1
|
Wu C, Yang B, Chu J. Periostin acts as an oncogene to promote laryngeal cancer progression by activating decorin. Histol Histopathol 2025; 40:687-696. [PMID: 39319525 DOI: 10.14670/hh-18-804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/26/2024]
Abstract
Laryngeal carcinoma (LC) is the second most common malignancy of the head and neck worldwide, with increasing incidence every year. However, the mechanism of its development is not completely clear. Periostin (POSTN) has been reported to be involved in various aspects of tumorigenesis. To determine the influence of POSTN on LC tumorigenesis, we first examined the expression of POSTN in tissues from patients with LC through immunohistochemistry, western blot, and qRT-PCR. Besides, we demonstrated that POSTN promoted LC cell migration, invasion, and proliferation in vitro by CCK-8, colony formation, and Transwell assays, and tumor growth in vivo by immunohistochemistry. Furthermore, the interaction between POSTN and decorin (DCN) was further verified by bioinformatics analysis and immunoprecipitation (IP), finding that POSTN promoted the malignant progression of LC by targeting DCN. Our findings support the idea that the level of POSTN expression and accumulation in tumors correlated with the malignancy degree of LC, suggesting that POSTN may play a potential role in improving laryngeal cancer treatment strategies.
Collapse
Affiliation(s)
- Chao Wu
- Department of Otolaryngology Head and Neck Surgery, Taizhou People's Hospital, Taizhou, Jiangsu Province, China.
| | - Bo Yang
- Department of Otolaryngology Head and Neck Surgery, Taizhou People's Hospital, Taizhou, Jiangsu Province, China
| | - Jiusheng Chu
- Department of Otolaryngology Head and Neck Surgery, Taizhou People's Hospital, Taizhou, Jiangsu Province, China
| |
Collapse
|
2
|
Sekulovski N, Carleton AE, Rengarajan AA, Lin CW, Juga LN, Whorton AE, Schmidt JK, Golos TG, Taniguchi K. Temporally resolved single cell transcriptomics in a human model of amniogenesis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.09.07.556553. [PMID: 39026707 PMCID: PMC11257495 DOI: 10.1101/2023.09.07.556553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/20/2024]
Abstract
Amniogenesis is triggered in a collection of pluripotent epiblast cells as the human embryo implants. To gain insights into the critical but poorly understood transcriptional machinery governing amnion fate determination, we examined the evolving transcriptome of a developing human pluripotent stem cell-derived amnion model at the single cell level. This analysis revealed several continuous amniotic fate progressing states with state-specific markers, which include a previously unrecognized CLDN10+ amnion progenitor state. Strikingly, we found that expression of CLDN10 is restricted to the amnion-epiblast boundary region in the human post-implantation amniotic sac model as well as in a peri-gastrula cynomolgus macaque embryo, bolstering the growing notion that, at this stage, the amnion-epiblast boundary is a site of active amniogenesis. Bioinformatic analysis of published primate peri-gastrula single cell sequencing data further confirmed that CLDN10 is expressed in cells progressing to amnion. Additionally, our loss of function analysis shows that CLDN10 promotes amniotic but suppresses primordial germ cell-like fate. Overall, this study presents a comprehensive amniogenic single cell transcriptomic resource and identifies a previously unrecognized CLDN10+ amnion progenitor population at the amnion-epiblast boundary of the primate peri-gastrula.
Collapse
Affiliation(s)
- Nikola Sekulovski
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Amber E. Carleton
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Anusha A. Rengarajan
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Chien-Wei Lin
- Division of Biostatistics, Institute for Health and Equity, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Lauren N. Juga
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Allison E. Whorton
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Jenna K. Schmidt
- Wisconsin National Primate Research Center (WNPRC), Madison, WI, USA
| | - Thaddeus G. Golos
- Wisconsin National Primate Research Center (WNPRC), Madison, WI, USA
- Department of Obstetrics and Gynecology, University of Wisconsin - Madison School of Medicine, Madison, WI USA
- Department of Comparative Biosciences, University of Wisconsin - Madison School of Veterinary Medicine, Madison, WI, USA
| | - Kenichiro Taniguchi
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Cancer Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| |
Collapse
|
3
|
Nakanoh S, Sham K, Ghimire S, Mohorianu I, Rayon T, Vallier L. Human surface ectoderm and amniotic ectoderm are sequentially specified according to cellular density. SCIENCE ADVANCES 2024; 10:eadh7748. [PMID: 38427729 PMCID: PMC10906920 DOI: 10.1126/sciadv.adh7748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Accepted: 01/29/2024] [Indexed: 03/03/2024]
Abstract
Mechanisms specifying amniotic ectoderm and surface ectoderm are unresolved in humans due to their close similarities in expression patterns and signal requirements. This lack of knowledge hinders the development of protocols to accurately model human embryogenesis. Here, we developed a human pluripotent stem cell model to investigate the divergence between amniotic and surface ectoderms. In the established culture system, cells differentiated into functional amnioblast-like cells. Single-cell RNA sequencing analyses of amnioblast differentiation revealed an intermediate cell state with enhanced surface ectoderm gene expression. Furthermore, when the differentiation started at the confluent condition, cells retained the expression profile of surface ectoderm. Collectively, we propose that human amniotic ectoderm and surface ectoderm are specified along a common nonneural ectoderm trajectory based on cell density. Our culture system also generated extraembryonic mesoderm-like cells from the primed pluripotent state. Together, this study provides an integrative understanding of the human nonneural ectoderm development and a model for embryonic and extraembryonic human development around gastrulation.
