1
|
Rudat C, Straube P, Hegermann J, Trowe MO, Thiesler H, Hildebrandt H, Witt L, Kispert A. PPARG contributes to urothelial integrity in the murine ureter by activating the expression of Shh and superficial cell-specific genes. Development 2025; 152:dev204324. [PMID: 40167323 PMCID: PMC12045629 DOI: 10.1242/dev.204324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 03/05/2025] [Indexed: 04/02/2025]
Abstract
The urothelium is a stratified epithelium with an important barrier function in the urinary drainage system. The differentiation and maintenance of the three major urothelial cell types (basal, intermediate and superficial cells) is incompletely understood. Here, we show that mice with a conditional deletion of the transcription factor gene peroxisome proliferator activated receptor gamma (Pparg) in the ureteric epithelium have a dilated ureter at postnatal stages with a urothelium consisting of a layer of undifferentiated luminal cells and a layer of proliferating basal cells. Molecular analysis of fetal stages revealed that the expression of a large number of genes is not activated in superficial cells and that of a few genes, including Shh, is not activated in intermediate and basal cells. Pharmacological activation of SHH signaling in explant cultures of perinatal Pparg-deficient ureters reduced ureteral width and urothelial cell number to normal levels, increased the number of intermediate cells and slightly reduced basal cell proliferation. Our data suggest that PPARG independently activates the expression of structural genes in superficial cells and of Shh in basal and intermediate cells, and that both functions contribute to urothelial integrity.
Collapse
Affiliation(s)
- Carsten Rudat
- Institute of Molecular Biology, Hannover Medical School, 30625 Hannover, Germany
| | - Philipp Straube
- Institute of Molecular Biology, Hannover Medical School, 30625 Hannover, Germany
| | - Jan Hegermann
- Institute of Functional and Applied Anatomy, Research Core Unit Electron Microscopy, Hannover Medical School, 30625 Hannover, Germany
| | - Mark-Oliver Trowe
- Institute of Molecular Biology, Hannover Medical School, 30625 Hannover, Germany
| | - Hauke Thiesler
- Institute of Clinical Biochemistry, Hannover Medical School, 30625 Hannover, Germany
- Center for Systems Neuroscience Hannover, Hannover, Germany
| | - Herbert Hildebrandt
- Institute of Clinical Biochemistry, Hannover Medical School, 30625 Hannover, Germany
- Center for Systems Neuroscience Hannover, Hannover, Germany
| | - Lisa Witt
- Institute of Molecular Biology, Hannover Medical School, 30625 Hannover, Germany
| | - Andreas Kispert
- Institute of Molecular Biology, Hannover Medical School, 30625 Hannover, Germany
| |
Collapse
|
2
|
Brinkmeier ML, Wang SQ, Pittman HA, Cheung LY, Prasov L. Myelin regulatory factor (MYRF) is a critical early regulator of retinal pigment epithelial development. PLoS Genet 2025; 21:e1011670. [PMID: 40233131 PMCID: PMC12052213 DOI: 10.1371/journal.pgen.1011670] [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: 06/18/2024] [Revised: 05/05/2025] [Accepted: 04/01/2025] [Indexed: 04/17/2025] Open
Abstract
Myelin regulatory factor (Myrf) is a critical transcription factor in early retinal and retinal pigment epithelial development, and human variants in MYRF are a cause for nanophthalmos. Single cell RNA sequencing (scRNAseq) was performed on Myrf conditional knockout mice (Rx > Cre Myrffl/fl) at 3 developmental timepoints. Myrf was expressed specifically in the RPE, and expression was abrogated in Rx > Cre Myrffl/fl eyes. scRNAseq analysis revealed a loss of RPE cells at all timepoints resulting from cell death. GO-term analysis in the RPE revealed downregulation of melanogenesis and anatomic structure morphogenesis pathways, which were supported by electron microscopy and histologic analysis. Novel structural target genes including Ermn and Upk3b, along with macular degeneration and inherited retinal disease genes were identified as downregulated, and a strong upregulation of TGFß/BMP signaling and effectors was observed. Regulon analysis placed Myrf downstream or parallel to Pax6 and Mitf and upstream of Sox10 in RPE differentiation. Together, these results suggest a strong role for MYRF in the RPE maturation by regulating melanogenesis, cell survival, and cell structure, in part acting through suppression of TGFß signaling and activation of Sox10.
Collapse
Affiliation(s)
- Michelle L. Brinkmeier
- Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, Michigan, United States of America
- Department of Human Genetics, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Su Qing Wang
- Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Hannah A. Pittman
- Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Leonard Y. Cheung
- Department of Human Genetics, University of Michigan, Ann Arbor, Michigan, United States of America
- Department of Physiology and Biophysics, State University of New York at Stony Brook, Stony Brook, New York, United States of America
| | - Lev Prasov
- Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, Michigan, United States of America
- Department of Human Genetics, University of Michigan, Ann Arbor, Michigan, United States of America
| |
Collapse
|
3
|
Luna G, Verheyden J, Tan C, Kim E, Hwa M, Sahi J, Shen Y, Chung W, McCulley D, Sun X. MYRF is Essential in Mesothelial Cells to Promote Lung Development and Maturation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.13.635155. [PMID: 39990361 PMCID: PMC11844445 DOI: 10.1101/2025.02.13.635155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/25/2025]
Abstract
The mesothelium is a squamous monolayer that ensheathes internal organs, lines the body cavity, and the diaphragm. It serves as a protective barrier, coated in glycocalyx, and secretes lubricants to facilitate tissue movement. How the mesothelium forms is poorly understood. Here, we investigate Myrf , a transcription factor gene expressed in the mesothelium, because it carries variants in patients with Congenital Diaphragmatic Hernia (CDH), a disorder that affects the diaphragm, lung, and other organs. In mice, inactivation of Myrf early in organogenesis resulted in CDH and defective mesothelial specification, compromising its function as a signaling center for lung growth. Inactivation of Myrf later led to enhanced mesothelium differentiation into mesenchymal cell types through partial epithelial-to-mesenchymal transition (EMT), resulting in a unique accumulation of smooth muscle encasing the lung. In this role, MYRF functions in parallel with YAP/TAZ. Together, these findings establish MYRF as a critical regulator of mesothelium development, and when mutated, causes CDH.
Collapse
|
4
|
Takahashi M, Isagawa T, Sato T, Takeda N, Kawakami K. Lineage tracing using Wnt2b-2A-CreERT2 knock-in mice reveals the contributions of Wnt2b-expressing cells to novel subpopulations of mesothelial/epicardial cell lineages during mouse development. Genes Cells 2024; 29:854-875. [PMID: 39109760 DOI: 10.1111/gtc.13147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 07/17/2024] [Accepted: 07/19/2024] [Indexed: 10/04/2024]
Abstract
Mesothelial and epicardial cells give rise to various types of mesenchymal cells via epithelial (mesothelial)-to-mesenchymal transition during development. However, the genes controlling the differentiation and diversification of mesothelial/epicardial cells remain unclear. Here, we examined Wnt2b expression in the embryonic mesothelium and epicardium and performed lineage tracing of Wnt2b-expressing cells by using novel Wnt2b-2A-CreERT2 knock-in and LacZ-reporter mice. Wnt2b was expressed in mesothelial cells covering visceral organs, but the expression was restricted in their subpopulations. Wnt2b-expressing cells labeled at embryonic day (E) 10.5 were distributed to the mesothelium and mesenchyme in the lungs, abdominal wall, stomach, and spleen in Wnt2b2A-CreERT2/+;R26RLacZ/+ mice at E13.0. Wnt2b was initially expressed in the proepicardial organ (PEO) at E9.5 and then in the epicardium after E10.0. Wnt2b-expressing PEO cells labeled at E9.5 differentiated into a small fraction of cardiac fibroblasts and preferentially localized at the left side of the postnatal heart. LacZ+ epicardium-derived cells labeled at E10.5 differentiated into a small fraction of fibroblasts and smooth muscle cells in the postnatal heart. Taken together, our results reveal novel subpopulations of PEO and mesothelial/epicardial cells that are distinguishable by Wnt2b expression and elucidate the unique contribution of Wnt2b-expressing PEO and epicardial cells to the postnatal heart.
Collapse
Affiliation(s)
- Masanori Takahashi
- Department of Anatomy, Division of Bioimaging and Neuro-cell Science, Jichi Medical University, Shimotsuke, Japan
- Division of Cardiology and Metabolism, Center for Molecular Medicine, Jichi Medical University, Shimotsuke, Japan
| | - Takayuki Isagawa
- Division of Cardiology and Metabolism, Center for Molecular Medicine, Jichi Medical University, Shimotsuke, Japan
- Data Science Center, Jichi Medical University, Shimotsuke, Japan
| | - Tatsuyuki Sato
- Division of Cardiology and Metabolism, Center for Molecular Medicine, Jichi Medical University, Shimotsuke, Japan
| | - Norihiko Takeda
- Division of Cardiology and Metabolism, Center for Molecular Medicine, Jichi Medical University, Shimotsuke, Japan
| | | |
Collapse
|
5
|
Brinkmeier ML, Wang SQ, Pittman H, Cheung LY, Prasov L. Myelin regulatory factor ( Myrf ) is a critical early regulator of retinal pigment epithelial development. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.26.591281. [PMID: 38746430 PMCID: PMC11092522 DOI: 10.1101/2024.04.26.591281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
Myelin regulatory factor (Myrf) is a critical transcription factor in early retinal and retinal pigment epithelial development, and human variants in MYRF are a cause for nanophthalmos. Single cell RNA sequencing (scRNAseq) was performed on Myrf conditional knockout mice ( Rx>Cre Myrf fl/fl ) at 3 developmental timepoints. Myrf was expressed specifically in the RPE, and expression was abrogated in Rx>Cre Myrf fl/fl eyes. scRNAseq analysis revealed a loss of RPE cells at all timepoints resulting from cell death. GO-term analysis in the RPE revealed downregulation of melanogenesis and anatomic structure morphogenesis pathways, which were supported by electron microscopy and histologic analysis. Novel structural target genes including Ermn and Upk3b , along with macular degeneration and inherited retinal disease genes were identified as downregulated, and a strong upregulation of TGFß/BMP signaling and effectors was observed. Regulon analysis placed Myrf downstream of Pax6 and Mitf and upstream of Sox10 in RPE differentiation. Together, these results suggest a strong role for Myrf in the RPE maturation by regulating melanogenesis, cell survival, and cell structure, in part acting through suppression of TGFß signaling and activation of Sox10 . SUMMARY STATEMENT Myrf regulates RPE development, melanogenesis, and is important for cell structure and survival, in part through regulation of Ermn , Upk3b and Sox10, and BMP/TGFb signaling.
Collapse
|
6
|
Qiu C, Martin BK, Welsh IC, Daza RM, Le TM, Huang X, Nichols EK, Taylor ML, Fulton O, O'Day DR, Gomes AR, Ilcisin S, Srivatsan S, Deng X, Disteche CM, Noble WS, Hamazaki N, Moens CB, Kimelman D, Cao J, Schier AF, Spielmann M, Murray SA, Trapnell C, Shendure J. A single-cell time-lapse of mouse prenatal development from gastrula to birth. Nature 2024; 626:1084-1093. [PMID: 38355799 PMCID: PMC10901739 DOI: 10.1038/s41586-024-07069-w] [Citation(s) in RCA: 57] [Impact Index Per Article: 57.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Accepted: 01/15/2024] [Indexed: 02/16/2024]
Abstract
The house mouse (Mus musculus) is an exceptional model system, combining genetic tractability with close evolutionary affinity to humans1,2. Mouse gestation lasts only 3 weeks, during which the genome orchestrates the astonishing transformation of a single-cell zygote into a free-living pup composed of more than 500 million cells. Here, to establish a global framework for exploring mammalian development, we applied optimized single-cell combinatorial indexing3 to profile the transcriptional states of 12.4 million nuclei from 83 embryos, precisely staged at 2- to 6-hour intervals spanning late gastrulation (embryonic day 8) to birth (postnatal day 0). From these data, we annotate hundreds of cell types and explore the ontogenesis of the posterior embryo during somitogenesis and of kidney, mesenchyme, retina and early neurons. We leverage the temporal resolution and sampling depth of these whole-embryo snapshots, together with published data4-8 from earlier timepoints, to construct a rooted tree of cell-type relationships that spans the entirety of prenatal development, from zygote to birth. Throughout this tree, we systematically nominate genes encoding transcription factors and other proteins as candidate drivers of the in vivo differentiation of hundreds of cell types. Remarkably, the most marked temporal shifts in cell states are observed within one hour of birth and presumably underlie the massive physiological adaptations that must accompany the successful transition of a mammalian fetus to life outside the womb.
Collapse
Affiliation(s)
- Chengxiang Qiu
- Department of Genome Sciences, University of Washington, Seattle, WA, USA.
| | - Beth K Martin
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
| | | | - Riza M Daza
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
| | - Truc-Mai Le
- Brotman Baty Institute for Precision Medicine, Seattle, WA, USA
| | - Xingfan Huang
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
- Paul G. Allen School of Computer Science and Engineering, University of Washington, Seattle, WA, USA
| | - Eva K Nichols
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
| | - Megan L Taylor
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
- Brotman Baty Institute for Precision Medicine, Seattle, WA, USA
| | - Olivia Fulton
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
| | - Diana R O'Day
- Brotman Baty Institute for Precision Medicine, Seattle, WA, USA
| | | | - Saskia Ilcisin
- Brotman Baty Institute for Precision Medicine, Seattle, WA, USA
| | - Sanjay Srivatsan
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
- Medical Scientist Training Program, University of Washington, Seattle, WA, USA
| | - Xinxian Deng
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
| | - Christine M Disteche
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
- Department of Medicine, University of Washington, Seattle, WA, USA
| | - William Stafford Noble
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
- Paul G. Allen School of Computer Science and Engineering, University of Washington, Seattle, WA, USA
| | - Nobuhiko Hamazaki
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
- Howard Hughes Medical Institute, Seattle, WA, USA
| | - Cecilia B Moens
- Division of Basic Sciences, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - David Kimelman
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
- Department of Biochemistry, University of Washington, Seattle, WA, USA
| | - Junyue Cao
- Laboratory of Single-Cell Genomics and Population dynamics, The Rockefeller University, New York, NY, USA
| | - Alexander F Schier
- Biozentrum, University of Basel, Basel, Switzerland
- Allen Discovery Center for Cell Lineage Tracing, Seattle, WA, USA
| | - Malte Spielmann
- Max Planck Institute for Molecular Genetics, Berlin, Germany
- Institute of Human Genetics, University Hospitals Schleswig-Holstein, University of Lübeck and Kiel University, Lübeck, Kiel, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg, Lübeck, Kiel, Lübeck, Germany
| | | | - Cole Trapnell
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
- Brotman Baty Institute for Precision Medicine, Seattle, WA, USA
- Allen Discovery Center for Cell Lineage Tracing, Seattle, WA, USA
- Seattle Hub for Synthetic Biology, Seattle, WA, USA
| | - Jay Shendure
- Department of Genome Sciences, University of Washington, Seattle, WA, USA.
