1
|
Kumar N, Prakash PG, Wentland C, Kurian SM, Jethva G, Brinkmann V, Mollenkopf HJ, Krammer T, Toussaint C, Saliba AE, Biebl M, Jürgensen C, Wiedenmann B, Meyer TF, Gurumurthy RK, Chumduri C. Decoding spatiotemporal transcriptional dynamics and epithelial fibroblast crosstalk during gastroesophageal junction development through single cell analysis. Nat Commun 2024; 15:3064. [PMID: 38594232 PMCID: PMC11004180 DOI: 10.1038/s41467-024-47173-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Accepted: 03/22/2024] [Indexed: 04/11/2024] Open
Abstract
The gastroesophageal squamocolumnar junction (GE-SCJ) is a critical tissue interface between the esophagus and stomach, with significant relevance in the pathophysiology of gastrointestinal diseases. Despite this, the molecular mechanisms underlying GE-SCJ development remain unclear. Using single-cell transcriptomics, organoids, and spatial analysis, we examine the cellular heterogeneity and spatiotemporal dynamics of GE-SCJ development from embryonic to adult mice. We identify distinct transcriptional states and signaling pathways in the epithelial and mesenchymal compartments of the esophagus and stomach during development. Fibroblast-epithelial interactions are mediated by various signaling pathways, including WNT, BMP, TGF-β, FGF, EGF, and PDGF. Our results suggest that fibroblasts predominantly send FGF and TGF-β signals to the epithelia, while epithelial cells mainly send PDGF and EGF signals to fibroblasts. We observe differences in the ligands and receptors involved in cell-cell communication between the esophagus and stomach. Our findings provide insights into the molecular mechanisms underlying GE-SCJ development and fibroblast-epithelial crosstalk involved, paving the way to elucidate mechanisms during adaptive metaplasia development and carcinogenesis.
Collapse
Affiliation(s)
- Naveen Kumar
- Laboratory of Infections, Carcinogenesis and Regeneration, Medical Biotechnology Section, Department of Biological and Chemical Engineering, Aarhus University, Aarhus, Denmark
- Department of Microbiology, University of Würzburg, Würzburg, Germany
| | | | | | | | - Gaurav Jethva
- Department of Microbiology, University of Würzburg, Würzburg, Germany
| | - Volker Brinkmann
- Department of Molecular Biology, Max Planck Institute for Infection Biology, Berlin, Germany
| | - Hans-Joachim Mollenkopf
- Department of Molecular Biology, Max Planck Institute for Infection Biology, Berlin, Germany
| | - Tobias Krammer
- Helmholtz Institute for RNA-based Infection Research (HIRI), Helmholtz-Center for Infection Research (HZI), Würzburg, Germany
| | - Christophe Toussaint
- Helmholtz Institute for RNA-based Infection Research (HIRI), Helmholtz-Center for Infection Research (HZI), Würzburg, Germany
| | - Antoine-Emmanuel Saliba
- Helmholtz Institute for RNA-based Infection Research (HIRI), Helmholtz-Center for Infection Research (HZI), Würzburg, Germany
- University of Würzburg, Faculty of Medicine, Institute of Molecular Infection Biology (IMIB), Würzburg, Germany
| | - Matthias Biebl
- Surgical Clinic Campus Charité Mitte, Charité University Medicine, Berlin, Germany
| | - Christian Jürgensen
- Department of Hepatology and Gastroenterology, Charité University Medicine, Berlin, Germany
| | - Bertram Wiedenmann
- Department of Hepatology and Gastroenterology, Charité University Medicine, Berlin, Germany
| | - Thomas F Meyer
- Department of Molecular Biology, Max Planck Institute for Infection Biology, Berlin, Germany
| | - Rajendra Kumar Gurumurthy
- Department of Microbiology, University of Würzburg, Würzburg, Germany
- Department of Molecular Biology, Max Planck Institute for Infection Biology, Berlin, Germany
| | - Cindrilla Chumduri
- Laboratory of Infections, Carcinogenesis and Regeneration, Medical Biotechnology Section, Department of Biological and Chemical Engineering, Aarhus University, Aarhus, Denmark.
- Department of Microbiology, University of Würzburg, Würzburg, Germany.
- Department of Molecular Biology, Max Planck Institute for Infection Biology, Berlin, Germany.
- Department of Hepatology and Gastroenterology, Charité University Medicine, Berlin, Germany.
| |
Collapse
|
2
|
Shi DL. Planar cell polarity regulators in asymmetric organogenesis during development and disease. J Genet Genomics 2023; 50:63-76. [PMID: 35809777 DOI: 10.1016/j.jgg.2022.06.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 06/22/2022] [Accepted: 06/23/2022] [Indexed: 12/22/2022]
Abstract
The phenomenon of planar cell polarity is critically required for a myriad of morphogenetic processes in metazoan and is accurately controlled by several conserved modules. Six "core" proteins, including Frizzled, Flamingo (Celsr), Van Gogh (Vangl), Dishevelled, Prickle, and Diego (Ankrd6), are major components of the Wnt/planar cell polarity pathway. The Fat/Dchs protocadherins and the Scrib polarity complex also function to instruct cellular polarization. In vertebrates, all these pathways are essential for tissue and organ morphogenesis, such as neural tube closure, left-right symmetry breaking, heart and gut morphogenesis, lung and kidney branching, stereociliary bundle orientation, and proximal-distal limb elongation. Mutations in planar polarity genes are closely linked to various congenital diseases. Striking advances have been made in deciphering their contribution to the establishment of spatially oriented pattern in developing organs and the maintenance of tissue homeostasis. The challenge remains to clarify the complex interplay of different polarity pathways in organogenesis and the link of cell polarity to cell fate specification. Interdisciplinary approaches are also important to understand the roles of mechanical forces in coupling cellular polarization and differentiation. This review outlines current advances on planar polarity regulators in asymmetric organ formation, with the aim to identify questions that deserve further investigation.
Collapse
Affiliation(s)
- De-Li Shi
- Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524001, China; Laboratory of Developmental Biology, CNRS-UMR7622, Institut de Biologie Paris-Seine (IBPS), Sorbonne University, 75005 Paris, France.
| |
Collapse
|
3
|
Auger NA, Medina-Feliciano JG, Quispe-Parra DJ, Colón-Marrero S, Ortiz-Zuazaga H, García-Arrarás JE. Characterization and Expression of Holothurian Wnt Signaling Genes during Adult Intestinal Organogenesis. Genes (Basel) 2023; 14:309. [PMID: 36833237 PMCID: PMC9957329 DOI: 10.3390/genes14020309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 01/18/2023] [Accepted: 01/19/2023] [Indexed: 01/27/2023] Open
Abstract
Wnt signaling has been shown to play multiple roles in regenerative processes, one of the most widely studied of which is the regeneration of the intestinal luminal epithelia. Most studies in this area have focused on self-renewal of the luminal stem cells; however, Wnt signaling may also have more dynamic functions, such as facilitating intestinal organogenesis. To explore this possibility, we employed the sea cucumber Holothuria glaberrima that can regenerate a full intestine over the course of 21 days after evisceration. We collected RNA-seq data from various intestinal tissues and regeneration stages and used these data to define the Wnt genes present in H. glaberrima and the differential gene expression (DGE) patterns during the regenerative process. Twelve Wnt genes were found, and their presence was confirmed in the draft genome of H. glaberrima. The expressions of additional Wnt-associated genes, such as Frizzled and Disheveled, as well as genes from the Wnt/β-catenin and Wnt/Planar Cell Polarity (PCP) pathways, were also analyzed. DGE showed unique distributions of Wnt in early- and late-stage intestinal regenerates, consistent with the Wnt/β-catenin pathway being upregulated during early-stages and the Wnt/PCP pathway being upregulated during late-stages. Our results demonstrate the diversity of Wnt signaling during intestinal regeneration, highlighting possible roles in adult organogenesis.
Collapse
Affiliation(s)
- Noah A. Auger
- Department of Biology, University of Puerto Rico, Rio Piedras Campus, San Juan 00925, Puerto Rico
| | | | - David J. Quispe-Parra
- Department of Biology, University of Puerto Rico, Rio Piedras Campus, San Juan 00925, Puerto Rico
| | - Stephanie Colón-Marrero
- Department of Biology, University of Puerto Rico, Rio Piedras Campus, San Juan 00925, Puerto Rico
| | - Humberto Ortiz-Zuazaga
- Department of Computer Science, University of Puerto Rico, Rio Piedras Campus, San Juan 00925, Puerto Rico
| | - José E. García-Arrarás
- Department of Biology, University of Puerto Rico, Rio Piedras Campus, San Juan 00925, Puerto Rico
| |
Collapse
|
4
|
Shi W, Filmus J. Glypican-6 and Glypican-4 stimulate embryonic stomach growth by regulating Hedgehog and noncanonical Wnt signaling. Dev Dyn 2022; 251:2015-2028. [PMID: 36057966 DOI: 10.1002/dvdy.533] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 07/28/2022] [Accepted: 08/15/2022] [Indexed: 01/30/2023] Open
Abstract
BACKGROUND Glypicans are a family of proteoglycans that play important roles in embryonic morphogenesis. The mammalian genome contains six glypicans (GPC1 to GPC6). GPC6 and GPC4 are the pair of glypicans that show the highest degree of homology within the family. GPC6-null embryos display bone abnormalities and severely shortened intestines. RESULTS We show that GPC6-null embryos display significantly smaller stomachs, and that Hedgehog and noncanonical Wnt signaling are dysregulated in GPC6-null stomachs. Like GPC6, GPC4 is expressed by the developing stomach. However, GPC4-null embryos have normal stomachs. To investigate whether GPC6 and GPC4 display functional overlap in the developing stomach, we crossed GPC4-null mice with GPC6 conditional mutants in which the expression of this glypican is severely reduced in the stomach. Notably, we found that the compound mutants display stomachs that are smaller than those of the GPC6 conditional mutants. We also found that this functional overlap between GPC6 and GPC4 is mediated by the noncanonical Wnt pathway. CONCLUSION This study demonstrates that GPC6 stimulates the growth of the embryonic stomach via Wnt and Hh signaling. In addition, we uncovered a Wnt-mediated functional overlap between GPC6 and GPC4 in the developing stomach.
Collapse
Affiliation(s)
- Wen Shi
- Department of Medical Biophysics, Sunnybrook Research Institute, University of Toronto, Toronto, Ontario, Canada
| | - Jorge Filmus
- Department of Medical Biophysics, Sunnybrook Research Institute, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
5
|
Nawae W, Sonthirod C, Yoocha T, Waiyamitra P, Soisook P, Tangphatsornruang S, Pootakham W. Genome assembly of the Pendlebury's roundleaf bat, Hipposideros pendleburyi, revealed the expansion of Tc1/Mariner DNA transposons in Rhinolophoidea. DNA Res 2022; 29:dsac026. [PMID: 36214371 PMCID: PMC9549598 DOI: 10.1093/dnares/dsac026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Indexed: 11/16/2022] Open
Abstract
Bats (Chiroptera) constitute the second largest order of mammals and have several distinctive features, such as true self-powered flight and strong immunity. The Pendlebury's roundleaf bat, Hipposideros pendleburyi, is endemic to Thailand and listed as a vulnerable species. We employed the 10× Genomics linked-read technology to obtain a genome assembly of H. pendleburyi. The assembly size was 2.17 Gb with a scaffold N50 length of 15,398,518 bases. Our phylogenetic analysis placed H. pendleburyi within the rhinolophoid clade of the suborder Yinpterochiroptera. A synteny analysis showed that H. pendleburyi shared conserved chromosome segments (up to 105 Mb) with Rhinolophus ferrumequinum and Phyllostomus discolor albeit having different chromosome numbers and belonging different families. We found positive selection signals in genes involved in inflammation, spermatogenesis and Wnt signalling. The analyses of transposable elements suggested the contraction of short interspersed nuclear elements (SINEs) and the accumulation of young mariner DNA transposons in the analysed hipposiderids. Distinct mariners were likely horizontally transferred to hipposiderid genomes over the evolution of this family. The lineage-specific profiles of SINEs and mariners might involve in the evolution of hipposiderids and be associated with the phylogenetic separations of these bats from other bat families.
Collapse
Affiliation(s)
- Wanapinun Nawae
- National Omics Center (NOC), National Science and Technology Development Agency, Thailand Science Park, Pathum Thani, Thailand
| | - Chutima Sonthirod
- National Omics Center (NOC), National Science and Technology Development Agency, Thailand Science Park, Pathum Thani, Thailand
| | - Thippawan Yoocha
- National Omics Center (NOC), National Science and Technology Development Agency, Thailand Science Park, Pathum Thani, Thailand
| | - Pitchaporn Waiyamitra
- National Omics Center (NOC), National Science and Technology Development Agency, Thailand Science Park, Pathum Thani, Thailand
| | - Pipat Soisook
- Princess Maha Chakri Sirindhorn Natural History Museum, Prince of Songkla University, Hat Yai, Thailand
| | - Sithichoke Tangphatsornruang
- National Omics Center (NOC), National Science and Technology Development Agency, Thailand Science Park, Pathum Thani, Thailand
| | - Wirulda Pootakham
- National Omics Center (NOC), National Science and Technology Development Agency, Thailand Science Park, Pathum Thani, Thailand
| |
Collapse
|
6
|
Chevalier NR. Physical organogenesis of the gut. Development 2022; 149:276365. [DOI: 10.1242/dev.200765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
ABSTRACT
The gut has been a central subject of organogenesis since Caspar Friedrich Wolff’s seminal 1769 work ‘De Formatione Intestinorum’. Today, we are moving from a purely genetic understanding of cell specification to a model in which genetics codes for layers of physical–mechanical and electrical properties that drive organogenesis such that organ function and morphogenesis are deeply intertwined. This Review provides an up-to-date survey of the extrinsic and intrinsic mechanical forces acting on the embryonic vertebrate gut during development and of their role in all aspects of intestinal morphogenesis: enteric nervous system formation, epithelium structuring, muscle orientation and differentiation, anisotropic growth and the development of myogenic and neurogenic motility. I outline numerous implications of this biomechanical perspective in the etiology and treatment of pathologies, such as short bowel syndrome, dysmotility, interstitial cells of Cajal-related disorders and Hirschsprung disease.