Collapse
Affiliation(s)
- Shota Nakanoh
- Wellcome-MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge CB2 0AW, UK
- Epigenetics & Signalling Programmes, Babraham Institute, Cambridge CB22 3AT, UK
| | - Kendig Sham
- Wellcome-MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge CB2 0AW, UK
| | - Sabitri Ghimire
- Wellcome-MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge CB2 0AW, UK
| | - Irina Mohorianu
- Wellcome-MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge CB2 0AW, UK
| | - Teresa Rayon
- Epigenetics & Signalling Programmes, Babraham Institute, Cambridge CB22 3AT, UK
| | - Ludovic Vallier
- Wellcome-MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge CB2 0AW, UK
- Berlin Institute of Health Centre for Regenerative Therapies, Charité - Universitätsmedizin Berlin, Berlin 13353, Germany
- Max Planck Institute for Molecular Genetics, Berlin 14195, Germany
| |
Collapse
|
4
|
Farkas K, Ferretti E. Derivation of Human Extraembryonic Mesoderm-like Cells from Primitive Endoderm. Int J Mol Sci 2023; 24:11366. [PMID: 37511125 PMCID: PMC10380231 DOI: 10.3390/ijms241411366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 06/22/2023] [Accepted: 06/29/2023] [Indexed: 07/30/2023] Open
Abstract
In vitro modeling of human peri-gastrulation development is a valuable tool for understanding embryogenetic mechanisms. The extraembryonic mesoderm (ExM) is crucial in supporting embryonic development by forming tissues such as the yolk sac, allantois, and chorionic villi. However, the origin of human ExM remains only partially understood. While evidence suggests a primitive endoderm (PrE) origin based on morphological findings, current in vitro models use epiblast-like cells. To address this gap, we developed a protocol to generate ExM-like cells from PrE-like cell line called naïve extraembryonic endoderm (nEnd). We identified the ExM-like cells by specific markers (LUM and ANXA1). Moreover, these in vitro-produced ExM cells displayed angiogenic potential on a soft matrix, mirroring their physiological role in vasculogenesis. By integrating single-cell RNA sequencing (scRNAseq) data, we found that the ExM-like cells clustered with the LUM/ANXA1-rich cell populations of the gastrulating embryo, indicating similarity between in vitro and ex utero cell populations. This study confirms the derivation of ExM from PrE and establishes a cell culture system that can be utilized to investigate ExM during human peri-gastrulation development, both in monolayer cultures and more complex models.
Collapse
Affiliation(s)
- Karin Farkas
- Novo Nordisk Foundation Center for Stem Cell Biology (DanStem), University of Copenhagen, 1165 Copenhagen, Denmark
- Department of Biomedical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Elisabetta Ferretti
- Novo Nordisk Foundation Center for Stem Cell Biology (DanStem), University of Copenhagen, 1165 Copenhagen, Denmark
- Department of Cellular and Molecular Medicine, University of Copenhagen, 2200 Copenhagen, Denmark
| |
Collapse
|
5
|
Translational Comparison of the Human and Mouse Yolk Sac Development and Function. Reprod Sci 2023; 30:41-53. [PMID: 35137348 DOI: 10.1007/s43032-022-00872-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2021] [Accepted: 01/29/2022] [Indexed: 01/06/2023]
Abstract
The yolk sac (YS) is the oldest of the extraembryonic membranes in vertebrates. Considered a transitory structure in the human species, the importance of the YS for a successful pregnancy is often overlooked. Due to the general inaccessibility of healthy human YS tissue for research, the use of experimental animal models is of great value. In order to better understand whether the mouse could be used as a translational model for the study of the human YS under normal and pathological conditions, this review comprehensively describes key developmental aspects of the human and mouse YS, detailing their development and function. YS major similarities in both species comprise the following: (1) histological composition (both being composed of endoderm, mesoderm, and mesothelium layers); (2) endoderm endocytosis, synthesis, secretion, and transport capabilities; and (3) mesoderm onset of haematopoiesis and angiogenesis. Examples of main dissimilarities include (1) persistence across pregnancy (i.e. early pregnancy in humans vs term pregnancy in mice); (2) the existence of a secondary YS in humans; (3) the presence of proliferative primordial germ cells (PGCs) in the human versus their absence in mice; and (4) eversion of histological layers in the mouse. Although these differences should be considered when interpreting data from mouse-based studies, the overall morphofunctional similarities in the YS between these species indicate that the mouse can be potentially used as a translational model for the study of the human YS.
Collapse
|
6
|
Chuva de Sousa Lopes SM, Roelen BAJ, Lawson KA, Zwijsen A. The development of the amnion in mice and other amniotes. Philos Trans R Soc Lond B Biol Sci 2022; 377:20210258. [PMID: 36252226 PMCID: PMC9574641 DOI: 10.1098/rstb.2021.0258] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
The amnion is an extraembryonic tissue that evolutionarily allowed embryos of all amniotes to develop in a transient and local aquatic environment. Despite the importance of this tissue, very little is known about its formation and its molecular characteristics. In this review, we have compared the basic organization of the extraembryonic membranes in amniotes and describe the two types of amniogenesis, folding and cavitation. We then zoom in on the atypical development of the amnion in mice that occurs via the formation of a single posterior amniochorionic fold. Moreover, we consolidate lineage tracing data to better understand the spatial and temporal origin of the progenitors of amniotic ectoderm, and visualize the behaviour of their descendants in the extraembryonic–embryonic junctional region. This analysis provides new insight on amnion development and expansion. Finally, using an online-available dataset of single-cell transcriptomics during the gastrulation period in mice, we provide bioinformatic analysis of the molecular signature of amniotic ectoderm and amniotic mesoderm. The amnion is a tissue with unique biomechanical properties that deserves to be better understood. This article is part of the theme issue ‘Extraembryonic tissues: exploring concepts, definitions and functions across the animal kingdom’.
