101
|
Id2a is required for hepatic outgrowth during liver development in zebrafish. Mech Dev 2015; 138 Pt 3:399-414. [PMID: 26022495 DOI: 10.1016/j.mod.2015.05.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2015] [Revised: 04/24/2015] [Accepted: 05/14/2015] [Indexed: 12/19/2022]
Abstract
During development, inhibitor of DNA binding (Id) proteins, a subclass of the helix-loop-helix family of proteins, regulate cellular proliferation, differentiation, and apoptosis in various organs. However, a functional role of Id2a in liver development has not yet been reported. Here, using zebrafish as a model organism, we provide in vivo evidence that Id2a regulates hepatoblast proliferation and cell death during liver development. Initially, in the liver, id2a is expressed in hepatoblasts and after their differentiation, id2a expression is restricted to biliary epithelial cells. id2a knockdown in zebrafish embryos had no effect on hepatoblast specification or hepatocyte differentiation. However, liver size was greatly reduced in id2a morpholino-injected embryos, indicative of a hepatic outgrowth defect attributable to the significant decrease in proliferating hepatoblasts concomitant with the significant increase in hepatoblast cell death. Altogether, these data support the role of Id2a as an important regulator of hepatic outgrowth via modulation of hepatoblast proliferation and survival during liver development in zebrafish.
Collapse
|
102
|
Di Maio A, Setar L, Tiozzo S, De Tomaso AW. Wnt affects symmetry and morphogenesis during post-embryonic development in colonial chordates. EvoDevo 2015; 6:17. [PMID: 26171140 PMCID: PMC4499891 DOI: 10.1186/s13227-015-0009-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2015] [Accepted: 04/09/2015] [Indexed: 12/26/2022] Open
Abstract
Background Wnt signaling is one of the earliest and most highly conserved regulatory pathways for the establishment of the body axes during regeneration and early development. In regeneration, body axes determination occurs independently of tissue rearrangement and early developmental cues. Modulation of the Wnt signaling in either process has shown to result in unusual body axis phenotypes. Botryllus schlosseri is a colonial ascidian that can regenerate its entire body through asexual budding. This processes leads to an adult body via a stereotypical developmental pathway (called blastogenesis), without proceeding through any embryonic developmental stages. Results In this study, we describe the role of the canonical Wnt pathway during the early stages of asexual development. We characterized expression of three Wnt ligands (Wnt2B, Wnt5A, and Wnt9A) by in situ hybridization and qRT-PCR. Chemical manipulation of the pathway resulted in atypical budding due to the duplication of the A/P axes, supernumerary budding, and loss of the overall cell apical-basal polarity. Conclusions Our results suggest that Wnt signaling is used for equivalent developmental processes both during embryogenesis and asexual development in an adult organism, suggesting that patterning mechanisms driving morphogenesis are conserved, independent of embryonic, or regenerative development. Electronic supplementary material The online version of this article (doi:10.1186/s13227-015-0009-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Alessandro Di Maio
- School of Bioscience, University of Birmingham, Edgbaston, Birmingham, B19 2TT UK.,Molecular Cellular and Developmental Biology, University of California Santa Barbara, Santa Barbara, CA 93106 USA
| | - Leah Setar
- Molecular Cellular and Developmental Biology, University of California Santa Barbara, Santa Barbara, CA 93106 USA
| | - Stefano Tiozzo
- CNRS, Sorbonne Universités, UPMC Univ Paris 06, Laboratoire de Biologie du Développement de Villefranche-sur-mer, Observatoire Océanographique, 06230 Villefranche-sur-mer, France
| | - Anthony W De Tomaso
- Molecular Cellular and Developmental Biology, University of California Santa Barbara, Santa Barbara, CA 93106 USA
| |
Collapse
|
103
|
Jiang F, Chen J, Ma X, Huang C, Zhu S, Wang F, Li L, Luo L, Ruan H, Huang H. Analysis of mutants from a genetic screening reveals the control of intestine and liver development by many common genes in zebrafish. Biochem Biophys Res Commun 2015; 460:838-44. [DOI: 10.1016/j.bbrc.2015.03.119] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2015] [Accepted: 03/21/2015] [Indexed: 01/10/2023]
|
104
|
Cox AG, Goessling W. The lure of zebrafish in liver research: regulation of hepatic growth in development and regeneration. Curr Opin Genet Dev 2015; 32:153-61. [PMID: 25863341 DOI: 10.1016/j.gde.2015.03.002] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2015] [Revised: 02/23/2015] [Accepted: 03/05/2015] [Indexed: 12/18/2022]
Abstract
The liver is an essential organ that plays a pivotal role in metabolism, digestion and nutrient storage. Major efforts have been made to develop zebrafish (Danio rerio) as a model system to study the pathways regulating hepatic growth during liver development and regeneration. Zebrafish offer unique advantages over other vertebrates including in vivo imaging at cellular resolution and the capacity for large-scale chemical and genetic screens. Here, we review the cellular and molecular mechanisms that regulate hepatic growth during liver development in zebrafish. We also highlight emerging evidence that developmental pathways are reactivated following liver injury to facilitate regeneration. Finally, we discuss how zebrafish have transformed drug discovery efforts and enabled the identification of drugs that stimulate hepatic growth and provide hepatoprotection in pre-clinical models of liver injury, with the ultimate goal of identifying novel therapeutic approaches to treat liver disease.
Collapse
Affiliation(s)
- Andrew G Cox
- Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| | - Wolfram Goessling
- Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States; Dana-Farber Cancer Institute, Boston, MA, United States; Harvard Stem Cell Institute, Cambridge, MA, United States; Broad Institute of MIT and Harvard, Cambridge, MA, United States.