Collapse
Affiliation(s)
- Nicolas R. Chevalier
- Laboratoire Matière et Systèmes Complexes, Université Paris Cité, CNRS UMR 7057 , 10 rue Alice Domon et Léonie Duquet, 75013 Paris , France
| |
Collapse
|
7
|
Zhao L, Song W, Chen YG. Mesenchymal-epithelial interaction regulates gastrointestinal tract development in mouse embryos. Cell Rep 2022; 40:111053. [PMID: 35830795 DOI: 10.1016/j.celrep.2022.111053] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Revised: 03/31/2022] [Accepted: 06/14/2022] [Indexed: 01/10/2023] Open
Abstract
After gut tube patterning in early embryos, the cellular and molecular changes of developing stomach and intestine remain largely unknown. Here, combining single-cell RNA sequencing and spatial RNA sequencing, we construct a spatiotemporal transcriptomic landscape of the mouse stomach and intestine during embryonic days E9.5-E15.5. Several subpopulations are identified, including Lox+ stomach mesenchyme, Aldh1a3+ small-intestinal mesenchyme, and Adamdec1+ large-intestinal mesenchyme. The regionalization and heterogeneity of both the epithelium and the mesenchyme can be traced back to E9.5. The spatiotemporal distributions of cell clusters and the mesenchymal-epithelial interaction analysis indicate that a coordinated development of the epithelium and mesenchyme contribute to the stomach regionalization, intestine segmentation, and villus formation. Using the gut tube-derived organoids, we find that the cell fate of the foregut and hindgut can be switched by the regional niche factors, including fibroblast growth factors (FGFs) and retinoic acid (RA). This work lays a foundation for further dissection of the mechanisms governing this process.
Collapse
Affiliation(s)
- Lianzheng Zhao
- The State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Wanlu Song
- The State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Ye-Guang Chen
- The State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China; Guangzhou Laboratory, Guangzhou, China.
| |
Collapse
|
8
|
Balaraju AK, Hu B, Rodriguez JJ, Murry M, Lin F. Glypican 4 regulates planar cell polarity of endoderm cells by controlling the localization of Cadherin 2. Development 2021; 148:dev199421. [PMID: 34131730 PMCID: PMC8313861 DOI: 10.1242/dev.199421] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 06/09/2021] [Indexed: 11/20/2022]
Abstract
Noncanonical Wnt/planar cell polarity (Wnt/PCP) signaling has been implicated in endoderm morphogenesis. However, the underlying cellular and molecular mechanisms of this process are unclear. We found that, during convergence and extension (C&E) in zebrafish, gut endodermal cells are polarized mediolaterally, with GFP-Vangl2 enriched at the anterior edges. Endoderm cell polarity is lost and intercalation is impaired in the absence of glypican 4 (gpc4), a heparan-sulfate proteoglycan that promotes Wnt/PCP signaling, suggesting that this signaling is required for endodermal cell polarity. Live imaging revealed that endoderm C&E is accomplished by polarized cell protrusions and junction remodeling, which are impaired in gpc4-deficient endodermal cells. Furthermore, in the absence of gpc4, Cadherin 2 expression on the endodermal cell surface is increased as a result of impaired Rab5c-mediated endocytosis, which partially accounts for the endodermal defects in these mutants. These findings indicate that Gpc4 regulates endodermal planar cell polarity during endoderm C&E by influencing the localization of Cadherin 2. Thus, our study uncovers a new mechanism by which Gpc4 regulates planar cell polarity and reveals the role of Wnt/PCP signaling in endoderm morphogenesis.
Collapse
Affiliation(s)
| | | | | | | | - Fang Lin
- Department of Anatomy and Cell Biology, Carver College of Medicine, The University of Iowa, Iowa City, IA 52242, USA
| |
Collapse
|
9
|
Christidi E, Brunham LR. Regulated cell death pathways in doxorubicin-induced cardiotoxicity. Cell Death Dis 2021; 12:339. [PMID: 33795647 PMCID: PMC8017015 DOI: 10.1038/s41419-021-03614-x] [Citation(s) in RCA: 298] [Impact Index Per Article: 99.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 03/10/2021] [Accepted: 03/11/2021] [Indexed: 02/01/2023]
Abstract
Doxorubicin is a chemotherapeutic drug used for the treatment of various malignancies; however, patients can experience cardiotoxic effects and this has limited the use of this potent drug. The mechanisms by which doxorubicin kills cardiomyocytes has been elusive and despite extensive research the exact mechanisms remain unknown. This review focuses on recent advances in our understanding of doxorubicin induced regulated cardiomyocyte death pathways including autophagy, ferroptosis, necroptosis, pyroptosis and apoptosis. Understanding the mechanisms by which doxorubicin leads to cardiomyocyte death may help identify novel therapeutic agents and lead to more targeted approaches to cardiotoxicity testing.
Collapse
Affiliation(s)
- Effimia Christidi
- grid.17091.3e0000 0001 2288 9830Centre for Heart Lung Innovation, Department of Medicine, University of British Columbia, Vancouver, BC Canada
| | - Liam R. Brunham
- grid.17091.3e0000 0001 2288 9830Centre for Heart Lung Innovation, Department of Medicine, University of British Columbia, Vancouver, BC Canada ,grid.17091.3e0000 0001 2288 9830Department of Medicine, University of British Columbia, Vancouver, BC Canada ,grid.17091.3e0000 0001 2288 9830Department of Medical Genetics, University of British Columbia, Vancouver, BC Canada
| |
Collapse
|
10
|
Ichinose M, Suzuki N, Wang T, Wright JA, Lannagan TRM, Vrbanac L, Kobayashi H, Gieniec KA, Ng JQ, Hayakawa Y, García-Gallastegui P, Monsalve EM, Bauer SR, Laborda J, García-Ramírez JJ, Ibarretxe G, Worthley DL, Woods SL. Stromal DLK1 promotes proliferation and inhibits differentiation of the intestinal epithelium during development. Am J Physiol Gastrointest Liver Physiol 2021; 320:G506-G520. [PMID: 33470182 DOI: 10.1152/ajpgi.00445.2020] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Accepted: 01/14/2021] [Indexed: 01/31/2023]
Abstract
The stem/progenitor cells of the developing intestine are biologically distinct from their adult counterparts. Here, we examine the microenvironmental cues that regulate the embryonic stem/progenitor population, focusing on the role of Notch pathway factor delta-like protein-1 (DLK1). mRNA-seq analyses of intestinal mesenchymal cells (IMCs) collected from embryonic day 14.5 (E14.5) or adult IMCs and a novel coculture system with E14.5 intestinal epithelial organoids were used. Following addition of recombinant DLK1 (rDLK) or Dlk1 siRNA (siDlk1), epithelial characteristics were compared using imaging, replating efficiency assays, qPCR, and immunocytochemistry. The intestinal phenotypes of littermate Dlk1+/+ and Dlk1-/- mice were compared using immunohistochemistry. Using transcriptomic analyses, we identified morphogens derived from the embryonic mesenchyme that potentially regulate the developing epithelial cells, to focus on Notch family candidate DLK1. Immunohistochemistry indicated that DLK1 was expressed exclusively in the intestinal stroma at E14.5 at the top of emerging villi, decreased after birth, and shifted to the intestinal epithelium in adulthood. In coculture experiments, addition of rDLK1 to adult IMCs inhibited organoid differentiation, whereas Dlk1 knockdown in embryonic IMCs increased epithelial differentiation to secretory lineage cells. Dlk1-/- mice had restricted Ki67+ cells in the villi base and increased secretory lineage cells compared with Dlk1+/+ embryos. Mesenchyme-derived DLK1 plays an important role in the promotion of epithelial stem/precursor expansion and prevention of differentiation to secretory lineages in the developing intestine.NEW & NOTEWORTHY Using a novel coculture system, transcriptomics, and transgenic mice, we investigated differential molecular signaling between the intestinal epithelium and mesenchyme during development and in the adult. We show that the Notch pathway factor delta-like protein-1 (DLK1) is stromally produced during development and uncover a new role for DLK1 in the regulation of intestinal epithelial stem/precursor expansion and differentiation to secretory lineages.
Collapse
Affiliation(s)
- Mari Ichinose
- School of Medicine, The University of Adelaide, School of Medicine, The University of Adelaide, Adelaide, South Australia, Australia
- South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
| | - Nobumi Suzuki
- School of Medicine, The University of Adelaide, School of Medicine, The University of Adelaide, Adelaide, South Australia, Australia
- South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
- Department of Gastroenterology, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| | - Tongtong Wang
- School of Medicine, The University of Adelaide, School of Medicine, The University of Adelaide, Adelaide, South Australia, Australia
- South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
| | - Josephine A Wright
- School of Medicine, The University of Adelaide, School of Medicine, The University of Adelaide, Adelaide, South Australia, Australia
- South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
| | - Tamsin R M Lannagan
- School of Medicine, The University of Adelaide, School of Medicine, The University of Adelaide, Adelaide, South Australia, Australia
- South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
| | - Laura Vrbanac
- School of Medicine, The University of Adelaide, School of Medicine, The University of Adelaide, Adelaide, South Australia, Australia
- South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
| | - Hiroki Kobayashi
- School of Medicine, The University of Adelaide, School of Medicine, The University of Adelaide, Adelaide, South Australia, Australia
- South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
| | - Krystyna A Gieniec
- School of Medicine, The University of Adelaide, School of Medicine, The University of Adelaide, Adelaide, South Australia, Australia
- South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
| | - Jia Q Ng
- School of Medicine, The University of Adelaide, School of Medicine, The University of Adelaide, Adelaide, South Australia, Australia
- South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
| | - Yoku Hayakawa
- Department of Gastroenterology, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| | - Patricia García-Gallastegui
- Department of Cell Biology and Histology, Faculty of Medicine and Nursing, University of the Basque Country, Bizkaia, Spain
| | - Eva M Monsalve
- Department of Inorganic and Organic Chemistry and Biochemistry, Medical School, Regional Center for Biomedical Research, University of Castilla-La Mancha, Albacete, Spain
| | - Steven R Bauer
- Division of Cellular and Gene Therapies, Center for Biologics Evaluation and Research, United States Food and Drug Administration, Silver Spring, Maryland
| | - Jorge Laborda
- Department of Inorganic and Organic Chemistry and Biochemistry, Medical School, Regional Center for Biomedical Research, University of Castilla-La Mancha, Albacete, Spain
| | - J J García-Ramírez
- Department of Inorganic and Organic Chemistry and Biochemistry, Medical School, Regional Center for Biomedical Research, University of Castilla-La Mancha, Albacete, Spain
| | - Gaskon Ibarretxe
- Department of Cell Biology and Histology, Faculty of Medicine and Nursing, University of the Basque Country, Bizkaia, Spain
| | - Daniel L Worthley
- South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
| | - Susan L Woods
- School of Medicine, The University of Adelaide, School of Medicine, The University of Adelaide, Adelaide, South Australia, Australia
- South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
| |
Collapse
|
11
|
Hung YH, Huang S, Dame MK, Yu Q, Yu QC, Zeng YA, Camp JG, Spence JR, Sethupathy P. Chromatin regulatory dynamics of early human small intestinal development using a directed differentiation model. Nucleic Acids Res 2021; 49:726-744. [PMID: 33406262 PMCID: PMC7826262 DOI: 10.1093/nar/gkaa1204] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 11/20/2020] [Accepted: 12/01/2020] [Indexed: 02/06/2023] Open
Abstract
The establishment of the small intestinal (SI) lineage during human embryogenesis ensures functional integrity of the intestine after birth. The chromatin dynamics that drive SI lineage formation and regional patterning in humans are essentially unknown. To fill this knowledge void, we apply a cutting-edge genomic technology to a state-of-the-art human model of early SI development. Specifically, we leverage chromatin run-on sequencing (ChRO-seq) to define the landscape of active promoters, enhancers and gene bodies across distinct stages of directed differentiation of human pluripotent stem cells into SI spheroids with regional specification. Through comprehensive ChRO-seq analysis we identify candidate stage-specific chromatin activity states, novel markers and enhancer hotspots during the directed differentiation. Moreover, we propose a detailed transcriptional network associated with SI lineage formation or regional patterning. Our ChRO-seq analyses uncover a previously undescribed pattern of enhancer activity and transcription at HOX gene loci underlying SI regional patterning. We also validated this unique HOX dynamics by the analysis of single cell RNA-seq data from human fetal SI. Overall, the results lead to a new proposed working model for the regulatory underpinnings of human SI development, thereby adding a novel dimension to the literature that has relied almost exclusively on non-human models.