- Brotman Baty Institute for Precision Medicine, Seattle, WA, USA.
- Howard Hughes Medical Institute, Seattle, WA, USA.
- Allen Discovery Center for Cell Lineage Tracing, Seattle, WA, USA.
- Seattle Hub for Synthetic Biology, Seattle, WA, USA.
| |
Collapse
|
7
|
Sanchez-Fernandez C, Rodriguez-Outeiriño L, Matias-Valiente L, Ramírez de Acuña F, Franco D, Aránega AE. Understanding Epicardial Cell Heterogeneity during Cardiogenesis and Heart Regeneration. J Cardiovasc Dev Dis 2023; 10:376. [PMID: 37754805 PMCID: PMC10531887 DOI: 10.3390/jcdd10090376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 08/30/2023] [Accepted: 08/31/2023] [Indexed: 09/28/2023] Open
Abstract
The outermost layer of the heart, the epicardium, is an essential cell population that contributes, through epithelial-to-mesenchymal transition (EMT), to the formation of different cell types and provides paracrine signals to the developing heart. Despite its quiescent state during adulthood, the adult epicardium reactivates and recapitulates many aspects of embryonic cardiogenesis in response to cardiac injury, thereby supporting cardiac tissue remodeling. Thus, the epicardium has been considered a crucial source of cell progenitors that offers an important contribution to cardiac development and injured hearts. Although several studies have provided evidence regarding cell fate determination in the epicardium, to date, it is unclear whether epicardium-derived cells (EPDCs) come from specific, and predetermined, epicardial cell subpopulations or if they are derived from a common progenitor. In recent years, different approaches have been used to study cell heterogeneity within the epicardial layer using different experimental models. However, the data generated are still insufficient with respect to revealing the complexity of this epithelial layer. In this review, we summarize the previous works documenting the cellular composition, molecular signatures, and diversity within the developing and adult epicardium.
Collapse
Affiliation(s)
- Cristina Sanchez-Fernandez
- Cardiovascular Development Group, Department of Experimental Biology, Faculty of Experimental Sciences, University of Jaén, 23071 Jaén, Spain; (C.S.-F.); (L.R.-O.); (L.M.-V.); (F.R.d.A.); (D.F.)
- Medina Foundation, Technology Park of Health Sciences, 18016 Granada, Spain
| | - Lara Rodriguez-Outeiriño
- Cardiovascular Development Group, Department of Experimental Biology, Faculty of Experimental Sciences, University of Jaén, 23071 Jaén, Spain; (C.S.-F.); (L.R.-O.); (L.M.-V.); (F.R.d.A.); (D.F.)
- Medina Foundation, Technology Park of Health Sciences, 18016 Granada, Spain
| | - Lidia Matias-Valiente
- Cardiovascular Development Group, Department of Experimental Biology, Faculty of Experimental Sciences, University of Jaén, 23071 Jaén, Spain; (C.S.-F.); (L.R.-O.); (L.M.-V.); (F.R.d.A.); (D.F.)
- Medina Foundation, Technology Park of Health Sciences, 18016 Granada, Spain
| | - Felicitas Ramírez de Acuña
- Cardiovascular Development Group, Department of Experimental Biology, Faculty of Experimental Sciences, University of Jaén, 23071 Jaén, Spain; (C.S.-F.); (L.R.-O.); (L.M.-V.); (F.R.d.A.); (D.F.)
- Medina Foundation, Technology Park of Health Sciences, 18016 Granada, Spain
| | - Diego Franco
- Cardiovascular Development Group, Department of Experimental Biology, Faculty of Experimental Sciences, University of Jaén, 23071 Jaén, Spain; (C.S.-F.); (L.R.-O.); (L.M.-V.); (F.R.d.A.); (D.F.)
- Medina Foundation, Technology Park of Health Sciences, 18016 Granada, Spain
| | - Amelia Eva Aránega
- Cardiovascular Development Group, Department of Experimental Biology, Faculty of Experimental Sciences, University of Jaén, 23071 Jaén, Spain; (C.S.-F.); (L.R.-O.); (L.M.-V.); (F.R.d.A.); (D.F.)
- Medina Foundation, Technology Park of Health Sciences, 18016 Granada, Spain
| |
Collapse
|
8
|
Du J, Yuan X, Deng H, Huang R, Liu B, Xiong T, Long X, Zhang L, Li Y, She Q. Single-cell and spatial heterogeneity landscapes of mature epicardial cells. J Pharm Anal 2023; 13:894-907. [PMID: 37719196 PMCID: PMC10499659 DOI: 10.1016/j.jpha.2023.07.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 07/13/2023] [Accepted: 07/18/2023] [Indexed: 09/19/2023] Open
Abstract
Tbx18, Wt1, and Tcf21 have been identified as epicardial markers during the early embryonic stage. However, the gene markers of mature epicardial cells remain unclear. Single-cell transcriptomic analysis was performed with the Seurat, Monocle, and CellphoneDB packages in R software with standard procedures. Spatial transcriptomics was performed on chilled Visium Tissue Optimization Slides (10x Genomics) and Visium Spatial Gene Expression Slides (10x Genomics). Spatial transcriptomics analysis was performed with Space Ranger software and R software. Immunofluorescence, whole-mount RNA in situ hybridization and X-gal staining were performed to validate the analysis results. Spatial transcriptomics analysis revealed distinct transcriptional profiles and functions between epicardial tissue and non-epicardial tissue. Several gene markers specific to postnatal epicardial tissue were identified, including Msln, C3, Efemp1, and Upk3b. Single-cell transcriptomic analysis revealed that cardiac cells from wildtype mouse hearts (from embryonic day 9.5 to postnatal day 9) could be categorized into six major cell types, which included epicardial cells. Throughout epicardial development, Wt1, Tbx18, and Upk3b were consistently expressed, whereas genes including Msln, C3, and Efemp1 exhibited increased expression during the mature stages of development. Pseudotime analysis further revealed two epicardial cell fates during maturation. Moreover, Upk3b, Msln, Efemp1, and C3 positive epicardial cells were enriched in extracellular matrix signaling. Our results suggested Upk3b, Efemp1, Msln, C3, and other genes were mature epicardium markers. Extracellular matrix signaling was found to play a critical role in the mature epicardium, thus suggesting potential therapeutic targets for heart regeneration in future clinical practice.
Collapse
Affiliation(s)
- Jianlin Du
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
| | - Xin Yuan
- Department of Nephrology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
| | - Haijun Deng
- Key Laboratory of Molecular Biology for Infectious Diseases, Ministry of Education, Institute for Viral Hepatitis, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
| | - Rongzhong Huang
- Precision Medicine Center, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
| | - Bin Liu
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
| | - Tianhua Xiong
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
| | - Xianglin Long
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
| | - Ling Zhang
- Basic Medicine Research and Innovation Center for Novel Target and Therapeutic Intervention, Ministry of Education, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
| | - Yingrui Li
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
| | - Qiang She
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
| |
Collapse
|
9
|
Chauvin M, Meinsohn MC, Dasari S, May P, Iyer S, Nguyen NMP, Oliva E, Lucchini Z, Nagykery N, Kashiwagi A, Mishra R, Maser R, Wells J, Bult CJ, Mitra AK, Donahoe PK, Pépin D. Cancer-associated mesothelial cells are regulated by the anti-Müllerian hormone axis. Cell Rep 2023; 42:112730. [PMID: 37453057 DOI: 10.1016/j.celrep.2023.112730] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 04/27/2023] [Accepted: 06/16/2023] [Indexed: 07/18/2023] Open
Abstract
Cancer-associated mesothelial cells (CAMCs) in the tumor microenvironment are thought to promote growth and immune evasion. We find that, in mouse and human ovarian tumors, cancer cells express anti-Müllerian hormone (AMH) while CAMCs express its receptor AMHR2, suggesting a paracrine axis. Factors secreted by cancer cells induce AMHR2 expression during their reprogramming into CAMCs in mouse and human in vitro models. Overexpression of AMHR2 in the Met5a mesothelial cell line is sufficient to induce expression of immunosuppressive cytokines and growth factors that stimulate ovarian cancer cell growth in an AMH-dependent way. Finally, syngeneic cancer cells implanted in transgenic mice with Amhr2-/- CAMCs grow significantly slower than in wild-type hosts. The cytokine profile of Amhr2-/- tumor-bearing mice is altered and their tumors express less immune checkpoint markers programmed-cell-death 1 (PD1) and cytotoxic T lymphocyte-associated protein 4 (CTLA4). Taken together, these data suggest that the AMH/AMHR2 axis plays a critical role in regulating the pro-tumoral function of CAMCs in ovarian cancer.
Collapse
Affiliation(s)
- M Chauvin
- Pediatric Surgical Research Laboratories, Massachusetts General Hospital, Boston, MA, USA; Department of Surgery, Harvard Medical School, Boston, MA, USA
| | - M-C Meinsohn
- Pediatric Surgical Research Laboratories, Massachusetts General Hospital, Boston, MA, USA; Department of Surgery, Harvard Medical School, Boston, MA, USA
| | - S Dasari
- Indiana University School of Medicine-Bloomington, Indiana University, Bloomington, IN, USA
| | - P May
- Pediatric Surgical Research Laboratories, Massachusetts General Hospital, Boston, MA, USA
| | - S Iyer
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA
| | - N M P Nguyen
- Pediatric Surgical Research Laboratories, Massachusetts General Hospital, Boston, MA, USA; Department of Surgery, Harvard Medical School, Boston, MA, USA
| | - E Oliva
- Department of Pathology, Massachusetts General Hospital, Boston, MA, USA
| | - Z Lucchini
- Pediatric Surgical Research Laboratories, Massachusetts General Hospital, Boston, MA, USA
| | - N Nagykery
- Pediatric Surgical Research Laboratories, Massachusetts General Hospital, Boston, MA, USA; Department of Surgery, Harvard Medical School, Boston, MA, USA
| | - A Kashiwagi
- Pediatric Surgical Research Laboratories, Massachusetts General Hospital, Boston, MA, USA; Department of Surgery, Harvard Medical School, Boston, MA, USA
| | - R Mishra
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA
| | - R Maser
- Mouse Genome Informatics, The Jackson Laboratory, Bar Harbor, ME, USA
| | - J Wells
- Mouse Genome Informatics, The Jackson Laboratory, Bar Harbor, ME, USA
| | - C J Bult
- Mouse Genome Informatics, The Jackson Laboratory, Bar Harbor, ME, USA
| | - A K Mitra
- Indiana University School of Medicine-Bloomington, Indiana University, Bloomington, IN, USA
| | - Patricia K Donahoe
- Pediatric Surgical Research Laboratories, Massachusetts General Hospital, Boston, MA, USA; Department of Surgery, Harvard Medical School, Boston, MA, USA
| | - D Pépin
- Pediatric Surgical Research Laboratories, Massachusetts General Hospital, Boston, MA, USA; Department of Surgery, Harvard Medical School, Boston, MA, USA; Mouse Genome Informatics, The Jackson Laboratory, Bar Harbor, ME, USA.
| |
Collapse
|
10
|
Xu M, Yang A, Xia J, Jiang J, Liu CF, Ye Z, Ma J, Yang S. Protein glycosylation in urine as a biomarker of diseases. Transl Res 2023; 253:95-107. [PMID: 35952983 DOI: 10.1016/j.trsl.2022.08.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 07/28/2022] [Accepted: 08/02/2022] [Indexed: 02/01/2023]
Abstract
Human body fluids have become an indispensable resource for clinical research, diagnosis and prognosis. Urine is widely used to discover disease-specific glycoprotein biomarkers because of its recurrently non-invasive collection and disease-indicating properties. While urine is an unstable fluid in that its composition changes with ingested nutrients and further as it is excreted through micturition, urinary proteins are more stable and their abnormal glycosylation is associated with diseases. It is known that aberrant glycosylation can define tumor malignancy and indicate disease initiation and progression. However, a thorough and translational survey of urinary glycosylation in diseases has not been performed. In this article, we evaluate the clinical applications of urine, introduce methods for urine glycosylation analysis, and discuss urine glycoprotein biomarkers. We emphasize the importance of mining urinary glycoproteins and searching for disease-specific glycosylation in various diseases (including cancer, neurodegenerative diseases, diabetes, and viral infections). With advances in mass spectrometry-based glycomics/glycoproteomics/glycopeptidomics, characterization of disease-specific glycosylation will optimistically lead to the discovery of disease-related urinary biomarkers with better sensitivity and specificity in the near future.
Collapse
Affiliation(s)
- Mingming Xu
- Center for Clinical Mass Spectrometry, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, China
| | - Arthur Yang
- Center for Clinical Mass Spectrometry, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, China
| | - Jun Xia
- Clinical Laboratory Center, Zhejiang Provincial People's Hospital, Hangzhou, Zhejiang, China
| | - Junhong Jiang
- Department of Pulmonary and Critical Care Medicine, Dushu Lake Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Chun-Feng Liu
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Zhenyu Ye
- Department of General Surgery, Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Junfeng Ma
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Georgetown University, Washington, District of Columbia.
| | - Shuang Yang
- Center for Clinical Mass Spectrometry, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, China.
| |
Collapse
|
11
|
Neirijnck Y, Sararols P, Kühne F, Mayère C, Weerasinghe Arachchige LC, Regard V, Nef S, Schedl A. Single-cell transcriptomic profiling redefines the origin and specification of early adrenogonadal progenitors. Cell Rep 2023; 42:112191. [PMID: 36862551 DOI: 10.1016/j.celrep.2023.112191] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 12/13/2022] [Accepted: 02/14/2023] [Indexed: 03/03/2023] Open
Abstract
Adrenal cortex and gonads represent the two major steroidogenic organs in mammals. Both tissues are considered to share a common developmental origin characterized by the expression of Nr5a1/Sf1. The precise origin of adrenogonadal progenitors and the processes driving differentiation toward the adrenal or gonadal fate remain, however, elusive. Here, we provide a comprehensive single-cell transcriptomic atlas of early mouse adrenogonadal development including 52 cell types belonging to twelve major cell lineages. Trajectory reconstruction reveals that adrenogonadal cells emerge from the lateral plate rather than the intermediate mesoderm. Surprisingly, we find that gonadal and adrenal fates have already diverged prior to Nr5a1 expression. Finally, lineage separation into gonadal and adrenal fates involves canonical versus non-canonical Wnt signaling and differential expression of Hox patterning genes. Thus, our study provides important insights into the molecular programs of adrenal and gonadal fate choice and will be a valuable resource for further research into adrenogonadal ontogenesis.