| |
Collapse
|
105
|
Schotanus BA, Kruitwagen HS, van den Ingh TSGAM, van Wolferen ME, Rothuizen J, Penning LC, Spee B. Enhanced Wnt/β-catenin and Notch signalling in the activated canine hepatic progenitor cell niche. BMC Vet Res 2014; 10:309. [PMID: 25551829 PMCID: PMC4302101 DOI: 10.1186/s12917-014-0309-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2014] [Accepted: 12/16/2014] [Indexed: 02/06/2023] Open
Abstract
Background The liver has a large regenerative capacity. Hepatocytes can replicate and regenerate a diseased liver. However, as is the case in severe liver diseases, this replication may become insufficient or exhausted and hepatic progenitor cells (HPCs) can be activated in an attempt to restore liver function. Due to their bi-potent differentiation capacity, these HPCs have great potential for regenerative approaches yet over-activation does pose potential health risks. Therefore the mechanisms leading to activation must be elucidated prior to safe implementation in the veterinary clinic. Wnt/β-catenin and Notch signalling have been implicated in the activation of HPCs in mouse models and in humans. Here we assessed the involvement in canine HPC activation. Gene-expression profiles were derived from laser microdissected HPC niches from lobular dissecting hepatitis (LDH) and normal liver tissue, with a focus on Wnt/β-catenin and Notch signalling. Immunohistochemical and immunofluorescent studies were combined to assess the role of the pathways in HPCs during LDH. Results Gene-expression confirmed higher expression of Wnt/β-catenin and Notch pathway components and target genes in activated HPC niches in diseased liver compared to quiescent HPC niches from normal liver. Immunofluorescence confirmed the activation of these pathways in the HPCs during disease. Immunohistochemistry showed proliferating HPCs during LDH, and double immunofluorescence showed downregulation of Wnt/β-catenin and Notch in differentiating HPCs. Vimentin, a mesenchymal marker, was expressed on a subset of undifferentiated HPCs. Conclusions Together these studies clearly revealed that both Wnt/β-catenin and Notch signalling pathways are enhanced in undifferentiated, proliferating and potentially migrating HPCs during severe progressive canine liver disease (LDH).
Collapse
Affiliation(s)
- Baukje A Schotanus
- Department of Clinical Sciences of Companion Animals, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands.
| | - Hedwig S Kruitwagen
- Department of Clinical Sciences of Companion Animals, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands.
| | | | - Monique E van Wolferen
- Department of Clinical Sciences of Companion Animals, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands.
| | - Jan Rothuizen
- Department of Clinical Sciences of Companion Animals, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands.
| | - Louis C Penning
- Department of Clinical Sciences of Companion Animals, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands.
| | - Bart Spee
- Department of Clinical Sciences of Companion Animals, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands.
| |
Collapse
|
106
|
Gao W, Zhou P, Ma X, Tschudy-Seney B, Chen J, Magner NL, Revzin A, Nolta JA, Zern MA, Duan Y. Ethanol negatively regulates hepatic differentiation of hESC by inhibition of the MAPK/ERK signaling pathway in vitro. PLoS One 2014; 9:e112698. [PMID: 25393427 PMCID: PMC4231066 DOI: 10.1371/journal.pone.0112698] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2014] [Accepted: 10/10/2014] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Alcohol insult triggers complex events in the liver, promoting fibrogenic/inflammatory signals and in more advanced cases, aberrant matrix deposition. It is well accepted that the regenerative capacity of the adult liver is impaired during alcohol injury. The liver progenitor/stem cells have been shown to play an important role in liver regeneration -in response to various chronic injuries; however, the effects of alcohol on stem cell differentiation in the liver are not well understood. METHODS We employed hepatic progenitor cells derived from hESCs to study the impact of ethanol on hepatocyte differentiation by exposure of these progenitor cells to ethanol during hepatocyte differentiation. RESULTS We found that ethanol negatively regulated hepatic differentiation of hESC-derived hepatic progenitor cells in a dose-dependent manner. There was also a moderate cell cycle arrest at G1/S checkpoint in the ethanol treated cells, which is associated with a reduced level of cyclin D1 in these cells. Ethanol treatment specifically inhibited the activation of the ERK but not JNK nor the p38 MAP signaling pathway. At the same time, the WNT signaling pathway was also reduced in the cells exposed to ethanol. Upon evaluating the effects of the inhibitors of these two signaling pathways, we determined that the Erk inhibitor replicated the effects of ethanol on the hepatocyte differentiation and attenuated the WNT/β-catenin signaling, however, inhibitors of WNT only partially replicated the effects of ethanol on the hepatocyte differentiation. CONCLUSION Our results demonstrated that ethanol negatively regulated hepatic differentiation of hESC-derived hepatic progenitors through inhibiting the MAPK/ERK signaling pathway, and subsequently attenuating the WNT signaling pathway. Thus, our finding provides a novel insight into the mechanism by which alcohol regulates cell fate selection of hESC-derived hepatic progenitor cells, and the identified pathways may provide therapeutic targets aimed at promoting liver repair and regeneration during alcoholic injury.
Collapse
Affiliation(s)
- Wei Gao
- Department of Biomedical Engineering, College of Biology, Hunan University, Changsha, Hunan, China
- Department of Internal Medicine, University of California Davis Medical Center, Sacramento, California, United States of America
- Institute for Regenerative Cures, University of California Davis Medical Center, Sacramento, California, United States of America
| | - Ping Zhou
- Department of Internal Medicine, University of California Davis Medical Center, Sacramento, California, United States of America
- Institute for Regenerative Cures, University of California Davis Medical Center, Sacramento, California, United States of America
- * E-mail: (YD); (PZ)
| | - Xiaocui Ma
- Department of Internal Medicine, University of California Davis Medical Center, Sacramento, California, United States of America
- Institute for Regenerative Cures, University of California Davis Medical Center, Sacramento, California, United States of America
| | - Benjamin Tschudy-Seney
- Department of Internal Medicine, University of California Davis Medical Center, Sacramento, California, United States of America
- Institute for Regenerative Cures, University of California Davis Medical Center, Sacramento, California, United States of America
| | - Jiamei Chen
- Department of Internal Medicine, University of California Davis Medical Center, Sacramento, California, United States of America
- Institute for Regenerative Cures, University of California Davis Medical Center, Sacramento, California, United States of America
- Institute of Liver Diseases, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Nataly L. Magner
