1
|
Tsutsumi H, Chiba T, Fujii Y, Matsushima T, Kimura T, Kanai A, Kishida A, Suzuki Y, Asahara H. Single-nucleus transcriptional and chromatin accessibility analyses of maturing mouse Achilles tendon uncover the molecular landscape of tendon stem/progenitor cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.24.619991. [PMID: 39484401 PMCID: PMC11527174 DOI: 10.1101/2024.10.24.619991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Tendons and ligaments are crucial connective tissues linking bones and muscles, yet achieving full functional recovery after injury remains challenging. We investigated the characteristics of tendon stem/progenitor cells (TSPCs) by focusing on the declining tendon repair capacity with growth. Using single-cell RNA sequencing on Achilles tendon cells from 2- and 6-week-old mice, we identified Cd55 and Cd248 as novel surface antigen markers for TSPCs. Combining single-nucleus ATAC and RNA sequencing analyses revealed that Cd55 and Cd248 positive fractions in tendon tissue are TSPCs, with this population decreasing at 1 weeks. We also identified candidate upstream transcription factors regulating these fractions. Functional analyses of isolated CD55/CD248 positive cells demonstrated high clonogenic potential and tendon differentiation capacity, forming functional tendon-like tissue in vitro . This study establishes CD55 and CD248 as novel TSPC surface antigens, potentially advancing tendon regenerative medicine and contributing to the development of new treatment strategies for tendon and ligament injuries.
Collapse
|
2
|
Schulte SJ, Fornace ME, Hall JK, Shin GJ, Pierce NA. HCR spectral imaging: 10-plex, quantitative, high-resolution RNA and protein imaging in highly autofluorescent samples. Development 2024; 151:dev202307. [PMID: 38415752 PMCID: PMC10941662 DOI: 10.1242/dev.202307] [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: 08/31/2023] [Accepted: 12/21/2023] [Indexed: 02/29/2024]
Abstract
Signal amplification based on the mechanism of hybridization chain reaction (HCR) provides a unified framework for multiplex, quantitative, high-resolution imaging of RNA and protein targets in highly autofluorescent samples. With conventional bandpass imaging, multiplexing is typically limited to four or five targets owing to the difficulty in separating signals generated by fluorophores with overlapping spectra. Spectral imaging has offered the conceptual promise of higher levels of multiplexing, but it has been challenging to realize this potential in highly autofluorescent samples, including whole-mount vertebrate embryos. Here, we demonstrate robust HCR spectral imaging with linear unmixing, enabling simultaneous imaging of ten RNA and/or protein targets in whole-mount zebrafish embryos and mouse brain sections. Further, we demonstrate that the amplified and unmixed signal in each of the ten channels is quantitative, enabling accurate and precise relative quantitation of RNA and/or protein targets with subcellular resolution, and RNA absolute quantitation with single-molecule resolution, in the anatomical context of highly autofluorescent samples.
Collapse
Affiliation(s)
- Samuel J. Schulte
- Division of Biology & Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Mark E. Fornace
- Division of Biology & Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - John K. Hall
- Division of Biology & Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Grace J. Shin
- Division of Biology & Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Niles A. Pierce
- Division of Biology & Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
- Division of Engineering & Applied Science, California Institute of Technology, Pasadena, CA 91125, USA
| |
Collapse
|
3
|
Yang X, Li X, Guo Z, Zhang Z, Song X, Zhang M, Han X, He L, Zhou B. Generation and characterization of PDGFRα-GFP knock-in mice for visualization of PDGFRα + fibroblasts in vivo. Biochem Biophys Res Commun 2023; 687:149215. [PMID: 37949027 DOI: 10.1016/j.bbrc.2023.149215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 10/27/2023] [Accepted: 11/03/2023] [Indexed: 11/12/2023]
Abstract
The platelet-derived growth factor (PDGF) and its receptor, PDGFRα, are critical for tissue development and injury repair. To track PDGFRα-expressing cells in vivo, we generated a knock-in mouse line that expresses green fluorescent protein (GFP) under the control of the PDGFRα promoter. This genetic tool enabled us to detect PDGFRα expression in various organs during both neonatal and adult stages. Additionally, we confirmed the correlation between endogenous PDGFRα and transgenic PDGFRα expression using mouse injury models, showing the potential of this genetic reporter for studying PDGFRα-mediated signaling pathways and developing therapeutic strategies. Overall, the PDGFRα-GFP knock-in mouse line serves as a valuable tool for investigating the biology of PDGFRα and its role in normal development and disease.
Collapse
Affiliation(s)
- Xueying Yang
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China
| | - Xufeng Li
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China
| | - Zhihou Guo
- School of Life Sciences, Westlake University, Hangzhou, Zhejiang, 310030, China
| | - Zhuonan Zhang
- School of Life Sciences, Westlake University, Hangzhou, Zhejiang, 310030, China
| | - Xin Song
- School of Life Sciences, Westlake University, Hangzhou, Zhejiang, 310030, China
| | - Mingjun Zhang
- New Cornerstone Science Laboratory, State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China
| | - Ximeng Han
- New Cornerstone Science Laboratory, State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China
| | - Lingjuan He
- School of Life Sciences, Westlake University, Hangzhou, Zhejiang, 310030, China.
| | - Bin Zhou
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China; New Cornerstone Science Laboratory, State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China; School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China.
| |
Collapse
|
4
|
Schulte SJ, Fornace ME, Hall JK, Pierce NA. HCR spectral imaging: 10-plex, quantitative, high-resolution RNA and protein imaging in highly autofluorescent samples. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.30.555626. [PMID: 37693627 PMCID: PMC10491186 DOI: 10.1101/2023.08.30.555626] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/12/2023]
Abstract
Signal amplification based on the mechanism of hybridization chain reaction (HCR) provides a unified framework for multiplex, quantitative, high-resolution imaging of RNA and protein targets in highly autofluorescent samples. With conventional bandpass imaging, multiplexing is typically limited to four or five targets due to the difficulty in separating signals generated by fluorophores with overlapping spectra. Spectral imaging has offered the conceptual promise of higher levels of multiplexing, but it has been challenging to realize this potential in highly autofluorescent samples including whole-mount vertebrate embryos. Here, we demonstrate robust HCR spectral imaging with linear unmixing, enabling simultaneous imaging of 10 RNA and/or protein targets in whole-mount zebrafish embryos and mouse brain sections. Further, we demonstrate that the amplified and unmixed signal in each of 10 channels is quantitative, enabling accurate and precise relative quantitation of RNA and/or protein targets with subcellular resolution, and RNA absolute quantitation with single-molecule resolution, in the anatomical context of highly autofluorescent samples. SUMMARY Spectral imaging with signal amplification based on the mechanism of hybridization chain reaction enables robust 10-plex, quantitative, high-resolution imaging of RNA and protein targets in whole-mount vertebrate embryos and brain sections.
Collapse
|
5
|
Schneider S, Hashmi SK, Thrasher AJ, Kothakapa DR, Wright CM, Heuckeroth RO. Single Nucleus Sequencing of Human Colon Myenteric Plexus-Associated Visceral Smooth Muscle Cells, Platelet Derived Growth Factor Receptor Alpha Cells, and Interstitial Cells of Cajal. GASTRO HEP ADVANCES 2023; 2:380-394. [PMID: 37206377 PMCID: PMC10194832 DOI: 10.1016/j.gastha.2022.12.004] [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] [Indexed: 05/21/2023]
Abstract
BACKGROUND AND AIMS Smooth muscle cells (SMCs), interstitial cells of Cajal (ICCs), and platelet-derived growth factor receptor alpha (PDGFRα+) cells (PαCs) form a functional syncytium in the bowel known as the "SIP syncytium." The SIP syncytium works in concert with the enteric nervous system (ENS) to coordinate bowel motility. However, our understanding of individual cell types that form this syncytium and how they interact with each other remains limited, with no prior single-cell RNAseq analyses focused on human SIP syncytium cells. METHODS We analyzed single-nucleus RNA sequencing data from 10,749 human colon SIP syncytium cells (5572 SMC, 372 ICC, and 4805 PαC nuclei) derived from 15 individuals. RESULTS Consistent with critical contractile and pacemaker functions and with known enteric nervous system interactions, SIP syncytium cell types express many ion channels, including mechanosensitive channels in ICCs and PαCs. PαCs also prominently express extracellular matrix-associated genes and the inhibitory neurotransmitter receptor for vasoactive intestinal peptide (VIPR2), a novel finding. We identified 2 PαC clusters that differ in the expression of many ion channels and transcriptional regulators. Interestingly, SIP syncytium cells co-express 6 transcription factors (FOS, MEIS1, MEIS2, PBX1, SCMH1, and ZBTB16) that may be part of a combinatorial signature that specifies these cells. Bowel region-specific differences in SIP syncytium gene expression may correlate with regional differences in function, with right (ascending) colon SMCs and PαCs expressing more transcriptional regulators and ion channels than SMCs and PαCs in left (sigmoid) colon. CONCLUSION These studies provide new insights into SIP syncytium biology that may be valuable for understanding bowel motility disorders and lead to future investigation of highlighted genes and pathways.
Collapse
Affiliation(s)
- Sabine Schneider
- Department of Pediatrics, The Children’s Hospital of Philadelphia Research Institute and the Perelman School of Medicine at the University of Pennsylvania, Abramson Research Center, Philadelphia, Pennsylvania
| | - Sohaib K. Hashmi
- Department of Pediatrics, The Children’s Hospital of Philadelphia Research Institute and the Perelman School of Medicine at the University of Pennsylvania, Abramson Research Center, Philadelphia, Pennsylvania
- Department of Bioengineering, The University of Pennsylvania School of Engineering and Applied Science, Philadelphia, Pennsylvania
| | - A. Josephine Thrasher
- Department of Pediatrics, The Children’s Hospital of Philadelphia Research Institute and the Perelman School of Medicine at the University of Pennsylvania, Abramson Research Center, Philadelphia, Pennsylvania
| | - Deepika R. Kothakapa
- Department of Pediatrics, The Children’s Hospital of Philadelphia Research Institute and the Perelman School of Medicine at the University of Pennsylvania, Abramson Research Center, Philadelphia, Pennsylvania
- Department of Biological Sciences, Rensselaer Polytechnic Institute, Troy, New York
- Albany Medical College, Albany, New York
| | - Christina M. Wright
- Department of Pediatrics, The Children’s Hospital of Philadelphia Research Institute and the Perelman School of Medicine at the University of Pennsylvania, Abramson Research Center, Philadelphia, Pennsylvania
| | - Robert O. Heuckeroth
- Department of Pediatrics, The Children’s Hospital of Philadelphia Research Institute and the Perelman School of Medicine at the University of Pennsylvania, Abramson Research Center, Philadelphia, Pennsylvania
| |
Collapse
|
6
|
The negative regulation of gene expression by microRNAs as key driver of inducers and repressors of cardiomyocyte differentiation. Clin Sci (Lond) 2022; 136:1179-1203. [PMID: 35979890 PMCID: PMC9411751 DOI: 10.1042/cs20220391] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 07/29/2022] [Accepted: 08/02/2022] [Indexed: 11/28/2022]
Abstract
Cardiac muscle damage-induced loss of cardiomyocytes (CMs) and dysfunction of the remaining ones leads to heart failure, which nowadays is the number one killer worldwide. Therapies fostering effective cardiac regeneration are the holy grail of cardiovascular research to stop the heart failure epidemic. The main goal of most myocardial regeneration protocols is the generation of new functional CMs through the differentiation of endogenous or exogenous cardiomyogenic cells. Understanding the cellular and molecular basis of cardiomyocyte commitment, specification, differentiation and maturation is needed to devise innovative approaches to replace the CMs lost after injury in the adult heart. The transcriptional regulation of CM differentiation is a highly conserved process that require sequential activation and/or repression of different genetic programs. Therefore, CM differentiation and specification have been depicted as a step-wise specific chemical and mechanical stimuli inducing complete myogenic commitment and cell-cycle exit. Yet, the demonstration that some microRNAs are sufficient to direct ESC differentiation into CMs and that four specific miRNAs reprogram fibroblasts into CMs show that CM differentiation must also involve negative regulatory instructions. Here, we review the mechanisms of CM differentiation during development and from regenerative stem cells with a focus on the involvement of microRNAs in the process, putting in perspective their negative gene regulation as a main modifier of effective CM regeneration in the adult heart.
Collapse
|
7
|
Chandrakanthan V, Rorimpandey P, Zanini F, Chacon D, Olivier J, Joshi S, Kang YC, Knezevic K, Huang Y, Qiao Q, Oliver RA, Unnikrishnan A, Carter DR, Lee B, Brownlee C, Power C, Brink R, Mendez-Ferrer S, Enikolopov G, Walsh W, Göttgens B, Taoudi S, Beck D, Pimanda JE. Mesoderm-derived PDGFRA + cells regulate the emergence of hematopoietic stem cells in the dorsal aorta. Nat Cell Biol 2022; 24:1211-1225. [PMID: 35902769 PMCID: PMC9359911 DOI: 10.1038/s41556-022-00955-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Accepted: 06/06/2022] [Indexed: 12/13/2022]
Abstract
Mouse haematopoietic stem cells (HSCs) first emerge at embryonic day 10.5 (E10.5), on the ventral surface of the dorsal aorta, by endothelial-to-haematopoietic transition. We investigated whether mesenchymal stem cells, which provide an essential niche for long-term HSCs (LT-HSCs) in the bone marrow, reside in the aorta-gonad-mesonephros and contribute to the development of the dorsal aorta and endothelial-to-haematopoietic transition. Here we show that mesoderm-derived PDGFRA+ stromal cells (Mesp1der PSCs) contribute to the haemogenic endothelium of the dorsal aorta and populate the E10.5-E11.5 aorta-gonad-mesonephros but by E13.5 were replaced by neural-crest-derived PSCs (Wnt1der PSCs). Co-aggregating non-haemogenic endothelial cells with Mesp1der PSCs but not Wnt1der PSCs resulted in activation of a haematopoietic transcriptional programme in endothelial cells and generation of LT-HSCs. Dose-dependent inhibition of PDGFRA or BMP, WNT and NOTCH signalling interrupted this reprogramming event. Together, aorta-gonad-mesonephros Mesp1der PSCs could potentially be harnessed to manufacture LT-HSCs from endothelium.