Collapse
Affiliation(s)
- Yasmine Neirijnck
- Department of Genetic Medicine and Development, University of Geneva, 1211 Geneva, Switzerland; Université Côte d'Azur, CNRS, INSERM, IBV, 06108 Nice, France.
| | - Pauline Sararols
- Department of Genetic Medicine and Development, University of Geneva, 1211 Geneva, Switzerland
| | - Françoise Kühne
- Department of Genetic Medicine and Development, University of Geneva, 1211 Geneva, Switzerland
| | - Chloé Mayère
- Department of Genetic Medicine and Development, University of Geneva, 1211 Geneva, Switzerland
| | | | - Violaine Regard
- Department of Genetic Medicine and Development, University of Geneva, 1211 Geneva, Switzerland
| | - Serge Nef
- Department of Genetic Medicine and Development, University of Geneva, 1211 Geneva, Switzerland.
| | - Andreas Schedl
- Université Côte d'Azur, CNRS, INSERM, IBV, 06108 Nice, France.
| |
Collapse
|
12
|
Zia S, Djalali-Cuevas A, Pflaum M, Hegermann J, Dipresa D, Kalozoumis P, Kouvaka A, Burgwitz K, Andriopoulou S, Repanas A, Will F, Grote K, Schrimpf C, Toumpaniari S, Mueller M, Glasmacher B, Haverich A, Morticelli L, Korossis S. Development of a dual-component infection-resistant arterial replacement for small-caliber reconstructions: A proof-of-concept study. Front Bioeng Biotechnol 2023; 11:957458. [PMID: 36741762 PMCID: PMC9889865 DOI: 10.3389/fbioe.2023.957458] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 01/02/2023] [Indexed: 01/19/2023] Open
Abstract
Introduction: Synthetic vascular grafts perform poorly in small-caliber (<6mm) anastomoses, due to intimal hyperplasia and thrombosis, whereas homografts are associated with limited availability and immunogenicity, and bioprostheses are prone to aneurysmal degeneration and calcification. Infection is another important limitation with vascular grafting. This study developed a dual-component graft for small-caliber reconstructions, comprising a decellularized tibial artery scaffold and an antibiotic-releasing, electrospun polycaprolactone (PCL)/polyethylene glycol (PEG) blend sleeve. Methods: The study investigated the effect of nucleases, as part of the decellularization technique, and two sterilization methods (peracetic acid and γ-irradiation), on the scaffold's biological and biomechanical integrity. It also investigated the effect of different PCL/PEG ratios on the antimicrobial, biological and biomechanical properties of the sleeves. Tibial arteries were decellularized using Triton X-100 and sodium-dodecyl-sulfate. Results: The scaffolds retained the general native histoarchitecture and biomechanics but were depleted of glycosaminoglycans. Sterilization with peracetic acid depleted collagen IV and produced ultrastructural changes in the collagen and elastic fibers. The two PCL/PEG ratios used (150:50 and 100:50) demonstrated differences in the structural, biomechanical and antimicrobial properties of the sleeves. Differences in the antimicrobial activity were also found between sleeves fabricated with antibiotics supplemented in the electrospinning solution, and sleeves soaked in antibiotics. Discussion: The study demonstrated the feasibility of fabricating a dual-component small-caliber graft, comprising a scaffold with sufficient biological and biomechanical functionality, and an electrospun PCL/PEG sleeve with tailored biomechanics and antibiotic release.
Collapse
Affiliation(s)
- Sonia Zia
- Lower Saxony Centre for Biomedical Engineering Implant Research and Development, Hannover Medical School, Hannover, Germany
| | - Adrian Djalali-Cuevas
- Lower Saxony Centre for Biomedical Engineering Implant Research and Development, Hannover Medical School, Hannover, Germany
| | - Michael Pflaum
- Lower Saxony Centre for Biomedical Engineering Implant Research and Development, Hannover Medical School, Hannover, Germany
| | - Jan Hegermann
- Institute of Functional and Applied Anatomy, Research Core Unit Electron Microscopy, Hannover Medical School, Hannover, Germany
| | - Daniele Dipresa
- Lower Saxony Centre for Biomedical Engineering Implant Research and Development, Hannover Medical School, Hannover, Germany
| | - Panagiotis Kalozoumis
- Lower Saxony Centre for Biomedical Engineering Implant Research and Development, Hannover Medical School, Hannover, Germany
| | - Artemis Kouvaka
- Lower Saxony Centre for Biomedical Engineering Implant Research and Development, Hannover Medical School, Hannover, Germany
| | - Karin Burgwitz
- Lower Saxony Centre for Biomedical Engineering Implant Research and Development, Hannover Medical School, Hannover, Germany
| | - Sofia Andriopoulou
- Lower Saxony Centre for Biomedical Engineering Implant Research and Development, Hannover Medical School, Hannover, Germany
| | - Alexandros Repanas
- Institute for Multiphase Processes, Leibniz University Hannover, Hannover, Germany
| | - Fabian Will
- LLS ROWIAK LaserLabSolutions GmbH, Hannover, Germany
| | - Karsten Grote
- Cardiology and Angiology, Philipps-University Marburg, Marburg, Germany
| | - Claudia Schrimpf
- Department of Cardiothoracic, Transplantation and Vascular Surgery, Hannover Medical School, Hannover, Germany
| | - Sotiria Toumpaniari
- Cardiopulmonary Regenerative Engineering Group (CARE), Centre for Biological Engineering, Loughborough University, Loughborough, United Kingdom,Wolfson School of Mechanical, Electrical and Manufacturing Engineering, Loughborough University, Loughborough, United Kingdom
| | - Marc Mueller
- Institute for Multiphase Processes, Leibniz University Hannover, Hannover, Germany
| | - Birgit Glasmacher
- Lower Saxony Centre for Biomedical Engineering Implant Research and Development, Hannover Medical School, Hannover, Germany,Institute for Multiphase Processes, Leibniz University Hannover, Hannover, Germany
| | - Axel Haverich
- Lower Saxony Centre for Biomedical Engineering Implant Research and Development, Hannover Medical School, Hannover, Germany,Department of Cardiothoracic, Transplantation and Vascular Surgery, Hannover Medical School, Hannover, Germany
| | - Lucrezia Morticelli
- Lower Saxony Centre for Biomedical Engineering Implant Research and Development, Hannover Medical School, Hannover, Germany
| | - Sotirios Korossis
- Lower Saxony Centre for Biomedical Engineering Implant Research and Development, Hannover Medical School, Hannover, Germany,Department of Cardiothoracic, Transplantation and Vascular Surgery, Hannover Medical School, Hannover, Germany,Cardiopulmonary Regenerative Engineering Group (CARE), Centre for Biological Engineering, Loughborough University, Loughborough, United Kingdom,Wolfson School of Mechanical, Electrical and Manufacturing Engineering, Loughborough University, Loughborough, United Kingdom,*Correspondence: Sotirios Korossis,
| |
Collapse
|
13
|
Al-Sharkawi M, Calonga-Solís V, Dressler FF, Busch H, Hiort O, Werner R. Persistence of foetal testicular features in patients with defective androgen signalling. Eur J Endocrinol 2023; 188:7017644. [PMID: 36721956 DOI: 10.1093/ejendo/lvad007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 01/09/2023] [Accepted: 01/26/2023] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Congenital defects of androgen synthesis or action in 46,XY individuals can result in impaired virilisation, despite the apparent testicular development. In a recent case, report of a young adult with complete androgen insensitivity syndrome (CAIS), tumourous gonadal tissue was shown to express HSD17B3 in Sertoli cells (SCs) and not in Leydig cells (LCs). This expression pattern differs from the typical adult human testis and resembles a foetal mouse testis, suggesting an underlying testicular development and function defect. Here, we investigate the effect of altered androgen signalling in gonads from five 46,XY individuals with defects in androgen synthesis or action. METHODS Gonadal tissue sections from four patients with CAIS, one with CYP17A1 deficiency, and one control were immunostained for LC developmental and steroidogenic markers. The expression of some of these markers during development was investigated by reanalysing previously published single-cell RNA sequencing (scRNA-seq) data from normal human testicular tissues. RESULTS All gonadal tissues from the patients show an exclusive expression of HSD17B3 in SCs and an expression of the foetal/immature LC marker DLK1 in a subset of LCs, suggesting an androgen-dependent differentiation defect of adult SCs and LCs. Furthermore, reanalysis of scRNA-seq data reveals an expression of HSD17B3 in foetal and neonatal SCs that is downregulated in adult SCs. CONCLUSIONS Androgen signalling may affect the differentiation of adults, but possibly not foetal SCs or LCs, and may induce a shift of testosterone production from the tubular compartment in the foetal phase to the interstitial compartment in the adult phase.
Collapse
Affiliation(s)
- Mostafa Al-Sharkawi
- Division of Paediatric Endocrinology and Diabetology, Department of Paediatrics, University of Lübeck, 23562 Lübeck, Germany
- Biochemical Genetics Department, Human Genetics and Genome Research Institute, 12622 Dokki, Cairo, Egypt
| | - Verónica Calonga-Solís
- Division of Paediatric Endocrinology and Diabetology, Department of Paediatrics, University of Lübeck, 23562 Lübeck, Germany
- Medical Systems Biology Division, Lübeck Institute of Experimental Dermatology and Institute for Cardiogenetics, University of Lübeck, 23562 Lübeck, Germany
| | - Franz F Dressler
- Institute of Pathology, University Hospital Schleswig-Holstein, Campus Lübeck, 23562 Lübeck, Germany
- Institute of Pathology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117, Berlin, Germany
| | - Hauke Busch
- Medical Systems Biology Division, Lübeck Institute of Experimental Dermatology and Institute for Cardiogenetics, University of Lübeck, 23562 Lübeck, Germany
| | - Olaf Hiort
- Division of Paediatric Endocrinology and Diabetology, Department of Paediatrics, University of Lübeck, 23562 Lübeck, Germany
| | - Ralf Werner
- Division of Paediatric Endocrinology and Diabetology, Department of Paediatrics, University of Lübeck, 23562 Lübeck, Germany
- Institute of Molecular Medicine, University of Lübeck, 23562 Lübeck, Germany
| |
Collapse
|
14
|
Li X, Su Q, Li W, Zhang X, Ran J. Analysis and identification of potential key genes in hepatic ischemia-reperfusion injury. ANNALS OF TRANSLATIONAL MEDICINE 2022; 10:1375. [PMID: 36660667 PMCID: PMC9843403 DOI: 10.21037/atm-22-6171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 12/20/2022] [Indexed: 01/01/2023]
Abstract
Background Hepatic ischemia-reperfusion injury (HIRI) is an unavoidable surgical complication after liver transplantation, but current HIRI treatments cannot achieve satisfactory clinical outcomes. Thus, safer and more effective prevention and treatment methods need to be explored. Methods Transcriptome messenger ribonucleic acid (mRNA) and long non-coding RNA (lncRNA) sequencing data were obtained from male Sprague-Dawley rats, and these data were used to identify the differentially expressed genes (DEGs) and differentially expressed lncRNAs (DE-lncRNAs) between the HIRI and control samples. A protein-protein interaction (PPI) network was also constructed for the DE-mRNAs to identify candidate genes, and the receiver operating characteristic curves of the 21 candidate genes were plotted to evaluate the diagnostic value of the candidate genes for HIRI. A random forest (RF) model, support vector machine model and generalized linear model were constructed based on the candidate genes. A gene set enrichment analysis (GSEA) of the key genes was conducted to determine the enriched pathways in the high expression groups. The miRWalk and miRanda database were used to constructed the lncRNA-miRNA-mRNA network. Finally, the expressions of the key genes were verified by quantitative real-time polymerase chain reaction (qRT-PCR). Results A total of 256 DEGs and 67 DE-lncRNAs were identified in the HIRI and control samples. To explore the interactions between the DE-mRNAs, a PPI network of 130 DEGs was constructed. Further, 21 genes were selected as the candidate genes. Subsequently, 6 genes [i.e., Keratin-14 (Krt14), Uroplakin 3B (Upk3b), Keratin 7 (Krt7), Cadherin 3 (Cdh3), mesothelin (Msln), and Glypican 3 (Gpc3)] in the RF model were defined as the key genes. The GSEA results indicated that these key genes were enriched in the terms of extracellular structure organization, and extracellular matrix organization. Moreover, a lncRNA-miRNA-mRNA network was constructed with 4 lncRNAs, 5 mRNAs, and 11 miRNAs. Finally, the results indicated that the expression of Krt14, Upk3b, Msln, and Gpc3 were more highly expressed in the control samples than the HIRI samples. Conclusions A total of 6 key genes (i.e., Krt14, Upk3b, Krt7, Cdh3, Msln, and Gpc3) were identified. Our findings provide novel ideas for the diagnosis and treatment of HIRI.