Collapse
Affiliation(s)
- Yu-Han Hung
- Department of Biomedical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853, USA
| | - Sha Huang
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA.,Department of Internal Medicine, Division of Gastroenterology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Michael K Dame
- Department of Internal Medicine, Division of Gastroenterology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Qianhui Yu
- Institute of Molecular and Clinical Ophthalmology Basal, Basel 4056, Switzerland
| | - Qing C Yu
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Yi A Zeng
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, 200031, China
| | - J Gray Camp
- Institute of Molecular and Clinical Ophthalmology Basal, Basel 4056, Switzerland.,Department of Ophthalmology, University of Basel, Basel 4001, Switzerland
| | - Jason R Spence
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA.,Department of Internal Medicine, Division of Gastroenterology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Praveen Sethupathy
- Department of Biomedical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853, USA
| |
Collapse
|
12
|
Grzymkowski J, Wyatt B, Nascone-Yoder N. The twists and turns of left-right asymmetric gut morphogenesis. Development 2020; 147:147/19/dev187583. [PMID: 33046455 DOI: 10.1242/dev.187583] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Many organs develop left-right asymmetric shapes and positions that are crucial for normal function. Indeed, anomalous laterality is associated with multiple severe birth defects. Although the events that initially orient the left-right body axis are beginning to be understood, the mechanisms that shape the asymmetries of individual organs remain less clear. Here, we summarize new evidence challenging century-old ideas about the development of stomach and intestine laterality. We compare classical and contemporary models of asymmetric gut morphogenesis and highlight key unanswered questions for future investigation.
Collapse
Affiliation(s)
- Julia Grzymkowski
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC 27607, USA
| | - Brent Wyatt
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC 27607, USA
| | - Nanette Nascone-Yoder
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC 27607, USA
| |
Collapse
|
13
|
Hu YH, Liu J, Lu J, Wang PX, Chen JX, Guo Y, Han FH, Wang JJ, Li W, Liu PQ. sFRP1 protects H9c2 cardiac myoblasts from doxorubicin-induced apoptosis by inhibiting the Wnt/PCP-JNK pathway. Acta Pharmacol Sin 2020; 41:1150-1157. [PMID: 32238888 PMCID: PMC7608092 DOI: 10.1038/s41401-020-0364-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2019] [Revised: 01/08/2020] [Accepted: 01/09/2020] [Indexed: 12/27/2022] Open
Abstract
Doxorubicin (Dox) is an effective chemotherapy drug against a wide range of cancers, including both hematological and solid tumors. However, the serious cardiotoxic effect restricted its clinical application. We previously have illuminated the protective role of canonical Wnt/β-catenin signaling in Dox-induced cardiotoxicity. Secreted frizzled-related protein 1 (sFRP1) is one of the endogenous inhibitors of both canonical and noncanonical Wnt signaling. In this study, we investigated the relationship between sFRP1 and noncanonical Wnt/PCP-JNK (Wnt/planar cell polarity-c-Jun N-terminal kinase) pathway in Dox-induced cardiotoxicity in vitro and in vivo. We showed that treatment of H9c2 cardiac myoblasts with Dox (1 μM) time-dependently suppressed cell viability accompanied by significantly decreased sFRP1 protein level and increased Wnt/PCP-JNK signaling. Pretreatment with SP600125, the Wnt/PCP-JNK signaling inhibitor, attenuated Dox-induced apoptosis of H9c2 cells. Overexpression of sFRP1 protected H9c2 cells from Dox-induced apoptosis by inhibiting the Wnt/PCP-JNK pathway. After intraperitoneal injection of a cumulative dose of 15 mg/kg Dox, rats displayed significant cardiac dysfunction; their heart showed inhibited Wnt/β-catenin signaling and activated Wnt/PCP-JNK signaling. These results suggest that sFRP1 may be a novel target for Dox-induced cardiotoxicity.
Collapse
|
14
|
Kostouros A, Koliarakis I, Natsis K, Spandidos DA, Tsatsakis A, Tsiaoussis J. Large intestine embryogenesis: Molecular pathways and related disorders (Review). Int J Mol Med 2020; 46:27-57. [PMID: 32319546 PMCID: PMC7255481 DOI: 10.3892/ijmm.2020.4583] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Accepted: 04/08/2020] [Indexed: 02/07/2023] Open
Abstract
The large intestine, part of the gastrointestinal tract (GI), is composed of all three germ layers, namely the endoderm, the mesoderm and the ectoderm, forming the epithelium, the smooth muscle layers and the enteric nervous system, respectively. Since gastrulation, these layers develop simultaneously during embryogenesis, signaling to each other continuously until adult age. Two invaginations, the anterior intestinal portal (AIP) and the caudal/posterior intestinal portal (CIP), elongate and fuse, creating the primitive gut tube, which is then patterned along the antero‑posterior (AP) axis and the radial (RAD) axis in the context of left‑right (LR) asymmetry. These events lead to the formation of three distinct regions, the foregut, midgut and hindgut. All the above‑mentioned phenomena are under strict control from various molecular pathways, which are critical for the normal intestinal development and function. Specifically, the intestinal epithelium constitutes a constantly developing tissue, deriving from the progenitor stem cells at the bottom of the intestinal crypt. Epithelial differentiation strongly depends on the crosstalk with the adjacent mesoderm. Major molecular pathways that are implicated in the embryogenesis of the large intestine include the canonical and non‑canonical wingless‑related integration site (Wnt), bone morphogenetic protein (BMP), Notch and hedgehog systems. The aberrant regulation of these pathways inevitably leads to several intestinal malformation syndromes, such as atresia, stenosis, or agangliosis. Novel theories, involving the regulation and homeostasis of intestinal stem cells, suggest an embryological basis for the pathogenesis of colorectal cancer (CRC). Thus, the present review article summarizes the diverse roles of these molecular factors in intestinal embryogenesis and related disorders.
Collapse
Affiliation(s)
- Antonios Kostouros
- Laboratory of Anatomy-Histology-Embryology, Medical School, University of Crete, 71110 Heraklion
| | - Ioannis Koliarakis
- Laboratory of Anatomy-Histology-Embryology, Medical School, University of Crete, 71110 Heraklion
| | - Konstantinos Natsis
- Department of Anatomy and Surgical Anatomy, Medical School, Aristotle University of Thessaloniki, 54124 Thessaloniki
| | | | - Aristidis Tsatsakis
- Laboratory of Toxicology, Medical School, University of Crete, 71409 Heraklion, Greece
| | - John Tsiaoussis
- Laboratory of Anatomy-Histology-Embryology, Medical School, University of Crete, 71110 Heraklion
| |
Collapse
|
15
|
Li X, Ortiz MA, Kotula L. The physiological role of Wnt pathway in normal development and cancer. Exp Biol Med (Maywood) 2020; 245:411-426. [PMID: 31996036 PMCID: PMC7082880 DOI: 10.1177/1535370220901683] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Over the decades, many studies have illustrated the critical roles of Wnt signaling pathways in both developmental processes as well as tumorigenesis. Due to the complexity of Wnt signaling regulation, there are still questions to be addressed about ways cells are able to manipulate different types of Wnt pathways in order to fulfill the requirements for normal or cancer development. In this review, we will describe different types of Wnt signaling pathways and their roles in both normal developmental processes and their role in cancer development and progression. Additionally, we will briefly introduce new strategies currently in clinical trials targeting Wnt signaling pathway components for cancer therapy.
Collapse
Affiliation(s)
- Xiang Li
- Department of Urology, SUNY Upstate Medical University, Syracuse, NY 13210, USA
- Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, NY 13210, USA
- Upstate Cancer Center, SUNY Upstate Medical University, Syracuse, NY 13210, USA
| | - Maria A Ortiz
- Department of Urology, SUNY Upstate Medical University, Syracuse, NY 13210, USA
- Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, NY 13210, USA
- Upstate Cancer Center, SUNY Upstate Medical University, Syracuse, NY 13210, USA
| | - Leszek Kotula
- Department of Urology, SUNY Upstate Medical University, Syracuse, NY 13210, USA
- Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, NY 13210, USA
- Upstate Cancer Center, SUNY Upstate Medical University, Syracuse, NY 13210, USA
| |
Collapse
|
16
|
Sunkara RR, Sarate RM, Setia P, Shah S, Gupta S, Chaturvedi P, Gera P, Waghmare SK. SFRP1 in Skin Tumor Initiation and Cancer Stem Cell Regulation with Potential Implications in Epithelial Cancers. Stem Cell Reports 2020; 14:271-284. [PMID: 31928951 PMCID: PMC7013199 DOI: 10.1016/j.stemcr.2019.12.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2018] [Revised: 12/05/2019] [Accepted: 12/09/2019] [Indexed: 12/30/2022] Open
Abstract
Wnt signaling is involved in the regulation of cancer stem cells (CSCs); however, the molecular mechanism involved is still obscure. SFRP1, a Wnt inhibitor, is downregulated in various human cancers; however, its role in tumor initiation and CSC regulation remains unexplored. Here, we used a skin carcinogenesis model, which showed early tumor initiation in Sfrp1−/− (Sfrp1 knockout) mice and increased tumorigenic potential of Sfrp1−/− CSCs. Expression profiling on Sfrp1−/− CSCs showed upregulation of genes involved in epithelial to mesenchymal transition, stemness, proliferation, and metastasis. Further, SOX-2 and SFRP1 expression was validated in human skin cutaneous squamous cell carcinoma, head and neck squamous cell carcinoma, and breast cancer. The data showed downregulation of SFRP1 and upregulation of SOX-2, establishing their inverse correlation. Importantly, we broadly uncover an inverse correlation of SFRP1 and SOX-2 in epithelial cancers that may be used as a potential prognostic marker in the management of cancer. Loss of Sfrp1 accelerates murine skin tumor initiation and SCC progression Sfrp1 loss enhances in vivo tumorigenic potential of murine skin CSCs We found enhanced EMT and Sox-2 in Sfrp1−/− murine skin SCC Sfrp1 and Sox-2 are inversely correlated in multiple human epithelial cancers
Collapse
Affiliation(s)
- Raghava R Sunkara
- Stem Cell Biology Group, Waghmare Lab, Cancer Research Institute, Advanced Centre for Treatment Research and Education in Cancer (ACTREC), Tata Memorial Centre, Kharghar, Navi Mumbai, Maharashtra 410210, India; Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai 400085, India
| | - Rahul M Sarate
- Stem Cell Biology Group, Waghmare Lab, Cancer Research Institute, Advanced Centre for Treatment Research and Education in Cancer (ACTREC), Tata Memorial Centre, Kharghar, Navi Mumbai, Maharashtra 410210, India
| | - Priyanka Setia
- Stem Cell Biology Group, Waghmare Lab, Cancer Research Institute, Advanced Centre for Treatment Research and Education in Cancer (ACTREC), Tata Memorial Centre, Kharghar, Navi Mumbai, Maharashtra 410210, India
| | - Sanket Shah
- Epigenetics and Chromatin Biology Group, Gupta Lab, Cancer Research Institute, Advanced Centre for Treatment Research and Education in Cancer (ACTREC), Tata Memorial Centre, Kharghar, Navi Mumbai, Maharashtra 410210, India; Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai 400085, India
| | - Sanjay Gupta
- Epigenetics and Chromatin Biology Group, Gupta Lab, Cancer Research Institute, Advanced Centre for Treatment Research and Education in Cancer (ACTREC), Tata Memorial Centre, Kharghar, Navi Mumbai, Maharashtra 410210, India; Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai 400085, India
| | | | - Poonam Gera
- Cancer Research Institute, ACTREC, Tata Memorial Centre, Kharghar, Navi Mumbai, Maharashtra 410210, India
| | - Sanjeev K Waghmare
- Stem Cell Biology Group, Waghmare Lab, Cancer Research Institute, Advanced Centre for Treatment Research and Education in Cancer (ACTREC), Tata Memorial Centre, Kharghar, Navi Mumbai, Maharashtra 410210, India; Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai 400085, India.
| |
Collapse
|
17
|
Shi W, Kaneiwa T, Cydzik M, Gariepy J, Filmus J. Glypican-6 stimulates intestinal elongation by simultaneously regulating Hedgehog and non-canonical Wnt signaling. Matrix Biol 2019; 88:19-32. [PMID: 31756413 DOI: 10.1016/j.matbio.2019.11.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Revised: 11/11/2019] [Accepted: 11/11/2019] [Indexed: 01/11/2023]
Abstract
We report here that Glypican-6 (GPC6)-null mice display at birth small intestines that are 75% shorter than those of normal littermates. Notably, we demonstrate that the role of GPC6 in intestinal elongation is mediated by both Hedgehog (Hh) and non-canonical Wnt signaling. Based on results from in vitro experiments, we had previously proposed that GPC6 stimulates Hh signaling by interacting with Hh and Patched1 (Ptc1), and facilitating/stabilizing their interaction. Here we provide strong support to this hypothesis by showing that GPC6 binds to Ptc1 in the mesenchymal layer of embryonic intestines. This study also provides experimental evidence that strongly suggests that GPC6 inhibits the activity of Wnt5a on the intestinal epithelium by binding to this growth factor, and reducing its release from the surrounding mesenchymal cells. Finally, we show that whereas the mesenchymal layer of GPC6-null intestines displays reduced cell proliferation and a thinner smooth muscle layer, epithelial cell differentiation is not altered in the mutant gut.