Collapse
Affiliation(s)
- Vashe Chandrakanthan
- Adult Cancer Program, Lowy Cancer Research Centre, UNSW Sydney, Sydney, NSW, Australia. .,Department of Pathology, School of Medical Sciences, UNSW Sydney, Sydney, NSW, Australia.
| | - Prunella Rorimpandey
- Adult Cancer Program, Lowy Cancer Research Centre, UNSW Sydney, Sydney, NSW, Australia.,Department of Pathology, School of Medical Sciences, UNSW Sydney, Sydney, NSW, Australia
| | - Fabio Zanini
- Adult Cancer Program, Lowy Cancer Research Centre, UNSW Sydney, Sydney, NSW, Australia.,School of Clinical Medicine, UNSW Sydney, Sydney, NSW, Australia.,Garvan-Weizmann Centre for Cellular Genomics, Sydney, Australia.,UNSW Futures Institute for Cellular Genomics, Sydney, Australia
| | - Diego Chacon
- Centre for Health Technologies and the School of Biomedical Engineering, University of Technology Sydney, Sydney, NSW, Australia
| | - Jake Olivier
- School of Mathematics and Statistics, UNSW Sydney, Sydney, NSW, Australia
| | - Swapna Joshi
- Adult Cancer Program, Lowy Cancer Research Centre, UNSW Sydney, Sydney, NSW, Australia.,Department of Pathology, School of Medical Sciences, UNSW Sydney, Sydney, NSW, Australia
| | - Young Chan Kang
- Adult Cancer Program, Lowy Cancer Research Centre, UNSW Sydney, Sydney, NSW, Australia.,Department of Pathology, School of Medical Sciences, UNSW Sydney, Sydney, NSW, Australia
| | - Kathy Knezevic
- Adult Cancer Program, Lowy Cancer Research Centre, UNSW Sydney, Sydney, NSW, Australia.,Department of Pathology, School of Medical Sciences, UNSW Sydney, Sydney, NSW, Australia
| | - Yizhou Huang
- School of Clinical Medicine, UNSW Sydney, Sydney, NSW, Australia.,School of Mathematics and Statistics, UNSW Sydney, Sydney, NSW, Australia.,Children's Cancer Institute Australia for Medical Research, Lowy Cancer Research Centre, UNSW Sydney, Sydney, NSW, Australia
| | - Qiao Qiao
- Adult Cancer Program, Lowy Cancer Research Centre, UNSW Sydney, Sydney, NSW, Australia
| | - Rema A Oliver
- Surgical & Orthopaedic Research Laboratories, Prince of Wales Clinical School, UNSW Sydney, Sydney, NSW, Australia
| | - Ashwin Unnikrishnan
- Adult Cancer Program, Lowy Cancer Research Centre, UNSW Sydney, Sydney, NSW, Australia.,School of Clinical Medicine, UNSW Sydney, Sydney, NSW, Australia
| | - Daniel R Carter
- School of Clinical Medicine, UNSW Sydney, Sydney, NSW, Australia.,School of Mathematics and Statistics, UNSW Sydney, Sydney, NSW, Australia.,Children's Cancer Institute Australia for Medical Research, Lowy Cancer Research Centre, UNSW Sydney, Sydney, NSW, Australia
| | - Brendan Lee
- Biological Resources Imaging Laboratory, Mark Wainwright Analytical Centre, Lowy Cancer Research Centre, UNSW Sydney, Sydney, NSW, Australia
| | - Chris Brownlee
- Biological Resources Imaging Laboratory, Mark Wainwright Analytical Centre, Lowy Cancer Research Centre, UNSW Sydney, Sydney, NSW, Australia
| | - Carl Power
- Biological Resources Imaging Laboratory, Mark Wainwright Analytical Centre, Lowy Cancer Research Centre, UNSW Sydney, Sydney, NSW, Australia
| | - Robert Brink
- Garvan Institute of Medical Research, Sydney, NSW, Australia.,UNSW Sydney, Sydney, NSW, Australia
| | - Simon Mendez-Ferrer
- Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute and Department of Haematology, University of Cambridge, Cambridge, UK
| | - Grigori Enikolopov
- Center for Developmental Genetics and Department of Anesthesiology, Stony Brook University, Stony Brook, NY, USA
| | - William Walsh
- Surgical & Orthopaedic Research Laboratories, Prince of Wales Clinical School, UNSW Sydney, Sydney, NSW, Australia
| | - Berthold Göttgens
- Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute and Department of Haematology, University of Cambridge, Cambridge, UK
| | - Samir Taoudi
- Epigenetics and development division, Walter and Eliza Hall Institute, Melbourne, VIC, Australia.,Department of Medical Biology, University of Melbourne, Melbourne, VIC, Australia
| | - Dominik Beck
- Centre for Health Technologies and the School of Biomedical Engineering, University of Technology Sydney, Sydney, NSW, Australia
| | - John E Pimanda
- Adult Cancer Program, Lowy Cancer Research Centre, UNSW Sydney, Sydney, NSW, Australia. .,Department of Pathology, School of Medical Sciences, UNSW Sydney, Sydney, NSW, Australia. .,Department of Haematology, The Prince of Wales Hospital, Sydney, NSW, Australia.
| |
Collapse
|
8
|
Ha SE, Jorgensen BG, Wei L, Jin B, Kim MS, Poudrier SM, Singh R, Bartlett A, Zogg H, Kim S, Baek G, Kurahashi M, Lee MY, Kim YS, Choi SC, Sasse KC, Rubin SJS, Gottfried-Blackmore A, Becker L, Habtezion A, Sanders KM, Ro S. Metalloendopeptidase ADAM-like Decysin 1 (ADAMDEC1) in Colonic Subepithelial PDGFRα + Cells Is a New Marker for Inflammatory Bowel Disease. Int J Mol Sci 2022; 23:5007. [PMID: 35563399 PMCID: PMC9103908 DOI: 10.3390/ijms23095007] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 04/19/2022] [Accepted: 04/22/2022] [Indexed: 12/25/2022] Open
Abstract
Metalloendopeptidase ADAM-Like Decysin 1 (ADAMDEC1) is an anti-inflammatory peptidase that is almost exclusively expressed in the gastrointestinal (GI) tract. We have recently found abundant and selective expression of Adamdec1 in colonic mucosal PDGFRα+ cells. However, the cellular origin for this gene expression is controversial as it is also known to be expressed in intestinal macrophages. We found that Adamdec1 mRNAs were selectively expressed in colonic mucosal subepithelial PDGFRα+ cells. ADAMDEC1 protein was mainly released from PDGFRα+ cells and accumulated in the mucosal layer lamina propria space near the epithelial basement membrane. PDGFRα+ cells significantly overexpressed Adamdec1 mRNAs and protein in DSS-induced colitis mice. Adamdec1 was predominantly expressed in CD45- PDGFRα+ cells in DSS-induced colitis mice, with only minimal expression in CD45+ CD64+ macrophages. Additionally, overexpression of both ADAMDEC1 mRNA and protein was consistently observed in PDGFRα+ cells, but not in CD64+ macrophages found in human colonic mucosal tissue affected by Crohn's disease. In summary, PDGFRα+ cells selectively express ADAMDEC1, which is localized to the colon mucosa layer. ADAMDEC1 expression significantly increases in DSS-induced colitis affected mice and Crohn's disease affected human tissue, suggesting that this gene can serve as a diagnostic and/or therapeutic target for intestinal inflammation and Crohn's disease.
Collapse
Affiliation(s)
- Se Eun Ha
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, NV 89557, USA; (S.E.H.); (B.G.J.); (L.W.); (B.J.); (S.M.P.); (R.S.); (A.B.); (H.Z.); (S.K.); (G.B.); (K.M.S.)
| | - Brian G. Jorgensen
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, NV 89557, USA; (S.E.H.); (B.G.J.); (L.W.); (B.J.); (S.M.P.); (R.S.); (A.B.); (H.Z.); (S.K.); (G.B.); (K.M.S.)
| | - Lai Wei
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, NV 89557, USA; (S.E.H.); (B.G.J.); (L.W.); (B.J.); (S.M.P.); (R.S.); (A.B.); (H.Z.); (S.K.); (G.B.); (K.M.S.)
| | - Byungchang Jin
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, NV 89557, USA; (S.E.H.); (B.G.J.); (L.W.); (B.J.); (S.M.P.); (R.S.); (A.B.); (H.Z.); (S.K.); (G.B.); (K.M.S.)
| | - Min-Seob Kim
- Department of Physiology, Wonkwang Digestive Disease Research Institute and Institute of Wonkwang Medical Science, School of Medicine, Wonkwang University, Iksan 54538, Korea; (M.-S.K.); (M.-Y.L.); (Y.-S.K.)
| | - Sandra M. Poudrier
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, NV 89557, USA; (S.E.H.); (B.G.J.); (L.W.); (B.J.); (S.M.P.); (R.S.); (A.B.); (H.Z.); (S.K.); (G.B.); (K.M.S.)
| | - Rajan Singh
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, NV 89557, USA; (S.E.H.); (B.G.J.); (L.W.); (B.J.); (S.M.P.); (R.S.); (A.B.); (H.Z.); (S.K.); (G.B.); (K.M.S.)
| | - Allison Bartlett
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, NV 89557, USA; (S.E.H.); (B.G.J.); (L.W.); (B.J.); (S.M.P.); (R.S.); (A.B.); (H.Z.); (S.K.); (G.B.); (K.M.S.)
| | - Hannah Zogg
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, NV 89557, USA; (S.E.H.); (B.G.J.); (L.W.); (B.J.); (S.M.P.); (R.S.); (A.B.); (H.Z.); (S.K.); (G.B.); (K.M.S.)
| | - Sei Kim
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, NV 89557, USA; (S.E.H.); (B.G.J.); (L.W.); (B.J.); (S.M.P.); (R.S.); (A.B.); (H.Z.); (S.K.); (G.B.); (K.M.S.)
| | - Gain Baek
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, NV 89557, USA; (S.E.H.); (B.G.J.); (L.W.); (B.J.); (S.M.P.); (R.S.); (A.B.); (H.Z.); (S.K.); (G.B.); (K.M.S.)
| | - Masaaki Kurahashi
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, University of Iowa, Iowa City, IA 52242, USA;
| | - Moon-Young Lee
- Department of Physiology, Wonkwang Digestive Disease Research Institute and Institute of Wonkwang Medical Science, School of Medicine, Wonkwang University, Iksan 54538, Korea; (M.-S.K.); (M.-Y.L.); (Y.-S.K.)
| | - Yong-Sung Kim
- Department of Physiology, Wonkwang Digestive Disease Research Institute and Institute of Wonkwang Medical Science, School of Medicine, Wonkwang University, Iksan 54538, Korea; (M.-S.K.); (M.-Y.L.); (Y.-S.K.)
| | - Suck-Chei Choi
- Department of gastroenterology, Wonkwang Digestive Disease Research Institute and Institute of Wonkwang Medical Science, School of Medicine, Wonkwang University, Iksan 54538, Korea;
| | | | - Samuel J. S. Rubin
- Gastroenterology and Hepatology, Stanford University School of Medicine, Stanford, CA 94305, USA; (S.J.S.R.); (A.G.-B.); (L.B.); (A.H.)
| | - Andres Gottfried-Blackmore
- Gastroenterology and Hepatology, Stanford University School of Medicine, Stanford, CA 94305, USA; (S.J.S.R.); (A.G.-B.); (L.B.); (A.H.)
| | - Laren Becker
- Gastroenterology and Hepatology, Stanford University School of Medicine, Stanford, CA 94305, USA; (S.J.S.R.); (A.G.-B.); (L.B.); (A.H.)
| | - Aida Habtezion
- Gastroenterology and Hepatology, Stanford University School of Medicine, Stanford, CA 94305, USA; (S.J.S.R.); (A.G.-B.); (L.B.); (A.H.)
| | - Kenton M. Sanders
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, NV 89557, USA; (S.E.H.); (B.G.J.); (L.W.); (B.J.); (S.M.P.); (R.S.); (A.B.); (H.Z.); (S.K.); (G.B.); (K.M.S.)
| | - Seungil Ro
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, NV 89557, USA; (S.E.H.); (B.G.J.); (L.W.); (B.J.); (S.M.P.); (R.S.); (A.B.); (H.Z.); (S.K.); (G.B.); (K.M.S.)
| |
Collapse
|
9
|
Solinc J, Raimbault‐Machado J, Dierick F, El Bernoussi L, Tu L, Thuillet R, Mougenot N, Hoareau‐Coudert B, Monceau V, Pavoine C, Atassi F, Sassoon D, Marazzi G, Harvey RP, Schofield P, Christ D, Humbert M, Guignabert C, Soubrier F, Nadaud S. Platelet‐Derived Growth Factor Receptor Type α Activation Drives Pulmonary Vascular Remodeling Via Progenitor Cell Proliferation and Induces Pulmonary Hypertension. J Am Heart Assoc 2022; 11:e023021. [PMID: 35348002 PMCID: PMC9075467 DOI: 10.1161/jaha.121.023021] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Background Platelet‐derived growth factor is a major regulator of the vascular remodeling associated with pulmonary arterial hypertension. We previously showed that protein widely 1 (PW1+) vascular progenitor cells participate in early vessel neomuscularization during experimental pulmonary hypertension (PH) and we addressed the role of the platelet‐derived growth factor receptor type α (PDGFRα) pathway in progenitor cell‐dependent vascular remodeling and in PH development. Methods and Results Remodeled pulmonary arteries from patients with idiopathic pulmonary arterial hypertension showed an increased number of perivascular and vascular PW1+ cells expressing PDGFRα. PW1nLacZ reporter mice were used to follow the fate of pulmonary PW1+ progenitor cells in a model of chronic hypoxia–induced PH development. Under chronic hypoxia, PDGFRα inhibition prevented the increase in PW1+ progenitor cell proliferation and differentiation into vascular smooth muscle cells and reduced pulmonary vessel neomuscularization, but did not prevent an increased right ventricular systolic pressure or the development of right ventricular hypertrophy. Conversely, constitutive PDGFRα activation led to neomuscularization via PW1+ progenitor cell differentiation into new smooth muscle cells and to PH development in male mice without fibrosis. In vitro, PW1+ progenitor cell proliferation, but not differentiation, was dependent on PDGFRα activity. Conclusions These results demonstrate a major role of PDGFRα signaling in progenitor cell–dependent lung vessel neomuscularization and vascular remodeling contributing to PH development, including in idiopathic pulmonary arterial hypertension patients. Our findings suggest that PDGFRα blockers may offer a therapeutic add‐on strategy to combine with current pulmonary arterial hypertension treatments to reduce vascular remodeling. Furthermore, our study highlights constitutive PDGFRα activation as a novel experimental PH model.