Collapse
Affiliation(s)
- Susana M Chuva de Sousa Lopes
- Department of Anatomy and Embryology, Leiden University Medical Centre, Einthovenweg 20, 2333 ZC Leiden, The Netherlands.,Ghent-Fertility and Stem Cell Team (G-FAST), Department of Reproductive Medicine, Ghent University Hospital, Corneel Heymanslaan 10, 9000 Ghent, Belgium
| | - Bernard A J Roelen
- Anatomy and Physiology, Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Yalelaan 1, 3584CL Utrecht, The Netherlands.,Department of Biosciences, Biotechnologies & Biopharmaceutics, University of Bari Aldo Moro, Bari, Italy
| | - Kirstie A Lawson
- MRC Human Genetics Unit, IGC, University of Edinburgh, Crewe Road South, Edinburgh EH4 2XU, UK
| | - An Zwijsen
- Cardiovascular Sciences, Center for Molecular and Vascular Biology, KU Leuven, Herestraat 49 box 911, 3000 Leuven, Belgium
| |
Collapse
|
7
|
Tyser RCV, Srinivas S. Recent advances in understanding cell types during human gastrulation. Semin Cell Dev Biol 2022; 131:35-43. [PMID: 35606274 PMCID: PMC7615356 DOI: 10.1016/j.semcdb.2022.05.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 04/20/2022] [Accepted: 05/04/2022] [Indexed: 12/14/2022]
Abstract
Gastrulation is a fundamental process during embryonic development, conserved across all multicellular animals [1]. In the majority of metazoans, gastrulation is characterised by large scale morphogenetic remodeling, leading to the conversion of an early pluripotent embryonic cell layer into the three primary 'germ layers': an outer ectoderm, inner endoderm and intervening mesoderm layer. The morphogenesis of these three layers of cells is closely coordinated with cellular diversification, laying the foundation for the generation of the hundreds of distinct specialized cell types in the animal body. The process of gastrulation has for a long time attracted tremendous attention in a broad range of experimental systems ranging from sponges to mice. In humans the process of gastrulation starts approximately 14 days after fertilization and continues for slightly over a week. However our understanding of this important process, as it pertains to human, is limited. Donations of human fetal material at these early stages are exceptionally rare, making it nearly impossible to study human gastrulation directly. Therefore, our understanding of human gastrulation is predominantly derived from animal models such as the mouse [2,3] and from studies of limited collections of fixed whole samples and histological sections of human gastrulae [4-7], some of which date back to over a century ago. More recently we have been gaining valuable molecular insights into human gastrulation using in vitro models of hESCs [8-12] and increasingly, in vitro cultured human and non-human primate embryos [13-16]. However, while methods have been developed to culture human embryos into this stage (and probably beyond), current ethical standards prohibit the culture of human embryos past 14 days again limiting our ability to experimentally probe human gastrulation. This review discusses recent molecular insights from the study of a rare CS 7 human gastrula obtained as a live sample and raises several questions arising from this recent study that it will be interesting to address in the future using emerging models of human gastrulation.
Collapse
Affiliation(s)
- Richard C V Tyser
- Department of Physiology, Anatomy and Genetics, South Parks Road, University of Oxford , Oxford OX1 3QX, UK
| | - Shankar Srinivas
- Department of Physiology, Anatomy and Genetics, South Parks Road, University of Oxford , Oxford OX1 3QX, UK
| |
Collapse
|
8
|
Amadei G, Handford CE, Qiu C, De Jonghe J, Greenfeld H, Tran M, Martin BK, Chen DY, Aguilera-Castrejon A, Hanna JH, Elowitz MB, Hollfelder F, Shendure J, Glover DM, Zernicka-Goetz M. Embryo model completes gastrulation to neurulation and organogenesis. Nature 2022; 610:143-153. [PMID: 36007540 PMCID: PMC9534772 DOI: 10.1038/s41586-022-05246-3] [Citation(s) in RCA: 141] [Impact Index Per Article: 47.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 08/17/2022] [Indexed: 11/23/2022]
Abstract
Embryonic stem (ES) cells can undergo many aspects of mammalian embryogenesis in vitro1-5, but their developmental potential is substantially extended by interactions with extraembryonic stem cells, including trophoblast stem (TS) cells, extraembryonic endoderm stem (XEN) cells and inducible XEN (iXEN) cells6-11. Here we assembled stem cell-derived embryos in vitro from mouse ES cells, TS cells and iXEN cells and showed that they recapitulate the development of whole natural mouse embryo in utero up to day 8.5 post-fertilization. Our embryo model displays headfolds with defined forebrain and midbrain regions and develops a beating heart-like structure, a trunk comprising a neural tube and somites, a tail bud containing neuromesodermal progenitors, a gut tube, and primordial germ cells. This complete embryo model develops within an extraembryonic yolk sac that initiates blood island development. Notably, we demonstrate that the neurulating embryo model assembled from Pax6-knockout ES cells aggregated with wild-type TS cells and iXEN cells recapitulates the ventral domain expansion of the neural tube that occurs in natural, ubiquitous Pax6-knockout embryos. Thus, these complete embryoids are a powerful in vitro model for dissecting the roles of diverse cell lineages and genes in development. Our results demonstrate the self-organization ability of ES cells and two types of extraembryonic stem cells to reconstitute mammalian development through and beyond gastrulation to neurulation and early organogenesis.