Collapse
Affiliation(s)
- Wen Shi
- Sunnybrook Research Institute and Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Tomoyuki Kaneiwa
- Sunnybrook Research Institute and Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Marzena Cydzik
- Sunnybrook Research Institute and Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Jean Gariepy
- Sunnybrook Research Institute and Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Jorge Filmus
- Sunnybrook Research Institute and Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
18
|
Peng JX, Liang SY, Li L. sFRP1 exerts effects on gastric cancer cells through GSK3β/Rac1‑mediated restraint of TGFβ/Smad3 signaling. Oncol Rep 2018; 41:224-234. [PMID: 30542739 PMCID: PMC6278527 DOI: 10.3892/or.2018.6838] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Accepted: 10/11/2018] [Indexed: 02/07/2023] Open
Abstract
Secreted frizzled-related protein 1 (sFRP1) is an inhibitor of canonical Wnt signaling; however, previous studies have determined a tumor-promoting function of sFRP1 in a number of different cancer types. A previous study demonstrated that sFRP1 overexpression was associated with an aggressive phenotype and the activation of transforming growth factor β (TGFβ) signaling. sFRP1 overexpression and sFRP1 knockdown cell models were established. Immunoblotting was conducted to examine the protein levels of the associated molecules. Immunofluorescence staining followed by confocal microscopy was performed to visualize the cytoskeleton alterations and subcellular localization of key proteins. sFRP1 overexpression restored glycogen synthase kinase 3β (GSK3β) activity, which activated Rac family small GTPase 1 (Rac1). GSK3β and Rac1 mediated the effect of sFRP1 on the positive regulation of cell growth and migration/invasion. Inhibition of GSK3β or Rac1 abolished the regulation of sFRP1 on TGFβ/SMAD family member 3 (Smad3) signaling and the aggressive phenotype; however, GSK3β or Rac1 overexpression increased cell migration/invasion and restrained Smad3 activity by preventing its nuclear translocation and limiting its transcriptional activity. The present study demonstrated a tumor-promoting function of sFRP1-overexpression by selectively activating TGFβ signaling in gastric cancer cells. GSK3β and Rac1 serve an important function in mediating the sFRP1-induced malignant alterations and signaling changes.
Collapse
Affiliation(s)
- Ji-Xiang Peng
- Department of Gastrointestinal Surgery, Guangzhou First People's Hospital, The Second Affiliated Hospital of South China University of Technology, Guangzhou, Guangdong 510180, P.R. China
| | - Shun-Yu Liang
- Department of Gastrointestinal Surgery, Guangzhou First Municipal People's Hospital, Affiliated Guangzhou Medical College, Guangzhou, Guangdong 510180, P.R. China
| | - Li Li
- Department of Gastrointestinal Surgery, Guangzhou First People's Hospital, The Second Affiliated Hospital of South China University of Technology, Guangzhou, Guangdong 510180, P.R. China
| |
Collapse
|
19
|
Sivakumar A, Kurpios NA. Transcriptional regulation of cell shape during organ morphogenesis. J Cell Biol 2018; 217:2987-3005. [PMID: 30061107 PMCID: PMC6122985 DOI: 10.1083/jcb.201612115] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Revised: 06/11/2018] [Accepted: 07/17/2018] [Indexed: 02/07/2023] Open
Abstract
The emerging field of transcriptional regulation of cell shape changes aims to address the critical question of how gene expression programs produce a change in cell shape. Together with cell growth, division, and death, changes in cell shape are essential for organ morphogenesis. Whereas most studies of cell shape focus on posttranslational events involved in protein organization and distribution, cell shape changes can be genetically programmed. This review highlights the essential role of transcriptional regulation of cell shape during morphogenesis of the heart, lungs, gastrointestinal tract, and kidneys. We emphasize the evolutionary conservation of these processes across different model organisms and discuss perspectives on open questions and research avenues that may provide mechanistic insights toward understanding birth defects.
Collapse
Affiliation(s)
- Aravind Sivakumar
- Department of Molecular Medicine, College of Veterinary Medicine, Cornell University, Ithaca, NY
| | - Natasza A Kurpios
- Department of Molecular Medicine, College of Veterinary Medicine, Cornell University, Ithaca, NY
| |
Collapse
|
20
|
Planar Cell Polarity Signaling in Mammalian Cardiac Morphogenesis. Pediatr Cardiol 2018; 39:1052-1062. [PMID: 29564519 PMCID: PMC5959767 DOI: 10.1007/s00246-018-1860-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Accepted: 03/06/2018] [Indexed: 01/16/2023]
Abstract
The mammalian heart is the first organ to form and is critical for embryonic survival and development. With an occurrence of 1%, congenital heart defects (CHDs) are also the most common birth defects in humans, and major cause of childhood morbidity and mortality (Hoffman and Kaplan in J Am Coll Cardiol 39(12):1890-1900, 2002; Samanek in Cardiol Young 10(3):179-185, 2000). Understanding how the heart forms will not only help to determine the etiology and to design diagnostic and therapeutic approaches for CHDs, but may also provide insight into regenerative medicine to repair injured adult hearts. Mammalian heart development requires precise orchestration of growth, differentiation, and morphogenesis to remodel a simple linear heart tube into an intricate, four-chambered heart with properly connected pulmonary artery and aorta, a structural basis for establishing the pulmonary and systemic circulation. Here we will review the recent advance in our understanding of how the planar cell polarity pathway, a highly conserved morphogenetic engine in vertebrates, regulates polarized morphogenetic processes to contribute to both the arterial and venous poles development of the heart.
Collapse
|
21
|
Huang M, He H, Belenkaya T, Lin X. Multiple roles of epithelial heparan sulfate in stomach morphogenesis. J Cell Sci 2018; 131:jcs.210781. [PMID: 29700203 DOI: 10.1242/jcs.210781] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Accepted: 04/19/2018] [Indexed: 12/12/2022] Open
Abstract
Heparan sulfate proteoglycans (HSPGs) have been shown to regulate various developmental processes. However, the function of heparan sulfate (HS) during the development of mammalian stomach has not been characterized yet. Here, we investigate the role of epithelial HS in embryonic stomach by examining mice deficient in the glycosyltransferase gene Ext1 We show that HS exhibits a specific and dynamic expression pattern in mouse embryonic stomach. Depletion of the epithelial HS leads to stomach hypoplasia, with phenotypic differences in the gastric mucosa between the forestomach and hindstomach. In the posterior stomach, HS depletion disrupts glandular stomach patterning and cytodifferentiation via attenuation of Fgf signaling activity. Inhibition of Fgf signaling in vitro recapitulates the patterning defect. Ligand and carbohydrate engagement assay (LACE) reveals a diminished assembly of Fgf10 and Fgfr2b in the mutant. In the anterior stomach, loss of epithelial HS leads to stratification and differentiation defects of the multilayered squamous epithelium, along with reduced Hh and Bmp signaling activity. Our data demonstrate that epithelial HS plays multiple roles in regulating mammalian stomach morphogenesis in a regional-specific manner.
Collapse
Affiliation(s)
- Meina Huang
- State Key Laboratory of Genetic Engineering, Institute of Genetics, Collaborative Innovation Center of Genetics and Development, School of Life Sciences, Fudan University, Shanghai 200438, China.,State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China.,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Hua He
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China.,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Tatyana Belenkaya
- Division of Developmental Biology, Perinatal Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Xinhua Lin
- State Key Laboratory of Genetic Engineering, Institute of Genetics, Collaborative Innovation Center of Genetics and Development, School of Life Sciences, Fudan University, Shanghai 200438, China .,Division of Developmental Biology, Perinatal Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| |
Collapse
|
22
|
Wang CP, Yu TH, Wu CC, Hung WC, Hsu CC, Tsai IT, Tang WH, Chung FM, Houng JY, Lee YJ, Lu YC. Circulating secreted frizzled-related protein 5 and chronic kidney disease in patients with acute ST-segment elevation myocardial infarction. Cytokine 2018; 110:367-373. [PMID: 29807686 DOI: 10.1016/j.cyto.2018.04.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Revised: 03/05/2018] [Accepted: 04/07/2018] [Indexed: 12/25/2022]
Abstract
Secreted frizzled-related protein-5 (Sfrp5) known as secreted antagonist binds to Wnt protein. It has been shown to be downregulated by histone acetylation and promoter methylation, and to function as a tumor suppressor gene by inducing apoptosis in renal cell cancer cells. However, its relationship with chronic kidney disease (CKD) has not been well studied. Our objective was to investigate the effect of plasma Sfrp5 levels in subjects with and without CKD. Plasma Sfrp5 levels were determined by enzyme-linked immunosorbent assays in 196 consecutive patients with acute ST-segment elevation myocardial infarction (STEMI). CKD was defined as an estimated glomerular filtration rate (eGFR) <60 ml/min per 1.73 m2. For the purpose of this study, stage 1 or 2 CKD patients (eGFR ≥ 60 ml/min per 1.73 m2) were classified as not having CKD. With increasing Sfrp5 tertiles, the patients had higher frequencies of hypertension, stage 4 or 5 CKD, and waist-to-hip ratio, incrementally lower eGFRs and serum hemoglobin levels, and higher levels of blood urine nitrogen (BUN), creatinine, and adiponectin. Multivariate logistic regression analysis showed that an increased plasma Sfrp5 level was independently associated with CKD for all subjects (adjusted odds ratio (OR), 1.08; 95% confidence interval (CI), 1.02-1.14; p = 0.011). Sfrp5 was also significantly positively related to BUN, creatinine, and adiponectin, and significantly negatively related to eGFR and hemoglobin. When the patients were stratified by age, plasma Sfrp5 level was independently related to CKD for patients >65 years old (adjusted OR, 1.10; 95% CI, 1.00-1.20; p = 0.045), however, the association was not significant for those <65 years old. In addition, Sfrp5 was significantly positively related to BUN, creatinine, and adiponectin, and significantly negatively related to eGFR and hemoglobin in patients >65 years old. Our results suggest that Sfrp5 may play a role in the pathogenesis of CKD in acute STEMI patients who are older than 65 years.
Collapse
Affiliation(s)
- Chao-Ping Wang
- Division of Cardiology, E-Da Hospital, I-Shou University, Kaohsiung 82445, Taiwan; School of Medicine for International Students, Institute of Biotechnology and Chemical Engineering, I-Shou University, Kaohsiung 82445, Taiwan.
| | - Teng-Hung Yu
- Division of Cardiology, E-Da Hospital, I-Shou University, Kaohsiung 82445, Taiwan.
| | - Cheng-Ching Wu
- Division of Cardiology, E-Da Hospital, I-Shou University, Kaohsiung 82445, Taiwan.
| | - Wei-Chin Hung
- Division of Cardiology, E-Da Hospital, I-Shou University, Kaohsiung 82445, Taiwan.
| | - Chia-Chang Hsu
- Division of Gastroenterology and Hepatology, E-Da Hospital, I-Shou University, Kaohsiung 82445, Taiwan.
| | - I-Ting Tsai
- Department of Emergency, E-Da Hospital, I-Shou University, Kaohsiung 82445, Taiwan.
| | - Wei-Hua Tang
- Division of Cardiology, Department of Internal Medicine, National Yang-Ming University Hospital, Yilan 26058, Taiwan.
| | - Fu-Mei Chung
- Division of Cardiology, E-Da Hospital, I-Shou University, Kaohsiung 82445, Taiwan.
| | - Jer-Yiing Houng
- Department of Nutrition, Institute of Biotechnology and Chemical Engineering, I-Shou University, Kaohsiung 82445, Taiwan.
| | - Yau-Jiunn Lee
- Lee's Endocrinologic Clinic, Pingtung 90000, Taiwan.
| | - Yung-Chuan Lu
- Division of Endocrinology and Metabolism, Department of Internal Medicine, E-Da Hospital, I-Shou University, Kaohsiung 82445, Taiwan; School of Medicine for International Students, Institute of Biotechnology and Chemical Engineering, I-Shou University, Kaohsiung 82445, Taiwan.
| |
Collapse
|
23
|
Vincent KM, Postovit LM. Matricellular proteins in cancer: a focus on secreted Frizzled-related proteins. J Cell Commun Signal 2018; 12:103-112. [PMID: 28589318 PMCID: PMC5842174 DOI: 10.1007/s12079-017-0398-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Accepted: 06/02/2017] [Indexed: 12/31/2022] Open
Abstract
Tumours are complex entities, wherein cancer cells interact with myriad soluble, insoluble and cell associated factors. These microenvironmental mediators regulate tumour growth, progression and metastasis, and are produced by cancer cells and by stromal components such as fibroblast, adipocytes and immune cells. Through their ability to bind to extracellular matrix proteins, cell surface receptors and growth factors, matricellular proteins enable a dynamic reciprocity between cancer cells and their microenvironment. Hence, matricellular proteins play a critical role in tumour progression by regulating where and when cancer cells are exposed to key growth factors and regulatory proteins. Recent studies suggest that, in addition to altering Wingless (Wnt) signalling, certain members of the Secreted Frizzled Related Protein (sFRP) family are matricellular in nature. In this review, we outline the importance of matricellular proteins in cancer, and discuss how sFRPs may function to both inhibit and promote cancer progression in a context-dependent manner. By considering the matricellular functionality of sFRPs, we may better understand their apparently paradoxical roles in cancers.