Collapse
Affiliation(s)
- Julien Solinc
- Sorbonne Université, INSERM, UMR_S 1166, Faculté de Médecine Pitié‐Salpêtrière Paris France
- ICAN Institute Paris France
| | - Jessica Raimbault‐Machado
- Sorbonne Université, INSERM, UMR_S 1166, Faculté de Médecine Pitié‐Salpêtrière Paris France
- ICAN Institute Paris France
| | - France Dierick
- Sorbonne Université, INSERM, UMR_S 1166, Faculté de Médecine Pitié‐Salpêtrière Paris France
- Lady Davis Institute for Medical Research, McGill University Montréal QC Canada
| | - Lamiaa El Bernoussi
- Sorbonne Université, INSERM, UMR_S 1166, Faculté de Médecine Pitié‐Salpêtrière Paris France
- ICAN Institute Paris France
| | - Ly Tu
- Université Paris‐Saclay, School of Medicine Le Kremlin‐Bicêtre France
- INSERM, Hôpital Marie Lannelongue, UMR_S 999 «Pulmonary Hypertension: Pathophysiology and Novel Therapies Le Plessis‐Robinson France
| | - Raphaël Thuillet
- Université Paris‐Saclay, School of Medicine Le Kremlin‐Bicêtre France
- INSERM, Hôpital Marie Lannelongue, UMR_S 999 «Pulmonary Hypertension: Pathophysiology and Novel Therapies Le Plessis‐Robinson France
| | - Nathalie Mougenot
- Sorbonne Universités, INSERM, UMS2, Faculté de Médecine Pitié‐Salpêtrière Paris France
| | | | | | - Catherine Pavoine
- Sorbonne Université, INSERM, UMR_S 1166, Faculté de Médecine Pitié‐Salpêtrière Paris France
- ICAN Institute Paris France
| | - Fabrice Atassi
- Sorbonne Université, INSERM, UMR_S 1166, Faculté de Médecine Pitié‐Salpêtrière Paris France
- ICAN Institute Paris France
| | - David Sassoon
- Université de Paris, INSERM, Paris Cardiovascular Research Center Paris France
| | - Giovanna Marazzi
- Université de Paris, INSERM, Paris Cardiovascular Research Center Paris France
| | - Richard P. Harvey
- Victor Chang Cardiac Research Institute Darlinghurst Australia
- St. Vincent’s Clinical School and School of Biotechnology and Biomolecular Science UNSW Sydney Sydney Australia
| | - Peter Schofield
- St. Vincent’s Clinical School and School of Biotechnology and Biomolecular Science UNSW Sydney Sydney Australia
- Immunology Division Garvan Institute of Medical Research Darlinghurst Australia
| | - Daniel Christ
- St. Vincent’s Clinical School and School of Biotechnology and Biomolecular Science UNSW Sydney Sydney Australia
- Immunology Division Garvan Institute of Medical Research Darlinghurst Australia
| | - Marc Humbert
- Université Paris‐Saclay, School of Medicine Le Kremlin‐Bicêtre France
- INSERM, Hôpital Marie Lannelongue, UMR_S 999 «Pulmonary Hypertension: Pathophysiology and Novel Therapies Le Plessis‐Robinson France
- Department of Respiratory and Intensive Care Medicine Assistance Publique–Hôpitaux de Paris (AP‐HP)Pulmonary Hypertension National Referral CenterHôpital Bicêtre Le Kremlin‐Bicêtre France
| | - Christophe Guignabert
- Université Paris‐Saclay, School of Medicine Le Kremlin‐Bicêtre France
- INSERM, Hôpital Marie Lannelongue, UMR_S 999 «Pulmonary Hypertension: Pathophysiology and Novel Therapies Le Plessis‐Robinson France
| | - Florent Soubrier
- Sorbonne Université, INSERM, UMR_S 1166, Faculté de Médecine Pitié‐Salpêtrière Paris France
- ICAN Institute Paris France
| | - Sophie Nadaud
- Sorbonne Université, INSERM, UMR_S 1166, Faculté de Médecine Pitié‐Salpêtrière Paris France
- ICAN Institute Paris France
| |
Collapse
|
10
|
Schwarzkopf M, Liu MC, Schulte SJ, Ives R, Husain N, Choi HMT, Pierce NA. Hybridization chain reaction enables a unified approach to multiplexed, quantitative, high-resolution immunohistochemistry and in situ hybridization. Development 2021; 148:dev199847. [PMID: 35020875 PMCID: PMC8645210 DOI: 10.1242/dev.199847] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Accepted: 10/12/2021] [Indexed: 12/20/2022]
Abstract
RNA in situ hybridization based on the mechanism of the hybridization chain reaction (HCR) enables multiplexed, quantitative, high-resolution RNA imaging in highly autofluorescent samples, including whole-mount vertebrate embryos, thick brain slices and formalin-fixed paraffin-embedded tissue sections. Here, we extend the benefits of one-step, multiplexed, quantitative, isothermal, enzyme-free HCR signal amplification to immunohistochemistry, enabling accurate and precise protein relative quantitation with subcellular resolution in an anatomical context. Moreover, we provide a unified framework for simultaneous quantitative protein and RNA imaging with one-step HCR signal amplification performed for all target proteins and RNAs simultaneously.
Collapse
Affiliation(s)
- Maayan Schwarzkopf
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Mike C. Liu
- Molecular Instruments, Los Angeles, CA 90041, USA
| | - Samuel J. Schulte
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Rachel Ives
- Molecular Instruments, Los Angeles, CA 90041, USA
| | - Naeem Husain
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | | | - Niles A. Pierce
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
- Division of Engineering & Applied Science, California Institute of Technology, Pasadena, CA 91125, USA
| |
Collapse
|
11
|
The Dorsal Integument of the Southern Long-Nosed Armadillo Dasypus hybridus (Cingulata, Xenarthra), and a Possible Neural Crest Origin of the Osteoderms. Discussing Evolutive Consequences for Amniota. J MAMM EVOL 2021. [DOI: 10.1007/s10914-021-09538-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
12
|
Lineage Contribution of PDGFR α-Expressing Cells in the Developing Mouse Eye. BIOMED RESEARCH INTERNATIONAL 2021; 2021:4982227. [PMID: 34285913 PMCID: PMC8275403 DOI: 10.1155/2021/4982227] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Revised: 06/16/2021] [Accepted: 06/21/2021] [Indexed: 02/02/2023]
Abstract
PDGFRα signaling is critically important in ocular development. Previous data on PDGFRα lacks an expression map with high spatial and temporal resolution and lineage information. In this study, we aim to present a detailed PDGFRα expression and lineage map from early embryogenesis to adulthood. PDGFRα-CreER; mT/mG reporter mice were analyzed. mEGFP-positive cells contributed to multiple ocular lineages in a spatiotemporally regulated manner. A dynamic PDGFRα expression was identified in corneal stromal cells, lens epithelial cells, lens fiber cells, and retinal astrocytes during the entire period of eye development, while PDGFRα expression in retinal astrocytes from E17.5 onwards and in Müller glial cells was identified within two weeks after birth. By revealing detailed characterization of gene expression and function, we present a comprehensive map of PDGFRα-expressing cells in the eye for a better understanding of PDGFRα signaling's role during eye development.
Collapse
|
13
|
Guo W, Spiller KV, Tang J, Karner CM, Hilton MJ, Wu C. Hypoxia depletes contaminating CD45 + hematopoietic cells from murine bone marrow stromal cell (BMSC) cultures: Methods for BMSC culture purification. Stem Cell Res 2021; 53:102317. [PMID: 33848794 DOI: 10.1016/j.scr.2021.102317] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 03/01/2021] [Accepted: 03/25/2021] [Indexed: 12/28/2022] Open
Abstract
Culture expanded bone marrow stromal cells (BMSCs) are easily isolated, can be grown rapidly en masse, and contain both skeletal stem cells (SSCs) and multipotent mesenchymal progenitors (MMPs). Despite this functional heterogeneity, BMSCs continue to be utilized for many applications due to the lack of definitive and universally accepted markers to prospectively identify and purify SSCs. Isolation is widely based on adherence to tissue culture plastic; however, high hematopoietic contamination is a significant impediment in murine models. Remarkably, when cultured at a physiological oxygen tension of 1% O2, a 10-fold reduction in CD45+ hematopoietic cells associated with a concomitant increase in PDGFRα+ stromal cells occur. This is due, in part, to a differential response of the two populations to hypoxia. In standard tissue culture conditions of 21% O2, CD45+ cells showed increased proliferation coupled with no changes in cell death compared to their counterparts grown at 1% O2. In contrast, PDGFR α+ stromal cells responded to hypoxia by increasing proliferation and exhibiting a 10-fold decrease in cell death. In summary, we describe a simple and reliable method exploiting the divergent biological response of hematopoietic and stromal cells to hypoxia to significantly increase the PDGFR α+ stromal cell population in murine BMSC cultures.
Collapse
Affiliation(s)
- Wendi Guo
- Department of Orthopaedic Surgery, Duke University School of Medicine, USA; Department of Pharmacology & Cancer Biology, Duke University School of Medicine, USA
| | | | - Jackie Tang
- Department of Orthopaedic Surgery, Duke University School of Medicine, USA
| | - Courtney M Karner
- Department of Orthopaedic Surgery, Duke University School of Medicine, USA; Department of Cell Biology, Duke University School of Medicine, USA
| | - Matthew J Hilton
- Department of Orthopaedic Surgery, Duke University School of Medicine, USA; Department of Cell Biology, Duke University School of Medicine, USA
| | - Colleen Wu
- Department of Orthopaedic Surgery, Duke University School of Medicine, USA; Department of Cell Biology, Duke University School of Medicine, USA; Department of Pharmacology & Cancer Biology, Duke University School of Medicine, USA.
| |
Collapse
|
14
|
Miura A, Shimbo T, Kitayama T, Ouchi Y, Yamazaki S, Nishida M, Takaki E, Yamamoto R, Wijaya E, Tamai K. Contribution of PDGFRα lineage cells in adult mouse hematopoiesis. Biochem Biophys Res Commun 2020; 534:186-192. [PMID: 33309273 DOI: 10.1016/j.bbrc.2020.11.114] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Accepted: 11/27/2020] [Indexed: 12/31/2022]
Abstract
Platelet-derived growth factor receptor alpha (PDGFRα) is a dominant marker of mesodermal mesenchymal cells in mice. Previous studies demonstrated that PDGFRα-positive (PDGFRα+) mesodermal cells develop not only into mesenchymal cells but also into a subset of total hematopoietic cells (HCs) in the limited period during mouse embryogenesis. However, the precise characteristics of the PDGFRα lineage positive (PDGFRα Lin+) HCs in adult mouse hematopoiesis are largely unknown. In this study, we systematically evaluated the characteristics of PDGFRα Lin+ HCs in the bone marrow and peripheral blood using PDGFRα-CRE; ROSAtdTomato mice. Flow cytometry analysis revealed that PDGFRα Lin+ HCs accounted for approximately 20% of total HCs in both the bone marrow and peripheral blood in adult mice. Compositions of myeloid and lymphoid subpopulations among CD45+ mononuclear cells were almost identical in both PDGFRα Lin+ and PDGFRα Lin- cells. Single-cell RNA-sequencing analysis also demonstrated that the transcriptomic signatures of the PDGFRα Lin+ HCs in the peripheral blood largely overlapped with those of the PDGFRα Lin- HCs, suggesting equivalent functions of the PDGFRα Lin+ and PDGFRα Lin- HCs. Although pathophysiological activities of the PDGFRα Lin + HCs were not evaluated, our data clearly demonstrate a significant role of the PDGFRα Lin + HCs in physiological hematopoiesis in adult mice.