Collapse
Affiliation(s)
- Gianluca Amadei
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
- Department of Biology, University of Padua, Padua, Italy
| | - Charlotte E Handford
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
- Centre for Trophoblast Research, University of Cambridge, Cambridge, UK
| | - Chengxiang Qiu
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
| | - Joachim De Jonghe
- Department of Biochemistry, University of Cambridge, Cambridge, UK
- Francis Crick Institute, London, UK
| | - Hannah Greenfeld
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Martin Tran
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Beth K Martin
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
| | - Dong-Yuan Chen
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | | | - Jacob H Hanna
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Michael B Elowitz
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
- Allen Discovery Center for Cell Lineage Tracing, Seattle, WA, USA
| | | | - Jay Shendure
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
- Allen Discovery Center for Cell Lineage Tracing, Seattle, WA, USA
- Brotman Baty Institute for Precision Medicine, Seattle, WA, USA
- Howard Hughes Medical Institute, Seattle, WA, USA
| | - David M Glover
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Magdalena Zernicka-Goetz
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK.
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA.
- Centre for Trophoblast Research, University of Cambridge, Cambridge, UK.
- Allen Discovery Center for Cell Lineage Tracing, Seattle, WA, USA.
| |
Collapse
|
9
|
Zhu Y, Wang H, Yin F, Guo Y, Li F, Gao D, Qin J. Amnion-on-a-chip: modeling human amniotic development in mid-gestation from pluripotent stem cells. LAB ON A CHIP 2020; 20:3258-3268. [PMID: 32749421 DOI: 10.1039/d0lc00268b] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
The amnion serves to create a protective environment for a growing fetus, and the study of amniotic development will greatly facilitate our understanding of normal and abnormal pregnancies. However, this remains a poorly studied field due to the lack of ideal human models. Herein, we present an integrative strategy to generate amnion-like cavity tissue from human pluripotent stem cells (hPSCs) in an amnion-on-a-chip device through combining a bioengineering approach and developmental biology principles. hPSCs could self-organize into an amnion epithelial cavity in a perfusable 3D culture microchip, resembling human amniotic development in mid-gestation. These cavities exhibited the critical features of amnion tissue based on morphological characteristics, marker expression, and transcriptome analysis. RNA-seq revealed that a set of amnion-specific genes were highly expressed in the obtained cavities, suggesting that the amnion epithelium was derived from hPSCs. Moreover, the amnion-specific mid-gestation marker KRT24 was highly expressed at the mRNA and protein levels, verifying the high maturation of amnion tissues after long-term 3D culturing and differentiation for up to 20 days. These new findings demonstrate the potential of this new amnion-on-a-chip model for investigating essential biological events in human amnions in normal and diseased states via integrating microengineering technology and stem cell biology.
Collapse
Affiliation(s)
- Yujuan Zhu
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, China. and University of Chinese Academy of Sciences, Beijing, China
| | - Hui Wang
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, China. and University of Chinese Academy of Sciences, Beijing, China
| | - Fangchao Yin
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, China. and University of Chinese Academy of Sciences, Beijing, China
| | - Yaqiong Guo
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, China. and University of Chinese Academy of Sciences, Beijing, China
| | - Fei Li
- Shanghai Institute of Biochemistry and Cell Biology, Shanghai, China.
| | - Dong Gao
- Shanghai Institute of Biochemistry and Cell Biology, Shanghai, China.
| | - Jianhua Qin
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, China. and University of Chinese Academy of Sciences, Beijing, China and Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China and CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| |
Collapse
|
10
|
Yin F, Zhu Y, Wang H, Wang Y, Li D, Qin J. Microengineered hiPSC-Derived 3D Amnion Tissue Model to Probe Amniotic Inflammatory Responses under Bacterial Exposure. ACS Biomater Sci Eng 2020; 6:4644-4652. [PMID: 33455183 DOI: 10.1021/acsbiomaterials.0c00592] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Intra-amniotic infection is a common cause of preterm birth that can lead to adverse neonatal outcomes. Despite the basic and clinical significance, the study in normal and diseased human amnion is highly challenging due to the limited use of human primary tissues and the distinct divergence between animal models and human. Here, we established a microengineered hiPSC-derived amnion tissue model on a chip to investigate the inflammatory responses of amnion tissues to bacterial exposure. The microdevice consisted of two parallel channels with a middle matrix channel, creating a permissive microenvironment for amnion differentiation. Dissociated hiPSCs efficiently self-organized into cell cavity and finally differentiated into a polarized squamous amniotic epithelium on the chip under perfused 3D culture. When exposed to E. coli, amnion tissue exhibited significant functional impairments compared to the control, including induced cell apoptosis, disrupted cell junction integrity, and increased inflammatory factor secretion, recapitulating a series of characteristic clinical signs of intra-amniotic infection at an early stage. Together, this amnion-on-a-chip model provides a promising platform to investigate intrauterine inflammation in early gestation, indicating its potential applications in human embryology and reproductive medicine.