- Department of Internal Medicine, University of California Davis Medical Center, Sacramento, California, United States of America
- Institute for Regenerative Cures, University of California Davis Medical Center, Sacramento, California, United States of America
| | - Alexander Revzin
- Department of Biomedical Engineering, University of California Davis, Davis, California, United States of America
| | - Jan A. Nolta
- Department of Internal Medicine, University of California Davis Medical Center, Sacramento, California, United States of America
- Institute for Regenerative Cures, University of California Davis Medical Center, Sacramento, California, United States of America
| | - Mark A. Zern
- Department of Internal Medicine, University of California Davis Medical Center, Sacramento, California, United States of America
- Institute for Regenerative Cures, University of California Davis Medical Center, Sacramento, California, United States of America
| | - Yuyou Duan
- Department of Internal Medicine, University of California Davis Medical Center, Sacramento, California, United States of America
- Institute for Regenerative Cures, University of California Davis Medical Center, Sacramento, California, United States of America
- * E-mail: (YD); (PZ)
| |
Collapse
|
107
|
Huang M, Chang A, Choi M, Zhou D, Anania FA, Shin CH. Antagonistic interaction between Wnt and Notch activity modulates the regenerative capacity of a zebrafish fibrotic liver model. Hepatology 2014; 60:1753-66. [PMID: 24995814 PMCID: PMC4211965 DOI: 10.1002/hep.27285] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2014] [Revised: 05/25/2014] [Accepted: 06/26/2014] [Indexed: 12/13/2022]
Abstract
UNLABELLED In chronic liver failure patients with sustained fibrosis, excessive accumulation of extracellular matrix proteins substantially dampens the regenerative capacity of the hepatocytes, resulting in poor prognosis and high mortality. Currently, the mechanisms and the strategies of inducing endogenous cellular sources such as hepatic progenitor cells (HPCs) to regenerate hepatocytes in various contexts of fibrogenic stimuli remain elusive. Here we aim to understand the molecular and cellular mechanisms that mediate the effects of sustained fibrosis on hepatocyte regeneration using the zebrafish as a model. In the ethanol-induced fibrotic zebrafish model, we identified a subset of HPCs, responsive to Notch signaling, that retains its capacity to regenerate as hepatocytes. Discrete levels of Notch signaling modulate distinct cellular outcomes of these Notch-responsive HPCs in hepatocyte regeneration. Lower levels of Notch signaling promote amplification and subsequent differentiation of these cells into hepatocytes, while high levels of Notch signaling suppress these processes. To identify small molecules facilitating hepatocyte regeneration in the fibrotic liver, we performed chemical screens and identified a number of Wnt agonists and Notch antagonists. Further analyses demonstrated that these Wnt agonists are capable of attenuating Notch signaling by inducing Numb, a membrane-associated protein that inhibits Notch signaling. This suggests that the antagonistic interplay between Wnt and Notch signaling crucially affects hepatocyte regeneration in the fibrotic liver. CONCLUSION Our findings not only elucidate how signaling pathways and cell-cell communications direct the cellular response of HPCs to fibrogenic stimuli, but also identify novel potential therapeutic strategies for chronic liver disease.
Collapse
Affiliation(s)
- Mianbo Huang
- School of Biology and the Parker H. Petit Institute for Bioengineering and Bioscience Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Angela Chang
- School of Biology and the Parker H. Petit Institute for Bioengineering and Bioscience Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Minna Choi
- School of Biology and the Parker H. Petit Institute for Bioengineering and Bioscience Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - David Zhou
- School of Biology and the Parker H. Petit Institute for Bioengineering and Bioscience Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Frank A. Anania
- Division of Digestive Diseases, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Chong Hyun Shin
- School of Biology and the Parker H. Petit Institute for Bioengineering and Bioscience Georgia Institute of Technology, Atlanta, GA 30332, USA,To whom correspondence should be addressed: Chong Hyun Shin, Ph.D., School of Biology and the Parker H. Petit Institute for Bioengineering and Bioscience, Interdisciplinary Bioengineering Graduate Program, Georgia Institute of Technology, 315 Ferst Drive NW, Room 1313, Atlanta, GA 30332, USA, , Telephone: 404-385-4211
| |
Collapse
|
108
|
O'Shields B, McArthur AG, Holowiecki A, Kamper M, Tapley J, Jenny MJ. Inhibition of endogenous MTF-1 signaling in zebrafish embryos identifies novel roles for MTF-1 in development. BIOCHIMICA ET BIOPHYSICA ACTA 2014; 1843:1818-33. [PMID: 24751692 PMCID: PMC4096078 DOI: 10.1016/j.bbamcr.2014.04.015] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/26/2013] [Revised: 04/09/2014] [Accepted: 04/11/2014] [Indexed: 02/07/2023]
Abstract
The metal responsive element-binding transcription factor-1 (MTF-1) responds to changes in cellular zinc levels caused by zinc exposure or disruption of endogenous zinc homeostasis by heavy metals or oxygen-related stress. Here we report the functional characterization of a complete zebrafish MTF-1 in comparison with the previously identified isoform lacking the highly conserved cysteine-rich motif (Cys-X-Cys-Cys-X-Cys) found in all other vertebrate MTF-1 orthologs. In an effort to develop novel molecular tools, a constitutively nuclear dominant-negative MTF-1 (dnMTF-1) was generated as tool for inhibiting endogenous MTF-1 signaling. The in vivo efficacy of the dnMTF-1 was determined by microinjecting in vitro transcribed dnMTF-1 mRNA into zebrafish embryos (1-2 cell stage) followed by transcriptomic profiling using an Agilent 4x44K array on 28- and 36-hpf embryos. A total of 594 and 560 probes were identified as differentially expressed at 28hpf and 36hpf, respectively, with interesting overlaps between timepoints. The main categories of genes affected by the inhibition of MTF-1 signaling were: nuclear receptors and genes involved in stress signaling, neurogenesis, muscle development and contraction, eye development, and metal homeostasis, including novel observations in iron and heme homeostasis. Finally, we investigate both the transcriptional activator and transcriptional repressor role of MTF-1 in potential novel target genes identified by transcriptomic profiling during early zebrafish development.
Collapse
Affiliation(s)
- Britton O'Shields
- Department of Biological Sciences, University of Alabama, Tuscaloosa, AL 35487, USA
| | | | - Andrew Holowiecki
- Department of Biological Sciences, University of Alabama, Tuscaloosa, AL 35487, USA
| | - Martin Kamper
- Department of Biological Sciences, University of Alabama, Tuscaloosa, AL 35487, USA
| | - Jeffrey Tapley
- Department of Biological Sciences, University of Alabama, Tuscaloosa, AL 35487, USA
| | - Matthew J Jenny
- Department of Biological Sciences, University of Alabama, Tuscaloosa, AL 35487, USA.