Collapse
Affiliation(s)
- Xiaokai Li
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Qiuming Su
- Department of Hepatopancreatobiliary Surgery, The Affiliated Calmette Hospital of Kunming Medical University, Kunming, China
| | - Wang Li
- Department of Hepatopancreatobiliary Surgery, The Affiliated Calmette Hospital of Kunming Medical University, Kunming, China
| | - Xibing Zhang
- Department of Hepatopancreatobiliary Surgery, The Affiliated Calmette Hospital of Kunming Medical University, Kunming, China
| | - Jianghua Ran
- Department of Hepatopancreatobiliary Surgery, The Affiliated Calmette Hospital of Kunming Medical University, Kunming, China
| |
Collapse
|
15
|
Lennartz M, Atug D, Dwertmann Rico S, Reiswich V, Viehweger F, Büscheck F, Kluth M, Hube-Magg C, Hinsch A, Bernreuther C, Sauter G, Burandt E, Marx AH, Krech T, Simon R, Minner S, Clauditz TS, Jacobsen F, Lebok P, Gorbokon N, Möller K, Steurer S, Fraune C. Analysis of More than 16,000 Human Tumor and Normal Tissues Identifies Uroplakin 3B as a Useful Diagnostic Marker for Mesothelioma and Normal Mesothelial Cells. Diagnostics (Basel) 2022; 12:2516. [PMID: 36292206 PMCID: PMC9600073 DOI: 10.3390/diagnostics12102516] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 09/20/2022] [Accepted: 10/10/2022] [Indexed: 10/02/2023] Open
Abstract
Uroplakin 3B (Upk3b) is involved in stabilizing and strengthening the urothelial cell layer of the bladder. Based on RNA expression studies, Upk3b is expressed in a limited number of normal and tumor tissues. The potential use of Upk3b as a diagnostic or prognostic marker in tumor diagnosis has not yet been extensively investigated. A tissue microarray containing 17,693 samples from 151 different tumor types/subtypes and 608 samples of 76 different normal tissue types was analyzed by immunohistochemistry. In normal tissues, Upk3b expression was largely limited to mesothelial cells, urothelial umbrella cells, and amnion cells. In tumor tissues, Upk3b was detectable in only 17 of 151 (11.3%) of tumor types. Upk3b expression was most frequent in mesotheliomas (82.1% of epithelioid and 30.8% of biphasic) and in urothelial tumors of the urinary bladder, where the positivity rate decreased from 61.9% in pTaG2 (low grade) to 58.0% in pTaG3 (high grade) and 14.6% in pT2-4 cancers. Among pT2-4 urothelial carcinomas, Upk3b staining was unrelated to tumor stage, lymph node status, and patient prognosis. Less commonly, Upk3b expression was also seen in Brenner tumors of the ovary (10.8%), as well as in four other subtypes of ovarian cancer (0.9-10.6%). Four additional tumor entities showed a weak to moderate Upk3b positivity in less than 5% of cases. In summary, Upk3b immunohistochemistry is a useful diagnostic tool for the distinction of mesotheliomas from other thoracic tumors and the visualization of normal mesothelial and umbrella cells.
Collapse
Affiliation(s)
- Maximilian Lennartz
- Department of Pathology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Dennis Atug
- Department of Pathology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | | | - Viktor Reiswich
- Department of Pathology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Florian Viehweger
- Department of Pathology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Franziska Büscheck
- Department of Pathology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Martina Kluth
- Department of Pathology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Claudia Hube-Magg
- Department of Pathology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Andrea Hinsch
- Department of Pathology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Christian Bernreuther
- Department of Pathology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Guido Sauter
- Department of Pathology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Eike Burandt
- Department of Pathology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Andreas H. Marx
- Department of Pathology, Academic Hospital Fuerth, 90766 Fuerth, Germany
| | - Till Krech
- Department of Pathology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
- Department of Pathology, Clinical Center Osnabrueck, 49076 Osnabrueck, Germany
| | - Ronald Simon
- Department of Pathology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Sarah Minner
- Department of Pathology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Till S. Clauditz
- Department of Pathology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Frank Jacobsen
- Department of Pathology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Patrick Lebok
- Department of Pathology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
- Department of Pathology, Clinical Center Osnabrueck, 49076 Osnabrueck, Germany
| | - Natalia Gorbokon
- Department of Pathology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Katharina Möller
- Department of Pathology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Stefan Steurer
- Department of Pathology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Christoph Fraune
- Department of Pathology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| |
Collapse
|
16
|
Chen T, Song S, Jiang H, Lian H, Hu S. Single Cell Sequencing Reveals Mechanisms of Persistent Truncus Arteriosus Formation after PDGFRα and PDGFRβ Double Knockout in Cardiac Neural Crest Cells. Genes (Basel) 2022; 13:genes13101708. [PMID: 36292593 PMCID: PMC9601305 DOI: 10.3390/genes13101708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 09/13/2022] [Accepted: 09/16/2022] [Indexed: 11/16/2022] Open
Abstract
Persistent truncus arteriosus (PTA) is an uncommon and complex congenital cardiac malformation accounting for about 1.2% of all congenital heart diseases (CHDs), which is caused by a deficiency in the embryonic heart outflow tract’s (OFT) septation and remodeling. PDGFRα and PDGFRβ double knockout (DKO) in cardiac neural crest cells (CNCCs) has been reported to cause PTA, but the underlying mechanisms remain unclear. Here, we constructed a PTA mouse model with PDGFRα and PDGFRβ double knockout in Pax3+ CNCCs and described the condensation failure into OFT septum of CNCC-derived cells due to disturbance of cell polarity in the DKO group. In addition, we further explored the mechanism with single-cell RNA sequencing. We found that two main cell differentiation trajectories into vascular smooth muscle cells (VSMCs) from cardiomyocytes (CMs) and mesenchymal cells (MSs), respectively, were interrupted in the DKO group. The process of CM differentiation into VSMC stagnated in a transitional CM I-like state, which contributed to the failure of OFT remodeling and muscular septum formation. On the other hand, a Penk+ transitional MS II cluster closely related to cell condensation into the OFT septum disappeared, which led to the OFT’s septation absence directly. In conclusion, the disturbance of CNCC-derived cells caused by PDGFRα and PDGFRβ knockout can lead to the OFT septation disorder and the occurrence of PTA.
Collapse
Affiliation(s)
- Tianyun Chen
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100006, China
| | - Shen Song
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100006, China
| | - Haobin Jiang
- Division of Thoracic Surgery, School of Medicine, First Affiliated Hospital, Zhejiang University, Hangzhou 310027, China
| | - Hong Lian
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100006, China
| | - Shengshou Hu
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100006, China
- Correspondence:
| |
Collapse
|
17
|
Calonga-Solís V, Fabbri-Scallet H, Ott F, Al-Sharkawi M, Künstner A, Wünsch L, Hiort O, Busch H, Werner R. MYRF: A New Regulator of Cardiac and Early Gonadal Development—Insights from Single Cell RNA Sequencing Analysis. J Clin Med 2022; 11:jcm11164858. [PMID: 36013096 PMCID: PMC9409872 DOI: 10.3390/jcm11164858] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 08/16/2022] [Accepted: 08/17/2022] [Indexed: 11/16/2022] Open
Abstract
De novo variants in the myelin regulatory factor (MYRF), a transcription factor involved in the differentiation of oligodendrocytes, have been linked recently to the cardiac and urogenital syndrome, while familiar variants are associated with nanophthalmos. Here, we report for the first time on a patient with a de novo stop-gain variant in MYRF (p.Q838*) associated with Scimitar syndrome, 46,XY partial gonadal dysgenesis (GD) and severe hyperopia. Since variants in MYRF have been described in both 46,XX and 46,XY GD, we assumed a role of MYRF in the early development of the bipotential gonad. We used publicly available single cell sequencing data of human testis and ovary from different developmental stages and analysed them for MYRF expression. We identified MYRF expression in the subset of coelomic epithelial cells at stages of gonadal ridge development in 46,XX and 46,XY individuals. Differential gene expression analysis revealed significantly upregulated genes. Within these, we identified CITED2 as a gene containing a MYRF binding site. It has been shown that Cited2−/− mice have gonadal defects in both testis and ovary differentiation, as well as defects in heart development and establishment of the left–right axis. This makes MYRF a potential candidate as an early regulator of gonadal and heart development via upregulation of the transcriptional cofactor CITED2.
Collapse
Affiliation(s)
- Verónica Calonga-Solís
- Division of Paediatric Endocrinology and Diabetes, Department of Paediatric and Adolescent Medicine, University of Lübeck, 23562 Lübeck, Germany
- Medical Systems Biology Division, Lübeck Institute of Experimental Dermatology, University of Lübeck, 23562 Lübeck, Germany
| | - Helena Fabbri-Scallet
- Division of Paediatric Endocrinology and Diabetes, Department of Paediatric and Adolescent Medicine, University of Lübeck, 23562 Lübeck, Germany
- Center for Molecular Biology and Genetic Engineering—CBMEG, State University of Campinas, Campinas 13083-875, Brazil
| | - Fabian Ott
- Medical Systems Biology Division, Lübeck Institute of Experimental Dermatology, University of Lübeck, 23562 Lübeck, Germany
| | - Mostafa Al-Sharkawi
- Division of Paediatric Endocrinology and Diabetes, Department of Paediatric and Adolescent Medicine, University of Lübeck, 23562 Lübeck, Germany
- Biochemical Genetics Department, Human Genetics and Genome Research Institute, National Research Centre, Dokki, Cairo 12622, Egypt
| | - Axel Künstner
- Medical Systems Biology Division, Lübeck Institute of Experimental Dermatology, University of Lübeck, 23562 Lübeck, Germany
| | - Lutz Wünsch
- Department of Pediatric Surgery, University of Lübeck, 23562 Lübeck, Germany
| | - Olaf Hiort
- Division of Paediatric Endocrinology and Diabetes, Department of Paediatric and Adolescent Medicine, University of Lübeck, 23562 Lübeck, Germany
| | - Hauke Busch
- Medical Systems Biology Division, Lübeck Institute of Experimental Dermatology, University of Lübeck, 23562 Lübeck, Germany
| | - Ralf Werner
- Division of Paediatric Endocrinology and Diabetes, Department of Paediatric and Adolescent Medicine, University of Lübeck, 23562 Lübeck, Germany
- Institute of Molecular Medicine, University of Lübeck, 23562 Lübeck, Germany
- Correspondence:
| |
Collapse
|
18
|
Capeling MM, Huang S, Childs CJ, Wu JH, Tsai YH, Wu A, Garg N, Holloway EM, Sundaram N, Bouffi C, Helmrath M, Spence JR. Suspension culture promotes serosal mesothelial development in human intestinal organoids. Cell Rep 2022; 38:110379. [PMID: 35172130 PMCID: PMC9002973 DOI: 10.1016/j.celrep.2022.110379] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 09/08/2021] [Accepted: 01/24/2022] [Indexed: 02/07/2023] Open
Abstract
Pluripotent-stem-cell-derived human intestinal organoids (HIOs) model some aspects of intestinal development and disease, but current culture methods do not fully recapitulate the diverse cell types and complex organization of the human intestine and are reliant on 3D extracellular matrix or hydrogel systems, which limit experimental control and translational potential for regenerative medicine. We describe suspension culture as a simple, low-maintenance method for culturing HIOs and for promoting in vitro differentiation of an organized serosal mesothelial layer that is similar to primary human intestinal serosal mesothelium based on single-cell RNA sequencing and histological analysis. Functionally, HIO serosal mesothelium has the capacity to differentiate into smooth-muscle-like cells and exhibits fibrinolytic activity. An inhibitor screen identifies Hedgehog and WNT signaling as regulators of human serosal mesothelial differentiation. Collectively, suspension HIOs represent a three-dimensional model to study the human serosal mesothelium.
Collapse
Affiliation(s)
- Meghan M Capeling
- Department of Biomedical Engineering, University of Michigan College of Engineering, Ann Arbor, MI 48109, USA
| | - Sha Huang
- Department of Internal Medicine, Gastroenterology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Charlie J Childs
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Joshua H Wu
- Department of Internal Medicine, Gastroenterology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Yu-Hwai Tsai
- Department of Internal Medicine, Gastroenterology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Angeline Wu
- Department of Internal Medicine, Gastroenterology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Neil Garg
- School of Kinesiology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Emily M Holloway
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Nambirajan Sundaram
- Division of Pediatric General and Thoracic Surgery Cincinnati Children's Hospital Research Foundation, Cincinnati, OH 45229, USA
| | - Carine Bouffi
- Division of Pediatric General and Thoracic Surgery Cincinnati Children's Hospital Research Foundation, Cincinnati, OH 45229, USA
| | - Michael Helmrath
- Division of Pediatric General and Thoracic Surgery Cincinnati Children's Hospital Research Foundation, Cincinnati, OH 45229, USA
| | - Jason R Spence
- Department of Biomedical Engineering, University of Michigan College of Engineering, Ann Arbor, MI 48109, USA; Department of Internal Medicine, Gastroenterology, University of Michigan Medical School, Ann Arbor, MI 48109, USA; Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA.
| |
Collapse
|
19
|
Li S, Chen LN, Zhu HJ, Feng X, Xie FY, Luo SM, Ou XH, Ma JY. Single-cell RNA sequencing analysis of mouse follicular somatic cells†. Biol Reprod 2021; 105:1234-1245. [PMID: 34467391 DOI: 10.1093/biolre/ioab163] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Revised: 06/29/2021] [Accepted: 08/19/2021] [Indexed: 12/31/2022] Open
Abstract
Within the development of ovarian follicle, in addition to cell proliferation and differentiation, sophisticated cell-cell cross talks are established among follicular somatic cells such as granulosa cells (GCs) and theca cells. To systematically reveal the cell differentiation and signal transductions in follicular somatic cells, we collected the mouse follicular somatic cells from secondary to ovulatory stage, and analyzed the single cell transcriptomes. Having data filtered and screened, we found 6883 high variable genes in 4888 single cells. Then follicular somatic cells were clustered into 26 cell clusters, including 18 GC clusters, 4 theca endocrine cell (TEC) clusters, and 4 other somatic cell clusters, which include immune cells and Acta2 positive theca externa cells. From our data, we found there was metabolic reprogramming happened during GC differentiation. We also found both Cyp19a1 and Cyp11a1 could be expressed in TECs. We analyzed the expression patterns of genes associated with cell-cell interactions such as steroid hormone receptor genes, insulin signaling genes, and cytokine/transformation growth factor beta associated genes in all cell clusters. Lastly, we clustered the highly variable genes into 300 gene clusters, which could be used to search new genes involved in follicle development. These transcriptomes of follicular somatic cells provide us potential clues to reveal how mammals regulating follicle development and could help us find targets to improve oocyte quality for women with low fertility.