Collapse
Affiliation(s)
- Fangchao Yin
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023 China.,University of Chinese Academy of Sciences, Beijing 100049 China
| | - Yujuan Zhu
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023 China.,University of Chinese Academy of Sciences, Beijing 100049 China
| | - Hui Wang
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023 China.,University of Chinese Academy of Sciences, Beijing 100049 China
| | - Yaqing Wang
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023 China.,University of Chinese Academy of Sciences, Beijing 100049 China
| | - Dong Li
- Dalian Municipal Women and Children's Medical Center, Dalian 116037 China
| | - Jianhua Qin
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023 China.,Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101 China.,CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031 China.,University of Chinese Academy of Sciences, Beijing 100049 China
| |
Collapse
|
11
|
Zheng Y, Xue X, Shao Y, Wang S, Esfahani SN, Li Z, Muncie JM, Lakins JN, Weaver VM, Gumucio DL, Fu J. Controlled modelling of human epiblast and amnion development using stem cells. Nature 2019; 573:421-425. [PMID: 31511693 PMCID: PMC8106232 DOI: 10.1038/s41586-019-1535-2] [Citation(s) in RCA: 323] [Impact Index Per Article: 53.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Accepted: 08/06/2019] [Indexed: 11/09/2022]
Abstract
Early human embryonic development involves extensive lineage diversification, cell-fate specification and tissue patterning1. Despite its basic and clinical importance, early human embryonic development remains relatively unexplained owing to interspecies divergence2,3 and limited accessibility to human embryo samples. Here we report that human pluripotent stem cells (hPSCs) in a microfluidic device recapitulate, in a highly controllable and scalable fashion, landmarks of the development of the epiblast and amniotic ectoderm parts of the conceptus, including lumenogenesis of the epiblast and the resultant pro-amniotic cavity, formation of a bipolar embryonic sac, and specification of primordial germ cells and primitive streak cells. We further show that amniotic ectoderm-like cells function as a signalling centre to trigger the onset of gastrulation-like events in hPSCs. Given its controllability and scalability, the microfluidic model provides a powerful experimental system to advance knowledge of human embryology and reproduction. This model could assist in the rational design of differentiation protocols of hPSCs for disease modelling and cell therapy, and in high-throughput drug and toxicity screens to prevent pregnancy failure and birth defects.
Collapse
Affiliation(s)
- Yi Zheng
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Xufeng Xue
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Yue Shao
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Sicong Wang
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, USA
| | | | - Zida Li
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Jonathon M Muncie
- Department of Surgery, Center for Bioengineering and Tissue Regeneration, University of California, San Francisco, San Francisco, CA, USA
- Graduate Program in Bioengineering, University of California, San Francisco and University of California, Berkeley, San Francisco, CA, USA
| | - Johnathon N Lakins
- Department of Surgery, Center for Bioengineering and Tissue Regeneration, University of California, San Francisco, San Francisco, CA, USA
| | - Valerie M Weaver
- Department of Surgery, Center for Bioengineering and Tissue Regeneration, University of California, San Francisco, San Francisco, CA, USA
- Department of Bioengineering and Therapeutic Sciences, Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA, USA
- Department of Radiation Oncology, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA
| | - Deborah L Gumucio
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Jianping Fu
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, USA.
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, USA.
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
12
|
Zhong H, Li X, Zhang J, Wu X. Overexpression of periostin is positively associated with gastric cancer metastasis through promoting tumor metastasis and invasion. J Cell Biochem 2019; 120:9927-9935. [PMID: 30637809 DOI: 10.1002/jcb.28275] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Accepted: 10/24/2018] [Indexed: 12/18/2022]
Abstract
BACKGROUND Gastric tumors generally have a poor prognosis and molecular markers to improve early detection and predict outcomes are greatly needed. The present study reports that periostin (POSTN), a secretory protein that can alter the remodeling of the extracellular matrix, is highly expressed in gastric tumors. MATERIALS AND METHODS Gastric tissues were collected from patients at the Department of Thoracic Surgery/Huiqiao Medical Center, Nanfang Hospital, Southern Medical University. These patients provided an informed consent and were approved by the institute. Normal, cancer, and metastatic gastric tissues from lymph nodes and tissues adjacent to the tumor were collected from patients diagnosed with gastric cancer. RESULTS Periostin expression gradually increased as the risk grade of the NIH classification increased, and this was closely correlated with disease-free survival and overall survival. Compared with adjacent normal gastric mucosa tissues, protein expression of POSTN in gastric cancer tissues and metastases was significantly higher by immunohistochemistry and Western blot analysis. In addition, POSTN was upregulated in advanced gastric cancer tissues than in early gastric cancer tissues. Moreover, the ectopic expression of POSTN in the immortalized human gastric cell line could increase the metastasis and invasion of gastric cancer cells. CONCLUSION The present results could establish the significance of POSTN in driving oncogenesis and metastasis in gastric tumors, with implications for its potential use as a diagnostic or prognostic biomarker, and as a candidate therapeutic target.