| |
Collapse
|
109
|
Zhang D, Golubkov VS, Han W, Correa RG, Zhou Y, Lee S, Strongin AY, Dong PDS. Identification of Annexin A4 as a hepatopancreas factor involved in liver cell survival. Dev Biol 2014; 395:96-110. [PMID: 25176043 DOI: 10.1016/j.ydbio.2014.08.025] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2014] [Revised: 08/19/2014] [Accepted: 08/20/2014] [Indexed: 01/27/2023]
Abstract
To gain insight into liver and pancreas development, we investigated the target of 2F11, a monoclonal antibody of unknown antigen, widely used in zebrafish studies for labeling hepatopancreatic ducts. Utilizing mass spectrometry and in vivo assays, we determined the molecular target of 2F11 to be Annexin A4 (Anxa4), a calcium binding protein. We further found that in both zebrafish and mouse endoderm, Anxa4 is broadly expressed in the developing liver and pancreas, and later becomes more restricted to the hepatopancreatic ducts and pancreatic islets, including the insulin producing ß-cells. Although Anxa4 is a known target of several monogenic diabetes genes and its elevated expression is associated with chemoresistance in malignancy, its in vivo role is largely unexplored. Knockdown of Anxa4 in zebrafish leads to elevated expression of caspase 8 and Δ113p53, and liver bud specific activation of Caspase 3 and apoptosis. Mosaic knockdown reveal that Anxa4 is required cell-autonomously in the liver bud for cell survival. This finding is further corroborated with mosaic anxa4 knockout studies using the CRISPR/Cas9 system. Collectively, we identify Anxa4 as a new, evolutionarily conserved hepatopancreatic factor that is required in zebrafish for liver progenitor viability, through inhibition of the extrinsic apoptotic pathway. A role for Anxa4 in cell survival may have implications for the mechanism of diabetic ß-cell apoptosis and cancer cell chemoresistance.
Collapse
Affiliation(s)
- Danhua Zhang
- Sanford Children's Health Research Center, Programs in Genetic Disease, Development and Aging, and Stem Cell and Regenerative Biology, Sanford-Burnham Medical Research Institute, La Jolla, CA, USA; Graduate School of Biomedical Sciences, Sanford-Burnham Medical Research Institute, La Jolla, CA, USA
| | - Vladislav S Golubkov
- Infectious and Inflammatory Disease Center, Sanford-Burnham Medical Research Institute, La Jolla, CA, USA
| | - Wenlong Han
- NCI-Designated Cancer Center, Tumor Microenvironment Program, Sanford-Burnham Medical Research Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Ricardo G Correa
- Infectious and Inflammatory Disease Center, Sanford-Burnham Medical Research Institute, La Jolla, CA, USA
| | - Ying Zhou
- Sanford Children's Health Research Center, Programs in Genetic Disease, Development and Aging, and Stem Cell and Regenerative Biology, Sanford-Burnham Medical Research Institute, La Jolla, CA, USA; Graduate School of Biomedical Sciences, Sanford-Burnham Medical Research Institute, La Jolla, CA, USA
| | - Sunyoung Lee
- NCI-Designated Cancer Center, Tumor Microenvironment Program, Sanford-Burnham Medical Research Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Alex Y Strongin
- Infectious and Inflammatory Disease Center, Sanford-Burnham Medical Research Institute, La Jolla, CA, USA
| | - P Duc Si Dong
- Sanford Children's Health Research Center, Programs in Genetic Disease, Development and Aging, and Stem Cell and Regenerative Biology, Sanford-Burnham Medical Research Institute, La Jolla, CA, USA; Graduate School of Biomedical Sciences, Sanford-Burnham Medical Research Institute, La Jolla, CA, USA.
| |
Collapse
|
110
|
Hypoxia-inducible factor 2 alpha is essential for hepatic outgrowth and functions via the regulation of leg1 transcription in the zebrafish embryo. PLoS One 2014; 9:e101980. [PMID: 25000307 PMCID: PMC4084947 DOI: 10.1371/journal.pone.0101980] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2014] [Accepted: 06/13/2014] [Indexed: 12/11/2022] Open
Abstract
The liver plays a vital role in metabolism, detoxification, digestion, and the maintenance of homeostasis. During development, the vertebrate embryonic liver undergoes a series of morphogenic processes known as hepatogenesis. Hepatogenesis can be separated into three interrelated processes: endoderm specification, hepatoblast differentiation, and hepatic outgrowth. Throughout this process, signaling molecules and transcription factors initiate and regulate the coordination of cell proliferation, apoptosis, differentiation, intercellular adhesion, and cell migration. Hifs are already recognized to be essential in embryonic development, but their role in hepatogenesis remains unknown. Using the zebrafish embryo as a model organism, we report that the lack of Hif2-alpha but not Hif1-alpha blocks hepatic outgrowth. While Hif2-alpha is not involved in hepatoblast specification, this transcription factor regulates hepatocyte cell proliferation during hepatic outgrowth. Furthermore, we demonstrated that the lack of Hif2-alpha can reduce the expression of liver-enriched gene 1 (leg1), which encodes a secretory protein essential for hepatic outgrowth. Additionally, exogenous mRNA expression of leg1 can rescue the small liver phenotype of hif2-alpha morphants. We also showed that Hif2-alpha directly binds to the promoter region of leg1 to control leg1 expression. Interestingly, we discovered overrepresented, high-density Hif-binding sites in the potential upstream regulatory sequences of leg1 in teleosts but not in terrestrial mammals. We concluded that hif2-alpha is a key factor required for hepatic outgrowth and regulates leg1 expression in zebrafish embryos. We also proposed that the hif2-alpha-leg1 axis in liver development may have resulted from the adaptation of teleosts to their environment.
Collapse
|
111
|
Yamashita T, Kaneko S. Orchestration of hepatocellular carcinoma development by diverse liver cancer stem cells. J Gastroenterol 2014; 49:1105-10. [PMID: 24647548 DOI: 10.1007/s00535-014-0951-1] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2013] [Accepted: 03/09/2014] [Indexed: 02/04/2023]
Abstract
Hepatocellular carcinoma (HCC) is one of the world's most aggressive diseases and carries a poor prognosis for patients. Recent evidence suggests that HCC is organized by cancer stem cells (CSCs), which are a subset of cells with stem cell-like features. CSCs are considered a pivotal target for the eradication of cancer, and liver CSCs have been investigated using various stem cell markers. Several hepatic stem/progenitor markers have been shown to be useful for isolating putative CSCs from HCC, although the expression patterns and phenotypic diversity of CSCs purified by these markers remain obscure. Recently, we found that liver CSCs defined by different markers show unique features of tumorigenicity and metastasis, with phenotypes closely associated with committed liver lineages. Furthermore, our data suggest that these distinct CSCs collaborate to orchestrate the tumorigenicity and metastasis of HCC. In this review article, we summarize the recent advances in understanding the pathogenesis and heterogeneity of liver CSCs.