Collapse
Affiliation(s)
- Krista Marie Vincent
- Department of Oncology, Faculty of Medicine and Dentistry, University of Alberta, 114th St and 87th Ave, Edmonton, AB T6G 2E1 Canada
- Department of Anatomy and Cell Biology, Faculty of Medicine and Dentistry, University of Western Ontario, 1151 Richmond St, London, ON N6A 3K7 Canada
| | - Lynne-Marie Postovit
- Department of Oncology, Faculty of Medicine and Dentistry, University of Alberta, 114th St and 87th Ave, Edmonton, AB T6G 2E1 Canada
| |
Collapse
|
24
|
Pickett MA, Dush MK, Nascone-Yoder NM. Acetylcholinesterase plays a non-neuronal, non-esterase role in organogenesis. Development 2017; 144:2764-2770. [PMID: 28684626 DOI: 10.1242/dev.149831] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Accepted: 06/19/2017] [Indexed: 01/10/2023]
Abstract
Acetylcholinesterase (AChE) is crucial for degrading acetylcholine at cholinergic synapses. In vitro studies suggest that, in addition to its role in nervous system signaling, AChE can also modulate non-neuronal cell properties, although it remains controversial whether AChE functions in this capacity in vivo Here, we show that AChE plays an essential non-classical role in vertebrate gut morphogenesis. Exposure of Xenopus embryos to AChE-inhibiting chemicals results in severe defects in intestinal development. Tissue-targeted loss-of-function assays (via microinjection of antisense morpholino or CRISPR-Cas9) confirm that AChE is specifically required in the gut endoderm tissue, a non-neuronal cell population, where it mediates adhesion to fibronectin and regulates cell rearrangement events that drive gut lengthening and digestive epithelial morphogenesis. Notably, the classical esterase activity of AChE is dispensable for this activity. As AChE is deeply conserved, widely expressed outside of the nervous system, and the target of many environmental chemicals, these results have wide-reaching implications for development and toxicology.
Collapse
Affiliation(s)
- Melissa A Pickett
- Department of Biology, Environmental and Molecular Toxicology Program, North Carolina State University, Raleigh, NC 27606, USA
| | - Michael K Dush
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC 27607, USA
| | - Nanette M Nascone-Yoder
- Department of Biology, Environmental and Molecular Toxicology Program, North Carolina State University, Raleigh, NC 27606, USA .,Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC 27607, USA
| |
Collapse
|
25
|
A Wnt5 Activity Asymmetry and Intercellular Signaling via PCP Proteins Polarize Node Cells for Left-Right Symmetry Breaking. Dev Cell 2017; 40:439-452.e4. [PMID: 28292423 DOI: 10.1016/j.devcel.2017.02.010] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2016] [Revised: 12/26/2016] [Accepted: 02/13/2017] [Indexed: 12/17/2022]
Abstract
Polarization of node cells along the anterior-posterior axis of mouse embryos is responsible for left-right symmetry breaking. How node cells become polarized has remained unknown, however. Wnt5a and Wnt5b are expressed posteriorly relative to the node, whereas genes for Sfrp inhibitors of Wnt signaling are expressed anteriorly. Here we show that polarization of node cells is impaired in Wnt5a-/-Wnt5b-/- and Sfrp mutant embryos, and also in the presence of a uniform distribution of Wnt5a or Sfrp1, suggesting that Wnt5 and Sfrp proteins act as instructive signals in this process. The absence of planar cell polarity (PCP) core proteins Prickle1 and Prickle2 in individual cells or local forced expression of Wnt5a perturbed polarization of neighboring wild-type cells. Our results suggest that opposing gradients of Wnt5a and Wnt5b and of their Sfrp inhibitors, together with intercellular signaling via PCP proteins, polarize node cells along the anterior-posterior axis for breaking of left-right symmetry.
Collapse
|
26
|
Agostino M, Pohl SÖG, Dharmarajan A. Structure-based prediction of Wnt binding affinities for Frizzled-type cysteine-rich domains. J Biol Chem 2017; 292:11218-11229. [PMID: 28533339 DOI: 10.1074/jbc.m117.786269] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Revised: 05/09/2017] [Indexed: 11/06/2022] Open
Abstract
Wnt signaling pathways are of significant interest in development and oncogenesis. The first step in these pathways typically involves the binding of a Wnt protein to the cysteine-rich domain (CRD) of a Frizzled receptor. Wnt-Frizzled interactions can be antagonized by secreted Frizzled-related proteins (SFRPs), which also contain a Frizzled-like CRD. The large number of Wnts, Frizzleds, and SFRPs, as well as the hydrophobic nature of Wnt, poses challenges to laboratory-based investigations of interactions involving Wnt. Here, utilizing structural knowledge of a representative Wnt-Frizzled CRD interaction, as well as experimentally determined binding affinities for a selection of Wnt-Frizzled CRD interactions, we generated homology models of Wnt-Frizzled CRD interactions and developed a quantitative structure-activity relationship for predicting their binding affinities. The derived model incorporates a small selection of terms derived from scoring functions used in protein-protein docking, as well as an energetic term considering the contribution made by the lipid of Wnt to the Wnt-Frizzled binding affinity. Validation with an external test set suggests that the model can accurately predict binding affinity for 75% of cases and that the error associated with the predictions is comparable with the experimental error. The model was applied to predict the binding affinities of the full range of mouse and human Wnt-Frizzled and Wnt-SFRP interactions, indicating trends in Wnt binding affinity for Frizzled and SFRP CRDs. The comprehensive predictions made in this study provide the basis for laboratory-based studies of previously unexplored Wnt-Frizzled and Wnt-SFRP interactions, which, in turn, may reveal further Wnt signaling pathways.
Collapse
Affiliation(s)
- Mark Agostino
- From the Stem Cell and Cancer Biology Laboratory, School of Biomedical Sciences and Curtin Health Innovation Research Institute and .,Curtin Institute of Computation, Curtin University, Kent Street, Bentley, Western Australia 6102, Australia
| | - Sebastian Öther-Gee Pohl
- From the Stem Cell and Cancer Biology Laboratory, School of Biomedical Sciences and Curtin Health Innovation Research Institute and
| | - Arun Dharmarajan
- From the Stem Cell and Cancer Biology Laboratory, School of Biomedical Sciences and Curtin Health Innovation Research Institute and
| |
Collapse
|
27
|
Miles LB, Mizoguchi T, Kikuchi Y, Verkade H. A role for planar cell polarity during early endoderm morphogenesis. Biol Open 2017; 6:531-539. [PMID: 28377456 PMCID: PMC5450312 DOI: 10.1242/bio.021899] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The zebrafish endoderm begins to develop at gastrulation stages as a monolayer of cells. The behaviour of the endoderm during gastrulation stages is well understood. However, knowledge of the morphogenic movements of the endoderm during somitogenesis stages, as it forms a mesenchymal rod, is lacking. Here we characterise endodermal development during somitogenesis stages, and describe the morphogenic movements as the endoderm transitions from a monolayer of cells into a mesenchymal endodermal rod. We demonstrate that, unlike the overlying mesoderm, endodermal cells are not polarised during their migration to the midline at early somitogenesis stages. Specifically, we describe the stage at which endodermal cells begin to leave the monolayer, a process we have termed 'midline aggregation'. The planar cell polarity (PCP) signalling pathway is known to regulate mesodermal and ectodermal cell convergence towards the dorsal midline. However, a role for PCP signalling in endoderm migration to the midline during somitogenesis stages has not been established. In this report, we investigate the role for PCP signalling in multiple phases of endoderm development during somitogenesis stages. Our data exclude involvement of PCP signalling in endodermal cells as they leave the monolayer.
Collapse
Affiliation(s)
- Lee B Miles
- School of Biological Sciences, Monash University, Clayton, Victoria 3800, Australia
| | - Takamasa Mizoguchi
- Graduate School of Pharmaceutical sciences, Chiba University, Chuo-ku 260-8675, Japan
| | - Yutaka Kikuchi
- Department of Biological Science, Graduate School of Science, Hiroshima University, Higashi-Hiroshima, Hiroshima 739-8526, Japan
| | - Heather Verkade
- School of Biological Sciences, Monash University, Clayton, Victoria 3800, Australia
| |
Collapse
|
28
|
McCracken KW, Wells JM. Mechanisms of embryonic stomach development. Semin Cell Dev Biol 2017; 66:36-42. [PMID: 28238948 DOI: 10.1016/j.semcdb.2017.02.004] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Accepted: 02/20/2017] [Indexed: 12/18/2022]
Abstract
The stomach is a digestive organ that has important roles in human physiology and pathophysiology. The developmental origin of the stomach is the embryonic foregut, which also gives rise a number of other structures. There are several signaling pathways and transcription factors that are known to regulate stomach development at different stages, including foregut patterning, stomach specification, and gastric regionalization. These developmental events have important implications in later homeostasis and disease in the adult stomach. Here we will review the literature that has shaped our current understanding of the molecular mechanisms that coordinate gastric organogenesis. Further we will discuss how developmental paradigms have guided recent efforts to differentiate stomach tissue from pluripotent stem cells.
Collapse
Affiliation(s)
- Kyle W McCracken
- Division of Developmental Biology, 3333 Burnet Avenue, Cincinnati, OH 45229-3039, USA; Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115, USA.
| | - James M Wells
- Division of Developmental Biology, 3333 Burnet Avenue, Cincinnati, OH 45229-3039, USA; Division of Endocrinology Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229-3039, USA.
| |
Collapse
|
29
|
Chin AM, Hill DR, Aurora M, Spence JR. Morphogenesis and maturation of the embryonic and postnatal intestine. Semin Cell Dev Biol 2017; 66:81-93. [PMID: 28161556 DOI: 10.1016/j.semcdb.2017.01.011] [Citation(s) in RCA: 124] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2016] [Revised: 01/28/2017] [Accepted: 01/30/2017] [Indexed: 12/12/2022]
Abstract
The intestine is a vital organ responsible for nutrient absorption, bile and waste excretion, and a major site of host immunity. In order to keep up with daily demands, the intestine has evolved a mechanism to expand the absorptive surface area by undergoing a morphogenetic process to generate finger-like units called villi. These villi house specialized cell types critical for both absorbing nutrients from food, and for protecting the host from commensal and pathogenic microbes present in the adult gut. In this review, we will discuss mechanisms that coordinate intestinal development, growth, and maturation of the small intestine, starting from the formation of the early gut tube, through villus morphogenesis and into early postnatal life when the intestine must adapt to the acquisition of nutrients through food intake, and to interactions with microbes.
Collapse
Affiliation(s)
- Alana M Chin
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, United States
| | - David R Hill
- Department of Internal Medicine, Division of Gastroenterology, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Megan Aurora
- Department of Pediatrics, Division of Neonatal-Perinatal Medicine, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Jason R Spence
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, United States; Department of Internal Medicine, Division of Gastroenterology, University of Michigan Medical School, Ann Arbor, MI, United States; Center for Organogenesis, University of Michigan Medical School, Ann Arbor, MI, United States.
| |
Collapse
|
30
|
Nigmatullina L, Norkin M, Dzama MM, Messner B, Sayols S, Soshnikova N. Id2 controls specification of Lgr5 + intestinal stem cell progenitors during gut development. EMBO J 2017; 36:869-885. [PMID: 28077488 DOI: 10.15252/embj.201694959] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Revised: 12/08/2016] [Accepted: 12/09/2016] [Indexed: 11/09/2022] Open
Abstract
The adult intestinal stem cells (ISCs), their hierarchies, mechanisms of maintenance and differentiation have been extensively studied. However, when and how ISCs are established during embryogenesis remains unknown. We show here that the transcription regulator Id2 controls the specification of embryonic Lgr5+ progenitors in the developing murine small intestine. Cell fate mapping analysis revealed that Lgr5+ progenitors emerge at E13.5 in wild-type embryos and differ from the rest on the intestinal epithelium by a characteristic ISC signature. In the absence of Id2, the intestinal epithelium differentiates into Lgr5+ cells already at E9.5. Furthermore, the size of the Lgr5+ cell pool is significantly increased. We show that Id2 restricts the activity of the Wnt signalling pathway at early stages and prevents precocious differentiation of the embryonic intestinal epithelium. Id2-deficient embryonic epithelial cells cultured ex vivo strongly activate Wnt target genes as well as markers of neoplastic transformation and form fast growing undifferentiated spheroids. Furthermore, adult ISCs from Id2-deficient mice display a distinct transcriptional signature, supporting an essential role for Id2 in the correct specification of ISCs.