Collapse
Affiliation(s)
- Hai Zhong
- Department of Thoracic Surgery, Huiqiao Medical Center, Nanfang Hospital, Southern Medical University, Guangzhou, People's Republic of China.,Department of Cardiothoracic Surgery, The second Hospital of Yinzhou District, Ningbo, People's Republic of China
| | - Xiang Li
- Department of Thoracic Surgery, Huiqiao Medical Center, Nanfang Hospital, Southern Medical University, Guangzhou, People's Republic of China
| | - Junhua Zhang
- Departmentof Operating Room, Nanfang Hospital, Southern Medical University, Guangzhou, People's Republic of China
| | - Xu Wu
- Department of Thoracic Surgery, Huiqiao Medical Center, Nanfang Hospital, Southern Medical University, Guangzhou, People's Republic of China
| |
Collapse
|
13
|
Dobreva MP, Abon Escalona V, Lawson KA, Sanchez MN, Ponomarev LC, Pereira PNG, Stryjewska A, Criem N, Huylebroeck D, Chuva de Sousa Lopes SM, Aerts S, Zwijsen A. Amniotic ectoderm expansion in mouse occurs via distinct modes and requires SMAD5-mediated signalling. Development 2018; 145:dev.157222. [PMID: 29884675 DOI: 10.1242/dev.157222] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Accepted: 05/30/2018] [Indexed: 12/18/2022]
Abstract
Upon gastrulation, the mammalian conceptus transforms rapidly from a simple bilayer into a multilayered embryo enveloped by its extra-embryonic membranes. Impaired development of the amnion, the innermost membrane, causes major malformations. To clarify the origin of the mouse amnion, we used single-cell labelling and clonal analysis. We identified four clone types with distinct clonal growth patterns in amniotic ectoderm. Two main types have progenitors in extreme proximal-anterior epiblast. Early descendants initiate and expand amniotic ectoderm posteriorly, while descendants of cells remaining anteriorly later expand amniotic ectoderm from its anterior side. Amniogenesis is abnormal in embryos deficient in the bone morphogenetic protein (BMP) signalling effector SMAD5, with delayed closure of the proamniotic canal, and aberrant amnion and folding morphogenesis. Transcriptomics of individual Smad5 mutant amnions isolated before visible malformations and tetraploid chimera analysis revealed two amnion defect sets. We attribute them to impairment of progenitors of the two main cell populations in amniotic ectoderm and to compromised cuboidal-to-squamous transition of anterior amniotic ectoderm. In both cases, SMAD5 is crucial for expanding amniotic ectoderm rapidly into a stretchable squamous sheet to accommodate exocoelom expansion, axial growth and folding morphogenesis.
Collapse
Affiliation(s)
- Mariya P Dobreva
- VIB-KU Leuven Center for Brain and Disease Research, Leuven 3000, Belgium .,Department of Human Genetics, KU Leuven, Leuven 3000, Belgium
| | - Vanesa Abon Escalona
- VIB-KU Leuven Center for Brain and Disease Research, Leuven 3000, Belgium.,Department of Human Genetics, KU Leuven, Leuven 3000, Belgium.,Department of Cardiovascular Sciences, KU Leuven, Leuven 3000, Belgium
| | - Kirstie A Lawson
- MRC Human Genetics Unit, MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh EH4 2XU, UK
| | | | - Ljuba C Ponomarev
- Department of Cardiovascular Sciences, KU Leuven, Leuven 3000, Belgium
| | - Paulo N G Pereira
- VIB-KU Leuven Center for Brain and Disease Research, Leuven 3000, Belgium.,Department of Human Genetics, KU Leuven, Leuven 3000, Belgium
| | - Agata Stryjewska
- Department of Development and Regeneration, KU Leuven, Leuven 3000, Belgium
| | - Nathan Criem
- VIB-KU Leuven Center for Brain and Disease Research, Leuven 3000, Belgium.,Department of Human Genetics, KU Leuven, Leuven 3000, Belgium.,Department of Cardiovascular Sciences, KU Leuven, Leuven 3000, Belgium
| | - Danny Huylebroeck
- Department of Development and Regeneration, KU Leuven, Leuven 3000, Belgium
| | | | - Stein Aerts
- Department of Human Genetics, KU Leuven, Leuven 3000, Belgium
| | - An Zwijsen
- VIB-KU Leuven Center for Brain and Disease Research, Leuven 3000, Belgium .,Department of Human Genetics, KU Leuven, Leuven 3000, Belgium.,Department of Cardiovascular Sciences, KU Leuven, Leuven 3000, Belgium
| |
Collapse
|
14
|
Freis A, Schlegel J, Kuon RJ, Doster A, Jauckus J, Strowitzki T, Germeyer A. Serum periostin levels in early in pregnancy are significantly altered in women with miscarriage. Reprod Biol Endocrinol 2017; 15:87. [PMID: 29096644 PMCID: PMC5667517 DOI: 10.1186/s12958-017-0307-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Accepted: 10/23/2017] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND Miscarriage is a common complication in pregnancy and there is still a lack of biomarkers usable in asymptomatic patients before the event occurs. Periostin (PER), whose levels rise particularly during injury or inflammation, has been shown to play an important local role in implantation and early embryonic development. As PER has been described as a biomarker in various medical conditions we intended to evaluate if changes in PER serum levels may help to identify women at risk for spontaneous abortion in the first trimester. METHODS Women between 18 and 42 years without confounding comorbidities who conceived by IVF/ICSI and ovarian hyperstimulation were analysed in the study after informed consent. Maternal serum samples from 41 patients were assessed at the time of pregnancy testing (PT) and the following first ultrasound checkup (US). Patients were subsequently divided in two groups: (1) patients with subsequent miscarriage in the first trimester (n = 18) and (2) patients with ongoing pregnancy (n = 23), allowing for statistical analysis and investigating the change of PER levels per individual. PER levels were measured using enzyme-linked immunosorbent assay. Statistical analysis was performed using the Fisher exact and Student's t test. p ≤ 0.05 was considered to be significant. RESULTS There was no significant difference concerning possible confounders between the two groups. We did not find any significant difference in PER levels at the time point of PT or US. By investigating the interindividual changes of PER between the two time points however, we observed that patients with a following miscarriage showed increasing levels of PER at the time point of PT compared to US in contrast to patients with an ongoing pregnancy who demonstrated a decrease in PER levels. These alterations were significant in the absolute as well as in the relative comparison. CONCLUSION The relative expression of PER between PT and US is significantly altered in asymptomatic women with subsequent miscarriage compared to women with ongoing pregnancy. Therefore systemic PER levels might represent a potential promising biomarker for the assessment of pregnancy outcome. TRIAL REGISTRATION Not applicable.