Collapse
Affiliation(s)
- Asaka Miura
- Department of Stem Cell Therapy Science, Graduate School of Medicine, Osaka University, Suita, 565-0871, Japan
| | - Takashi Shimbo
- Department of Stem Cell Therapy Science, Graduate School of Medicine, Osaka University, Suita, 565-0871, Japan; StemRIM Institute of Regeneration-Inducing Medicine, Osaka University, Suita, 565-0871, Japan
| | - Tomomi Kitayama
- Department of Stem Cell Therapy Science, Graduate School of Medicine, Osaka University, Suita, 565-0871, Japan; StemRIM Co., Ltd., Ibaraki, Osaka, 567-0085, Japan
| | - Yuya Ouchi
- Department of Stem Cell Therapy Science, Graduate School of Medicine, Osaka University, Suita, 565-0871, Japan; StemRIM Co., Ltd., Ibaraki, Osaka, 567-0085, Japan
| | - Sho Yamazaki
- Department of Stem Cell Therapy Science, Graduate School of Medicine, Osaka University, Suita, 565-0871, Japan; StemRIM Co., Ltd., Ibaraki, Osaka, 567-0085, Japan
| | - Mami Nishida
- Department of Stem Cell Therapy Science, Graduate School of Medicine, Osaka University, Suita, 565-0871, Japan; StemRIM Co., Ltd., Ibaraki, Osaka, 567-0085, Japan
| | - Eiichi Takaki
- Department of Stem Cell Therapy Science, Graduate School of Medicine, Osaka University, Suita, 565-0871, Japan; StemRIM Co., Ltd., Ibaraki, Osaka, 567-0085, Japan
| | - Ryoma Yamamoto
- Department of Stem Cell Therapy Science, Graduate School of Medicine, Osaka University, Suita, 565-0871, Japan; StemRIM Co., Ltd., Ibaraki, Osaka, 567-0085, Japan
| | - Edward Wijaya
- Department of Stem Cell Therapy Science, Graduate School of Medicine, Osaka University, Suita, 565-0871, Japan; StemRIM Co., Ltd., Ibaraki, Osaka, 567-0085, Japan
| | - Katsuto Tamai
- Department of Stem Cell Therapy Science, Graduate School of Medicine, Osaka University, Suita, 565-0871, Japan.
| |
Collapse
|
15
|
Fukushima H, Yoshioka M, Kawatou M, López-Dávila V, Takeda M, Kanda Y, Sekino Y, Yoshida Y, Yamashita JK. Specific induction and long-term maintenance of high purity ventricular cardiomyocytes from human induced pluripotent stem cells. PLoS One 2020; 15:e0241287. [PMID: 33137106 PMCID: PMC7605685 DOI: 10.1371/journal.pone.0241287] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Accepted: 10/13/2020] [Indexed: 12/28/2022] Open
Abstract
Currently, cardiomyocyte (CM) differentiation methods require a purification step after CM induction to ensure the high purity of the cell population. Here we show an improved human CM differentiation protocol with which high-purity ventricular-type CMs can be obtained and maintained without any CM purification process. We induced and collected a mesodermal cell population (platelet-derived growth factor receptor-α (PDGFRα)-positive cells) that can respond to CM differentiation cues, and then stimulated CM differentiation by means of Wnt inhibition. This method reproducibly generated CMs with purities above 95% in several human pluripotent stem cell lines. Furthermore, these CM populations were maintained in culture at such high purity without any further CM purification step for over 200 days. The majority of these CMs (>95%) exhibited a ventricular-like phenotype with a tendency to structural and electrophysiological maturation, including T-tubule-like structure formation and the ability to respond to QT prolongation drugs. This is a simple and valuable method to stably generate CM populations suitable for cardiac toxicology testing, disease modeling and regenerative medicine.
Collapse
Affiliation(s)
- Hiroyuki Fukushima
- Department of Cell Growth and Differentiation, Laboratory of Stem Cell Differentiation, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Miki Yoshioka
- Department of Cell Growth and Differentiation, Laboratory of Stem Cell Differentiation, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Masahide Kawatou
- Department of Cell Growth and Differentiation, Laboratory of Stem Cell Differentiation, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
- Department of Cardiovascular Surgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
- Institute for Advancement of Clinical and Translational Science (iACT), Kyoto University Hospital, Kyoto, Japan
| | - Víctor López-Dávila
- Department of Cell Growth and Differentiation, Laboratory of Stem Cell Differentiation, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Masafumi Takeda
- Department of Cell Growth and Differentiation, Laboratory of Stem Cell Differentiation, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
- Institute for Advancement of Clinical and Translational Science (iACT), Kyoto University Hospital, Kyoto, Japan
| | - Yasunari Kanda
- Division of Pharmacology, National Institute of Health Sciences, Kawasaki, Kanagawa, Japan
| | - Yuko Sekino
- Division of Pharmacology, National Institute of Health Sciences, Kawasaki, Kanagawa, Japan
| | - Yoshinori Yoshida
- Department of Cell Growth and Differentiation, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Jun K. Yamashita
- Department of Cell Growth and Differentiation, Laboratory of Stem Cell Differentiation, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
- * E-mail:
| |
Collapse
|
16
|
Thorup AS, Dell'Accio F, Eldridge SE. Lessons from joint development for cartilage repair in the clinic. Dev Dyn 2020; 250:360-376. [PMID: 32738003 DOI: 10.1002/dvdy.228] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 07/23/2020] [Accepted: 07/24/2020] [Indexed: 12/19/2022] Open
Abstract
More than 250 years ago, William Hunter stated that when cartilage is destroyed it never recovers. In the last 20 years, the understanding of the mechanisms that lead to joint formation and the knowledge that some of these mechanisms are reactivated in the homeostatic responses of cartilage to injury has offered an unprecedented therapeutic opportunity to achieve cartilage regeneration. Very large investments in ambitious clinical trials are finally revealing that, although we do not have perfect medicines yet, disease modification is a feasible possibility for human osteoarthritis.
Collapse
Affiliation(s)
- Anne-Sophie Thorup
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Francesco Dell'Accio
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Suzanne E Eldridge
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| |
Collapse
|
17
|
Eisner C, Cummings M, Johnston G, Tung LW, Groppa E, Chang C, Rossi FM. Murine Tissue-Resident PDGFRα+ Fibro-Adipogenic Progenitors Spontaneously Acquire Osteogenic Phenotype in an Altered Inflammatory Environment. J Bone Miner Res 2020; 35:1525-1534. [PMID: 32251540 DOI: 10.1002/jbmr.4020] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Revised: 03/05/2020] [Accepted: 03/24/2020] [Indexed: 01/11/2023]
Abstract
Acquired heterotopic ossifications (HO) arising as a result of various traumas, including injury or surgical interventions, often result in pain and loss of motion. Though triggers for HO have been identified, the cellular source of these heterotopic lesions as well as the underlying mechanisms that drive the formation of acquired HO remain poorly understood, and treatment options, including preventative treatments, remain limited. Here, we explore the cellular source of HO and a possible underlying mechanism for their spontaneous osteogenic differentiation. We demonstrate that HO lesions arise from tissue-resident PDGFRα+ fibro/adipogenic progenitors (FAPs) in skeletal muscle and not from circulating bone marrow-derived progenitors. Further, we show that accumulation of these cells in the tissue after damage due to alterations in the inflammatory environment can result in activation of their inherent osteogenic potential. This work suggests a mechanism by which an altered inflammatory cell and FAP interactions can lead to the formation of HO after injury and presents potential targets for therapeutics in acquired HO. © 2020 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Christine Eisner
- Department of Medical Genetics, University of British Columbia, Vancouver, Canada.,Faculty of Medicine, The University of British Columbia, Vancouver, Canada
| | - Michael Cummings
- Department of Biochemistry, University of British Columbia, Vancouver, Canada
| | | | - Lin Wei Tung
- Department of Medical Genetics, University of British Columbia, Vancouver, Canada.,Faculty of Medicine, The University of British Columbia, Vancouver, Canada
| | - Elena Groppa
- Department of Medical Genetics, University of British Columbia, Vancouver, Canada.,Faculty of Medicine, The University of British Columbia, Vancouver, Canada
| | - Chihkai Chang
- Department of Medical Genetics, University of British Columbia, Vancouver, Canada
| | - Fabio Mv Rossi
- Department of Medical Genetics, University of British Columbia, Vancouver, Canada.,Faculty of Medicine, The University of British Columbia, Vancouver, Canada
| |
Collapse
|
18
|
Greicius G, Virshup DM. Stromal control of intestinal development and the stem cell niche. Differentiation 2019; 108:8-16. [DOI: 10.1016/j.diff.2019.01.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Accepted: 01/07/2019] [Indexed: 02/07/2023]
|
19
|
Reinhardt R, Gullotta F, Nusspaumer G, Ünal E, Ivanek R, Zuniga A, Zeller R. Molecular signatures identify immature mesenchymal progenitors in early mouse limb buds that respond differentially to morphogen signaling. Development 2019; 146:dev.173328. [PMID: 31076486 PMCID: PMC6550019 DOI: 10.1242/dev.173328] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Accepted: 05/01/2019] [Indexed: 12/31/2022]
Abstract
The key molecular interactions governing vertebrate limb bud development are a paradigm for studying the mechanisms controlling progenitor cell proliferation and specification during vertebrate organogenesis. However, little is known about the cellular heterogeneity of the mesenchymal progenitors in early limb buds that ultimately contribute to the chondrogenic condensations prefiguring the skeleton. We combined flow cytometric and transcriptome analyses to identify the molecular signatures of several distinct mesenchymal progenitor cell populations present in early mouse forelimb buds. In particular, jagged 1 (JAG1)-positive cells located in the posterior-distal mesenchyme were identified as the most immature limb bud mesenchymal progenitors (LMPs), which crucially depend on SHH and FGF signaling in culture. The analysis of gremlin 1 (Grem1)-deficient forelimb buds showed that JAG1-expressing LMPs are protected from apoptosis by GREM1-mediated BMP antagonism. At the same stage, the osteo-chondrogenic progenitors (OCPs) located in the core mesenchyme are already actively responding to BMP signaling. This analysis sheds light on the cellular heterogeneity of the early mouse limb bud mesenchyme and on the distinct response of LMPs and OCPs to morphogen signaling.
Collapse
Affiliation(s)
- Robert Reinhardt
- Developmental Genetics, Department of Biomedicine, University of Basel, 4058 Basel, Switzerland
| | - Fabiana Gullotta
- Developmental Genetics, Department of Biomedicine, University of Basel, 4058 Basel, Switzerland
| | - Gretel Nusspaumer
- Developmental Genetics, Department of Biomedicine, University of Basel, 4058 Basel, Switzerland.,Development and Evolution, Centro Andaluz de Biología del Desarrollo, Universidad Pablo de Olavide, 41013 Sevilla, Spain
| | - Erkan Ünal
- Developmental Genetics, Department of Biomedicine, University of Basel, 4058 Basel, Switzerland.,Swiss Institute of Bioinformatics, 4058 Basel, Switzerland.,Bioinformatics Core Facility, Department of Biomedicine, University of Basel, 4056 Basel, Switzerland
| | - Robert Ivanek
- Swiss Institute of Bioinformatics, 4058 Basel, Switzerland.,Bioinformatics Core Facility, Department of Biomedicine, University of Basel, 4056 Basel, Switzerland
| | - Aimée Zuniga
- Developmental Genetics, Department of Biomedicine, University of Basel, 4058 Basel, Switzerland
| | - Rolf Zeller
- Developmental Genetics, Department of Biomedicine, University of Basel, 4058 Basel, Switzerland
| |
Collapse
|
20
|
Lan SY, Tan MA, Yang SH, Cai JZ, Chen B, Li PW, Fan DM, Liu FB, Yu T, Chen QK. Musashi 1-positive cells derived from mouse embryonic stem cells treated with LY294002 are prone to differentiate into intestinal epithelial-like tissues. Int J Mol Med 2019; 43:2471-2480. [PMID: 30942388 DOI: 10.3892/ijmm.2019.4145] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Accepted: 03/13/2019] [Indexed: 11/06/2022] Open
Abstract
The majority of Musashi 1 (Msi1)‑positive cells derived from mouse embryonic stem cells (mESCs) are prone to differentiate into neural epithelial‑like cells, and only a small proportion of Msi1‑positive cells differentiate into intestinal epithelial‑like cells. Whether inhibiting the phosphatidylinositol 3‑kinase (PI3K) signaling of mESCs can promote the differentiation of Msi1‑positive cells into intestinal epithelial‑like cells remains to be fully elucidated. In the present study, to inhibit PI3K signaling, mESCs were treated with LY294002. A pMsi1‑green fluorescence protein reporter plasmid was used to sort the Msi1‑positive cells from mESCs treated and untreated with LY294002 (5 µmol/l). The Msi1‑positive cells were hypodermically engrafted into the backs of non‑obese diabetic/severe combined immunodeficient mice. The presence of neural and intestinal epithelial‑like cells in the grafts was detected by reverse transcription‑quantitative polymerase chain reaction analysis and immunohistochemistry. Compared with the Msi1‑positive cells derived from mESCs without LY294002 treatment, Msi1‑positive cells derived from mESCs treated with LY294002 expressed higher levels of leucine‑rich repeat‑containing G‑protein coupled receptor, a marker of intestinal epithelial stem cells, and lower levels of Nestin, a marker of neural epithelial stem cells. The grafts from Msi1‑positive cells treated with LY294002 contained more intestinal epithelial‑like tissues and fewer neural epithelial‑like tissues, compared with those from untreated Msi1‑positive cells. LY294002 had the ability to promote the differentiation of mESCs into intestinal epithelial‑like tissues. The Msi1‑positive cells selected from the cell population derived from mESCs treated with LY294002 exhibited more characteristics of intestinal epithelial stem cells than those from the untreated group.