Collapse
|
31
|
Miyoshi H. Wnt-expressing cells in the intestines: guides for tissue remodeling. J Biochem 2016; 161:19-25. [PMID: 28013225 DOI: 10.1093/jb/mvw070] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Accepted: 09/28/2016] [Indexed: 01/07/2023] Open
Abstract
The crypt is a minimal functional unit in the intestinal epithelium. This unique structure is maintained by surrounding mesenchymal cells that focally interact with associated epithelial cells. Canonical and non-canonical Wnt ligands enable specific microenvironments localized to each end of the crypt major axis. While canonical Wnt-expressing cells are localized near the crypt bottom where intestinal stem cells reside, non-canonical Wnt-expressing cells are positioned beneath the luminal surface of epithelial cells. During wound healing, propagation and appropriate relocation of each cell population are thought to ensure subsequent crypt regeneration. In this review, I integrate information from recent studies on Wnt-expressing cells and intestinal fibroblast lineages and discuss their roles in homeostasis and wound healing. More information on the lineages of Wnt-expressing cells will help clarify the mechanisms of epithelial tissue formation.
Collapse
Affiliation(s)
- Hiroyuki Miyoshi
- Division of Experimental Therapeutics, Department of Gastrointestinal Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| |
Collapse
|
32
|
Dass RA, Sarshad AA, Carson BB, Feenstra JM, Kaur A, Obrdlik A, Parks MM, Prakash V, Love DK, Pietras K, Serra R, Blanchard SC, Percipalle P, Brown AMC, Vincent CT. Wnt5a Signals through DVL1 to Repress Ribosomal DNA Transcription by RNA Polymerase I. PLoS Genet 2016; 12:e1006217. [PMID: 27500936 PMCID: PMC4976976 DOI: 10.1371/journal.pgen.1006217] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Accepted: 07/05/2016] [Indexed: 11/19/2022] Open
Abstract
Ribosome biogenesis is essential for cell growth and proliferation and is commonly elevated in cancer. Accordingly, numerous oncogene and tumor suppressor signaling pathways target rRNA synthesis. In breast cancer, non-canonical Wnt signaling by Wnt5a has been reported to antagonize tumor growth. Here, we show that Wnt5a rapidly represses rDNA gene transcription in breast cancer cells and generates a chromatin state with reduced transcription of rDNA by RNA polymerase I (Pol I). These effects were specifically dependent on Dishevelled1 (DVL1), which accumulates in nucleolar organizer regions (NORs) and binds to rDNA regions of the chromosome. Upon DVL1 binding, the Pol I transcription activator and deacetylase Sirtuin 7 (SIRT7) releases from rDNA loci, concomitant with disassembly of Pol I transcription machinery at the rDNA promoter. These findings reveal that Wnt5a signals through DVL1 to suppress rRNA transcription. This provides a novel mechanism for how Wnt5a exerts tumor suppressive effects and why disruption of Wnt5a signaling enhances mammary tumor growth in vivo.
Collapse
Affiliation(s)
- Randall A. Dass
- Department of Cell and Developmental Biology, Weill Cornell Medical College, New York, New York, United States of America
- Department of Physiology and Biophysics, Weill Cornell Medical College, New York, New York, United States of America
| | - Aishe A. Sarshad
- Department of Cell and Molecular Biology, Karolinska Institute, Stockholm, Sweden
- Biology Program, New York University Abu Dhabi, Abu Dhabi, United Arab Emirates
| | - Brittany B. Carson
- Department of Physiology and Pharmacology, Karolinska Institute, Stockholm, Sweden
| | - Jennifer M. Feenstra
- Department of Physiology and Pharmacology, Karolinska Institute, Stockholm, Sweden
| | - Amanpreet Kaur
- Department of Cell and Developmental Biology, Weill Cornell Medical College, New York, New York, United States of America
| | - Ales Obrdlik
- Department of Cell and Molecular Biology, Karolinska Institute, Stockholm, Sweden
- Biology Program, New York University Abu Dhabi, Abu Dhabi, United Arab Emirates
| | - Matthew M. Parks
- Department of Physiology and Biophysics, Weill Cornell Medical College, New York, New York, United States of America
| | - Varsha Prakash
- Department of Physiology and Pharmacology, Karolinska Institute, Stockholm, Sweden
| | - Damon K. Love
- Department of Cell and Developmental Biology, Weill Cornell Medical College, New York, New York, United States of America
| | - Kristian Pietras
- Department of Laboratory Medicine, Center for Molecular Pathology, Lund University, Lund, Sweden
| | - Rosa Serra
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Scott C. Blanchard
- Department of Physiology and Biophysics, Weill Cornell Medical College, New York, New York, United States of America
- Tri-Institutional PhD program in Chemical Biology, Weill Cornell Medical College, New York, New York, United States of America
| | - Piergiorgio Percipalle
- Department of Cell and Molecular Biology, Karolinska Institute, Stockholm, Sweden
- Biology Program, New York University Abu Dhabi, Abu Dhabi, United Arab Emirates
- * E-mail: (PP); (AMCB); (CTV)
| | - Anthony M. C. Brown
- Department of Cell and Developmental Biology, Weill Cornell Medical College, New York, New York, United States of America
- * E-mail: (PP); (AMCB); (CTV)
| | - C. Theresa Vincent
- Department of Cell and Developmental Biology, Weill Cornell Medical College, New York, New York, United States of America
- Department of Physiology and Biophysics, Weill Cornell Medical College, New York, New York, United States of America
- Department of Physiology and Pharmacology, Karolinska Institute, Stockholm, Sweden
- * E-mail: (PP); (AMCB); (CTV)
| |
Collapse
|
33
|
Abstract
The stomach, an organ derived from foregut endoderm, secretes acid and enzymes and plays a key role in digestion. During development, mesenchymal-epithelial interactions drive stomach specification, patterning, differentiation and growth through selected signaling pathways and transcription factors. After birth, the gastric epithelium is maintained by the activity of stem cells. Developmental signals are aberrantly activated and stem cell functions are disrupted in gastric cancer and other disorders. Therefore, a better understanding of stomach development and stem cells can inform approaches to treating these conditions. This Review highlights the molecular mechanisms of stomach development and discusses recent findings regarding stomach stem cells and organoid cultures, and their roles in investigating disease mechanisms.
Collapse
Affiliation(s)
- Tae-Hee Kim
- Program in Developmental and Stem Cell Biology, The Hospital for Sick Children, Toronto, Ontario, Canada M5G 0A4 Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada M5S 1A8
| | - Ramesh A Shivdasani
- Department of Medical Oncology and Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA 02215, USA Department of Medicine, Brigham & Women's Hospital and Harvard Medical School, Boston, MA 02215, USA
| |
Collapse
|
34
|
Abstract
The vertebrate small intestine requires an enormous surface area to effectively absorb nutrients from food. Morphological adaptations required to establish this extensive surface include generation of an extremely long tube and convolution of the absorptive surface of the tube into villi and microvilli. In this Review, we discuss recent findings regarding the morphogenetic and molecular processes required for intestinal tube elongation and surface convolution, examine shared and unique aspects of these processes in different species, relate these processes to known human maladies that compromise absorptive function and highlight important questions for future research.
Collapse
Affiliation(s)
- Katherine D Walton
- Cell and Developmental Biology Department, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Andrew M Freddo
- Cell and Developmental Biology Department, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Sha Wang
- Cell and Developmental Biology Department, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Deborah L Gumucio
- Cell and Developmental Biology Department, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| |
Collapse
|
35
|
Hasebe T, Fujimoto K, Kajita M, Ishizuya-Oka A. Thyroid hormone activates Wnt/β-catenin signaling involved in adult epithelial development during intestinal remodeling in Xenopus laevis. Cell Tissue Res 2016; 365:309-18. [PMID: 27068920 DOI: 10.1007/s00441-016-2396-8] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Accepted: 03/09/2016] [Indexed: 02/08/2023]
Abstract
During amphibian intestinal remodeling, thyroid hormone (TH) induces some larval epithelial cells to dedifferentiate into adult stem cells, which newly generate the absorptive epithelium analogous to the mammalian epithelium. To clarify molecular mechanisms underlying adult epithelial development, we here focus on TH response genes that are associated with the canonical Wnt pathway. Our quantitative reverse transcription plus polymerase chain reaction and immunohistochemical analyses indicate that all of the genes examined, including β-catenin, c-Myc and secreted frizzle-related protein 2 (SFRP2), are up-regulated in Xenopus laevis intestine during both natural and TH-induced metamorphosis. Moreover, immunoreactivity for nuclear β-catenin becomes detectable in adult stem cells from the start of their appearance and then increases in intensity in adult epithelial primordia derived from the stem cells, which actively proliferate and coexpress Wnt target genes c-Myc and LGR5. These expression profiles strongly suggest the involvement of the canonical Wnt pathway in the maintenance and/or proliferation of adult stem/progenitor cells. More importantly, by using organ cultures of the tadpole intestine, we have experimentally shown that the addition of exogenous SFRP2 protein to the culture medium promotes cell proliferation of the adult epithelial primordia, whereas inhibition of endogenous SFRP2 by its antibody suppresses their proliferation. The inhibition of SFRP2 suppresses larval epithelial changes in shape from simple columnar to stem-cell-like roundish cells, resulting in the failure of epithelial dedifferentiation. Thus, TH-up-regulated SFRP2 in the postembryonic intestine promotes adult stem cell development, possibly by acting as an agonist of both canonical and non-canonical Wnt signaling.
Collapse
Affiliation(s)
- Takashi Hasebe
- Department of Biology, Nippon Medical School, 1-7-1 Kyonan-cho, Musashino, Tokyo, 180-0023, Japan
| | - Kenta Fujimoto
- Department of Biology, Nippon Medical School, 1-7-1 Kyonan-cho, Musashino, Tokyo, 180-0023, Japan
| | - Mitsuko Kajita
- Department of Molecular Biology, Institute for Advanced Medical Sciences, Nippon Medical School, Kawasaki, Kanagawa, Japan
| | - Atsuko Ishizuya-Oka
- Department of Biology, Nippon Medical School, 1-7-1 Kyonan-cho, Musashino, Tokyo, 180-0023, Japan.
| |
Collapse
|
36
|
Womble M, Pickett M, Nascone-Yoder N. Frogs as integrative models for understanding digestive organ development and evolution. Semin Cell Dev Biol 2016; 51:92-105. [PMID: 26851628 PMCID: PMC4798877 DOI: 10.1016/j.semcdb.2016.02.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2015] [Accepted: 02/01/2016] [Indexed: 12/16/2022]
Abstract
The digestive system comprises numerous cells, tissues and organs that are essential for the proper assimilation of nutrients and energy. Many aspects of digestive organ function are highly conserved among vertebrates, yet the final anatomical configuration of the gut varies widely between species, especially those with different diets. Improved understanding of the complex molecular and cellular events that orchestrate digestive organ development is pertinent to many areas of biology and medicine, including the regeneration or replacement of diseased organs, the etiology of digestive organ birth defects, and the evolution of specialized features of digestive anatomy. In this review, we highlight specific examples of how investigations using Xenopus laevis frog embryos have revealed insight into the molecular and cellular dynamics of digestive organ patterning and morphogenesis that would have been difficult to obtain in other animal models. Additionally, we discuss recent studies of gut development in non-model frog species with unique feeding strategies, such as Lepidobatrachus laevis and Eleutherodactylous coqui, which are beginning to provide glimpses of the evolutionary mechanisms that may generate morphological variation in the digestive tract. The unparalleled experimental versatility of frog embryos make them excellent, integrative models for studying digestive organ development across multiple disciplines.
Collapse
Affiliation(s)
- Mandy Womble
- Department of Molecular Biomedical Sciences, North Carolina State University, Raleigh, NC 27607, United States
| | - Melissa Pickett
- Department of Molecular Biomedical Sciences, North Carolina State University, Raleigh, NC 27607, United States
| | - Nanette Nascone-Yoder
- Department of Molecular Biomedical Sciences, North Carolina State University, Raleigh, NC 27607, United States.
| |
Collapse
|
37
|
Kohlnhofer BM, Thompson CA, Walker EM, Battle MA. GATA4 regulates epithelial cell proliferation to control intestinal growth and development in mice. Cell Mol Gastroenterol Hepatol 2015; 2:189-209. [PMID: 27066525 PMCID: PMC4823006 DOI: 10.1016/j.jcmgh.2015.11.010] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND & AIMS The embryonic small intestinal epithelium is highly proliferative, and although much is known about mechanisms regulating proliferation in the adult intestine, the mechanisms controlling epithelial cell proliferation in the developing intestine are less clear. GATA4, a transcription factor that regulates proliferation in other developing tissues, is first expressed early in the developing gut in midgut endoderm. GATA4 function within midgut endoderm and the early intestinal epithelium has not been investigated. METHODS Using Sonic Hedgehog Cre to eliminate GATA4 in the midgut endoderm of mouse embryos, we determined the impact of loss of GATA4 on intestinal development, including epithelial cell proliferation, between E9.5-E18.5. RESULTS We found that intestinal length and width were decreased in GATA4 mutants compared with controls. GATA4-deficient intestinal epithelium contained fewer cells, and epithelial girth was decreased. We further observed a decreased proportion of proliferating cells at E10.5 and E11.5 in GATA4 mutants. We demonstrated that GATA4 binds to chromatin containing GATA4 consensus binding sites within Cyclin D2 (Ccnd2), Cyclin dependent kinase 6 (Cdk6), and Frizzled 5 (Fzd5). Moreover, Ccnd2, Cdk6, and Fzd5 transcripts were reduced at E11.5 in GATA4 mutant tissue. Villus morphogenesis was delayed, and villus structure was abnormal in GATA4 mutant intestine. CONCLUSIONS Our data identify GATA4 as an essential regulator of early intestinal epithelial cell proliferation. We propose that GATA4 controls proliferation in part by directly regulating transcription of cell cycle mediators. Our data further suggest that GATA4 affects proliferation through transcriptional regulation of Fzd5, perhaps by influencing the response of the epithelium to WNT signaling.