Collapse
Affiliation(s)
- Taro Yamashita
- Departments of General Medicine, Kanazawa University Hospital, Kanazawa, Ishikawa, Japan,
| | | |
Collapse
|
112
|
Dickover M, Hegarty JM, Ly K, Lopez D, Yang H, Zhang R, Tedeschi N, Hsiai TK, Chi NC. The atypical Rho GTPase, RhoU, regulates cell-adhesion molecules during cardiac morphogenesis. Dev Biol 2014; 389:182-91. [PMID: 24607366 DOI: 10.1016/j.ydbio.2014.02.014] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2013] [Revised: 01/28/2014] [Accepted: 02/18/2014] [Indexed: 11/29/2022]
Abstract
The vertebrate heart undergoes early complex morphologic events in order to develop key cardiac structures that regulate its overall function (Fahed et al., 2013). Although many genetic factors that participate in patterning the heart have been elucidated (Tu and Chi, 2012), the cellular events that drive cardiac morphogenesis have been less clear. From a chemical genetic screen to identify cellular pathways that control cardiac morphogenesis in zebrafish, we observed that inhibition of the Rho signaling pathways resulted in failure to form the atrioventricular canal and loop the linear heart tube. To identify specific Rho proteins that may regulate this process, we analyzed cardiac expression profiling data and discovered that RhoU was expressed at the atrioventricular canal during the time when it forms. Loss of RhoU function recapitulated the atrioventricular canal and cardiac looping defects observed in the ROCK inhibitor treated zebrafish. Similar to its family member RhoV/Chp (Tay et al., 2010), we discovered that RhoU regulates the cell junctions between cardiomyocytes through the Arhgef7b/Pak kinase pathway in order to guide atrioventricular canal development and cardiac looping. Inhibition of this pathway resulted in similar underlying cardiac defects and conversely, overexpression of a PAK kinase was able to rescue the loss of RhoU cardiac defect. Finally, we found that Wnt signaling, which has been implicated in atrioventricular canal development (Verhoeven et al., 2011), may regulate the expression of RhoU at the atrioventricular canal. Overall, these findings reveal a cardiac developmental pathway involving RhoU/Arhgef7b/Pak signaling, which helps coordinate cell junction formation between atrioventricular cardiomyocytes to promote cell adhesiveness and cell shapes during cardiac morphogenesis. Failure to properly form these cell adhesions during cardiac development may lead to structural heart defects and mechanistically account for the cellular events that occur in certain human congenital heart diseases.
Collapse
Affiliation(s)
- Michael Dickover
- Department of Medicine, Division of Cardiology, University of California, San Diego, La Jolla, CA 92093-0613J, USA
| | - Jeffrey M Hegarty
- Department of Medicine, Division of Cardiology, University of California, San Diego, La Jolla, CA 92093-0613J, USA
| | - Kim Ly
- Department of Medicine, Division of Cardiology, University of California, San Diego, La Jolla, CA 92093-0613J, USA
| | - Diana Lopez
- Department of Medicine, Division of Cardiology, University of California, San Diego, La Jolla, CA 92093-0613J, USA
| | - Hongbo Yang
- Department of Medicine, Division of Cardiology, University of California, San Diego, La Jolla, CA 92093-0613J, USA
| | - Ruilin Zhang
- Department of Medicine, Division of Cardiology, University of California, San Diego, La Jolla, CA 92093-0613J, USA
| | - Neil Tedeschi
- Department of Medicine, Division of Cardiology, University of California, San Diego, La Jolla, CA 92093-0613J, USA
| | - Tzung K Hsiai
- Department of Medicine, Division of Cardiology, University of Southern California, Los Angeles, CA, USA
| | - Neil C Chi
- Department of Medicine, Division of Cardiology, University of California, San Diego, La Jolla, CA 92093-0613J, USA; Institute of Genomic Medicine, University of California, San Diego, La Jolla, CA, USA.
| |
Collapse
|
113
|
Loh SL, Teh C, Muller J, Guccione E, Hong W, Korzh V. Zebrafish yap1 plays a role in differentiation of hair cells in posterior lateral line. Sci Rep 2014; 4:4289. [PMID: 24598795 PMCID: PMC3944368 DOI: 10.1038/srep04289] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2013] [Accepted: 02/13/2014] [Indexed: 11/25/2022] Open
Abstract
The evolutionarily conserved Hippo signaling pathway controls organ size by regulating cell proliferation and apoptosis and this process involves Yap1. The zebrafish Yap1 acts during neural differentiation, but its function is not fully understood. The detailed analysis of yap1 expression in proliferative regions, revealed it in the otic placode that gives rise to the lateral line system affected by the morpholino-mediated knockdown of Yap1. The comparative microarray analysis of transcriptome of Yap1-deficient embryos demonstrated changes in expression of many genes, including the Wnt signaling pathway and, in particular, prox1a known for its role in development of mechanoreceptors in the lateral line. The knockdown of Yap1 causes a deficiency of differentiation of mechanoreceptors, and this defect can be rescued by prox1a mRNA. Our studies revealed a role of Yap1 in regulation of Wnt signaling pathway and its target Prox1a during differentiation of mechanosensory cells.
Collapse
Affiliation(s)
- Siau-Lin Loh
- Institute of Molecular and Cell Biology, Singapore
| | - Cathleen Teh
- Institute of Molecular and Cell Biology, Singapore
| | | | | | - Wanjin Hong
- 1] Institute of Molecular and Cell Biology, Singapore [2] Department of Biochemistry, National University of Singapore, Singapore
| | - Vladimir Korzh
- 1] Institute of Molecular and Cell Biology, Singapore [2] Department of Biological Sciences, National University of Singapore, Singapore
| |
Collapse
|
114
|
Affiliation(s)
- Catherine M Verfaillie
- Stem Cell Institute, Stem Cell and Developmental Biology, Department of Development and Regeneration, Group of Biomedical Science, KU Leuven, Leuven, Belgium.
| |
Collapse
|
115
|
Choi TY, Ninov N, Stainier DY, Shin D. Extensive conversion of hepatic biliary epithelial cells to hepatocytes after near total loss of hepatocytes in zebrafish. Gastroenterology 2014; 146:776-88. [PMID: 24148620 PMCID: PMC3943869 DOI: 10.1053/j.gastro.2013.10.019] [Citation(s) in RCA: 175] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2013] [Revised: 09/10/2013] [Accepted: 10/16/2013] [Indexed: 01/26/2023]
Abstract
BACKGROUND & AIMS Biliary epithelial cells (BECs) are considered to be a source of regenerating hepatocytes when hepatocyte proliferation is compromised. However, there is still controversy about the extent to which BECs can contribute to the regenerating hepatocyte population, and thereby to liver recovery. To investigate this issue, we established a zebrafish model of liver regeneration in which the extent of hepatocyte ablation can be controlled. METHODS Hepatocytes were depleted by administration of metronidazole to Tg(fabp10a:CFP-NTR) animals. We traced the origin of regenerating hepatocytes using short-term lineage-tracing experiments, as well as the inducible Cre/loxP system; specifically, we utilized both a BEC tracer line Tg(Tp1:CreER(T2)) and a hepatocyte tracer line Tg(fabp10a:CreER(T2)). We also examined BEC and hepatocyte proliferation and liver marker gene expression during liver regeneration. RESULTS BECs gave rise to most of the regenerating hepatocytes in larval and adult zebrafish after severe hepatocyte depletion. After hepatocyte loss, BECs proliferated as they dedifferentiated into hepatoblast-like cells; they subsequently differentiated into highly proliferative hepatocytes that restored the liver mass. This process was impaired in zebrafish wnt2bb mutants; in these animals, hepatocytes regenerated but their proliferation was greatly reduced. CONCLUSIONS BECs contribute to regenerating hepatocytes after substantial hepatocyte depletion in zebrafish, thereby leading to recovery from severe liver damage.