Collapse
Affiliation(s)
- Sen Li
- Fertility Preservation Lab, Guangdong-Hong Kong Metabolism & Reproduction Joint Laboratory, Guangdong Second Provincial General Hospital, Guangzhou, China.,Fertility Preservation Lab, Reproductive Medicine Center, Guangdong Second Provincial General Hospital, Guangzhou, China
| | - Lei-Ning Chen
- Fertility Preservation Lab, Guangdong-Hong Kong Metabolism & Reproduction Joint Laboratory, Guangdong Second Provincial General Hospital, Guangzhou, China.,Fertility Preservation Lab, Reproductive Medicine Center, Guangdong Second Provincial General Hospital, Guangzhou, China
| | - Hai-Jing Zhu
- Fertility Preservation Lab, Reproductive Medicine Center, Guangdong Second Provincial General Hospital, Guangzhou, China.,Teaching Center in Guangdong Second Provincial General Hospital, University of South China, Guangzhou, China
| | - Xie Feng
- Fertility Preservation Lab, Guangdong-Hong Kong Metabolism & Reproduction Joint Laboratory, Guangdong Second Provincial General Hospital, Guangzhou, China.,Fertility Preservation Lab, Reproductive Medicine Center, Guangdong Second Provincial General Hospital, Guangzhou, China.,The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Feng-Yun Xie
- Fertility Preservation Lab, Guangdong-Hong Kong Metabolism & Reproduction Joint Laboratory, Guangdong Second Provincial General Hospital, Guangzhou, China.,Fertility Preservation Lab, Reproductive Medicine Center, Guangdong Second Provincial General Hospital, Guangzhou, China
| | - Shi-Ming Luo
- Fertility Preservation Lab, Guangdong-Hong Kong Metabolism & Reproduction Joint Laboratory, Guangdong Second Provincial General Hospital, Guangzhou, China.,Fertility Preservation Lab, Reproductive Medicine Center, Guangdong Second Provincial General Hospital, Guangzhou, China
| | - Xiang-Hong Ou
- Fertility Preservation Lab, Guangdong-Hong Kong Metabolism & Reproduction Joint Laboratory, Guangdong Second Provincial General Hospital, Guangzhou, China.,Fertility Preservation Lab, Reproductive Medicine Center, Guangdong Second Provincial General Hospital, Guangzhou, China.,Teaching Center in Guangdong Second Provincial General Hospital, University of South China, Guangzhou, China.,Bioland Laboratory, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, China
| | - Jun-Yu Ma
- Fertility Preservation Lab, Guangdong-Hong Kong Metabolism & Reproduction Joint Laboratory, Guangdong Second Provincial General Hospital, Guangzhou, China.,Fertility Preservation Lab, Reproductive Medicine Center, Guangdong Second Provincial General Hospital, Guangzhou, China
| |
Collapse
|
20
|
Beckers A, Fuhl F, Ott T, Boldt K, Brislinger MM, Walentek P, Schuster-Gossler K, Hegermann J, Alten L, Kremmer E, Przykopanski A, Serth K, Ueffing M, Blum M, Gossler A. The highly conserved FOXJ1 target CFAP161 is dispensable for motile ciliary function in mouse and Xenopus. Sci Rep 2021; 11:13333. [PMID: 34172766 PMCID: PMC8233316 DOI: 10.1038/s41598-021-92495-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Accepted: 06/08/2021] [Indexed: 12/14/2022] Open
Abstract
Cilia are protrusions of the cell surface and composed of hundreds of proteins many of which are evolutionary and functionally well conserved. In cells assembling motile cilia the expression of numerous ciliary components is under the control of the transcription factor FOXJ1. Here, we analyse the evolutionary conserved FOXJ1 target CFAP161 in Xenopus and mouse. In both species Cfap161 expression correlates with the presence of motile cilia and depends on FOXJ1. Tagged CFAP161 localises to the basal bodies of multiciliated cells of the Xenopus larval epidermis, and in mice CFAP161 protein localises to the axoneme. Surprisingly, disruption of the Cfap161 gene in both species did not lead to motile cilia-related phenotypes, which contrasts with the conserved expression in cells carrying motile cilia and high sequence conservation. In mice mutation of Cfap161 stabilised the mutant mRNA making genetic compensation triggered by mRNA decay unlikely. However, genes related to microtubules and cilia, microtubule motor activity and inner dyneins were dysregulated, which might buffer the Cfap161 mutation.
Collapse
Affiliation(s)
- Anja Beckers
- Institute for Molecular Biology, OE5250, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - Franziska Fuhl
- Institute of Biology, University of Hohenheim, Garbenstraße 30, 70593, Stuttgart, Germany
| | - Tim Ott
- Institute of Biology, University of Hohenheim, Garbenstraße 30, 70593, Stuttgart, Germany
| | - Karsten Boldt
- Institute of Ophthalmic Research, Center for Ophthalmology, University of Tübingen, Elfriede-Aulhorn-Strasse 7, 72076, Tübingen, Germany
| | - Magdalena Maria Brislinger
- Institute of Biology, University of Hohenheim, Garbenstraße 30, 70593, Stuttgart, Germany.,Renal Division, Department of Medicine, University Hospital Freiburg, Freiburg University Faculty of Medicine & CIBSS-Centre for Integrative Biological Signalling Studies, University of Freiburg, Habsburger Str. 49, 79104, Freiburg, Germany
| | - Peter Walentek
- Renal Division, Department of Medicine, University Hospital Freiburg, Freiburg University Faculty of Medicine & CIBSS-Centre for Integrative Biological Signaling Studies, University of Freiburg, Habsburger Str. 49, 79104, Freiburg, Germany
| | - Karin Schuster-Gossler
- Institute for Molecular Biology, OE5250, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - Jan Hegermann
- Institute of Functional and Applied Anatomy, Research Core Unit Electron Microscopy, OE8840, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - Leonie Alten
- Institute for Molecular Biology, OE5250, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany.,Twist Bioscience, 681 Gateway Blvd South, South San Francisco, CA, 94080, USA
| | - Elisabeth Kremmer
- Institute of Molecular Immunology, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Core Facility Monoclonal Antibodies, Marchioninistr. 25, 81377, München, Germany.,Department of Biology II, Ludwig-Maximilians University, Großhaderner Straße 2, 82152, Martinsried, Germany
| | - Adina Przykopanski
- Institute for Molecular Biology, OE5250, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany.,Institute for Toxicology, OE 5340, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - Katrin Serth
- Institute for Molecular Biology, OE5250, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - Marius Ueffing
- Institute of Ophthalmic Research, Center for Ophthalmology, University of Tübingen, Elfriede-Aulhorn-Strasse 7, 72076, Tübingen, Germany
| | - Martin Blum
- Institute of Biology, University of Hohenheim, Garbenstraße 30, 70593, Stuttgart, Germany.
| | - Achim Gossler
- Institute for Molecular Biology, OE5250, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany.
| |
Collapse
|
21
|
Zhang Q, Carlin D, Zhu F, Cattaneo P, Ideker T, Evans SM, Bloomekatz J, Chi NC. Unveiling Complexity and Multipotentiality of Early Heart Fields. Circ Res 2021; 129:474-487. [PMID: 34162224 DOI: 10.1161/circresaha.121.318943] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Qingquan Zhang
- Medicine, Division of Cardiology (Q.Z., D.C., F.Z., P.C., S.M.E., J.B., N.C.C.)
| | - Daniel Carlin
- Medicine, Division of Cardiology (Q.Z., D.C., F.Z., P.C., S.M.E., J.B., N.C.C.)
| | - Fugui Zhu
- Medicine, Division of Cardiology (Q.Z., D.C., F.Z., P.C., S.M.E., J.B., N.C.C.)
| | - Paola Cattaneo
- Medicine, Division of Cardiology (Q.Z., D.C., F.Z., P.C., S.M.E., J.B., N.C.C.)
| | - Trey Ideker
- Medicine, Division of Genetics (T.I.).,Department of Computer Science and Engineering (T.I.).,Department of Bioengineering (T.I.).,Institute of Genomic Medicine (T.I., N.C.C.)
| | - Sylvia M Evans
- Medicine, Division of Cardiology (Q.Z., D.C., F.Z., P.C., S.M.E., J.B., N.C.C.).,Pharmacology, Skaggs School of Pharmacy and Pharmaceutical Sciences (S.M.E.), University of California San Diego, La Jolla, CA
| | - Joshua Bloomekatz
- Medicine, Division of Cardiology (Q.Z., D.C., F.Z., P.C., S.M.E., J.B., N.C.C.).,Now with Department of Biology, University of Mississippi, Oxford, MS (J.B.)
| | - Neil C Chi
- Medicine, Division of Cardiology (Q.Z., D.C., F.Z., P.C., S.M.E., J.B., N.C.C.).,Institute of Genomic Medicine (T.I., N.C.C.)
| |
Collapse
|
22
|
Post-Surgical Peritoneal Scarring and Key Molecular Mechanisms. Biomolecules 2021; 11:biom11050692. [PMID: 34063089 PMCID: PMC8147932 DOI: 10.3390/biom11050692] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 04/27/2021] [Accepted: 04/29/2021] [Indexed: 02/06/2023] Open
Abstract
Post-surgical adhesions are internal scar tissue and a major health and economic burden. Adhesions affect and involve the peritoneal lining of the abdominal cavity, which consists of a continuous mesothelial covering of the cavity wall and majority of internal organs. Our understanding of the full pathophysiology of adhesion formation is limited by the fact that the mechanisms regulating normal serosal repair and regeneration of the mesothelial layer are still being elucidated. Emerging evidence suggests that mesothelial cells do not simply form a passive barrier but perform a wide range of important regulatory functions including maintaining a healthy peritoneal homeostasis as well as orchestrating events leading to normal repair or pathological outcomes following injury. Here, we summarise recent advances in our understanding of serosal repair and adhesion formation with an emphasis on molecular mechanisms and novel gene expression signatures associated with these processes. We discuss changes in mesothelial biomolecular marker expression during peritoneal development, which may help, in part, to explain findings in adults from lineage tracing studies using experimental adhesion models. Lastly, we highlight examples of where local tissue specialisation may determine a particular response of peritoneal cells to injury.
Collapse
|
23
|
Redpath AN, Lupu IE, Smart N. Analysis of epicardial genes in embryonic mouse hearts with flow cytometry. STAR Protoc 2021; 2:100359. [PMID: 33718887 PMCID: PMC7921713 DOI: 10.1016/j.xpro.2021.100359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022] Open
Abstract
Genetic markers used to define discrete cell populations are seldom expressed exclusively in the population of interest and are, thus, unsuitable when evaluated individually, especially in the absence of spatial and morphological information. Here, we present fluorescence in situ hybridization for flow cytometry to allow simultaneous analysis of multiple marker genes at the single whole-cell level, exemplified by application to the embryonic epicardium. The protocol facilitates multiplexed quantification of gene and protein expression and temporal changes across specific cell populations. For complete details on the use and execution of this protocol, please refer to Lupu et al. (2020).
Collapse
Affiliation(s)
- Andia Nicole Redpath
- British Heart Foundation Centre of Regenerative Medicine, Burdon Sanderson Cardiac Science Centre, Department of Physiology, Anatomy & Genetics, University of Oxford, South Parks Road, Oxford OX1 3PT, UK
| | - Irina-Elena Lupu
- British Heart Foundation Centre of Regenerative Medicine, Burdon Sanderson Cardiac Science Centre, Department of Physiology, Anatomy & Genetics, University of Oxford, South Parks Road, Oxford OX1 3PT, UK
| | - Nicola Smart
- British Heart Foundation Centre of Regenerative Medicine, Burdon Sanderson Cardiac Science Centre, Department of Physiology, Anatomy & Genetics, University of Oxford, South Parks Road, Oxford OX1 3PT, UK
| |
Collapse
|
24
|
Telomerase therapy attenuates cardiotoxic effects of doxorubicin. Mol Ther 2021; 29:1395-1410. [PMID: 33388418 PMCID: PMC8058493 DOI: 10.1016/j.ymthe.2020.12.035] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 11/17/2020] [Accepted: 12/23/2020] [Indexed: 12/15/2022] Open
Abstract
Doxorubicin is one of the most potent chemotherapeutic agents. However, its clinical use is restricted due to the severe risk of cardiotoxicity, partially attributed to elevated production of reactive oxygen species (ROS). Telomerase canonically maintains telomeres during cell division but is silenced in adult hearts. In non-dividing cells such as cardiomyocytes, telomerase confers pro-survival traits, likely owing to the detoxification of ROS. Therefore, we hypothesized that pharmacological overexpression of telomerase may be used as a therapeutic strategy for the prevention of doxorubicin-induced cardiotoxicity. We used adeno-associated virus (AAV)-mediated gene therapy for long-term expression of telomerase in in vitro and in vivo models of doxorubicin-induced cardiotoxicity. Overexpression of telomerase protected the heart from doxorubicin-mediated apoptosis and rescued cardiac function, which was accompanied by preserved cardiomyocyte size. At the mechanistic level, we observed altered mitochondrial morphology and dynamics in response to telomerase expression. Complementary in vitro experiments confirmed the anti-apoptotic effects of telomerase overexpression in human induced pluripotent stem cell-derived cardiomyocytes after doxorubicin treatment. Strikingly, elevated levels of telomerase translocated to the mitochondria upon doxorubicin treatment, which helped to maintain mitochondrial function. Thus, telomerase gene therapy could be a novel preventive strategy for cardiotoxicity by chemotherapy agents such as the anthracyclines.
Collapse
|
25
|
Redpath AN, Smart N. Recapturing embryonic potential in the adult epicardium: Prospects for cardiac repair. Stem Cells Transl Med 2020; 10:511-521. [PMID: 33222384 PMCID: PMC7980211 DOI: 10.1002/sctm.20-0352] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 10/07/2020] [Accepted: 10/25/2020] [Indexed: 12/12/2022] Open
Abstract
Research into potential targets for cardiac repair encompasses recognition of tissue‐resident cells with intrinsic regenerative properties. The adult vertebrate heart is covered by mesothelium, named the epicardium, which becomes active in response to injury and contributes to repair, albeit suboptimally. Motivation to manipulate the epicardium for treatment of myocardial infarction is deeply rooted in its central role in cardiac formation and vasculogenesis during development. Moreover, the epicardium is vital to cardiac muscle regeneration in lower vertebrate and neonatal mammalian‐injured hearts. In this review, we discuss our current understanding of the biology of the mammalian epicardium in development and injury. Considering present challenges in the field, we further contemplate prospects for reinstating full embryonic potential in the adult epicardium to facilitate cardiac regeneration.