Collapse
Affiliation(s)
- A. Freis
- Department of Gynecological Endocrinology and Fertility Disorders, University Hospital Heidelberg, INF 440, 69120 Heidelberg, Germany
| | - J. Schlegel
- Department of Gynecological Endocrinology and Fertility Disorders, University Hospital Heidelberg, INF 440, 69120 Heidelberg, Germany
| | - R. J. Kuon
- Department of Gynecological Endocrinology and Fertility Disorders, University Hospital Heidelberg, INF 440, 69120 Heidelberg, Germany
| | - A. Doster
- Department of Gynecological Endocrinology and Fertility Disorders, University Hospital Heidelberg, INF 440, 69120 Heidelberg, Germany
| | - J. Jauckus
- Department of Gynecological Endocrinology and Fertility Disorders, University Hospital Heidelberg, INF 440, 69120 Heidelberg, Germany
| | - T. Strowitzki
- Department of Gynecological Endocrinology and Fertility Disorders, University Hospital Heidelberg, INF 440, 69120 Heidelberg, Germany
| | - A. Germeyer
- Department of Gynecological Endocrinology and Fertility Disorders, University Hospital Heidelberg, INF 440, 69120 Heidelberg, Germany
| |
Collapse
|
15
|
Shao Y, Taniguchi K, Townshend RF, Miki T, Gumucio DL, Fu J. A pluripotent stem cell-based model for post-implantation human amniotic sac development. Nat Commun 2017; 8:208. [PMID: 28785084 PMCID: PMC5547056 DOI: 10.1038/s41467-017-00236-w] [Citation(s) in RCA: 192] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Accepted: 06/12/2017] [Indexed: 01/24/2023] Open
Abstract
Development of the asymmetric amniotic sac-with the embryonic disc and amniotic ectoderm occupying opposite poles-is a vital milestone during human embryo implantation. Although essential to embryogenesis and pregnancy, amniotic sac development in humans remains poorly understood. Here, we report a human pluripotent stem cell (hPSC)-based model, termed the post-implantation amniotic sac embryoid (PASE), that recapitulates multiple post-implantation embryogenic events centered around amniotic sac development. Without maternal or extraembryonic tissues, the PASE self-organizes into an epithelial cyst with an asymmetric amniotic ectoderm-epiblast pattern that resembles the human amniotic sac. Upon further development, the PASE initiates a process that resembles posterior primitive streak development in a SNAI1-dependent manner. Furthermore, we observe asymmetric BMP-SMAD signaling concurrent with PASE development, and establish that BMP-SMAD activation/inhibition modulates stable PASE development. This study reveals a previously unrecognized fate potential of human pluripotent stem cells and provides a platform for advancing human embryology.Early in human embryonic development, it is unclear how amniotic sac formation is regulated. Here, the authors use a human pluripotent stem cell-based model, termed the post-implantation amniotic sac embryoid, to recapitulate early embryogenic events of human amniotic sac development.
Collapse
Affiliation(s)
- Yue Shao
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Kenichiro Taniguchi
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
| | - Ryan F Townshend
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
| | - Toshio Miki
- Department of Biochemistry and Molecular Biology, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90089, USA
| | - Deborah L Gumucio
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, 48109, USA.
| | - Jianping Fu
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA.
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, 48109, USA.
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA.
| |
Collapse
|
16
|
Shao Y, Taniguchi K, Gurdziel K, Townshend RF, Xue X, Yong KMA, Sang J, Spence JR, Gumucio DL, Fu J. Self-organized amniogenesis by human pluripotent stem cells in a biomimetic implantation-like niche. NATURE MATERIALS 2017; 16:419-425. [PMID: 27941807 PMCID: PMC5374007 DOI: 10.1038/nmat4829] [Citation(s) in RCA: 170] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Accepted: 11/22/2016] [Indexed: 05/05/2023]
Abstract
Amniogenesis-the development of amnion-is a critical developmental milestone for early human embryogenesis and successful pregnancy. However, human amniogenesis is poorly understood due to limited accessibility to peri-implantation embryos and a lack of in vitro models. Here we report an efficient biomaterial system to generate human amnion-like tissue in vitro through self-organized development of human pluripotent stem cells (hPSCs) in a bioengineered niche mimicking the in vivo implantation environment. We show that biophysical niche factors act as a switch to toggle hPSC self-renewal versus amniogenesis under self-renewal-permissive biochemical conditions. We identify a unique molecular signature of hPSC-derived amnion-like cells and show that endogenously activated BMP-SMAD signalling is required for the amnion-like tissue development by hPSCs. This study unveils the self-organizing and mechanosensitive nature of human amniogenesis and establishes the first hPSC-based model for investigating peri-implantation human amnion development, thereby helping advance human embryology and reproductive medicine.