Collapse
Affiliation(s)
- Tae-Young Choi
- Department of Developmental Biology, McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Nikolay Ninov
- Department of Biochemistry and Biophysics, Programs in Developmental and Stem Cell Biology, Genetics and Human Genetics, Diabetes Center, and Liver Center, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Didier Y.R. Stainier
- Department of Biochemistry and Biophysics, Programs in Developmental and Stem Cell Biology, Genetics and Human Genetics, Diabetes Center, and Liver Center, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Donghun Shin
- Department of Developmental Biology, McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania.
| |
Collapse
|
116
|
Minor class splicing shapes the zebrafish transcriptome during development. Proc Natl Acad Sci U S A 2014; 111:3062-7. [PMID: 24516132 DOI: 10.1073/pnas.1305536111] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Minor class or U12-type splicing is a highly conserved process required to remove a minute fraction of introns from human pre-mRNAs. Defects in this splicing pathway have recently been linked to human disease, including a severe developmental disorder encompassing brain and skeletal abnormalities known as Taybi-Linder syndrome or microcephalic osteodysplastic primordial dwarfism 1, and a hereditary intestinal polyposis condition, Peutz-Jeghers syndrome. Although a key mechanism for regulating gene expression, the impact of impaired U12-type splicing on the transcriptome is unknown. Here, we describe a unique zebrafish mutant, caliban (clbn), with arrested development of the digestive organs caused by an ethylnitrosourea-induced recessive lethal point mutation in the rnpc3 [RNA-binding region (RNP1, RRM) containing 3] gene. rnpc3 encodes the zebrafish ortholog of human RNPC3, also known as the U11/U12 di-snRNP 65-kDa protein, a unique component of the U12-type spliceosome. The biochemical impact of the mutation in clbn is the formation of aberrant U11- and U12-containing small nuclear ribonucleoproteins that impair the efficiency of U12-type splicing. Using RNA sequencing and microarrays, we show that multiple genes involved in various steps of mRNA processing, including transcription, splicing, and nuclear export are disrupted in clbn, either through intron retention or differential gene expression. Thus, clbn provides a useful and specific model of aberrant U12-type splicing in vivo. Analysis of its transcriptome reveals efficient mRNA processing as a critical process for the growth and proliferation of cells during vertebrate development.
Collapse
|
117
|
Arterbery AS, Bogue CW. Endodermal and mesenchymal cross talk: a crossroad for the maturation of foregut organs. Pediatr Res 2014; 75:120-6. [PMID: 24192700 DOI: 10.1038/pr.2013.201] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2013] [Accepted: 08/27/2013] [Indexed: 01/30/2023]
Abstract
The developmental stages of each foregut organ are intimately linked to the development of the other foregut organs such that the ultimate function of any one foregut organ, such as the metabolic function of the liver, depends on organizational changes associated with the maturation of multiple foregut organs. These changes include: (i) proliferation of the intrahepatic bile ducts and hepatoblasts within the liver coinciding with parenchymal expansion, (ii) elongation of extrahepatic bile ducts, which allows for proper gallbladder (GB) formation, and (iii) duodenal elongation and rotation, which coincides with all of the above to connect the intrahepatic, extrahepatic, and pancreatic ductal systems with the intestine. It is well established that cross talk between endodermal and mesenchymal components of the foregut occurs, particularly regarding the vascularization of developing organs. Furthermore, genetic mutations in mesenchymal and hepatic compartments of the developing foregut result in similar foregut pathologies: hypoplastic liver, absence of GB, biliary atresia (intrahepatic and/or extrahepatic), and failure of gut elongation and rotation. Finally, these shared pathologies can be linked to deficiencies in genes specific to the septum transversum mesenchyme (Hes1, Hlx, and Foxf1) or liver (Hhex and Hnf6), illustrating the complexity of such cross talk.
Collapse
Affiliation(s)
- Adam S Arterbery
- Department of Pediatrics, Yale School of Medicine, New Haven, Connecticut
| | - Clifford W Bogue
- Department of Pediatrics, Yale School of Medicine, New Haven, Connecticut
| |
Collapse
|
118
|
Abstract
Wnt/β-catenin signaling plays key roles not only during development but also in adult tissue homeostasis. This is also evident in liver biology where many temporal roles of β-catenin have been identified during hepatic development, where, in hepatic progenitors or hepatoblasts, it is a key determinant of proliferation and eventually differentiation to mature hepatocytes, while also playing an important role in bile duct homeostasis. β-Catenin signaling cascade is mostly quiescent in hepatocytes in an adult liver except in the centrizonal region of a hepatic lobule. This small rim of hepatocytes around the central vein show constitutive β-catenin activation that in turn regulates expression of genes whose products play an important role in ammonia and xenobiotic metabolism. Intriguingly, β-catenin can also undergo activation in hepatocytes after acute liver loss secondary to surgical or toxicant insult. Such activation of this progrowth protein is observed as nuclear translocation of β-catenin and formation of its complex with the T-cell factor (TCF) family of transcription factors. Expression of cyclin-D1, a key inducer of transition from the G1 to S phase of cell cycle, is regulated by β-catenin-TCF complex. Thus, β-catenin activation is absolutely critical in the normal regeneration process of the liver as shown by studies in several models across various species. In the current review, the temporal role and regulation of β-catenin in liver development, metabolic zonation in a basal adult liver, and during the liver regeneration process will be discussed. In addition, the probability of therapeutically regulating β-catenin activity as a possible future treatment strategy for liver insufficiency will also be discussed.