Collapse
Affiliation(s)
- Shao-Yang Lan
- Department of Spleen and Stomach Diseases, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510405, P.R. China
| | - Mei-Ao Tan
- First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510405, P.R. China
| | - Shu-Hui Yang
- First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510405, P.R. China
| | - Jia-Zhong Cai
- Pi‑Wei Institute, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510405, P.R. China
| | - Bin Chen
- Department of Spleen and Stomach Diseases, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510405, P.R. China
| | - Pei-Wu Li
- Department of Spleen and Stomach Diseases, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510405, P.R. China
| | - Dong-Mei Fan
- Department of Spleen and Stomach Diseases, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510405, P.R. China
| | - Feng-Bin Liu
- Department of Spleen and Stomach Diseases, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510405, P.R. China
| | - Tao Yu
- Department of Gastroenterology, The Second Affiliated Hospital, Sun Yat‑Sen University, Guangzhou, Guangdong 510120, P.R. China
| | - Qi-Kui Chen
- Department of Gastroenterology, The Second Affiliated Hospital, Sun Yat‑Sen University, Guangzhou, Guangdong 510120, P.R. China
| |
Collapse
|
21
|
Wattez JS, Qiao L, Lee S, Natale DRC, Shao J. The platelet-derived growth factor receptor alpha promoter-directed expression of cre recombinase in mouse placenta. Dev Dyn 2019; 248:363-374. [PMID: 30843624 PMCID: PMC6488356 DOI: 10.1002/dvdy.21] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Revised: 02/09/2019] [Accepted: 02/22/2019] [Indexed: 12/12/2022] Open
Abstract
Background Numerous pathologies of pregnancy originate from placental dysfunction. It is essential to understand the functions of key genes in the placenta in order to discern the etiology of placental pathologies. A paucity of animal models that allow conditional and inducible expression of a target gene in the placenta is a major limitation for studying placental development and function. Results To study the platelet‐derived growth factor receptor alpha (PDGFRα)‐directed and tamoxifen‐induced Cre recombinase expression in the placenta, PDGFRα‐CreER mice were crossed with mT/mG dual‐fluorescent reporter mice. The expression of endogenous membrane‐localized enhanced green fluorescent protein (mEGFP) and/or dTomato in the placenta was examined to identify PDGFRα promoter‐directed Cre expression. Pregnant PDGFRα‐CreER;mT/mG mice were treated with tamoxifen at various gestational ages. Upon tamoxifen treatment, reporter protein mEGFP was observed in the junctional zone (JZ) and chorionic plate (CP). Furthermore, a single dose of tamoxifen was sufficient to induce the recombination. Conclusions PDGFRα‐CreER expression is restricted to the JZ and CP of mouse placentas. PDGFRα‐CreER mice provide a useful tool to conditionally knock out or overexpress a target gene in these regions of the mouse placenta. Inducible PDGFRα‐directed Cre expression trophoblasts cells. A single tamoxifen treatment is sufficient to induce the recombination. Valuable tool to temporary knockout or over‐express a target gene in the placenta. Do not require sophisticated system and suitable for ordinary laboratory setting.
Collapse
Affiliation(s)
| | - Liping Qiao
- Department of Pediatrics, University of California San Diego, La Jolla, California
| | - Samuel Lee
- Department of Pediatrics, University of California San Diego, La Jolla, California
| | | | - Jianhua Shao
- Department of Pediatrics, University of California San Diego, La Jolla, California
| |
Collapse
|
22
|
Koyano-Nakagawa N, Garry DJ. Etv2 as an essential regulator of mesodermal lineage development. Cardiovasc Res 2018; 113:1294-1306. [PMID: 28859300 DOI: 10.1093/cvr/cvx133] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Accepted: 07/24/2017] [Indexed: 11/14/2022] Open
Abstract
The 'master regulatory factors' that position at the top of the genetic hierarchy of lineage determination have been a focus of intense interest, and have been investigated in various systems. Etv2/Etsrp71/ER71 is such a factor that is both necessary and sufficient for the development of haematopoietic and endothelial lineages. As such, genetic ablation of Etv2 leads to complete loss of blood and vessels, and overexpression can convert non-endothelial cells to the endothelial lineage. Understanding such master regulatory role of a lineage is not only a fundamental quest in developmental biology, but also holds immense possibilities in regenerative medicine. To harness its activity and utility for therapeutic interventions, it is essential to understand the regulatory mechanisms, molecular function, and networks that surround Etv2. In this review, we provide a comprehensive overview of Etv2 biology focused on mouse and human systems.
Collapse
Affiliation(s)
- Naoko Koyano-Nakagawa
- Lillehei Heart Institute, Department of Medicine, University of Minnesota, 2231 6th st. SE, Minneapolis, MN 55455, USA
| | - Daniel J Garry
- Lillehei Heart Institute, Department of Medicine, University of Minnesota, 2231 6th st. SE, Minneapolis, MN 55455, USA
| |
Collapse
|
23
|
Dorraji SE, Hovd AMK, Kanapathippillai P, Bakland G, Eilertsen GØ, Figenschau SL, Fenton KA. Mesenchymal stem cells and T cells in the formation of Tertiary Lymphoid Structures in Lupus Nephritis. Sci Rep 2018; 8:7861. [PMID: 29777158 PMCID: PMC5959845 DOI: 10.1038/s41598-018-26265-z] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Accepted: 05/09/2018] [Indexed: 12/13/2022] Open
Abstract
Tertiary lymphoid structures (TLS) develop in the kidneys of lupus-prone mice and systemic lupus erythematosus (SLE) patients with lupus nephritis (LN). Here we investigated the presence of mesenchymal stem cells (MSCs) in the development of TLS in murine LN, as well as the role of human MSCs as lymphoid tissue organizer (LTo) cells on the activation of CD4+ T cells from three groups of donors including Healthy, SLE and LN patients. Mesenchymal stem like cells were detected within the pelvic wall and TLS in kidneys of lupus-prone mice. An increase in LTβ, CXCL13, CCL19, VCAM1 and ICAM1 gene expressions were detected during the development of murine LN. Human MSCs stimulated with the pro-inflammatory cytokines TNF-α and IL-1β significantly increased the expression of CCL19, VCAM1, ICAM1, TNF-α, and IL-1β. Stimulated MSCs induced proliferation of CD4+ T cells, but an inhibitory effect was observed when in co-culture with non-stimulated MSCs. A contact dependent increase in Th2 and Th17 subsets were observed for T cells from the Healthy group after co-culture with stimulated MSCs. Our data suggest that tissue-specific or/and migratory MSCs could have pivotal roles as LTo cells in accelerating early inflammatory processes and initiating the formation of kidney specific TLS in chronic inflammatory conditions.
Collapse
Affiliation(s)
- S Esmaeil Dorraji
- RNA and Molecular Pathology Research Group, Institute of Medical Biology, Faculty of Health Sciences, UiT, The Arctic University of Norway, Tromsø, Norway
| | - Aud-Malin K Hovd
- RNA and Molecular Pathology Research Group, Institute of Medical Biology, Faculty of Health Sciences, UiT, The Arctic University of Norway, Tromsø, Norway
| | - Premasany Kanapathippillai
- RNA and Molecular Pathology Research Group, Institute of Medical Biology, Faculty of Health Sciences, UiT, The Arctic University of Norway, Tromsø, Norway
| | - Gunnstein Bakland
- University Hospital of Northern Norway, Tromsø, Norway.,Molecular Inflammatory Research Group, Institute of Clinical Medicine, Faculty of Health Sciences, UiT, The Arctic University of Norway, Tromsø, Norway
| | - Gro Østli Eilertsen
- University Hospital of Northern Norway, Tromsø, Norway.,Molecular Inflammatory Research Group, Institute of Clinical Medicine, Faculty of Health Sciences, UiT, The Arctic University of Norway, Tromsø, Norway
| | - Stine L Figenschau
- RNA and Molecular Pathology Research Group, Institute of Medical Biology, Faculty of Health Sciences, UiT, The Arctic University of Norway, Tromsø, Norway
| | - Kristin A Fenton
- RNA and Molecular Pathology Research Group, Institute of Medical Biology, Faculty of Health Sciences, UiT, The Arctic University of Norway, Tromsø, Norway.
| |
Collapse
|
24
|
Miwa H, Era T. Tracing the destiny of mesenchymal stem cells from embryo to adult bone marrow and white adipose tissue via Pdgfrα expression. Development 2018; 145:145/2/dev155879. [DOI: 10.1242/dev.155879] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Accepted: 12/21/2017] [Indexed: 12/13/2022]
Abstract
ABSTRACT
Mesenchymal stem cells (MSCs) are somatic stem cells that can be derived from adult bone marrow (BM) and white adipose tissue (WAT), and that display multipotency and self-renewal capacity. Although MSCs are essential for tissue formation and have already been used in clinical therapy, the origins and markers of these cells remain unknown. In this study, we first investigated the developmental process of MSCs in mouse embryos using the gene encoding platelet-derived growth factor receptor α (Pdgfra) as a marker. We then traced cells expressing Pdgfra and other genes (brachyury, Sox1 and Pmx1) in various mutant mouse embryos until the adult stage. This tracing of MSC origins and destinies indicates that embryonic MSCs emerge in waves and that almost all adult BM MSCs and WAT MSCs originate from mesoderm and embryonic Pdgfrα-positive cells. Furthermore, we demonstrate that adult Pdgfrα-positive cells are involved in some pathological conditions.
Collapse
Affiliation(s)
- Hiroyuki Miwa
- Department of Cell Modulation, Institute of Molecular Embryology and Genetics, Kumamoto University, 2-2-1 Honjo, Chuo-ku, Kumamoto 860-0811, Japan
| | - Takumi Era
- Department of Cell Modulation, Institute of Molecular Embryology and Genetics, Kumamoto University, 2-2-1 Honjo, Chuo-ku, Kumamoto 860-0811, Japan
| |
Collapse
|
25
|
Mohamed FF, Franceschi RT. Skeletal Stem Cells: Origins, Functions and Uncertainties. CURRENT MOLECULAR BIOLOGY REPORTS 2017; 3:236-246. [PMID: 29430387 PMCID: PMC5802417 DOI: 10.1007/s40610-017-0075-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The development and maintenance of the skeleton requires a steady source of skeletal progenitors to provide the osteoblasts and chondrocytes necessary for bone and cartilage growth and development. The current model for skeletal stem cells (SSCs) posits that SSC/progenitor cells are present in bone marrow (BM) and other osteogenic sites such as cranial sutures where they undergo self-renewal and differentiation to give rise to the main skeletal tissues. SSCs hold great promise for understanding skeletal biology and genetic diseases of bone as well as for the advancement of bone tissue engineering and regenerative medicine strategies. In the past few years, a considerable effort has been devoted to identifying and purifying skeletal stem cells and determining their contribution to bone formation and homeostasis. Here, we review recent progress in this area with particular emphasis on the discovery of specific SSC markers, their use in tracking the progression of cell populations along specific lineages and the regulation of SSCs in both the appendicular and cranial skeleton.
Collapse
Affiliation(s)
- Fatma F. Mohamed
- Departments of Periodontics and Oral Medicine, University of Michigan School of Medicine, Ann Arbor, MI 48109-0600
| | - Renny T. Franceschi
- Departments of Periodontics and Oral Medicine, University of Michigan School of Medicine, Ann Arbor, MI 48109-0600
- Department of Biological Chemistry, University of Michigan School of Medicine, Ann Arbor, MI 48109-0600
| |
Collapse
|
26
|
Abstract
New sequencing technologies have made it possible to interrogate with unprecedented depth the intrinsic changes experienced by cells as they transit the arena of development. Recently in Cell, Loh, Chen, and colleagues investigated early lineage-restricted human mesoderm cell types and their precursors going back to pluripotency (Loh et al., 2016).
Collapse
Affiliation(s)
- Michael Kyba
- Department of Pediatrics and Lillehei Heart Institute, University of Minnesota, Minneapolis, MN 55455, USA.
| |
Collapse
|
27
|
Bahm I, Barriga EH, Frolov A, Theveneau E, Frankel P, Mayor R. PDGF controls contact inhibition of locomotion by regulating N-cadherin during neural crest migration. Development 2017; 144:2456-2468. [PMID: 28526750 PMCID: PMC5536867 DOI: 10.1242/dev.147926] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2016] [Accepted: 05/15/2017] [Indexed: 12/20/2022]
Abstract
A fundamental property of neural crest (NC) migration is contact inhibition of locomotion (CIL), a process by which cells change their direction of migration upon cell contact. CIL has been proven to be essential for NC migration in amphibians and zebrafish by controlling cell polarity in a cell contact-dependent manner. Cell contact during CIL requires the participation of the cell adhesion molecule N-cadherin, which starts to be expressed by NC cells as a consequence of the switch between E- and N-cadherins during epithelial-to-mesenchymal transition (EMT). However, the mechanism that controls the upregulation of N-cadherin remains unknown. Here, we show that platelet-derived growth factor receptor alpha (PDGFRα) and its ligand platelet-derived growth factor A (PDGF-A) are co-expressed in migrating cranial NC. Inhibition of PDGF-A/PDGFRα blocks NC migration by inhibiting N-cadherin and, consequently, impairing CIL. Moreover, we identify phosphatidylinositol-3-kinase (PI3K)/AKT as a downstream effector of the PDGFRα cellular response during CIL. Our results lead us to propose PDGF-A/PDGFRα signalling as a tissue-autonomous regulator of CIL by controlling N-cadherin upregulation during EMT. Finally, we show that once NC cells have undergone EMT, the same PDGF-A/PDGFRα works as an NC chemoattractant, guiding their directional migration. Summary: PDGF-A and its receptor control Xenopus neural crest migration by promoting EMT and contact inhibition of locomotion, acting via N-cadherin regulation at early stages of development and working as chemoattractant later.
