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2014] [Revised: 03/05/2014] [Accepted: 03/10/2014] [Indexed: 11/16/2022] Open
Abstract
Marfan syndrome (MFS) is a dominantly inherited connective tissue disorder caused by mutations in the gene encoding fibrillin-1 (FBN1) and is characterized by aortic dilatation and dissection, which is the primary cause of death in untreated MFS patients. However, disease progression-associated changes in gene expression in the aortic lesions of MFS patients remained unknown. Using a mouse model of MFS, FBN1 hypomorphic mouse (mgR/mgR), we characterized the aortic gene expression profiles during the progression of the MFS. Homozygous mgR mice exhibited MFS-like phenotypic features, such as fragmentation of elastic fibers throughout the vessel wall and were graded into mgR1–4 based on the pathological severity in aortic walls. Comparative gene expression profiling of WT and four mgR mice using microarrays revealed that the changes in the transcriptome were a direct reflection of the severity of aortic pathological features. Gene ontology analysis showed that genes related to oxidation/reduction, myofibril assembly, cytoskeleton organization, and cell adhesion were differentially expressed in the mgR mice. Further analysis of differentially expressed genes identified several candidate genes whose known roles were suggestive of their involvement in the progressive destruction of aorta during MFS. This study is the first genome-wide analysis of the aortic gene expression profiles associated with the progression of MFS. Our findings provide valuable information regarding the molecular pathogenesis during MFS progression and contribute to the development of new biomarkers as well as improved therapeutic strategies.
Collapse
Affiliation(s)
- Koung Li Kim
- College of Pharmacy, Ajou University, Suwon 443-749, Republic of Korea
| | - Chanmi Choi
- College of Pharmacy, Ajou University, Suwon 443-749, Republic of Korea
| | - Wonhee Suh
- College of Pharmacy, Ajou University, Suwon 443-749, Republic of Korea
| |
Collapse
|
31
|
Wilkinson RN, Jopling C, van Eeden FJM. Zebrafish as a model of cardiac disease. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2014; 124:65-91. [PMID: 24751427 DOI: 10.1016/b978-0-12-386930-2.00004-5] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The zebrafish has been rapidly adopted as a model for cardiac development and disease. The transparency of the embryo, its limited requirement for active oxygen delivery, and ease of use in genetic manipulations and chemical exposure have made it a powerful alternative to rodents. Novel technologies like TALEN/CRISPR-mediated genome engineering and advanced imaging methods will only accelerate its use. Here, we give an overview of heart development and function in the fish and highlight a number of areas where it is most actively contributing to the understanding of cardiac development and disease. We also review the current state of research on a feature that we only could wish to be conserved between fish and human; cardiac regeneration.
Collapse
Affiliation(s)
- Robert N Wilkinson
- Department of Cardiovascular Science, Medical School, University of Sheffield, Sheffield, United Kingdom
| | - Chris Jopling
- CNRS, UMR-5203, Institut de Génomique Fonctionnelle, Département de Physiologie, Labex Ion Channel Science and Therapeutics, Montpellier, France; INSERM, U661, Montpellier, France; Universités de Montpellier 1&2, UMR-5203, Montpellier, France
| | - Fredericus J M van Eeden
- MRC Centre for Biomedical Genetics, Department of Biomedical Science, University of Sheffield, Sheffield, United Kingdom
| |
Collapse
|
32
|
Tao G, Miller LJ, Lincoln J. Snai1 is important for avian epicardial cell transformation and motility. Dev Dyn 2013; 242:699-708. [PMID: 23553854 DOI: 10.1002/dvdy.23967] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2012] [Revised: 03/21/2013] [Accepted: 03/25/2013] [Indexed: 11/08/2022] Open
Abstract
BACKGROUND Formation of the epicardium requires several cellular processes including migration, transformation, invasion, and differentiation in order to give rise to fibroblast, smooth muscle, coronary endothelial and myocyte cell lineages within the developing myocardium. Snai1 is a zinc finger transcription factor that plays an important role in regulating cell survival and fate during embryonic development and under pathological conditions. However, its role in avian epicardial development has not been examined. RESULTS Here we show that Snai1 is highly expressed in epicardial cells from as early as the proepicardial cell stage and its expression is maintained as proepicardial cells migrate and spread over the surface of the myocardium and undergo epicardial-to-mesenchymal transformation in the generation of epicardial-derived cells. Using multiple in vitro assays, we show that Snai1 overexpression in chick explants enhances proepicardial cell migration at Hamburger Hamilton Stage (HH St.) 16, and epicardial-to-mesenchymal transformation, cell migration, and invasion at HH St. 24. Further, we demonstrate that Snai1-mediated cell migration requires matrix metalloproteinase activity, and MMP15 is sufficient for this process. CONCLUSIONS Together our data provide new insights into the multiple roles that Snai1 has in regulating avian epicardial development.
Collapse
Affiliation(s)
- Ge Tao
- Molecular Cell and Developmental Biology Graduate Program, Leonard M. Miller School of Medicine, Miami, Florida, USA
| | | | | |
Collapse
|