Collapse
|
119
|
Opitz R, Antonica F, Costagliola S. New model systems to illuminate thyroid organogenesis. Part I: an update on the zebrafish toolbox. Eur Thyroid J 2013; 2:229-42. [PMID: 24783054 PMCID: PMC3923603 DOI: 10.1159/000357079] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2013] [Revised: 11/07/2013] [Indexed: 12/16/2022] Open
Abstract
Thyroid dysgenesis (TD) resulting from defects during embryonic thyroid development represents a major cause of congenital hypothyroidism. The pathogenetic mechanisms of TD in human newborns, however, are still poorly understood and disease-causing genetic variants have been identified in only a small percentage of TD cases. This limited understanding of the pathogenesis of TD is partly due to a lack of knowledge on how intrinsic factors and extrinsic signalling cues orchestrate the differentiation of thyroid follicular cells and the morphogenesis of thyroid tissue. Recently, embryonic stem cells and zebrafish embryos emerged as novel model systems that allow for innovative experimental approaches in order to decipher cellular and molecular mechanisms of thyroid development and to unravel pathogenic mechanisms of TD. Zebrafish embryos offer several salient properties for studies on thyroid organogenesis including rapid and external development, optical transparency, ease of breeding, relative short generation time and amenability for genome editing. In this review, we will highlight recent advances in the zebrafish toolkit to visualize cellular dynamics of organ development and discuss specific prospects of the zebrafish model for studies on vertebrate thyroid development and human congenital thyroid diseases.
Collapse
Affiliation(s)
- Robert Opitz
- Institute of Interdisciplinary Research in Molecular Human Biology, Université Libre de Bruxelles, Brussels, Belgium
| | - Francesco Antonica
- Institute of Interdisciplinary Research in Molecular Human Biology, Université Libre de Bruxelles, Brussels, Belgium
| | - Sabine Costagliola
- Institute of Interdisciplinary Research in Molecular Human Biology, Université Libre de Bruxelles, Brussels, Belgium
| |
Collapse
|
120
|
Liu WH, Ren LN, Chen T, You N, Liu LY, Wang T, Yan HT, Luo H, Tang LJ. Unbalanced distribution of materials: the art of giving rise to hepatocytes from liver stem/progenitor cells. J Cell Mol Med 2013; 18:1-14. [PMID: 24286303 PMCID: PMC3916112 DOI: 10.1111/jcmm.12183] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2013] [Accepted: 10/08/2013] [Indexed: 12/12/2022] Open
Abstract
Liver stem/progenitor cells (LSPCs) are able to duplicate themselves and differentiate into each type of cells in the liver, including mature hepatocytes and cholangiocytes. Understanding how to accurately control the hepatic differentiation of LSPCs is a challenge in many fields from preclinical to clinical treatments. This review summarizes the recent advances made to control the hepatic differentiation of LSPCs over the last few decades. The hepatic differentiation of LSPCs is a gradual process consisting of three main steps: initiation, progression and accomplishment. The unbalanced distribution of the affecting materials in each step results in the hepatic maturation of LSPCs. As the innovative and creative works for generating hepatocytes with full functions from LSPCs are gradually accumulated, LSPC therapies will soon be a new choice for treating liver diseases.
Collapse
Affiliation(s)
- Wei-Hui Liu
- General Surgery Center of PLA, Chengdu Military General Hospital, Chengdu, Sichuan Province, China
| | | | | | | | | | | | | | | | | |
Collapse
|
121
|
Abstract
The liver performs a large number of essential synthetic and regulatory functions that are acquired during fetal development and persist throughout life. Their disruption underlies a diverse group of heritable and acquired diseases that affect both pediatric and adult patients. Although experimental analyses used to study liver development and disease are typically performed in cell culture models or rodents, the zebrafish is increasingly used to complement discoveries made in these systems. Forward and reverse genetic analyses over the past two decades have shown that the molecular program for liver development is largely conserved between zebrafish and mammals, and that the zebrafish can be used to model heritable human liver disorders. Recent work has demonstrated that zebrafish can also be used to study the mechanistic basis of acquired liver diseases. Here, we provide a comprehensive summary of how the zebrafish has contributed to our understanding of human liver development and disease.
Collapse
Affiliation(s)
- Benjamin J Wilkins
- Department of Pathology and Laboratory Medicine, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | | |
Collapse
|
122
|
Abstract
Liver regeneration is perhaps the most studied example of compensatory growth aimed to replace loss of tissue in an organ. Hepatocytes, the main functional cells of the liver, manage to proliferate to restore mass and to simultaneously deliver all functions hepatic functions necessary to maintain body homeostasis. They are the first cells to respond to regenerative stimuli triggered by mitogenic growth factor receptors MET (the hepatocyte growth factor receptor] and epidermal growth factor receptor and complemented by auxiliary mitogenic signals induced by other cytokines. Termination of liver regeneration is a complex process affected by integrin mediated signaling and it restores the organ to its original mass as determined by the needs of the body (hepatostat function). When hepatocytes cannot proliferate, progenitor cells derived from the biliary epithelium transdifferentiate to restore the hepatocyte compartment. In a reverse situation, hepatocytes can also transdifferentiate to restore the biliary compartment. Several hormones and xenobiotics alter the hepatostat directly and induce an increase in liver to body weight ratio (augmentative hepatomegaly). The complex challenges of the liver toward body homeostasis are thus always preserved by complex but unfailing responses involving orchestrated signaling and affecting growth and differentiation of all hepatic cell types.
Collapse
Affiliation(s)
- George K Michalopoulos
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA.
| |
Collapse
|
123
|
Rodríguez-Seguel E, Mah N, Naumann H, Pongrac IM, Cerdá-Esteban N, Fontaine JF, Wang Y, Chen W, Andrade-Navarro MA, Spagnoli FM. Mutually exclusive signaling signatures define the hepatic and pancreatic progenitor cell lineage divergence. Genes Dev 2013; 27:1932-46. [PMID: 24013505 PMCID: PMC3778245 DOI: 10.1101/gad.220244.113] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
A key question in stem cell biology is how distinct cell types arise from common multipotent progenitor cells. It is unknown how liver and pancreas cells diverge from a common endoderm progenitor population and adopt specific fates. Using RNA-seq, Spagnoli and colleagues define the gene expression programs of liver and pancreas progenitors and identify the noncanonical Wnt pathway as a potential developmental regulator of this fate decision. Furthermore, this study provides a framework for lineage-reprogramming strategies to convert adult hepatic cells into pancreatic cells. Understanding how distinct cell types arise from multipotent progenitor cells is a major quest in stem cell biology. The liver and pancreas share many aspects of their early development and possibly originate from a common progenitor. However, how liver and pancreas cells diverge from a common endoderm progenitor population and adopt specific fates remains elusive. Using RNA sequencing (RNA-seq), we defined the molecular identity of liver and pancreas progenitors that were isolated from the mouse embryo at two time points, spanning the period when the lineage decision is made. The integration of temporal and spatial gene expression profiles unveiled mutually exclusive signaling signatures in hepatic and pancreatic progenitors. Importantly, we identified the noncanonical Wnt pathway as a potential developmental regulator of this fate decision and capable of inducing the pancreas program in endoderm and liver cells. Our study offers an unprecedented view of gene expression programs in liver and pancreas progenitors and forms the basis for formulating lineage-reprogramming strategies to convert adult hepatic cells into pancreatic cells.