Collapse
Affiliation(s)
- Yue Shao
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Kenichiro Taniguchi
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Katherine Gurdziel
- Institute of Environmental Health Sciences, Wayne State University, Detroit, MI 48202, USA
| | - Ryan F. Townshend
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Xufeng Xue
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Koh Meng Aw Yong
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Jianming Sang
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Jason R. Spence
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Deborah L. Gumucio
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Correspondence should be addressed to J. F. () or D. L. G. ()
| | - Jianping Fu
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
- Correspondence should be addressed to J. F. () or D. L. G. ()
| |
Collapse
|
17
|
Seubbuk S, Sritanaudomchai H, Kasetsuwan J, Surarit R. High glucose promotes the osteogenic differentiation capability of human periodontal ligament fibroblasts. Mol Med Rep 2017; 15:2788-2794. [PMID: 28447734 DOI: 10.3892/mmr.2017.6333] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Accepted: 01/16/2017] [Indexed: 11/06/2022] Open
Abstract
Periodontal ligament fibroblasts (PDLFs) are important cells, which are involved in maintaining tooth integrity. Diabetes has been found to be associated with periodontal disease in a bidirectional manner. The aim of the present study was to investigate the stemness properties of human PDLFs (HPDLFs) in high glucose conditions. HPDLFs were analyzed for their osteogenic differentiation capacity by inducing the cells with osteogenic medium in various glucose concentrations. The gene expression was then examined using reverse transcription‑quantitative polymerase chain reaction analysis, and examinations of alkaline phosphatase activity and nodule formation were performed. The results of the gene expression analysis revealed that high glucose media induced the expression of NANOG, octamer-binding transcription factor 4, (sex determining region Y)‑box 2, cluster of differentiation 166 (CD166), PERIOSTIN and β‑CATENIN following culture of the cells for 3 days. Alkaline phosphatase activity increased following 14 days in the high glucose condition. In addition, higher numbers of calcified nodules were formed on day 28 in the group cultured with high glucose. The results showed that high glucose induced bone formation by elevating the expression of stem cell markers, particularly CD166, and this induction may be regulated through β-CATENIN.
Collapse
Affiliation(s)
- Sujiwan Seubbuk
- Molecular Medicine Program, Faculty of Science, Mahidol University, Ratchthewi, Bangkok 10400, Thailand
| | - Hathaitip Sritanaudomchai
- Department of Oral Biology, Faculty of Dentistry, Mahidol University, Ratchthewi, Bangkok 10400, Thailand
| | - Julalux Kasetsuwan
- Department of Oral Medicine and Periodontology, Faculty of Dentistry, Mahidol University, Ratchthewi, Bangkok 10400, Thailand
| | - Rudee Surarit
- Department of Oral Biology, Faculty of Dentistry, Mahidol University, Ratchthewi, Bangkok 10400, Thailand
| |
Collapse
|
18
|
Xu HY, Nie EM, Deng G, Lai LZ, Sun FY, Tian H, Fang FC, Zou YG, Wu BL, Ou-Yang J. Periostin is essential for periodontal ligament remodeling during orthodontic treatment. Mol Med Rep 2017; 15:1800-1806. [DOI: 10.3892/mmr.2017.6200] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Accepted: 12/09/2016] [Indexed: 11/06/2022] Open
|
19
|
Cajal M, Creuzet SE, Papanayotou C, Sabéran-Djoneidi D, Chuva de Sousa Lopes SM, Zwijsen A, Collignon J, Camus A. A conserved role for non-neural ectoderm cells in early neural development. Development 2014; 141:4127-38. [DOI: 10.1242/dev.107425] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
During the early steps of head development, ectodermal patterning leads to the emergence of distinct non-neural and neural progenitor cells. The induction of the preplacodal ectoderm and the neural crest depends on well-studied signalling interactions between the non-neural ectoderm fated to become epidermis and the prospective neural plate. By contrast, the involvement of the non-neural ectoderm in the morphogenetic events leading to the development and patterning of the central nervous system has been studied less extensively. Here, we show that the removal of the rostral non-neural ectoderm abutting the prospective neural plate at late gastrulation stage leads, in mouse and chick embryos, to morphological defects in forebrain and craniofacial tissues. In particular, this ablation compromises the development of the telencephalon without affecting that of the diencephalon. Further investigations of ablated mouse embryos established that signalling centres crucial for forebrain regionalization, namely the axial mesendoderm and the anterior neural ridge, form normally. Moreover, changes in cell death or cell proliferation could not explain the specific loss of telencephalic tissue. Finally, we provide evidence that the removal of rostral tissues triggers misregulation of the BMP, WNT and FGF signalling pathways that may affect telencephalon development. This study opens new perspectives on the role of the neural/non-neural interface and reveals its functional relevance across higher vertebrates.
Collapse
Affiliation(s)
- Marieke Cajal
- Université Paris Diderot, Sorbonne Paris Cité, Institut Jacques Monod, UMR7592 CNRS, Paris F-75013, France
| | - Sophie E. Creuzet
- Institut de Neurobiologie, Laboratoire Neurobiologie et Développement, CNRS-UPR3294, avenue de la Terrasse, Gif-sur-Yvette 91198, France
| | - Costis Papanayotou
- Université Paris Diderot, Sorbonne Paris Cité, Institut Jacques Monod, UMR7592 CNRS, Paris F-75013, France
| | - Délara Sabéran-Djoneidi
- Université Paris Diderot, Sorbonne Paris Cité, Institut Jacques Monod, UMR7592 CNRS, Paris F-75013, France
| | | | - An Zwijsen
- Laboratory of Developmental Signaling, VIB Center for the Biology of Disease, and KU Leuven, Department for Human Genetics, Leuven 3000, Belgium
| | - Jérôme Collignon
- Université Paris Diderot, Sorbonne Paris Cité, Institut Jacques Monod, UMR7592 CNRS, Paris F-75013, France
| | - Anne Camus
- Université Paris Diderot, Sorbonne Paris Cité, Institut Jacques Monod, UMR7592 CNRS, Paris F-75013, France
| |
Collapse
|
20
|
Francisco JC, Cunha RC, Simeoni RB, Guarita-Souza LC, Ferreira RJ, Irioda AC, Souza CMCO, Srikanth GVN, Nityanand S, Chachques JC, de Carvalho KAT. Amniotic membrane as a potent source of stem cells and a matrix for engineering heart tissue. ACTA ACUST UNITED AC 2013. [DOI: 10.4236/jbise.2013.612147] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|