Collapse
Affiliation(s)
| | | | | | - Michele A. Battle
- Correspondence Address correspondence to: Michele A. Battle, PhD, Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, Wisconsin 53226. fax: (414) 955-6517.Department of Cell BiologyNeurobiology and AnatomyMedical College of Wisconsin8701 Watertown Plank RoadMilwaukeeWisconsin 53226
| |
Collapse
|
38
|
Rodrigo Albors A, Tazaki A, Rost F, Nowoshilow S, Chara O, Tanaka EM. Planar cell polarity-mediated induction of neural stem cell expansion during axolotl spinal cord regeneration. eLife 2015; 4:e10230. [PMID: 26568310 PMCID: PMC4755742 DOI: 10.7554/elife.10230] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Accepted: 11/12/2015] [Indexed: 02/07/2023] Open
Abstract
Axolotls are uniquely able to mobilize neural stem cells to regenerate all missing regions of the spinal cord. How a neural stem cell under homeostasis converts after injury to a highly regenerative cell remains unknown. Here, we show that during regeneration, axolotl neural stem cells repress neurogenic genes and reactivate a transcriptional program similar to embryonic neuroepithelial cells. This dedifferentiation includes the acquisition of rapid cell cycles, the switch from neurogenic to proliferative divisions, and the re-expression of planar cell polarity (PCP) pathway components. We show that PCP induction is essential to reorient mitotic spindles along the anterior-posterior axis of elongation, and orthogonal to the cell apical-basal axis. Disruption of this property results in premature neurogenesis and halts regeneration. Our findings reveal a key role for PCP in coordinating the morphogenesis of spinal cord outgrowth with the switch from a homeostatic to a regenerative stem cell that restores missing tissue. DOI:http://dx.doi.org/10.7554/eLife.10230.001 Stem cells found in adult tissues are vitally important for tissue repair and maintenance. These cells divide in two main ways: equally to create two new stem cells, or unequally to create a stem cell and a cell that can develop into one of the cell types in the tissue. A key challenge for biologists is to understand how these tissue-resident stem cells are activated and organized to regenerate injured or missing tissue. Throughout the life of the axolotl salamander, neural stem cells in the spinal cord occasionally divide to add new nerve cells to the healthy spinal cord. However, the axolotl can also regenerate part of its spinal cord, for example if its tail is lost. Under these conditions, the neural stem cells can convert into a highly regenerative stem cell that can produce all the different cell types needed to regrow the spinal cord. As a stem cell becomes a new cell type, it activates different sets of genes. Therefore, Rodrigo Albors, Tazaki et al. measured gene activity in the neural stem cells involved in axolotl spinal cord regeneration to uncover how these cells develop into a more regenerative form. This revealed that when an axolotl tail is amputated, resident stem cells turn off the genes that are specifically active in neuron-generating cells. In addition, they activate a similar set of genes to that seen in the embryonic cells that form the developing nervous system. These genes speed up cell division and activate an important signaling pathway. This pathway – the Wnt/PCP pathway – fulfils various developmental roles, one being to orient cell divisions, particularly in elongating tissues. In axolotls, this pathway causes the stem cells to divide equally to increase the number of available stem cells, and orients the direction of these divisions to ensure that the regenerating spinal cord elongates correctly. If this pathway is disrupted, the cells return to dividing unequally, generating nerve cells prematurely and halting the growth of the spinal cord. Such insights could help develop methods of repairing damaged nervous tissue in other animals that cannot regenerate to the extent that axolotls can. DOI:http://dx.doi.org/10.7554/eLife.10230.002
Collapse
Affiliation(s)
- Aida Rodrigo Albors
- Deutsche Forschungsgemeinschaft - Center for Regenerative Therapies Dresden, Dresden, Germany.,Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany.,Technische Universität Dresden, Dresden, Germany
| | - Akira Tazaki
- Deutsche Forschungsgemeinschaft - Center for Regenerative Therapies Dresden, Dresden, Germany.,Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany.,Technische Universität Dresden, Dresden, Germany
| | - Fabian Rost
- Center for Information Services and High Performance Computing, Technische Universität Dresden, Dresden, Germany
| | - Sergej Nowoshilow
- Deutsche Forschungsgemeinschaft - Center for Regenerative Therapies Dresden, Dresden, Germany.,Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany.,Technische Universität Dresden, Dresden, Germany
| | - Osvaldo Chara
- Center for Information Services and High Performance Computing, Technische Universität Dresden, Dresden, Germany.,Institute of Physics of Liquids and Biological Systems, National Scientific and Technical Research Council, University of La Plata, La Plata, Argentina
| | - Elly M Tanaka
- Deutsche Forschungsgemeinschaft - Center for Regenerative Therapies Dresden, Dresden, Germany.,Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany.,Technische Universität Dresden, Dresden, Germany
| |
Collapse
|
39
|
van der Werf CS, Halim D, Verheij JB, Alves MM, Hofstra RM. Congenital Short Bowel Syndrome: from clinical and genetic diagnosis to the molecular mechanisms involved in intestinal elongation. Biochim Biophys Acta Mol Basis Dis 2015; 1852:2352-61. [DOI: 10.1016/j.bbadis.2015.08.007] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2015] [Revised: 08/11/2015] [Accepted: 08/12/2015] [Indexed: 12/16/2022]
|
40
|
Song J, Song J, Xue K, Han H, Ding F, Li S, Dai Y, Li N. Rapid atraumatic sex identification of developmental day 14-16 mice. Biotech Histochem 2015; 90:309-14. [PMID: 25801294 DOI: 10.3109/10520295.2015.1004557] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Embryonic mice have been used widely to study organ development. Days 14-16 are critical for sex organ development and differentiation in mice. Current methods for sex identification are limited. Even the simplest polymerase chain reaction method may injure the embryo. We determined that morphologic analysis of embryonic mammary anlagen could be used for rapid atraumatic sex identification of day 14-16 mice. The accuracy of our method was verified by molecular and anatomical approaches.
Collapse
Affiliation(s)
- J Song
- College of Basic Medical Sciences, Dalian Medical University , Dalian
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Xiong F, Ma W, Hiscock TW, Mosaliganti KR, Tentner AR, Brakke KA, Rannou N, Gelas A, Souhait L, Swinburne IA, Obholzer ND, Megason SG. Interplay of cell shape and division orientation promotes robust morphogenesis of developing epithelia. Cell 2015; 159:415-27. [PMID: 25303534 DOI: 10.1016/j.cell.2014.09.007] [Citation(s) in RCA: 89] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2014] [Revised: 05/29/2014] [Accepted: 09/03/2014] [Indexed: 11/17/2022]
Abstract
Epithelial cells acquire functionally important shapes (e.g., squamous, cuboidal, columnar) during development. Here, we combine theory, quantitative imaging, and perturbations to analyze how tissue geometry, cell divisions, and mechanics interact to shape the presumptive enveloping layer (pre-EVL) on the zebrafish embryonic surface. We find that, under geometrical constraints, pre-EVL flattening is regulated by surface cell number changes following differentially oriented cell divisions. The division pattern is, in turn, determined by the cell shape distribution, which forms under geometrical constraints by cell-cell mechanical coupling. An integrated mathematical model of this shape-division feedback loop recapitulates empirical observations. Surprisingly, the model predicts that cell shape is robust to changes of tissue surface area, cell volume, and cell number, which we confirm in vivo. Further simulations and perturbations suggest the parameter linking cell shape and division orientation contributes to epithelial diversity. Together, our work identifies an evolvable design logic that enables robust cell-level regulation of tissue-level development.
Collapse
Affiliation(s)
- Fengzhu Xiong
- Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Wenzhe Ma
- Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Tom W Hiscock
- Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA
| | | | - Andrea R Tentner
- Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Kenneth A Brakke
- Mathematics Department, Susquehanna University, Selinsgrove, PA 17870, USA
| | - Nicolas Rannou
- Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Arnaud Gelas
- Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Lydie Souhait
- Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Ian A Swinburne
- Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Nikolaus D Obholzer
- Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Sean G Megason
- Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
42
|
Inhibition of phosphodiesterase 5 reduces bone mass by suppression of canonical Wnt signaling. Cell Death Dis 2014; 5:e1544. [PMID: 25429621 PMCID: PMC4260761 DOI: 10.1038/cddis.2014.510] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2014] [Revised: 10/22/2014] [Accepted: 10/23/2014] [Indexed: 01/21/2023]
Abstract
Inhibitors of phosphodiesterase 5 (PDE5) are widely used to treat erectile
dysfunction and pulmonary hypertension in clinics. PDE5, cyclic guanosine
monophosphate (cGMP), and protein kinase G (PKG) are important components of the
non-canonical Wnt signaling. This study aimed to investigate the effect of PDE5
inhibition on canonical Wnt signaling and osteoblastogenesis, using both in
vitro cell culture and in vivo animal models. In the in
vitro experiments, PDE5 inhibition resulted in activation of cGMP-dependent
protein kinase 2 and consequent inhibition of glycogen synthase kinase
3β phosphorylation, destabilization of cytosolic
β-catenin and the ultimate suppression of canonical Wnt signaling and
reduced osteoblastic differentiation in HEK293T and C3H10T1/2 cells. In animal
experiments, systemic inhibition of PDE5 suppressed the activity of canonical Wnt
signaling and osteoblastogenesis in bone marrow-derived stromal cells, resulting in
the reduction of bone mass in wild-type adult C57B/6 mice, significantly
attenuated secreted Frizzled-related protein-1 (SFRP1) deletion-induced activation of
canonical Wnt signaling and excessive bone growth in adult
SFRP1−/− mice. Together, these results uncover a
hitherto uncharacterized role of PDE5/cGMP/PKG signaling in bone homeostasis
and provide the evidence that long-term treatment with PDE5 inhibitors at a high
dosage may potentially cause bone catabolism.
Collapse
|
43
|
Matsuyama M, Nomori A, Nakakuni K, Shimono A, Fukushima M. Secreted Frizzled-related protein 1 (Sfrp1) regulates the progression of renal fibrosis in a mouse model of obstructive nephropathy. J Biol Chem 2014; 289:31526-33. [PMID: 25253698 DOI: 10.1074/jbc.m114.584565] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Renal fibrosis is responsible for progressive renal diseases that cause chronic renal failure. Sfrp1 (secreted Frizzled-related protein 1) is highly expressed in kidney, although little is known about connection between the protein and renal diseases. Here, we focused on Sfrp1 to investigate its roles in renal fibrosis using a mouse model of unilateral ureteral obstruction (UUO). In wild-type mice, the expression of Sfrp1 protein was markedly increased after UUO. The kidneys from Sfrp1 knock-out mice showed significant increase in expression of myofibrobast markers, α-smooth muscle actin (αSMA). Sfrp1 deficiency also increased protein levels of the fibroblast genes, vimentin, and decreased those of the epithelial genes, E-cadherin, indicated that enhanced epithelial-to-mesenchymal transition. There was no difference in the levels of canonical Wnt signaling; rather, the levels of phosphorylated c-Jun and JNK were more increased in the Sfrp1(-/-) obstructed kidney. Moreover, the apoptotic cell population was significantly elevated in the obstructed kidneys from Sfrp1(-/-) mice following UUO but was slightly increased in those from wild-type mice. These results indicate that Sfrp1 is required for inhibition of renal damage through the non-canonical Wnt/PCP pathway.
Collapse
Affiliation(s)
- Makoto Matsuyama
- From the Division of Molecular Genetics, Shigei Medical Research Institute, and
| | - Akane Nomori
- From the Division of Molecular Genetics, Shigei Medical Research Institute, and
| | - Kyomi Nakakuni
- Shigei Medical Research Hospital, Minami-ku, Okayama 701-0202, Japan and
| | | | - Masaki Fukushima
- Shigei Medical Research Hospital, Minami-ku, Okayama 701-0202, Japan and
| |
Collapse
|
44
|
Kwon YW, Chung YJ, Kim J, Lee HJ, Park J, Roh TY, Cho HJ, Yoon CH, Koo BK, Kim HS. Comparative study of efficacy of dopaminergic neuron differentiation between embryonic stem cell and protein-based induced pluripotent stem cell. PLoS One 2014; 9:e85736. [PMID: 24465672 PMCID: PMC3899054 DOI: 10.1371/journal.pone.0085736] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2013] [Accepted: 12/01/2013] [Indexed: 11/29/2022] Open
Abstract
In patients with Parkinson's disease (PD), stem cells can serve as therapeutic agents to restore or regenerate injured nervous system. Here, we differentiated two types of stem cells; mouse embryonic stem cells (mESCs) and protein-based iPS cells (P-iPSCs) generated by non-viral methods, into midbrain dopaminergic (mDA) neurons, and then compared the efficiency of DA neuron differentiation from these two cell types. In the undifferentiated stage, P-iPSCs expressed pluripotency markers as ES cells did, indicating that protein-based reprogramming was stable and authentic. While both stem cell types were differentiated to the terminally-matured mDA neurons, P-iPSCs showed higher DA neuron-specific markers' expression than ES cells. To investigate the mechanism of the superior induction capacity of DA neurons observed in P-iPSCs compared to ES cells, we analyzed histone modifications by genome-wide ChIP sequencing analysis and their corresponding microarray results between two cell types. We found that Wnt signaling was up-regulated, while SFRP1, a counter-acting molecule of Wnt, was more suppressed in P-iPSCs than in mESCs. In PD rat model, transplantation of neural precursor cells derived from both cell types showed improved function. The present study demonstrates that P-iPSCs could be a suitable cell source to provide patient-specific therapy for PD without ethical problems or rejection issues.