Collapse
Affiliation(s)
- Andia N Redpath
- Department of Physiology, Anatomy and Genetics, British Heart Foundation Centre of Regenerative Medicine, Burdon Sanderson Cardiac Science Centre, University of Oxford, Oxford, UK
| | - Nicola Smart
- Department of Physiology, Anatomy and Genetics, British Heart Foundation Centre of Regenerative Medicine, Burdon Sanderson Cardiac Science Centre, University of Oxford, Oxford, UK
| |
Collapse
|
26
|
Dalghi MG, Montalbetti N, Carattino MD, Apodaca G. The Urothelium: Life in a Liquid Environment. Physiol Rev 2020; 100:1621-1705. [PMID: 32191559 PMCID: PMC7717127 DOI: 10.1152/physrev.00041.2019] [Citation(s) in RCA: 112] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 03/02/2020] [Accepted: 03/14/2020] [Indexed: 02/08/2023] Open
Abstract
The urothelium, which lines the renal pelvis, ureters, urinary bladder, and proximal urethra, forms a high-resistance but adaptable barrier that surveils its mechanochemical environment and communicates changes to underlying tissues including afferent nerve fibers and the smooth muscle. The goal of this review is to summarize new insights into urothelial biology and function that have occurred in the past decade. After familiarizing the reader with key aspects of urothelial histology, we describe new insights into urothelial development and regeneration. This is followed by an extended discussion of urothelial barrier function, including information about the roles of the glycocalyx, ion and water transport, tight junctions, and the cellular and tissue shape changes and other adaptations that accompany expansion and contraction of the lower urinary tract. We also explore evidence that the urothelium can alter the water and solute composition of urine during normal physiology and in response to overdistension. We complete the review by providing an overview of our current knowledge about the urothelial environment, discussing the sensor and transducer functions of the urothelium, exploring the role of circadian rhythms in urothelial gene expression, and describing novel research tools that are likely to further advance our understanding of urothelial biology.
Collapse
Affiliation(s)
- Marianela G Dalghi
- Department of Medicine, Renal-Electrolyte Division, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Nicolas Montalbetti
- Department of Medicine, Renal-Electrolyte Division, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Marcelo D Carattino
- Department of Medicine, Renal-Electrolyte Division, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Gerard Apodaca
- Department of Medicine, Renal-Electrolyte Division, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| |
Collapse
|
27
|
Liu X, Chen W, Li W, Li Y, Priest JR, Zhou B, Wang J, Zhou Z. Single-Cell RNA-Seq of the Developing Cardiac Outflow Tract Reveals Convergent Development of the Vascular Smooth Muscle Cells. Cell Rep 2020; 28:1346-1361.e4. [PMID: 31365875 DOI: 10.1016/j.celrep.2019.06.092] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Revised: 04/17/2019] [Accepted: 06/25/2019] [Indexed: 02/07/2023] Open
Abstract
Cardiac outflow tract (OFT) is a major hotspot for congenital heart diseases. A thorough understanding of the cellular diversity, transitions, and regulatory networks of normal OFT development is essential to decipher the etiology of OFT malformations. We performed single-cell transcriptomic sequencing of 55,611 mouse OFT cells from three developmental stages that generally correspond to the early, middle, and late stages of OFT remodeling and septation. Known cellular transitions, such as endothelial-to-mesenchymal transition, have been recapitulated. In particular, we identified convergent development of the vascular smooth muscle cell (VSMC) lineage where intermediate cell subpopulations were found to be involved in either myocardial-to-VSMC trans-differentiation or mesenchymal-to-VSMC transition. Finally, we uncovered transcriptional regulators potentially governing cellular transitions. Our study provides a single-cell reference map of cell states for normal OFT development and paves the way for further studies of the etiology of OFT malformations at the single-cell level.
Collapse
Affiliation(s)
- Xuanyu Liu
- State Key Laboratory of Cardiovascular Disease, Beijing Key Laboratory for Molecular Diagnostics of Cardiovascular Diseases, Center of Laboratory Medicine, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
| | - Wen Chen
- State Key Laboratory of Cardiovascular Disease, Beijing Key Laboratory for Molecular Diagnostics of Cardiovascular Diseases, Center of Laboratory Medicine, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
| | - Wenke Li
- State Key Laboratory of Cardiovascular Disease, Beijing Key Laboratory for Molecular Diagnostics of Cardiovascular Diseases, Center of Laboratory Medicine, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
| | - Yan Li
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - James R Priest
- Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Bin Zhou
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China; Key Laboratory of Regenerative Medicine of Ministry of Education, Jinan University. School of Life Science and Technology, ShanghaiTech University, Shanghai 200031, China
| | - Jikui Wang
- Henan Key Laboratory for Medical Tissue Regeneration, School of Basic Medical Sciences, Xinxiang Medical University. Xinxiang 453003, China.
| | - Zhou Zhou
- State Key Laboratory of Cardiovascular Disease, Beijing Key Laboratory for Molecular Diagnostics of Cardiovascular Diseases, Center of Laboratory Medicine, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China.
| |
Collapse
|
28
|
Jackson AR, Ching CB, McHugh KM, Becknell B. Roles for urothelium in normal and aberrant urinary tract development. Nat Rev Urol 2020; 17:459-468. [PMID: 32647226 DOI: 10.1038/s41585-020-0348-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/03/2020] [Indexed: 12/11/2022]
Abstract
Congenital anomalies of the kidney and urinary tract (CAKUTs) represent the leading cause of chronic kidney disease and end-stage kidney disease in children. Increasing evidence points to critical roles for the urothelium in the developing urinary tract and in the genesis of CAKUTs. The involvement of the urothelium in patterning the urinary tract is supported by evidence that CAKUTs can arise as a result of abnormal urothelial development. Emerging evidence indicates that congenital urinary tract obstruction triggers urothelial remodelling that stabilizes the obstructed kidney and limits renal injury. Finally, the diagnostic potential of radiological findings and urinary biomarkers derived from the urothelium of patients with CAKUTs might aid their contribution to clinical care.
Collapse
Affiliation(s)
- Ashley R Jackson
- Nephrology and Urology Research Affinity Group, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, USA.,Center for Clinical and Translational Research, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
| | - Christina B Ching
- Nephrology and Urology Research Affinity Group, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, USA.,Center for Clinical and Translational Research, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, USA.,Division of Pediatric Urology, Department of Surgery, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Kirk M McHugh
- Nephrology and Urology Research Affinity Group, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, USA.,Center for Clinical and Translational Research, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, USA.,Department of Anatomy, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Brian Becknell
- Nephrology and Urology Research Affinity Group, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, USA. .,Center for Clinical and Translational Research, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, USA. .,Nephrology Division, Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH, USA.
| |
Collapse
|
29
|
Beckers A, Adis C, Schuster-Gossler K, Tveriakhina L, Ott T, Fuhl F, Hegermann J, Boldt K, Serth K, Rachev E, Alten L, Kremmer E, Ueffing M, Blum M, Gossler A. The FOXJ1 target Cfap206 is required for sperm motility, mucociliary clearance of the airways and brain development. Development 2020; 147:dev.188052. [PMID: 32376681 DOI: 10.1242/dev.188052] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Accepted: 04/15/2020] [Indexed: 12/11/2022]
Abstract
Cilia are complex cellular protrusions consisting of hundreds of proteins. Defects in ciliary structure and function, many of which have not been characterised molecularly, cause ciliopathies: a heterogeneous group of human syndromes. Here, we report on the FOXJ1 target gene Cfap206, orthologues of which so far have only been studied in Chlamydomonas and Tetrahymena In mouse and Xenopus, Cfap206 was co-expressed with and dependent on Foxj1 CFAP206 protein localised to the basal body and to the axoneme of motile cilia. In Xenopus crispant larvae, the ciliary beat frequency of skin multiciliated cells was enhanced and bead transport across the epidermal mucociliary epithelium was reduced. Likewise, Cfap206 knockout mice revealed ciliary phenotypes. Electron tomography of immotile knockout mouse sperm flagella indicated a role in radial spoke formation reminiscent of FAP206 function in Tetrahymena Male infertility, hydrocephalus and impaired mucociliary clearance of the airways in the absence of laterality defects in Cfap206 mutant mice suggests that Cfap206 may represent a candidate for the subgroup of human primary ciliary dyskinesias caused by radial spoke defects.
Collapse
Affiliation(s)
- Anja Beckers
- Institute for Molecular Biology, OE5250, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - Christian Adis
- Institute for Molecular Biology, OE5250, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - Karin Schuster-Gossler
- Institute for Molecular Biology, OE5250, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - Lena Tveriakhina
- Institute for Molecular Biology, OE5250, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - Tim Ott
- Institute of Zoology, University of Hohenheim, Garbenstraße 30, 70593 Stuttgart, Germany
| | - Franziska Fuhl
- Institute of Zoology, University of Hohenheim, Garbenstraße 30, 70593 Stuttgart, Germany
| | - Jan Hegermann
- Institute of Functional and Applied Anatomy, OE8840, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - Karsten Boldt
- Institute of Ophthalmic Research, Center for Ophthalmology, University of Tübingen, Röntgenweg 11, 72076 Tübingen, Germany
| | - Katrin Serth
- Institute for Molecular Biology, OE5250, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - Ev Rachev
- Institute for Molecular Biology, OE5250, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - Leonie Alten
- Institute for Molecular Biology, OE5250, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - Elisabeth Kremmer
- Institute of Molecular Immunology, Helmholtz Zentrum München, German Research Center for Environmental Health, Core Facility Monoclonal Antibodies, Marchioninistr. 25, 81377 München, Germany
| | - Marius Ueffing
- Institute of Ophthalmic Research, Center for Ophthalmology, University of Tübingen, Röntgenweg 11, 72076 Tübingen, Germany
| | - Martin Blum
- Institute of Zoology, University of Hohenheim, Garbenstraße 30, 70593 Stuttgart, Germany
| | - Achim Gossler
- Institute for Molecular Biology, OE5250, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| |
Collapse
|
30
|
Lupu IE, Redpath AN, Smart N. Spatiotemporal Analysis Reveals Overlap of Key Proepicardial Markers in the Developing Murine Heart. Stem Cell Reports 2020; 14:770-787. [PMID: 32359445 PMCID: PMC7221110 DOI: 10.1016/j.stemcr.2020.04.002] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 04/02/2020] [Accepted: 04/02/2020] [Indexed: 01/05/2023] Open
Abstract
The embryonic epicardium, originating from the proepicardial organ (PEO), provides a source of multipotent progenitors for cardiac lineages, including pericytes, fibroblasts, and vascular smooth muscle cells. Maximizing the regenerative capacity of the adult epicardium depends on recapitulating embryonic cell fates. The potential of the epicardium to contribute coronary endothelium is unclear, due to conflicting Cre-based lineage trace data. Controversy also surrounds when epicardial cell fate becomes restricted. Here, we systematically investigate expression of five widely used epicardial markers, Wt1, Tcf21, Tbx18, Sema3d, and Scx, over the course of development. We show overlap of markers in all PEO and epicardial cells until E13.5, and find no evidence for discrete proepicardial sub-compartments that might contribute coronary endothelium via the epicardial layer. Our findings clarify a number of prevailing discrepancies and support the notion that epicardium-derived cell fate, to form fibroblasts or mural cells, is specified after epithelial-mesenchymal transition, not pre-determined within the PEO.
Collapse
Affiliation(s)
- Irina-Elena Lupu
- Department of Physiology, Anatomy & Genetics, University of Oxford, Oxford OX1 3PT, UK
| | - Andia N Redpath
- Department of Physiology, Anatomy & Genetics, University of Oxford, Oxford OX1 3PT, UK
| | - Nicola Smart
- Department of Physiology, Anatomy & Genetics, University of Oxford, Oxford OX1 3PT, UK.
| |
Collapse
|
31
|
Hamanaka K, Takata A, Uchiyama Y, Miyatake S, Miyake N, Mitsuhashi S, Iwama K, Fujita A, Imagawa E, Alkanaq AN, Koshimizu E, Azuma Y, Nakashima M, Mizuguchi T, Saitsu H, Wada Y, Minami S, Katoh-Fukui Y, Masunaga Y, Fukami M, Hasegawa T, Ogata T, Matsumoto N. MYRF haploinsufficiency causes 46,XY and 46,XX disorders of sex development: bioinformatics consideration. Hum Mol Genet 2020; 28:2319-2329. [PMID: 30985895 DOI: 10.1093/hmg/ddz066] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Revised: 03/06/2019] [Accepted: 03/21/2019] [Indexed: 12/30/2022] Open
Abstract
Disorders of sex development (DSDs) are defined as congenital conditions in which chromosomal, gonadal or anatomical sex is atypical. In many DSD cases, genetic causes remain to be elucidated. Here, we performed a case-control exome sequencing study comparing gene-based burdens of rare damaging variants between 26 DSD cases and 2625 controls. We found exome-wide significant enrichment of rare heterozygous truncating variants in the MYRF gene encoding myelin regulatory factor, a transcription factor essential for oligodendrocyte development. All three variants occurred de novo. We identified an additional 46,XY DSD case of a de novo damaging missense variant in an independent cohort. The clinical symptoms included hypoplasia of Müllerian derivatives and ovaries in 46,XX DSD patients, defective development of Sertoli and Leydig cells in 46,XY DSD patients and congenital diaphragmatic hernia in one 46,XY DSD patient. As all of these cells and tissues are or partly consist of coelomic epithelium (CE)-derived cells (CEDC) and CEDC developed from CE via proliferaiton and migration, MYRF might be related to these processes. Consistent with this hypothesis, single-cell RNA sequencing of foetal gonads revealed high expression of MYRF in CE and CEDC. Reanalysis of public chromatin immunoprecipitation sequencing data for rat Myrf showed that genes regulating proliferation and migration were enriched among putative target genes of Myrf. These results suggested that MYRF is a novel causative gene of 46,XY and 46,XX DSD and MYRF is a transcription factor regulating CD and/or CEDC proliferation and migration, which is essential for development of multiple organs.