Collapse
Affiliation(s)
- Isabel Bahm
- Department of Cell and Developmental Biology, University College London, London WC1E 6BT, UK
| | - Elias H Barriga
- Department of Cell and Developmental Biology, University College London, London WC1E 6BT, UK.,London Centre for Nanotechnology, University College London, London WC1H 0AH, UK
| | - Antonina Frolov
- Centre for Cardiovascular Biology and Medicine, Division of Medicine, University College London, London WC1E 6JJ, UK
| | - Eric Theveneau
- Department of Cell and Developmental Biology, University College London, London WC1E 6BT, UK
| | - Paul Frankel
- Centre for Cardiovascular Biology and Medicine, Division of Medicine, University College London, London WC1E 6JJ, UK
| | - Roberto Mayor
- Department of Cell and Developmental Biology, University College London, London WC1E 6BT, UK
| |
Collapse
|
28
|
Hepler C, Gupta RK. The expanding problem of adipose depot remodeling and postnatal adipocyte progenitor recruitment. Mol Cell Endocrinol 2017; 445:95-108. [PMID: 27743993 PMCID: PMC5346481 DOI: 10.1016/j.mce.2016.10.011] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Revised: 10/08/2016] [Accepted: 10/11/2016] [Indexed: 02/07/2023]
Abstract
The rising incidence of obesity and associated metabolic diseases has increased the urgency in understanding all aspects of adipose tissue biology. This includes the function of adipocytes, how adipose tissue expands in obesity, and how expanded adipose tissues in adults can impact physiology. Here, we highlight the growing appreciation for the importance of de novo adipocyte differentiation to adipose tissue expansion in adult humans and animals. We detail recent efforts to identify adipose precursor populations that contribute to the physiological postnatal recruitment of white, brown, and beige adipocytes in mice, and summarize new data that reveal the complexity of adipose tissue development in vivo.
Collapse
Affiliation(s)
- Chelsea Hepler
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Rana K Gupta
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| |
Collapse
|
29
|
Foxa2 identifies a cardiac progenitor population with ventricular differentiation potential. Nat Commun 2017; 8:14428. [PMID: 28195173 PMCID: PMC5316866 DOI: 10.1038/ncomms14428] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2016] [Accepted: 12/22/2016] [Indexed: 12/18/2022] Open
Abstract
The recent identification of progenitor populations that contribute to the developing heart in a distinct spatial and temporal manner has fundamentally improved our understanding of cardiac development. However, the mechanisms that direct atrial versus ventricular specification remain largely unknown. Here we report the identification of a progenitor population that gives rise primarily to cardiovascular cells of the ventricles and only to few atrial cells (<5%) of the differentiated heart. These progenitors are specified during gastrulation, when they transiently express Foxa2, a gene not previously implicated in cardiac development. Importantly, Foxa2+ cells contribute to previously identified progenitor populations in a defined pattern and ratio. Lastly, we describe an analogous Foxa2+ population during differentiation of embryonic stem cells. Together, these findings provide insight into the developmental origin of ventricular and atrial cells, and may lead to the establishment of new strategies for generating chamber-specific cell types from pluripotent stem cells.
Collapse
|
30
|
Kim BC, Jun SM, Kim SY, Kwon YD, Choe SC, Kim EC, Lee JH, Kim J, Suh JKF, Hwang YS. Engineering three dimensional micro nerve tissue using postnatal stem cells from human dental apical papilla. Biotechnol Bioeng 2016; 114:903-914. [PMID: 27775170 DOI: 10.1002/bit.26205] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Revised: 10/16/2016] [Accepted: 10/19/2016] [Indexed: 12/26/2022]
Abstract
The in vitro generation of cell-based three dimensional (3D) nerve tissue is an attractive subject to improve graft survival and integration into host tissue for neural tissue regeneration or to model biological events in stem cell differentiation. Although 3D organotypic culture strategies are well established for 3D nerve tissue formation of pluripotent stem cells to study underlying biology in nerve development, cell-based nerve tissues have not been developed using human postnatal stem cells with therapeutic potential. Here, we established a culture strategy for the generation of in vitro cell-based 3D nerve tissue from postnatal stem cells from apical papilla (SCAPs) of teeth, which originate from neural crest-derived ectomesenchyme cells. A stem cell population capable of differentiating into neural cell lineages was generated during the ex vivo expansion of SCAPs in the presence of EGF and bFGF, and SCAPs differentiated into neural cells, showing neural cell lineage-related molecular and gene expression profiles, morphological changes and electrophysical property under neural-inductive culture conditions. Moreover, we showed the first evidence that 3D cell-based nerve-like tissue with axons and myelin structures could be generated from SCAPs via 3D organotypic culture using an integrated bioprocess composed of polyethylene glycol (PEG) microwell-mediated cell spheroid formation and subsequent dynamic culture in a high aspect ratio vessel (HARV) bioreactor. In conclusion, the culture strategy in our study provides a novel approach to develop in vitro engineered nerve tissue using SCAPs and a foundation to study biological events in the neural differentiation of postnatal stem cells. Biotechnol. Bioeng. 2017;114: 903-914. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Byung-Chul Kim
- Department of Maxillofacial Biomedical Engineering and Institute of Oral Biology, School of Dentistry, Kyung Hee University, 1 Hoegi-dong, Dongdaemun-gu, Seoul 130-701, Seoul, Korea
| | - Sung-Min Jun
- Department of Maxillofacial Biomedical Engineering and Institute of Oral Biology, School of Dentistry, Kyung Hee University, 1 Hoegi-dong, Dongdaemun-gu, Seoul 130-701, Seoul, Korea.,The Center for Bionics, Korea Institute of Science and Technology, Hwarang-ro 14-gil 5, Seongbuk-gu, Seoul 136-791, Seoul, Korea
| | - So Yeon Kim
- Department of Maxillofacial Biomedical Engineering and Institute of Oral Biology, School of Dentistry, Kyung Hee University, 1 Hoegi-dong, Dongdaemun-gu, Seoul 130-701, Seoul, Korea
| | - Yong-Dae Kwon
- Department of Oral Maxillofacial Surgery, Kyung Hee University, Seoul, Korea
| | - Sung Chul Choe
- Department of Pediatric Dentistry, Kyung Hee University, Seoul, Korea
| | - Eun-Chul Kim
- Department of Oral and Maxillofacial Pathology, School of Dentistry, Kyung Hee University, Seoul, Korea
| | - Jae-Hyung Lee
- Department of Maxillofacial Biomedical Engineering and Institute of Oral Biology, School of Dentistry, Kyung Hee University, 1 Hoegi-dong, Dongdaemun-gu, Seoul 130-701, Seoul, Korea.,Department of Life and Nanopharmaceutical Sciences, Kyung Hee University, Seoul, Korea
| | - Jinseok Kim
- The Center for Bionics, Korea Institute of Science and Technology, Hwarang-ro 14-gil 5, Seongbuk-gu, Seoul 136-791, Seoul, Korea
| | - Jun-Kyo Francis Suh
- The Center for Bionics, Korea Institute of Science and Technology, Hwarang-ro 14-gil 5, Seongbuk-gu, Seoul 136-791, Seoul, Korea
| | - Yu-Shik Hwang
- Department of Maxillofacial Biomedical Engineering and Institute of Oral Biology, School of Dentistry, Kyung Hee University, 1 Hoegi-dong, Dongdaemun-gu, Seoul 130-701, Seoul, Korea
| |
Collapse
|
31
|
Chemotaxis during neural crest migration. Semin Cell Dev Biol 2016; 55:111-8. [DOI: 10.1016/j.semcdb.2016.01.031] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Accepted: 01/22/2016] [Indexed: 01/12/2023]
|
32
|
Siavashi V, Sariri R, Nassiri SM, Esmaeilivand M, Asadian S, Cheraghi H, Barekati-Mowahed M, Rahbarghazi R. Angiogenic activity of endothelial progenitor cells through angiopoietin-1 and angiopoietin-2. Anim Cells Syst (Seoul) 2016. [DOI: 10.1080/19768354.2016.1189961] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
|
33
|
Küspert M, Wegner M. SomethiNG 2 talk about-Transcriptional regulation in embryonic and adult oligodendrocyte precursors. Brain Res 2015; 1638:167-182. [PMID: 26232072 DOI: 10.1016/j.brainres.2015.07.024] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Revised: 07/14/2015] [Accepted: 07/18/2015] [Indexed: 12/26/2022]
Abstract
Glial cells that express the chondroitin sulfate proteoglycan NG2 represent an inherently heterogeneous population. These so-called NG2-glia are present during development and in the adult CNS, where they are referred to as embryonic oligodendrocyte precursors and adult NG2-glia, respectively. They give rise to myelinating oligodendrocytes at all times of life. Over the years much has been learnt about the transcriptional network in embryonic oligodendrocyte precursors, and several transcription factors from the HLH, HMG-domain, zinc finger and homeodomain protein families have been identified as main constituents. Much less is known about the corresponding network in adult NG2-glia. Here we summarize and discuss current knowledge on functions of each of these transcription factor families in NG2-glia, and where possible compare transcriptional regulation in embryonic oligodendrocyte precursors and adult NG2-glia. This article is part of a Special Issue entitled SI:NG2-glia (Invited only).
Collapse
Affiliation(s)
- Melanie Küspert
- Institut für Biochemie, Emil-Fischer-Zentrum, Friedrich-Alexander-Universität Erlangen-Nürnberg, Fahrstrasse 17, Erlangen D-91054, Germany.
| | - Michael Wegner
- Institut für Biochemie, Emil-Fischer-Zentrum, Friedrich-Alexander-Universität Erlangen-Nürnberg, Fahrstrasse 17, Erlangen D-91054, Germany.
| |
Collapse
|
34
|
Platelet-Derived Growth Factor Receptor Alpha as a Marker of Mesenchymal Stem Cells in Development and Stem Cell Biology. Stem Cells Int 2015; 2015:362753. [PMID: 26257789 PMCID: PMC4519552 DOI: 10.1155/2015/362753] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2015] [Revised: 05/24/2015] [Accepted: 06/17/2015] [Indexed: 11/17/2022] Open
Abstract
Three decades on, the mesenchymal stem cells (MSCs) have been intensively researched on the bench top and used clinically. However, ambiguity still exists in regard to their anatomical locations, identities, functions, and extent of their differentiative abilities. One of the major impediments in the quest of the MSC research has been lack of appropriate in vivo markers. In recent years, this obstacle has been resolved to some degree as PDGFRα emerges as an important mesenchymal stem cell marker. Accumulating lines of evidence are showing that the PDGFRα (+) cells reside in the perivascular locations of many adult interstitium and fulfil the classic concepts of MSCs in vitro and in vivo. PDGFRα has long been recognised for its roles in the mesoderm formation and connective tissue development during the embryogenesis. Current review describes the lines of evidence regarding the role of PDGFRα in morphogenesis and differentiation and its implications for MSC biology.
Collapse
|
35
|
Miwa H, Era T. Generation and characterization of PDGFRα-GFPCreERT2 knock-In mouse line. Genesis 2015; 53:329-36. [PMID: 25884589 DOI: 10.1002/dvg.22853] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2015] [Revised: 04/12/2015] [Accepted: 04/13/2015] [Indexed: 12/18/2022]
Abstract
Platelet-derived growth factor (PDGF) and its receptor play an important role in embryogenesis. PDGF receptor α (PDGFRα) is expressed specifically in the embryonic day 7.5 (E7.5) mesoderm and in the E9.5 neural crest among other tissues. PDGFRα-expressing cells and their descendants are involved in the formation of various tissues. To trace PDGFRα-expressing cells in vivo, we generated a knock-in mouse line that expressed a fusion protein of green fluorescent protein (GFP), Cre recombinase (Cre), and mutated estrogen receptor ligand-binding domain (ERT2) under the control of the PDGFRα promoter. In these mice, Cre activity in PDGFRα-expressing cells could be induced by tamoxifen treatment. Taken together, our results suggest that the knock-in mouse line generated here could be useful for studying PDGFRα-expressing cells and their descendants in vivo at various stages of development.
Collapse
Affiliation(s)
- Hiroyuki Miwa
- Department of Cell Modulation, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto, Japan
| | - Takumi Era
- Department of Cell Modulation, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto, Japan
| |
Collapse
|
36
|
MiR-24 is required for hematopoietic differentiation of mouse embryonic stem cells. PLoS Genet 2015; 11:e1004959. [PMID: 25634354 PMCID: PMC4310609 DOI: 10.1371/journal.pgen.1004959] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2014] [Accepted: 12/16/2014] [Indexed: 11/19/2022] Open
Abstract
Overexpression of miRNA, miR-24, in mouse hematopoietic progenitors increases monocytic/ granulocytic differentiation and inhibits B cell development. To determine if endogenous miR-24 is required for hematopoiesis, we antagonized miR-24 in mouse embryonic stem cells (ESCs) and performed in vitro differentiations. Suppression of miR-24 resulted in an inability to produce blood and hematopoietic progenitors (HPCs) from ESCs. The phenotype is not a general defect in mesoderm production since we observe production of nascent mesoderm as well as mesoderm derived cardiac muscle and endothelial cells. Results from blast colony forming cell (BL-CFC) assays demonstrate that miR-24 is not required for generation of the hemangioblast, the mesoderm progenitor that gives rise to blood and endothelial cells. However, expression of the transcription factors Runx1 and Scl is greatly reduced, suggesting an impaired ability of the hemangioblast to differentiate. Lastly, we observed that known miR-24 target, Trib3, is upregulated in the miR-24 antagonized embryoid bodies (EBs). Overexpression of Trib3 alone in ESCs was able to decrease HPC production, though not as great as seen with miR-24 knockdown. These results demonstrate an essential role for miR-24 in the hematopoietic differentiation of ESCs. Although many miRNAs have been implicated in regulation of hematopoiesis, this is the first miRNA observed to be required for the specification of mammalian blood progenitors from early mesoderm. Studies of mouse embryos and embryonic stem cells (ESCs) have defined the ontogeny of mammalian embryonic hematopoietic cells. The ESC differentiation system has been valuable for dissecting the molecular regulation of the development of mesoderm into HPCs. Extracellular signals regulate a complex network of transcription factors to direct embryonic hematopoietic development. Mammalian miRNAs have previously not been described to regulate this genetic network during embryonic hematopoiesis. However, a role for miRNAs in producing the hemangioblast, and hemogenic endothelium in Xenopus has been described. Our work with ESCs demonstrates a specific requirement for the miRNA, miR-24, in the development of hematopoietic progenitors cells (HPCs). Antagonizing miR-24 in ESCs does not affect generation of BL-CFCs, the in vitro equivalent of the hemangioblast, but does compromise the ability of those BL-CFCs to produced HPCs. Expression of transcription factors required for HPC production downstream of the hemangioblast, Scl, and Runx1, is greatly reduced by antagonizing miR-24. These results identify miR-24, as a mammalian miRNA required for the development of blood from newly formed mesoderm.