Collapse
Affiliation(s)
- Yoo-Wook Kwon
- National Research Laboratory for Stem Cell Niche, Seoul National University Hospital, Seoul, Korea
- Innovative Research Institute for Cell Therapy, Seoul National University Hospital, Seoul, Korea
| | - Yeon-Ju Chung
- National Research Laboratory for Stem Cell Niche, Seoul National University Hospital, Seoul, Korea
- Innovative Research Institute for Cell Therapy, Seoul National University Hospital, Seoul, Korea
| | - Joonoh Kim
- National Research Laboratory for Stem Cell Niche, Seoul National University Hospital, Seoul, Korea
- Innovative Research Institute for Cell Therapy, Seoul National University Hospital, Seoul, Korea
| | - Ho-Jae Lee
- National Research Laboratory for Stem Cell Niche, Seoul National University Hospital, Seoul, Korea
- Innovative Research Institute for Cell Therapy, Seoul National University Hospital, Seoul, Korea
| | - Jihwan Park
- Division of Molecular and Life Sciences,Pohang University of Science and Technology, Pohang, Korea
| | - Tae-Young Roh
- Division of Molecular and Life Sciences,Pohang University of Science and Technology, Pohang, Korea
| | - Hyun-Jai Cho
- National Research Laboratory for Stem Cell Niche, Seoul National University Hospital, Seoul, Korea
- Innovative Research Institute for Cell Therapy, Seoul National University Hospital, Seoul, Korea
- Department of Internal Medicine, Seoul National University, Seoul, Korea
| | - Chang-Hwan Yoon
- Cardiovascular center, Seoul National University Bundang Hospital, Seoul National University, Seoul, Korea
| | - Bon-Kwon Koo
- Department of Internal Medicine, Seoul National University, Seoul, Korea
| | - Hyo-Soo Kim
- National Research Laboratory for Stem Cell Niche, Seoul National University Hospital, Seoul, Korea
- Innovative Research Institute for Cell Therapy, Seoul National University Hospital, Seoul, Korea
- Department of Internal Medicine, Seoul National University, Seoul, Korea
- Molecular Medicine and Biopharmaceutical Sciences, Seoul National University, Seoul, Korea
- * E-mail:
| |
Collapse
|
45
|
Gon H, Fumoto K, Ku Y, Matsumoto S, Kikuchi A. Wnt5a signaling promotes apical and basolateral polarization of single epithelial cells. Mol Biol Cell 2013; 24:3764-74. [PMID: 24088568 PMCID: PMC3843002 DOI: 10.1091/mbc.e13-07-0357] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Wnt signal plays important roles in polarization. Here intestinal epithelial cells are shown to form apicobasal polarization at a single-cell level in an extracellular matrix adhesion–dependent manner. Wnt5a signaling promotes single-cell polarization through balanced control between Rac1 and RhoA activities spatially. Single epithelial-derived tumor cells have been shown to induce apical and basolateral (AB) polarity by expression of polarization-related proteins. However, physiological cues and molecular mechanisms for AB polarization of single normal epithelial cells are unclear. When intestinal epithelial cells 6 (IEC6 cells) were seeded on basement membrane proteins (Matrigel), single cells formed an F-actin cap on the upper cell surface, where apical markers accumulated, and a basolateral marker was localized to the rest of the cell surface region, in a Wnt5a signaling–dependent manner. However, these phenotypes were not induced by type I collagen. Rac1 activity in the noncap region was higher than that in the cap region, whereas Rho activity increased toward the cap region. Wnt5a signaling activated and inhibited Rac1 and RhoA, respectively, independently through Tiam1 and p190RhoGAP-A, which formed a tertiary complex with Dishevelled. Furthermore, Wnt5a signaling through Rac1 and RhoA was required for cystogenesis of IEC6 cells. These results suggest that Wnt5a promotes the AB polarization of IEC6 cells through regulation of Rac and Rho activities in a manner dependent on adhesion to specific extracellular matrix proteins.
Collapse
Affiliation(s)
- Hidetoshi Gon
- Department of Molecular Biology and Biochemistry, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan Division of Hepato-Biliary-Pancreatic Surgery, Department of Surgery, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe 650-0017, Japan
| | | | | | | | | |
Collapse
|
46
|
Roles of cilia, fluid flow, and Ca2+ signaling in breaking of left-right symmetry. Trends Genet 2013; 30:10-7. [PMID: 24091059 DOI: 10.1016/j.tig.2013.09.001] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2013] [Revised: 08/18/2013] [Accepted: 09/03/2013] [Indexed: 11/23/2022]
Abstract
The emergence of left-right (L-R) asymmetry during embryogenesis is a classic problem in developmental biology. It is only since the 1990s, however, that substantial insight into this problem has been achieved by molecular and genetic approaches. Various genes required for L-R asymmetric morphogenesis in vertebrates have now been identified, and many of these genes are required for the formation and motility of cilia. Breaking of L-R symmetry in the mouse embryo occurs in the ventral node, where two types of cilia are present. Whereas centrally located motile cilia generate a leftward fluid flow, peripherally located immotile cilia sense a flow-dependent signal, which is either chemical or mechanical in nature. Although Ca2+ signaling is implicated in flow sensing, the precise mechanism remains unknown. Here we summarize current knowledge of L-R symmetry breaking in vertebrates (focusing on the mouse), with a special emphasis on the roles of cilia, fluid flow, and Ca2+ signaling.
Collapse
|
47
|
Welsh IC, Thomsen M, Gludish DW, Alfonso-Parra C, Bai Y, Martin JF, Kurpios NA. Integration of left-right Pitx2 transcription and Wnt signaling drives asymmetric gut morphogenesis via Daam2. Dev Cell 2013; 26:629-44. [PMID: 24091014 PMCID: PMC3965270 DOI: 10.1016/j.devcel.2013.07.019] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2012] [Revised: 06/23/2013] [Accepted: 07/26/2013] [Indexed: 02/06/2023]
Abstract
A critical aspect of gut morphogenesis is initiation of a leftward tilt, and failure to do so leads to gut malrotation and volvulus. The direction of tilt is specified by asymmetric cell behaviors within the dorsal mesentery (DM), which suspends the gut tube, and is downstream of Pitx2, the key transcription factor responsible for the transfer of left-right (L-R) information from early gastrulation to morphogenesis. Although Pitx2 is a master regulator of L-R organ development, its cellular targets that drive asymmetric morphogenesis are not known. Using laser microdissection and targeted gene misexpression in the chicken DM, we show that Pitx2-specific effectors mediate Wnt signaling to activate the formin Daam2, a key Wnt effector and itself a Pitx2 target, linking actin dynamics to cadherin-based junctions to ultimately generate asymmetric cell behaviors. Our work highlights how integration of two conserved cascades may be the ultimate force through which Pitx2 sculpts L-R organs.
Collapse
Affiliation(s)
- Ian C. Welsh
- Department of Molecular Medicine, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853, USA
| | - Michael Thomsen
- Department of Molecular Medicine, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853, USA
| | - David W. Gludish
- Department of Molecular Medicine, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853, USA
| | - Catalina Alfonso-Parra
- Department of Molecular Medicine, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853, USA
| | - Yan Bai
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX 77030, USA
| | - James F. Martin
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Natasza A. Kurpios
- Department of Molecular Medicine, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853, USA
| |
Collapse
|
48
|
Qu Y, Ray PS, Li J, Cai Q, Bagaria SP, Moran C, Sim MS, Zhang J, Turner RR, Zhu Z, Cui X, Liu B. High levels of secreted frizzled-related protein 1 correlate with poor prognosis and promote tumourigenesis in gastric cancer. Eur J Cancer 2013; 49:3718-28. [PMID: 23927957 DOI: 10.1016/j.ejca.2013.07.011] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2013] [Revised: 06/17/2013] [Accepted: 07/15/2013] [Indexed: 01/15/2023]
Abstract
BACKGROUND Secreted frizzled-related protein 1 (sFRP1), Wnt signalling regulator, can positively or negatively regulate tumourigenesis and progression. We sought to determine the clinical relevance and the role of sFRP1 in gastric cancer development and progression. METHODS We investigated the sFRP1 protein expression levels and its clinicopathological correlations using 85 cases of human gastric samples with survival information (JWCI cohort). mRNA levels of sFRP1 and coexpressed genes were analysed using 131-sample cDNA microarray data (Ruijin cohort). The effects of sFRP1 alteration were investigated using cell proliferation, colony formation, migration, and invasion and xenograft models. RESULTS We show that sFRP1 is overexpressed in some human cancers and is significantly associated with lymph node metastasis and decreased overall survival in gastric cancer patients. Using gastric cancer cell models, we demonstrate that sFRP1 overexpression is correlated with the activation of TGFβ (transforming growth factor-beta) signalling pathway and thereby induces cell proliferation, epithelial-mesenchymal transition (EMT), and invasion. Conversely, sFRP1 knockdown shows the opposite effects. Furthermore, sFRP1 overexpression promotes tumourigenesis and metastasis in a xenograft model. CONCLUSION Our studies demonstrate that sFRP1 is a biomarker for aggressive subgroups of human gastric cancer and a prognostic biomarker for patients with poor survival. Our data provide insight into a crosstalk between Wnt and TGFβ pathways which underlies gastric cancer development and progression.
Collapse
Affiliation(s)
- Ying Qu
- Shanghai Key Laboratory of Gastric Neoplasms, Department of Surgery, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Zuo L, Iordanou E, Chandran RR, Jiang L. Novel mechanisms of tube-size regulation revealed by the Drosophila trachea. Cell Tissue Res 2013; 354:343-54. [PMID: 23824100 DOI: 10.1007/s00441-013-1673-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2013] [Accepted: 05/27/2013] [Indexed: 12/17/2022]
Abstract
The size of various tubes within tubular organs such as the lung, vascular system and kidney must be finely tuned for the optimal delivery of gases, nutrients, waste and cells within the entire organism. Aberrant tube sizes lead to devastating human illnesses, such as polycystic kidney disease, fibrocystic breast disease, pancreatic cystic neoplasm and thyroid nodules. However, the underlying mechanisms that are responsible for tube-size regulation have yet to be fully understood. Therefore, no effective treatments are available for disorders caused by tube-size defects. Recently, the Drosophila tracheal system has emerged as an excellent in vivo model to explore the fundamental mechanisms of tube-size regulation. Here, we discuss the role of the apical luminal matrix, cell polarity and signaling pathways in regulating tube size in Drosophila trachea. Previous studies of the Drosophila tracheal system have provided general insights into epithelial tube morphogenesis. Mechanisms that regulate tube size in Drosophila trachea could be well conserved in mammalian tubular organs. This knowledge should greatly aid our understanding of tubular organogenesis in vertebrates and potentially lead to new avenues for the treatment of human disease caused by tube-size defects.
Collapse
Affiliation(s)
- Li Zuo
- Department of Biological Sciences, Oakland University, Rochester, MI 48309, USA; School of Health and Rehabilitation Sciences, The Ohio State College of Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Ekaterini Iordanou
- Department of Biological Sciences, Oakland University, Rochester, MI 48309, USA
| | - Rachana R Chandran
- Department of Biological Sciences, Oakland University, Rochester, MI 48309, USA
| | - Lan Jiang
- Department of Biological Sciences, Oakland University, Rochester, MI 48309, USA; Dodge Hall of Engineering 322, 2200 N. Squirrel Road, Rochester, MI 48309, USA
| |
Collapse
|
50
|
Abstract
The centrosome, a major organizer of microtubules, has important functions in regulating cell shape, polarity, cilia formation and intracellular transport as well as the position of cellular structures, including the mitotic spindle. By means of these activities, centrosomes have important roles during animal development by regulating polarized cell behaviors, such as cell migration or neurite outgrowth, as well as mitotic spindle orientation. In recent years, the pace of discovery regarding the structure and composition of centrosomes has continuously accelerated. At the same time, functional studies have revealed the importance of centrosomes in controlling both morphogenesis and cell fate decision during tissue and organ development. Here, we review examples of centrosome and centriole positioning with a particular emphasis on vertebrate developmental systems, and discuss the roles of centrosome positioning, the cues that determine positioning and the mechanisms by which centrosomes respond to these cues. The studies reviewed here suggest that centrosome functions extend to the development of tissues and organs in vertebrates.
Collapse
Affiliation(s)
- Nan Tang
- Department of Anatomy, Cardiovascular Research Institute, The University of California, San Francisco, USA.
| | | |
Collapse
|