Collapse
Affiliation(s)
| | | | - Yuri Uchiyama
- Department of Human Genetics.,Department of Oncology
| | - Satoko Miyatake
- Department of Human Genetics.,Clinical Genetics Department, Yokohama City University Hospital, Yokohama, Kanagawa, Japan
| | | | | | | | | | | | | | | | - Yoshiki Azuma
- Department of Human Genetics.,Department of Pediatrics, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Mitsuko Nakashima
- Department of Biochemistry, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka, Japan
| | | | - Hirotomo Saitsu
- Department of Biochemistry, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka, Japan
| | - Yuka Wada
- Department of Neonatology, National Center for Child Health and Development, Setagaya, Tokyo, Japan
| | - Sawako Minami
- Deparment of Obstetrics and Gynecology, Wakayama Medical University, Wakayama, Wakayama, Japan
| | - Yuko Katoh-Fukui
- Department of Molecular Endocrinology, National Center for Child Health and Development, Setagaya, Tokyo, Japan
| | - Yohei Masunaga
- Department of Pediatrics, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka, Japan
| | - Maki Fukami
- Department of Molecular Endocrinology, National Center for Child Health and Development, Setagaya, Tokyo, Japan
| | - Tomonobu Hasegawa
- Department of Pediatrics, Keio University School of Medicine, Shinjuku, Tokyo, Japan
| | - Tsutomu Ogata
- Department of Pediatrics, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka, Japan
| | | |
Collapse
|
32
|
Rachev E, Schuster-Gossler K, Fuhl F, Ott T, Tveriakhina L, Beckers A, Hegermann J, Boldt K, Mai M, Kremmer E, Ueffing M, Blum M, Gossler A. CFAP43 modulates ciliary beating in mouse and Xenopus. Dev Biol 2020; 459:109-125. [DOI: 10.1016/j.ydbio.2019.12.010] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 12/14/2019] [Accepted: 12/18/2019] [Indexed: 11/26/2022]
|
33
|
Gupta SK, Garg A, Avramopoulos P, Engelhardt S, Streckfuss-Bömeke K, Batkai S, Thum T. miR-212/132 Cluster Modulation Prevents Doxorubicin-Mediated Atrophy and Cardiotoxicity. Mol Ther 2018; 27:17-28. [PMID: 30527757 DOI: 10.1016/j.ymthe.2018.11.004] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Revised: 11/01/2018] [Accepted: 11/07/2018] [Indexed: 10/27/2022] Open
Abstract
Improved therapy of cancer has significantly increased the lifespan of patients. However, cancer survivors face an increased risk of cardiovascular complications due to adverse effects of cancer therapies. The chemotherapy drug doxorubicin is well known to induce myofibril damage and cardiac atrophy. Our aim was to test potential counteracting effects of the pro-hypertrophic miR-212/132 family in doxorubicin-induced cardiotoxicity. In vitro, overexpression of the pro-hypertrophic miR-212/132 cluster in primary rodent and human iPSC-derived cardiomyocytes inhibited doxorubicin-induced toxicity. Next, a disease model of doxorubicin-induced cardiotoxicity was established in male C57BL/6N mice. Mice were administered either adeno-associated virus (AAV)9-control or AAV9-miR-212/132 to achieve myocardial overexpression of the miR-212/132 cluster. AAV9-mediated overexpression limited cardiac atrophy by increasing left ventricular mass and wall thickness, decreased doxorubicin-mediated apoptosis, and prevented myofibril damage. Based on a transcriptomic profiling we identified fat storage-inducing transmembrane protein 2 (Fitm2) as a novel target and downstream effector molecule responsible, at least in part, for the observed miR-212/132 anti-cardiotoxic effects. Overexpression of Fitm2 partially reversed the effects of miR-212/132. Overexpression of the miR-212/132 family reduces development of doxorubicin-induced cardiotoxicity and thus could be a therapeutic entry point to limit doxorubicin-mediated adverse cardiac effects.
Collapse
Affiliation(s)
- Shashi Kumar Gupta
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Hannover, Germany.
| | - Ankita Garg
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Hannover, Germany
| | - Petros Avramopoulos
- Institute of Pharmacology and Toxicology, Technical University of Munich, Munich, Germany; German Center for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany
| | - Stefan Engelhardt
- Institute of Pharmacology and Toxicology, Technical University of Munich, Munich, Germany; German Center for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany
| | - Katrin Streckfuss-Bömeke
- Clinic for Cardiology and Pneumology, Stem Cell Laboratory, University Medical Center, Gottingen, Germany
| | - Sandor Batkai
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Hannover, Germany; Cardior Pharmaceuticals GmbH, Hannover Medical School Campus, Hannover, Germany
| | - Thomas Thum
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Hannover, Germany; Cardior Pharmaceuticals GmbH, Hannover Medical School Campus, Hannover, Germany; Excellence Cluster REBIRTH, Hannover Medical School, Hannover, Germany; National Heart and Lung Institute, Imperial College London, London, UK.
| |
Collapse
|
34
|
Liao Y, Chang HC, Liang FX, Chung PJ, Wei Y, Nguyen TP, Zhou G, Talebian S, Krey LC, Deng FM, Wong TW, Chicote JU, Grifo JA, Keefe DL, Shapiro E, Lepor H, Wu XR, DeSalle R, Garcia-España A, Kim SY, Sun TT. Uroplakins play conserved roles in egg fertilization and acquired additional urothelial functions during mammalian divergence. Mol Biol Cell 2018; 29:3128-3143. [PMID: 30303751 PMCID: PMC6340209 DOI: 10.1091/mbc.e18-08-0496] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Uroplakin (UP) tetraspanins and their associated proteins are major mammalian urothelial differentiation products that form unique two-dimensional crystals of 16-nm particles (“urothelial plaques”) covering the apical urothelial surface. Although uroplakins are highly expressed only in mammalian urothelium and are often referred to as being urothelium specific, they are also expressed in several mouse nonurothelial cell types in stomach, kidney, prostate, epididymis, testis/sperms, and ovary/oocytes. In oocytes, uroplakins colocalize with CD9 on cell-surface and multivesicular body-derived exosomes, and the cytoplasmic tail of UPIIIa undergoes a conserved fertilization-dependent, Fyn-mediated tyrosine phosphorylation that also occurs in Xenopus laevis eggs. Uroplakin knockout and antibody blocking reduce mouse eggs’ fertilization rate in in vitro fertilization assays, and UPII/IIIa double-knockout mice have a smaller litter size. Phylogenetic analyses showed that uroplakin sequences underwent significant mammal-specific changes. These results suggest that, by mediating signal transduction and modulating membrane stability that do not require two-dimensional-crystal formation, uroplakins can perform conserved and more ancestral fertilization functions in mouse and frog eggs. Uroplakins acquired the ability to form two-dimensional-crystalline plaques during mammalian divergence, enabling them to perform additional functions, including umbrella cell enlargement and the formation of permeability and mechanical barriers, to protect/modify the apical surface of the modern-day mammalian urothelium.
Collapse
Affiliation(s)
- Yi Liao
- Department of Cell Biology, New York University School of Medicine, New York, NY 10016
| | - Hung-Chi Chang
- Department of Obstetrics and Gynecology, New York University School of Medicine, New York, NY 10016.,Department of Obstetrics and Gynecology, National Taiwan University, Taipei 10617, Taiwan
| | - Feng-Xia Liang
- Department of Cell Biology, New York University School of Medicine, New York, NY 10016
| | | | - Yuan Wei
- Department of Cell Biology, New York University School of Medicine, New York, NY 10016
| | - Tuan-Phi Nguyen
- Department of Cell Biology, New York University School of Medicine, New York, NY 10016
| | - Ge Zhou
- Regeneron, Tarrytown, NY 10591
| | - Sheeva Talebian
- Department of Obstetrics and Gynecology, New York University School of Medicine, New York, NY 10016
| | - Lewis C Krey
- Department of Obstetrics and Gynecology, New York University School of Medicine, New York, NY 10016
| | - Fang-Ming Deng
- Department of Pathology, New York University School of Medicine, New York, NY 10016.,Department of Urology, New York University School of Medicine, New York, NY 10016
| | - Tak-Wah Wong
- Department of Dermatology, National Cheng Kung University, Tainan 701, Taiwan
| | - Javier U Chicote
- Unitat De Recerca, Hospital Joan XXIII, Institut de Investigacio Sanitaria Pere Virgili (IISPV), Universitat Rovira i Virgili, Tarragona 43007, Spain
| | - James A Grifo
- Department of Obstetrics and Gynecology, New York University School of Medicine, New York, NY 10016
| | - David L Keefe
- Department of Obstetrics and Gynecology, New York University School of Medicine, New York, NY 10016
| | - Ellen Shapiro
- Department of Urology, New York University School of Medicine, New York, NY 10016
| | - Herbert Lepor
- Department of Urology, New York University School of Medicine, New York, NY 10016.,Sackler Institute of Comparative Genomics, American Museum of Natural History, New York, NY 10024
| | - Xue-Ru Wu
- Department of Pathology, New York University School of Medicine, New York, NY 10016.,Department of Urology, New York University School of Medicine, New York, NY 10016.,Department of Biochemistry and Molecular Pharmacology, New York University School of Medicine, New York, NY 10016
| | - Robert DeSalle
- Veterans Affairs New York Harbor Healthcare System, New York, NY 10010
| | - Antonio Garcia-España
- Unitat De Recerca, Hospital Joan XXIII, Institut de Investigacio Sanitaria Pere Virgili (IISPV), Universitat Rovira i Virgili, Tarragona 43007, Spain
| | - Sang Yong Kim
- Department of Pathology, New York University School of Medicine, New York, NY 10016
| | - Tung-Tien Sun
- Department of Cell Biology, New York University School of Medicine, New York, NY 10016.,Department of Urology, New York University School of Medicine, New York, NY 10016.,The Ronald O. Perelman Department of Dermatology, New York University School of Medicine, New York, NY 10016.,Sackler Institute of Comparative Genomics, American Museum of Natural History, New York, NY 10024
| |
Collapse
|
35
|
Hustler A, Eardley I, Hinley J, Pearson J, Wezel F, Radvanyi F, Baker SC, Southgate J. Differential transcription factor expression by human epithelial cells of buccal and urothelial derivation. Exp Cell Res 2018; 369:284-294. [PMID: 29842880 PMCID: PMC6092173 DOI: 10.1016/j.yexcr.2018.05.031] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Revised: 05/24/2018] [Accepted: 05/25/2018] [Indexed: 12/29/2022]
Abstract
Identification of transcription factors expressed by differentiated cells is informative not only of tissue-specific pathways, but to help identify master regulators for cellular reprogramming. If applied, such an approach could generate healthy autologous tissue-specific cells for clinical use where cells from the homologous tissue are unavailable due to disease. Normal human epithelial cells of buccal and urothelial derivation maintained in identical culture conditions that lacked significant instructive or permissive signaling cues were found to display inherent similarities and differences of phenotype. Investigation of transcription factors implicated in driving urothelial-type differentiation revealed buccal epithelial cells to have minimal or absent expression of PPARG, GATA3 and FOXA1 genes. Retroviral overexpression of protein coding sequences for GATA3 or PPARy1 in buccal epithelial cells resulted in nuclear immunolocalisation of the respective proteins, with both transductions also inducing expression of the urothelial differentiation-associated claudin 3 tight junction protein. PPARG1 overexpression alone entrained expression of nuclear FOXA1 and GATA3 proteins, providing objective evidence of its upstream positioning in a transcription factor network and identifying it as a candidate factor for urothelial-type transdifferentiation or reprogramming.
Collapse
Affiliation(s)
- Arianna Hustler
- Jack Birch Unit for Molecular Carcinogenesis, Department of Biology, University of York, York YO10 5DD, United Kingdom
| | - Ian Eardley
- Pyrah Department of Urology, St. James's University Hospital, Leeds LS9 7TF, United Kingdom
| | - Jennifer Hinley
- Jack Birch Unit for Molecular Carcinogenesis, Department of Biology, University of York, York YO10 5DD, United Kingdom
| | - Joanna Pearson
- Jack Birch Unit for Molecular Carcinogenesis, Department of Biology, University of York, York YO10 5DD, United Kingdom
| | - Felix Wezel
- Jack Birch Unit for Molecular Carcinogenesis, Department of Biology, University of York, York YO10 5DD, United Kingdom
| | - Francois Radvanyi
- Oncologie Moléculaire, Institut Curie, Centre de Recherche, 75248 Paris cedex 05, France
| | - Simon C Baker
- Jack Birch Unit for Molecular Carcinogenesis, Department of Biology, University of York, York YO10 5DD, United Kingdom
| | - Jennifer Southgate
- Jack Birch Unit for Molecular Carcinogenesis, Department of Biology, University of York, York YO10 5DD, United Kingdom.
| |
Collapse
|
36
|
Hippo Signaling Plays an Essential Role in Cell State Transitions during Cardiac Fibroblast Development. Dev Cell 2018; 45:153-169.e6. [PMID: 29689192 DOI: 10.1016/j.devcel.2018.03.019] [Citation(s) in RCA: 133] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Revised: 02/02/2018] [Accepted: 03/26/2018] [Indexed: 12/14/2022]
Abstract
During development, progenitors progress through transition states. The cardiac epicardium contains progenitors of essential non-cardiomyocytes. The Hippo pathway, a kinase cascade that inhibits the Yap transcriptional co-factor, controls organ size in developing hearts. Here, we investigated Hippo kinases Lats1 and Lats2 in epicardial diversification. Epicardial-specific deletion of Lats1/2 was embryonic lethal, and mutant embryos had defective coronary vasculature remodeling. Single-cell RNA sequencing revealed that Lats1/2 mutant cells failed to activate fibroblast differentiation but remained in an intermediate cell state with both epicardial and fibroblast characteristics. Lats1/2 mutant cells displayed an arrested developmental trajectory with persistence of epicardial markers and expanded expression of Yap targets Dhrs3, an inhibitor of retinoic acid synthesis, and Dpp4, a protease that modulates extracellular matrix (ECM) composition. Genetic and pharmacologic manipulation revealed that Yap inhibits fibroblast differentiation, prolonging a subepicardial-like cell state, and promotes expression of matricellular factors, such as Dpp4, that define ECM characteristics.
Collapse
|
37
|
Development and Characterization of a Porcine Mitral Valve Scaffold for Tissue Engineering. J Cardiovasc Transl Res 2017; 10:374-390. [PMID: 28462436 DOI: 10.1007/s12265-017-9747-z] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Accepted: 04/10/2017] [Indexed: 10/19/2022]
Abstract
Decellularized scaffolds represent a promising alternative for mitral valve (MV) replacement. This work developed and characterized a protocol for the decellularization of whole MVs. Porcine MVs were decellularized with 0.5% (w/v) SDS and 0.5% (w/v) SD and sterilized with 0.1% (v/v) PAA. Decellularized samples were seeded with human foreskin fibroblasts and human adipose-derived stem cells to investigate cellular repopulation and infiltration, and with human colony-forming endothelial cells to investigate collagen IV formation. Histology revealed an acellular scaffold with a generally conserved histoarchitecture, but collagen IV loss. Following decellularization, no significant changes were observed in the hydroxyproline content, but there was a significant reduction in the glycosaminoglycan content. SEM/TEM analysis confirmed cellular removal and loss of some extracellular matrix components. Collagen and elastin were generally preserved. The endothelial cells produced newly formed collagen IV on the non-cytotoxic scaffold. The protocol produced acellular scaffolds with generally preserved histoarchitecture, biochemistry, and biomechanics.