Collapse
|
37
|
Schrimpf C, Teebken OE, Wilhelmi M, Duffield JS. The role of pericyte detachment in vascular rarefaction. J Vasc Res 2014; 51:247-58. [PMID: 25195856 DOI: 10.1159/000365149] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2013] [Accepted: 06/07/2014] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Pericytes surround endothelial cells at the perivascular interface. Signaling between endothelial cells and pericytes is crucial for capillary homeostasis, as pericytes stabilize vessels and regulate many microvascular functions. Recently it has been shown that pericytes are able to detach from the vascular wall and contribute to fibrosis by becoming scar-forming myofibroblasts in many organs including the kidney. At the same time, the loss of pericytes within the perivascular compartment results in vulnerable capillaries which are prone to instability, pathological angiogenesis, and, ultimately, rarefaction. AIMS This review will give an overview of pericyte-endothelial cell interactions, summarize the signaling pathways that have been identified to be involved in pericyte detachment from the vascular wall, and present pathological endothelial responses in the context of disease of the kidney.
Collapse
Affiliation(s)
- Claudia Schrimpf
- Division of Vascular and Endovascular Surgery, Department of Cardiothoracic, Transplantation and Vascular Surgery, Hannover Medical School, Hannover, Germany
| | | | | | | |
Collapse
|
38
|
Sahara M, Hansson EM, Wernet O, Lui KO, Später D, Chien KR. Manipulation of a VEGF-Notch signaling circuit drives formation of functional vascular endothelial progenitors from human pluripotent stem cells. Cell Res 2014; 24:820-41. [PMID: 24810299 PMCID: PMC4085760 DOI: 10.1038/cr.2014.59] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2013] [Revised: 03/10/2014] [Accepted: 03/31/2014] [Indexed: 12/13/2022] Open
Abstract
Human pluripotent stem cell (hPSC)-derived endothelial lineage cells constitutes a promising source for therapeutic revascularization, but progress in this arena has been hampered by a lack of clinically-scalable differentiation protocols and inefficient formation of a functional vessel network integrating with the host circulation upon transplantation. Using a human embryonic stem cell reporter cell line, where green fluorescent protein expression is driven by an endothelial cell-specific VE-cadherin (VEC) promoter, we screened for > 60 bioactive small molecules that would promote endothelial differentiation, and found that administration of BMP4 and a GSK-3β inhibitor in an early phase and treatment with VEGF-A and inhibition of the Notch signaling pathway in a later phase led to efficient differentiation of hPSCs to the endothelial lineage within six days. This sequential approach generated > 50% conversion of hPSCs to endothelial cells (ECs), specifically VEC+CD31+CD34+CD14−KDRhigh endothelial progenitors (EPs) that exhibited higher angiogenic and clonogenic proliferation potential among endothelial lineage cells. Pharmaceutical inhibition or genetical knockdown of Notch signaling, in combination with VEGF-A treatment, resulted in efficient formation of EPs via KDR+ mesodermal precursors and blockade of the conversion of EPs to mature ECs. The generated EPs successfully formed functional capillary vessels in vivo with anastomosis to the host vessels when transplanted into immunocompromised mice. Manipulation of this VEGF-A-Notch signaling circuit in our protocol leads to rapid large-scale production of the hPSC-derived EPs by 12- to 20-fold vs current methods, which may serve as an attractive cell population for regenerative vascularization with superior vessel forming capability compared to mature ECs.
Collapse
Affiliation(s)
- Makoto Sahara
- 1] Department of Stem Cell and Regenerative Biology, 7 Divinity Avenue, Cambridge, MA 02138, USA [2] Harvard Stem Cell Institute, Harvard University, 7 Divinity Avenue, Cambridge, MA 02138, USA [3] Center for Regenerative Medicine, Massachusetts General Hospital, 185 Cambridge Street, Boston, MA 021141, USA [4] Department of Medicine-Cardiology/Cell and Molecular Biology, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| | - Emil M Hansson
- 1] Department of Stem Cell and Regenerative Biology, 7 Divinity Avenue, Cambridge, MA 02138, USA [2] Harvard Stem Cell Institute, Harvard University, 7 Divinity Avenue, Cambridge, MA 02138, USA
| | - Oliver Wernet
- Department of Anesthesiology and Intensive Care Medicine, Charité-University Medicine Berlin, Campus Charité Mitte, Charitéplatz 1, 10117 Berlin, Germany
| | - Kathy O Lui
- 1] Department of Stem Cell and Regenerative Biology, 7 Divinity Avenue, Cambridge, MA 02138, USA [2] Harvard Stem Cell Institute, Harvard University, 7 Divinity Avenue, Cambridge, MA 02138, USA [3] Center for Regenerative Medicine, Massachusetts General Hospital, 185 Cambridge Street, Boston, MA 021141, USA
| | - Daniela Später
- 1] Department of Stem Cell and Regenerative Biology, 7 Divinity Avenue, Cambridge, MA 02138, USA [2] Harvard Stem Cell Institute, Harvard University, 7 Divinity Avenue, Cambridge, MA 02138, USA [3] Center for Regenerative Medicine, Massachusetts General Hospital, 185 Cambridge Street, Boston, MA 021141, USA
| | - Kenneth R Chien
- 1] Department of Stem Cell and Regenerative Biology, 7 Divinity Avenue, Cambridge, MA 02138, USA [2] Harvard Stem Cell Institute, Harvard University, 7 Divinity Avenue, Cambridge, MA 02138, USA [3] Department of Medicine-Cardiology/Cell and Molecular Biology, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| |
Collapse
|
39
|
Wang QA, Scherer PE, Gupta RK. Improved methodologies for the study of adipose biology: insights gained and opportunities ahead. J Lipid Res 2014; 55:605-24. [PMID: 24532650 PMCID: PMC3966696 DOI: 10.1194/jlr.r046441] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2013] [Revised: 02/10/2014] [Indexed: 12/14/2022] Open
Abstract
Adipocyte differentiation and function have become areas of intense focus in the field of energy metabolism; however, understanding the role of specific genes in the establishment and maintenance of fat cell function can be challenging and complex. In this review, we offer practical guidelines for the study of adipocyte development and function. We discuss improved cellular and genetic systems for the study of adipose biology and highlight recent insights gained from these new approaches.
Collapse
Affiliation(s)
- Qiong A. Wang
- Department of Internal Medicine, Touchstone Diabetes Center, and University of Texas Southwestern Medical Center, Dallas, TX 75287
| | - Philipp E. Scherer
- Department of Internal Medicine, Touchstone Diabetes Center, and University of Texas Southwestern Medical Center, Dallas, TX 75287
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX 75287
| | - Rana K. Gupta
- Department of Internal Medicine, Touchstone Diabetes Center, and University of Texas Southwestern Medical Center, Dallas, TX 75287
| |
Collapse
|
40
|
Inoue-Yokoo T, Tani K, Sugiyama D. Mesodermal and hematopoietic differentiation from ES and iPS cells. Stem Cell Rev Rep 2014; 9:422-34. [PMID: 22684542 DOI: 10.1007/s12015-012-9388-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Embryonic stem (ES) and induced pluripotent stem (iPS) cells can differentiate into any type of tissue when grown in a suitable culture environment and are considered valuable tools for regenerative medicine. In the field of hematology, generation of hematopoietic stem cells (HSCs) and mature hematopoietic cells (HCs) from ES and iPS cells through mesodermal cells, the ancestors of HCs, can facilitate transplantation and transfusion therapy. Several studies report generation of functional HCs from both mouse and human ES and iPS cells. This approach will likely be applied to individual patient-derived iPS cells for regenerative medicine approaches and drug screening in the future. Here, we summarize current studies of HC-generation from ES and iPS cells.
Collapse
Affiliation(s)
- Tomoko Inoue-Yokoo
- Division of Hematopoietic Stem Cells, Advanced Medical Initiatives, Department of Advanced Medical Initiatives, Kyushu University Faculty of Medical Sciences, Fukuoka 812-8582, Japan
| | | | | |
Collapse
|
41
|
The role of the non-canonical Wnt-planar cell polarity pathway in neural crest migration. Biochem J 2014; 457:19-26. [PMID: 24325550 DOI: 10.1042/bj20131182] [Citation(s) in RCA: 70] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
The neural crest is an embryonic stem cell population whose migratory behaviour has been likened to malignant invasion. The neural crest, as does cancer, undergoes an epithelial-to-mesenchymal transition and migrates to colonize almost all the tissues of the embryo. Neural crest cells exhibit collective cell migration, moving in streams of high directionality. The migratory neural crest streams are kept in shape by the presence of negative signals in their vicinity. The directionality of the migrating neural crest is achieved by contact-dependent cell polarization, in a phenomenon called contact inhibition of locomotion. Two cells experiencing contact inhibition of locomotion move away from each other after collision. However, if the cell density is high only cells exposed to a free edge can migrate away from the cluster leading to the directional migration of the whole group. Recent work performed in chicks, zebrafish and frogs has shown that the non-canonical Wnt-PCP (planar cell polarity) pathway plays a major role in neural crest migration. PCP signalling controls contact inhibition of locomotion between neural crest cells by localizing different PCP proteins at the site of cell contact during collision and locally regulating the activity of Rho GTPases. Upon collision RhoA (ras homologue family member A) is activated, whereas Rac1 is inhibited at the contact between two migrating neural crest cells, leading to the collapse of protrusions and the migration of cells away from one another. The present review summarizes the mechanisms that control neural crest migration and focuses on the role of non-canonical Wnt or PCP signalling in this process.
Collapse
|
42
|
Lajiness JD, Conway SJ. Origin, development, and differentiation of cardiac fibroblasts. J Mol Cell Cardiol 2013; 70:2-8. [PMID: 24231799 DOI: 10.1016/j.yjmcc.2013.11.003] [Citation(s) in RCA: 96] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2013] [Revised: 10/23/2013] [Accepted: 11/04/2013] [Indexed: 01/14/2023]
Abstract
Cardiac fibroblasts are the most abundant cell in the mammalian heart. While they have been historically underappreciated in terms of their functional contributions to cardiac development and physiology, they and their activated form, myofibroblasts, are now known to play key roles in both development and disease through structural, paracrine, and electrical interactions with cardiomyocytes. The lack of specific markers for fibroblasts currently convolutes the study of this dynamic cell lineage, but advances in marker analysis and lineage mapping technologies are continuously being made. Understanding how to best utilize these tools, both individually and in combination, will help to elucidate the functional significance of fibroblast-cardiomyocyte interactions in vivo. Here we review what is currently known about the diverse roles played by cardiac fibroblasts and myofibroblasts throughout development and periods of injury with the intent of emphasizing the duality of their nature. This article is part of a Special Issue entitled "Myocyte-Fibroblast Signalling in Myocardium ".
Collapse
Affiliation(s)
- Jacquelyn D Lajiness
- Developmental Biology and Neonatal Medicine Program, HB Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Simon J Conway
- Developmental Biology and Neonatal Medicine Program, HB Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| |
Collapse
|
43
|
Babic AM, Jang S, Nicolov E, Voicu H, Luckey CJ. Culture of mouse amniotic fluid-derived cells on irradiated STO feeders results in the generation of primitive endoderm cell lines capable of self-renewal in vitro. Cells Tissues Organs 2013; 198:111-26. [PMID: 24060676 DOI: 10.1159/000353942] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/24/2013] [Indexed: 12/14/2022] Open
Abstract
The cells present in amniotic fluid (AF) are currently used for prenatal diagnosis of fetal anomalies but are also a potential source of cells for cell therapy. To better characterize putative progenitor cell populations present in AF, we used culture conditions that support self-renewal to determine if these promoted the generation of stable cell lines from AF-derived cells (AFC). Cells isolated from E11.5 mouse were cultured on irradiated STO fibroblast feeder layers in human embryonic germ cell derivation conditions. The cultures grew multicellular epithelial colonies that could be repropagated from single cells. Reverse transcription semiquantitative polymerase chain reaction of established cell lines revealed that they belonged to the extraembryonic endoderm (ExEn) expressing high levels of Gata6, Gata4, Sox17, Foxa2 and Sox7 mRNA. Hierarchical clustering based on the whole transcriptome expression profile of the AFC lines (AFCL) shows significant correlation between transcription profiles of AFCL and blastocyst-derived XEN, an ExEn cell line. In vitro differentiation of AFCL results in the generation of cells expressing albumin and α-fetoprotein (AFP), while intramuscular injection of AFCL into immunodeficient mice produced AFP-positive tumors with primitive endodermal appearance. Hence, E11.5 mouse AF contains cells that efficiently produce XEN lines. These AF-derived XEN lines do not spontaneously differentiate into embryonic-type cells but are phenotypically stable and have the capacity for extensive expansion. The lack of requirement for reprogramming factors to turn AF-derived progenitor cells into stable cell lines capable of massive expansion together with the known ability of ExEn to contribute to embryonic tissue suggests that this cell type may be a candidate for banking for cell therapies.
Collapse
Affiliation(s)
- Aleksandar M Babic
- Department of Pathology and Genomic Medicine, The Methodist Hospital, Houston, Tex., USA
| | | | | | | | | |
Collapse
|
44
|
Craft AM, Ahmed N, Rockel JS, Baht GS, Alman BA, Kandel RA, Grigoriadis AE, Keller GM. Specification of chondrocytes and cartilage tissues from embryonic stem cells. Development 2013; 140:2597-610. [PMID: 23715552 DOI: 10.1242/dev.087890] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Osteoarthritis primarily affects the articular cartilage of synovial joints. Cell and/or cartilage replacement is a promising therapy, provided there is access to appropriate tissue and sufficient numbers of articular chondrocytes. Embryonic stem cells (ESCs) represent a potentially unlimited source of chondrocytes and tissues as they can generate a broad spectrum of cell types under appropriate conditions in vitro. Here, we demonstrate that mouse ESC-derived chondrogenic mesoderm arises from a Flk-1(-)/Pdgfrα(+) (F(-)P(+)) population that emerges in a defined temporal pattern following the development of an early cardiogenic F(-)P(+) population. Specification of the late-arising F(-)P(+) population with BMP4 generated a highly enriched population of chondrocytes expressing genes associated with growth plate hypertrophic chondrocytes. By contrast, specification with Gdf5, together with inhibition of hedgehog and BMP signaling pathways, generated a population of non-hypertrophic chondrocytes that displayed properties of articular chondrocytes. The two chondrocyte populations retained their hypertrophic and non-hypertrophic properties when induced to generate spatially organized proteoglycan-rich cartilage-like tissue in vitro. Transplantation of either type of chondrocyte, or tissue generated from them, into immunodeficient recipients resulted in the development of cartilage tissue and bone within an 8-week period. Significant ossification was not observed when the tissue was transplanted into osteoblast-depleted mice or into diffusion chambers that prevent vascularization. Thus, through stage-specific manipulation of appropriate signaling pathways it is possible to efficiently and reproducibly derive hypertrophic and non-hypertrophic chondrocyte populations from mouse ESCs that are able to generate distinct cartilage-like tissue in vitro and maintain a cartilage tissue phenotype within an avascular and/or osteoblast-free niche in vivo.
Collapse
Affiliation(s)
- April M Craft
- McEwen Centre for Regenerative Medicine, University Health Network, Toronto, ON, M5G 1L7, Canada
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Ishimine H, Yamakawa N, Sasao M, Tadokoro M, Kami D, Komazaki S, Tokuhara M, Takada H, Ito Y, Kuno S, Yoshimura K, Umezawa A, Ohgushi H, Asashima M, Kurisaki A. N-Cadherin is a prospective cell surface marker of human mesenchymal stem cells that have high ability for cardiomyocyte differentiation. Biochem Biophys Res Commun 2013; 438:753-9. [PMID: 23899519 DOI: 10.1016/j.bbrc.2013.07.081] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2013] [Accepted: 07/19/2013] [Indexed: 11/28/2022]
Abstract
Mesenchymal stem cells (MSCs) are among the most promising sources of stem cells for regenerative medicine. However, the range of their differentiation ability is very limited. In this study, we explored prospective cell surface markers of human MSCs that readily differentiate into cardiomyocytes. When the cardiomyogenic differentiation potential and the expression of cell surface markers involved in heart development were analyzed using various immortalized human MSC lines, the MSCs with high expression of N-cadherin showed a higher probability of differentiation into beating cardiomyocytes. The differentiated cardiomyocytes expressed terminally differentiated cardiomyocyte-specific markers such as α-actinin, cardiac troponin T, and connexin-43. A similar correlation was observed with primary human MSCs derived from bone marrow and adipose tissue. Moreover, N-cadherin-positive MSCs isolated with N-cadherin antibody-conjugated magnetic beads showed an apparently higher ability to differentiate into cardiomyocytes than the N-cadherin-negative population. Quantitative polymerase chain reaction analyses demonstrated that the N-cadherin-positive population expressed significantly elevated levels of cardiomyogenic progenitor-specific transcription factors, including Nkx2.5, Hand1, and GATA4 mRNAs. Our results suggest that N-cadherin is a novel prospective cell surface marker of human MSCs that show a better ability for cardiomyocyte differentiation.
Collapse
Affiliation(s)
- Hisako Ishimine
- Research Center for Stem Cell Engineering, National Institute of Advanced Industrial Science and Technology, Tsukuba, Ibaraki, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Dandapat A, Hartweck LM, Bosnakovski D, Kyba M. Expression of the human FSHD-linked DUX4 gene induces neurogenesis during differentiation of murine embryonic stem cells. Stem Cells Dev 2013; 22:2440-8. [PMID: 23560660 DOI: 10.1089/scd.2012.0643] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Misexpression of the double homeodomain protein DUX4 in muscle is believed to cause facioscapulohumeral muscular dystrophy (FSHD). Although strategies are being devised to inhibit DUX4 activity in FSHD, there is little known about the normal function of this protein. Expression of DUX4 has been reported in pluripotent cells and testis. To test the idea that DUX4 may be involved in initiating a germ lineage program in pluripotent cells, we interrogated the effect of expressing the human DUX4 gene at different stages during in vitro differentiation of murine embryonic stem (ES) cells. We find that expression of even low levels of DUX4 is incompatible with pluripotency: DUX4-expressing ES cells downregulate pluripotency markers and rapidly differentiate even in the presence of leukemia inhibitory factor (LIF) and bone morphogenetic protein 4 (BMP4). Transcriptional profiling revealed unexpectedly that DUX4 induced a neurectodermal program. Embryoid bodies exposed to a pulse of DUX4 expression displayed severely inhibited mesodermal differentiation, but acquired neurogenic potential. In a serum-containing medium in which neurogenic differentiation is minimal, DUX4 expression served as a neural-inducing factor, enabling the differentiation of Tuj1+ neurites. These data suggest that besides effects in muscle and germ cells, the involvement of DUX4 in neurogenesis should be considered as anti-DUX4 therapies are developed.
Collapse
Affiliation(s)
- Abhijit Dandapat
- Department of Pediatrics, Lillehei Heart Institute, University of Minnesota, Minneapolis, Minnesota 55455, USA
| | | | | | | |
Collapse
|
47
|
Ding G, Tanaka Y, Hayashi M, Nishikawa SI, Kataoka H. PDGF receptor alpha+ mesoderm contributes to endothelial and hematopoietic cells in mice. Dev Dyn 2013; 242:254-68. [PMID: 23335233 PMCID: PMC3597973 DOI: 10.1002/dvdy.23923] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2012] [Revised: 12/12/2012] [Accepted: 12/20/2012] [Indexed: 11/08/2022] Open
Abstract
BACKGROUND Early mesoderm can be classified into Flk-1+ or PDGF receptor alpha (PDGFRα)+ population, grossly representing lateral and paraxial mesoderm, respectively. It has been demonstrated that all endothelial (EC) and hematopoietic (HPC) cells are derived from Flk-1+ cells. Although PDGFRα+ cells give rise to ECs/HPCs in in vitro ES differentiation, whether PDGFRα+ population can become hemato-endothelial lineages has not been proved in mouse embryos. RESULTS Using PDGFRαMerCreMer mice, PDGFRα+ early mesoderm was shown to contribute to endothelial cells including hemogenic ECs, fetal liver B lymphocytes, and Lin-Kit+Sca-1+ (KSL) cells. Contribution of PDGFRα+ mesoderm into ECs and HPCs was limited until E8.5, indicating that PDGFRα+/Flk-1+ population that exists until E8.5 may be the source for hemato-endothelial lineages from PDGFRα+ population. The functional significance of PDGFRα+ mesoderm in vascular development and hematopoiesis was confirmed by genetic deletion of Etv2 or restoration of Runx1 in PDGFRα+ cells. Etv2 deletion and Runx1 restoration in PDGFRα+ cells resulted in abnormal vascular remodeling and rescue of fetal liver CD45+ and Lin-Kit+Sca-1+ (KSL) cells, respectively. CONCLUSIONS Endothelial and hematopoietic cells can be derived from PDGFRα+ early mesoderm in mice. PDGFRα+ mesoderm is functionally significant in vascular development and hematopoiesis from phenotype analysis of genetically modified embryos.
Collapse
Affiliation(s)
- Guo Ding
- Laboratory for Stem Cell Biology, RIKEN Center for Developmental Biology, Kobe, Japan
| | | | | | | | | |
Collapse
|
48
|
Nakajima Y, Imanaka-Yoshida K. New insights into the developmental mechanisms of coronary vessels and epicardium. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2013; 303:263-317. [PMID: 23445813 DOI: 10.1016/b978-0-12-407697-6.00007-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
During heart development, the epicardium, which originates from the proepicardial organ (PE), is a source of coronary vessels. The PE develops from the posterior visceral mesoderm of the pericardial coelom after stimulation with a combination of weak bone morphogenetic protein and strong fibroblast growth factor (FGF) signaling. PE-derived cells migrate across the heart surface to form the epicardial sheet, which subsequently seeds multipotent subepicardial mesenchymal cells via epithelial-mesenchymal transition, which is regulated by several signaling pathways including retinoic acid, FGF, sonic hedgehog, Wnt, transforming growth factor-β, and platelet-derived growth factor. Subepicardial endothelial progenitors eventually generate the coronary vascular plexus, which acquires an arterial or venous phenotype, connects with the sinus venosus and aortic sinuses, and then matures through the recruitment of vascular smooth muscle cells under the regulation of complex growth factor signaling pathways. These developmental programs might be activated in the adult heart after injury and play a role in the regeneration/repair of the myocardium.
Collapse
Affiliation(s)
- Yuji Nakajima
- Department of Anatomy and Cell Biology, Graduate School of Medicine, Osaka City University, Osaka, Japan.
| | | |
Collapse
|
49
|
Salani S, Donadoni C, Rizzo F, Bresolin N, Comi GP, Corti S. Generation of skeletal muscle cells from embryonic and induced pluripotent stem cells as an in vitro model and for therapy of muscular dystrophies. J Cell Mol Med 2012; 16:1353-64. [PMID: 22129481 PMCID: PMC3823206 DOI: 10.1111/j.1582-4934.2011.01498.x] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Muscular dystrophies (MDs) are a heterogeneous group of inherited disorders characterized by progressive muscle wasting and weakness likely associated with exhaustion of muscle regeneration potential. At present, no cures or efficacious treatments are available for these diseases, but cell transplantation could be a potential therapeutic strategy. Transplantation of myoblasts using satellite cells or other myogenic cell populations has been attempted to promote muscle regeneration, based on the hypothesis that the donor cells repopulate the muscle and contribute to its regeneration. Embryonic stem cells (ESCs) and more recently induced pluripotent stem cells (iPSCs) could generate an unlimited source of differentiated cell types, including myogenic cells. Here we review the literature regarding the generation of myogenic cells considering the main techniques employed to date to elicit efficient differentiation of human and murine ESCs or iPSCs into skeletal muscle. We also critically analyse the possibility of using these cellular populations as an alternative source of myogenic cells for cell therapy of MDs.
Collapse
Affiliation(s)
- Sabrina Salani
- Department of Neurological Sciences, Dino Ferrari Centre, University of Milan, IRCCS Fondazione Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy.
| | | | | | | | | | | |
Collapse
|
50
|
Komada Y, Yamane T, Kadota D, Isono K, Takakura N, Hayashi SI, Yamazaki H. Origins and properties of dental, thymic, and bone marrow mesenchymal cells and their stem cells. PLoS One 2012. [PMID: 23185234 PMCID: PMC3504117 DOI: 10.1371/journal.pone.0046436] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Mesenchymal cells arise from the neural crest (NC) or mesoderm. However, it is difficult to distinguish NC-derived cells from mesoderm-derived cells. Using double-transgenic mouse systems encoding P0-Cre, Wnt1-Cre, Mesp1-Cre, and Rosa26EYFP, which enabled us to trace NC-derived or mesoderm-derived cells as YFP-expressing cells, we demonstrated for the first time that both NC-derived (P0- or Wnt1-labeled) and mesoderm-derived (Mesp1-labeled) cells contribute to the development of dental, thymic, and bone marrow (BM) mesenchyme from the fetal stage to the adult stage. Irrespective of the tissues involved, NC-derived and mesoderm-derived cells contributed mainly to perivascular cells and endothelial cells, respectively. Dental and thymic mesenchyme were composed of either NC-derived or mesoderm-derived cells, whereas half of the BM mesenchyme was composed of cells that were not derived from the NC or mesoderm. However, a colony-forming unit-fibroblast (CFU-F) assay indicated that CFU-Fs in the dental pulp, thymus, and BM were composed of NC-derived and mesoderm-derived cells. Secondary CFU-F assays were used to estimate the self-renewal potential, which showed that CFU-Fs in the teeth, thymus, and BM were entirely NC-derived cells, entirely mesoderm-derived cells, and mostly NC-derived cells, respectively. Colony formation was inhibited drastically by the addition of anti-platelet-derived growth factor receptor-β antibody, regardless of the tissue and its origin. Furthermore, dental mesenchyme expressed genes encoding critical hematopoietic factors, such as interleukin-7, stem cell factor, and cysteine-X-cysteine (CXC) chemokine ligand 12, which supports the differentiation of B lymphocytes and osteoclasts. Therefore, the mesenchymal stem cells found in these tissues had different origins, but similar properties in each organ.
Collapse
Affiliation(s)
- Yukiya Komada
- Department of Stem Cell and Developmental Biology, Mie University Graduate School of Medicine, Tsu, Japan
| | - Toshiyuki Yamane
- Department of Stem Cell and Developmental Biology, Mie University Graduate School of Medicine, Tsu, Japan
| | - Daiji Kadota
- Department of Stem Cell and Developmental Biology, Mie University Graduate School of Medicine, Tsu, Japan
| | - Kana Isono
- Department of Stem Cell and Developmental Biology, Mie University Graduate School of Medicine, Tsu, Japan
| | - Nobuyuki Takakura
- Department of Signal Transduction, Research Institute for Microbial Disease, Osaka University, Suita, Japan
| | - Shin-Ichi Hayashi
- Division of Immunology, School of Life Science, Faculty of Medicine, Tottori University, Yonago, Japan
| | - Hidetoshi Yamazaki
- Department of Stem Cell and Developmental Biology, Mie University Graduate School of Medicine, Tsu, Japan
- * E-mail:
| |
Collapse
|