Collapse
|
38
|
Fan J, Liu Y, Wang S, Liu Y, Li S, Long R, Zhang R, Kankala RK. Synthesis and characterization of innovative poly(lactide-co-glycolide)-(poly-l-ornithine/fucoidan) core–shell nanocarriers by layer-by-layer self-assembly. RSC Adv 2017. [DOI: 10.1039/c7ra04908k] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Layer-by-Layer (LbL) self-assembly of nanocarriers has garnered the interest of researchers for a wide variety of biomedical applications.
Collapse
Affiliation(s)
- Jingqian Fan
- College of Chemical Engineering
- Huaqiao University
- Xiamen
- China
| | - Yuangang Liu
- College of Chemical Engineering
- Huaqiao University
- Xiamen
- China
- Institute of Pharmaceutical Engineering
| | - Shibin Wang
- College of Chemical Engineering
- Huaqiao University
- Xiamen
- China
- Institute of Pharmaceutical Engineering
| | - Yulu Liu
- College of Chemical Engineering
- Huaqiao University
- Xiamen
- China
| | - Siming Li
- College of Chemical Engineering
- Huaqiao University
- Xiamen
- China
| | - Ruimin Long
- College of Chemical Engineering
- Huaqiao University
- Xiamen
- China
| | - Ran Zhang
- College of Chemical Engineering
- Huaqiao University
- Xiamen
- China
| | - Ranjith Kumar Kankala
- College of Chemical Engineering
- Huaqiao University
- Xiamen
- China
- Institute of Pharmaceutical Engineering
| |
Collapse
|
39
|
Weidemann M, Schuster-Gossler K, Stauber M, Wrede C, Hegermann J, Ott T, Boldt K, Beyer T, Serth K, Kremmer E, Blum M, Ueffing M, Gossler A. CFAP157 is a murine downstream effector of FOXJ1 that is specifically required for flagellum morphogenesis and sperm motility. Development 2016; 143:4736-4748. [DOI: 10.1242/dev.139626] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Accepted: 11/09/2016] [Indexed: 12/21/2022]
Abstract
Motile cilia move extracellular fluids or mediate cellular motility. Their function is essential for embryonic development, adult tissue homeostasis and reproduction throughout vertebrates. FOXJ1 is a key transcription factor for the formation of motile cilia but its downstream genetic programme is only partially understood. Here, we characterise a novel FOXJ1 target, Cfap157, that is specifically expressed in motile ciliated tissues in mouse and Xenopus in a FOXJ1-dependent manner. CFAP157 protein localises to basal bodies and interacts with tubulin and the centrosomal protein CEP350. Cfap157 knockout mice appear normal but homozygous males are infertile. Spermatozoa display impaired motility and a novel phenotype: Cfap157-deficient sperm exhibit axonemal loops, supernumerary axonemal profiles with ectopic accessory structures, excess cytoplasm and clustered mitochondria in the midpiece regions, and defective axonemes along the flagella. Our study thus demonstrates an essential sperm-specific function for CFAP157 and suggests that this novel FOXJ1 effector is part of a mechanism that acts during spermiogenesis to suppress the formation of supernumerary axonemes and ensures a correct ultrastructure.
Collapse
Affiliation(s)
- Marina Weidemann
- Institute for Molecular Biology, OE5250, Hannover Medical School, Carl-Neuberg-Str. 1, Hannover 30625, Germany
| | - Karin Schuster-Gossler
- Institute for Molecular Biology, OE5250, Hannover Medical School, Carl-Neuberg-Str. 1, Hannover 30625, Germany
| | - Michael Stauber
- Institute for Molecular Biology, OE5250, Hannover Medical School, Carl-Neuberg-Str. 1, Hannover 30625, Germany
| | - Christoph Wrede
- Institute of Functional and Applied Anatomy, OE8840, Hannover Medical School, Carl-Neuberg-Str. 1, Hannover 30625, Germany
| | - Jan Hegermann
- Institute of Functional and Applied Anatomy, OE8840, Hannover Medical School, Carl-Neuberg-Str. 1, Hannover 30625, Germany
| | - Tim Ott
- Institute of Zoology, University of Hohenheim, Garbenstraße 30, Stuttgart 70593, Germany
| | - Karsten Boldt
- Institute of Ophthalmic Research, Center for Ophthalmology, University of Tübingen, Röntgenweg 11, Tübingen 72076, Germany
| | - Tina Beyer
- Institute of Ophthalmic Research, Center for Ophthalmology, University of Tübingen, Röntgenweg 11, Tübingen 72076, Germany
| | - Katrin Serth
- Institute for Molecular Biology, OE5250, Hannover Medical School, Carl-Neuberg-Str. 1, Hannover 30625, Germany
| | - Elisabeth Kremmer
- Institute of Molecular Immunology, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Core Facility Monoclonal Antibodies, Marchioninistr. 25, München 81377, Germany
| | - Martin Blum
- Institute of Zoology, University of Hohenheim, Garbenstraße 30, Stuttgart 70593, Germany
| | - Marius Ueffing
- Institute of Ophthalmic Research, Center for Ophthalmology, University of Tübingen, Röntgenweg 11, Tübingen 72076, Germany
| | - Achim Gossler
- Institute for Molecular Biology, OE5250, Hannover Medical School, Carl-Neuberg-Str. 1, Hannover 30625, Germany
| |
Collapse
|
40
|
de Graaf P, van der Linde EM, Rosier PFWM, Izeta A, Sievert KD, Bosch JLHR, de Kort LMO. Systematic Review to Compare Urothelium Differentiation with Urethral Epithelium Differentiation in Fetal Development, as a Basis for Tissue Engineering of the Male Urethra. TISSUE ENGINEERING PART B-REVIEWS 2016; 23:257-267. [PMID: 27809709 DOI: 10.1089/ten.teb.2016.0352] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND Tissue-engineered (TE) urethra is desirable in men with urethral disease (stricture or hypospadias) and shortage of local tissue. Although ideally a TE graft would contain urethral epithelium cells, currently, bladder epithelium (urothelium) is widely used, but morphologically different. Understanding the differences and similarities of urothelium and urethral epithelium could help design a protocol for in vitro generation of urethral epithelium to be used in TE grafts for the urethra. PURPOSE To understand the development toward urethral epithelium or urothelium to improve TE of the urethra. METHODS A literature search was done following PRISMA guidelines. Articles describing urethral epithelium and bladder urothelium development in laboratory animals and humans were selected. RESULTS Twenty-nine studies on development of urethral epithelium and 29 studies on development of urothelium were included. Both tissue linings derive from endoderm and although adult urothelium and urethral epithelium are characterized by different gene expression profiles, the signaling pathways underlying their development are similar, including Shh, BMP, Wnt, and FGF. The progenitor of the urothelium and the urethral epithelium is the early fetal urogenital sinus (UGS). The urethral plate and the urothelium are both formed from the p63+ cells of the UGS. Keratin 20 and uroplakins are exclusively expressed in urothelium, not in the urethral epithelium. Further research has to be done on unique markers for the urethral epithelium. CONCLUSION This review has summarized the current knowledge about embryonic development of urothelium versus urethral epithelium and especially focuses on the influencing factors that are potentially specific for the eventual morphological differences of both cell linings, to be a basis for developmental or tissue engineering of urethral tissue.
Collapse
Affiliation(s)
- Petra de Graaf
- 1 Department of Urology, University Medical Centre Utrecht , Utrecht, The Netherlands .,2 Regenerative Medicine Center Utrecht , Utrecht, The Netherlands
| | | | - Peter F W M Rosier
- 1 Department of Urology, University Medical Centre Utrecht , Utrecht, The Netherlands
| | - Ander Izeta
- 3 Tissue Engineering Laboratory, Bioengineering Area, Instituto Biodonostia, Hospital Universitario Donostia , San Sebastián, Spain .,4 Department of Biomedical Engineering, School of Engineering, Tecnun-University of Navarra , San Sebastián, Spain
| | | | - J L H Ruud Bosch
- 1 Department of Urology, University Medical Centre Utrecht , Utrecht, The Netherlands
| | - Laetitia M O de Kort
- 1 Department of Urology, University Medical Centre Utrecht , Utrecht, The Netherlands
| |
Collapse
|
41
|
Kuriyama S, Tamiya Y, Tanaka M. Spatiotemporal expression of UPK3B and its promoter activity during embryogenesis and spermatogenesis. Histochem Cell Biol 2016; 147:17-26. [PMID: 27577269 DOI: 10.1007/s00418-016-1486-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/24/2016] [Indexed: 01/14/2023]
Abstract
Uroplakin (Upk) 3 is one of the main structural components of the urothelium tissue. Although expression of UPK3B is seen in a wider variety of the tissues and organs than UPK3A, tissue-specific expression has not yet been analyzed. Here, we analyzed the Cre recombinase activity driven by the Upk3b promoter in transgenic mice and the endogenous localization of UPK3B. We generated Tg(Upk3b-Cre)/R26tdTomato mice by crossing ROSA26tm14(CAG-tdTomato) (R26tdTomato) mice with Tg(Upk3b-Cre) mice and investigated the spatiotemporal distribution of tdTomato in embryonic and adult mice. In embryos, we detected Cre recombinase activity in neural crest cells and the heart, liver, kidneys, and lungs. In adult mice, Cre recombinase activity was detected in male and female genital organs; however, the activity was absent in the bladder. Histological analyses revealed that both tdTomato and UPK3B were present in testicular and epididymal sperm; however, tdTomato was not present in the ductus epididymis, where the endogenous expression of UPK3B was detected. In female siblings, both tdTomato and UPK3B expressions were detected in the follicles of the ovary, whereas no tdTomato expression was found in the mucosal epithelium of the fallopian tubes, where the endogenous UPK3B was expressed. These data suggest that UPK3B may play a pivotal role in the maturation of gametes and gamete-delivery organs.
Collapse
Affiliation(s)
- Sei Kuriyama
- Department of Molecular Biochemistry, Graduate School Medicine Akita University, Hondo 1-1-1, Akita City, Akita, 010-8543, Japan.
| | - Yuutaro Tamiya
- Department of Molecular Biochemistry, Graduate School Medicine Akita University, Hondo 1-1-1, Akita City, Akita, 010-8543, Japan.,Department of Lifescience, Faculty and Graduate School of Engineering and Resource Science, Akita University, 1-1 Tegata Gakuenmachi, Akita City, Akita, 010-8502, Japan
| | - Masamitsu Tanaka
- Department of Molecular Biochemistry, Graduate School Medicine Akita University, Hondo 1-1-1, Akita City, Akita, 010-8543, Japan
| |
Collapse
|
42
|
Lua I, Li Y, Zagory JA, Wang KS, French SW, Sévigny J, Asahina K. Characterization of hepatic stellate cells, portal fibroblasts, and mesothelial cells in normal and fibrotic livers. J Hepatol 2016; 64:1137-1146. [PMID: 26806818 PMCID: PMC4834254 DOI: 10.1016/j.jhep.2016.01.010] [Citation(s) in RCA: 118] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Revised: 01/06/2016] [Accepted: 01/11/2016] [Indexed: 02/07/2023]
Abstract
BACKGROUND & AIMS Contribution of hepatic stellate cells (HSCs), portal fibroblasts (PFs), and mesothelial cells (MCs) to myofibroblasts is not fully understood due to insufficient availability of markers and isolation methods. The present study aimed to isolate these cells, characterize their phenotypes, and examine their contribution to myofibroblasts in liver fibrosis. METHODS Liver fibrosis was induced in Collagen1a1-green fluorescent protein (Col1a1(GFP)) mice by bile duct ligation (BDL), 3,5-diethoxycarbonyl-1,4-dihydrocollidine (DDC) diet, or CCl4 injections. Combining vitamin A (VitA) lipid autofluorescence and expression of GFP and glycoprotein M6a (GPM6A), we separated HSCs, PFs, and MCs from normal and fibrotic livers by fluorescence-activated cell sorting (FACS). RESULTS Normal Col1a1(GFP) livers broadly expressed GFP in HSCs, PFs, and MCs. Isolated VitA+ HSCs expressed reelin, whereas VitA-GFP+GPM6A- PFs expressed ectonucleoside triphosphate diphosphohydrolase-2 and elastin. VitA-GFP+GPM6A+ MCs expressed keratin 19, mesothelin, and uroplakin 1b. Transforming growth factor (TGF)-β1 treatment induced the transformation of HSCs, PFs, and MCs into myofibroblasts in culture. TGF-β1 suppressed cyclin D1 mRNA expression in PFs but not in HSCs and MCs. In biliary fibrosis, PFs adjacent to the bile duct expressed α-smooth muscle actin. FACS analysis revealed that HSCs are the major source of GFP+ myofibroblasts in the injured Col1a1(GFP) mice after DDC or CCl4 treatment. Although PFs partly contributed to GFP+ myofibroblasts in the BDL model, HSCs were still dominant source of myofibroblasts. CONCLUSION HSCs, PFs, and MCs have distinct phenotypes, and PFs partly contribute to myofibroblasts in the portal triad in biliary fibrosis.
Collapse
Affiliation(s)
- Ingrid Lua
- Southern California Research Center for ALPD and Cirrhosis, Department of Pathology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Yuchang Li
- Southern California Research Center for ALPD and Cirrhosis, Department of Pathology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Jessica A. Zagory
- Developmental Biology, Regenerative Medicine and Stem Cell Program, Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, CA, USA
| | - Kasper S. Wang
- Developmental Biology, Regenerative Medicine and Stem Cell Program, Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, CA, USA
| | - Samuel W. French
- Department of Pathology, Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Jean Sévigny
- Département de Microbiologie-Infectiologie et d'Immunologie, Faculté de Médecine, Université Laval, Québec, QC G1V 0A6, Canada,Centre de Recherche du CHU de Québec - Université Laval, CHUL, Québec, QC G1V 4G2, Canada
| | - Kinji Asahina
- Southern California Research Center for ALPD and Cirrhosis, Department of Pathology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.
| |
Collapse
|