Collapse
|
124
|
Tsedensodnom O, Sadler KC. ROS: redux and paradox in fatty liver disease. Hepatology 2013; 58:1210-2. [PMID: 23703804 PMCID: PMC4072499 DOI: 10.1002/hep.26497] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2013] [Accepted: 04/23/2013] [Indexed: 12/16/2022]
Affiliation(s)
- Orkhontuya Tsedensodnom
- Department of Medicine, Division of Liver Diseases, Icahn School of Medicine at Mount Sinai, New York, NY,Department of Developmental and Regenerative, Biology, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Kirsten C. Sadler
- Department of Medicine, Division of Liver Diseases, Icahn School of Medicine at Mount Sinai, New York, NY,Department of Developmental and Regenerative, Biology, Icahn School of Medicine at Mount Sinai, New York, NY,Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY
| |
Collapse
|
125
|
Nussbaum JM, Liu LJ, Hasan SA, Schaub M, McClendon A, Stainier DY, Sakaguchi TF. Homeostatic generation of reactive oxygen species protects the zebrafish liver from steatosis. Hepatology 2013; 58:1326-38. [PMID: 23744565 PMCID: PMC3791216 DOI: 10.1002/hep.26551] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2012] [Accepted: 05/21/2013] [Indexed: 12/11/2022]
Abstract
UNLABELLED Nonalcoholic fatty liver disease is the most common liver disease in both adults and children. The earliest stage of this disease is hepatic steatosis, in which triglycerides are deposited as cytoplasmic lipid droplets in hepatocytes. Through a forward genetic approach in zebrafish, we found that guanosine monophosphate (GMP) synthetase mutant larvae develop hepatic steatosis. We further demonstrate that activity of the small GTPase Rac1 and Rac1-mediated production of reactive oxygen species (ROS) are down-regulated in GMP synthetase mutant larvae. Inhibition of Rac1 activity or ROS production in wild-type larvae by small molecule inhibitors was sufficient to induce hepatic steatosis. More conclusively, treating larvae with hydrogen peroxide, a diffusible ROS that has been implicated as a signaling molecule, alleviated hepatic steatosis in both GMP synthetase mutant and Rac1 inhibitor-treated larvae, indicating that homeostatic production of ROS is required to prevent hepatic steatosis. We further found that ROS positively regulate the expression of the triglyceride hydrolase gene, which is responsible for the mobilization of stored triglycerides in hepatocytes. Consistently, inhibition of triglyceride hydrolase activity in wild-type larvae by a small molecule inhibitor was sufficient to induce hepatic steatosis. CONCLUSION De novo GMP synthesis influences the activation of the small GTPase Rac1, which controls hepatic lipid dynamics through ROS-mediated regulation of triglyceride hydrolase expression in hepatocytes.
Collapse
Affiliation(s)
- Justin M. Nussbaum
- Department of Stem Cell Biology and Regenerative Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195, USA
| | - Liuhong J. Liu
- Department of Stem Cell Biology and Regenerative Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195, USA
| | - Syeda A. Hasan
- Department of Stem Cell Biology and Regenerative Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195, USA
| | - Madeline Schaub
- Department of Stem Cell Biology and Regenerative Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195, USA
| | - Allyson McClendon
- Department of Stem Cell Biology and Regenerative Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195, USA
| | - Didier Y.R. Stainier
- Department of Biochemistry and Biophysics, Programs in Developmental and Stem Cell Biology, Genetics, and Human Genetics, and Liver Center, University of California, San Francisco, San Francisco, CA 94158
| | - Takuya F. Sakaguchi
- Department of Stem Cell Biology and Regenerative Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195, USA,Department of Biochemistry and Biophysics, Programs in Developmental and Stem Cell Biology, Genetics, and Human Genetics, and Liver Center, University of California, San Francisco, San Francisco, CA 94158,Author for correspondence: ()
| |
Collapse
|
126
|
Hochgreb-Hägele T, Yin C, Koo DES, Bronner ME, Stainier DYR. Laminin β1a controls distinct steps during the establishment of digestive organ laterality. Development 2013; 140:2734-45. [PMID: 23757411 DOI: 10.1242/dev.097618] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Visceral organs, including the liver and pancreas, adopt asymmetric positions to ensure proper function. Yet the molecular and cellular mechanisms controlling organ laterality are not well understood. We identified a mutation affecting zebrafish laminin β1a (lamb1a) that disrupts left-right asymmetry of the liver and pancreas. In these mutants, the liver spans the midline and the ventral pancreatic bud remains split into bilateral structures. We show that lamb1a regulates asymmetric left-right gene expression in the lateral plate mesoderm (LPM). In particular, lamb1a functions in Kupffer's vesicle (KV), a ciliated organ analogous to the mouse node, to control the length and function of the KV cilia. Later during gut-looping stages, dynamic expression of Lamb1a is required for the bilayered organization and asymmetric migration of the LPM. Loss of Lamb1a function also results in aberrant protrusion of LPM cells into the gut. Collectively, our results provide cellular and molecular mechanisms by which extracellular matrix proteins regulate left-right organ morphogenesis.
Collapse
Affiliation(s)
- Tatiana Hochgreb-Hägele
- Department of Biochemistry and Biophysics, Programs in Developmental and Stem Cell Biology, Genetics and Human Genetics, Liver Center and Diabetes Center, Institute for Regeneration Medicine, University of California, San Francisco, CA 94158, USA.
| | | | | | | | | |
Collapse
|
127
|
Nolan DJ, Ginsberg M, Israely E, Palikuqi B, Poulos MG, James D, Ding BS, Schachterle W, Liu Y, Rosenwaks Z, Butler JM, Xiang J, Rafii A, Shido K, Rabbany SY, Elemento O, Rafii S. Molecular signatures of tissue-specific microvascular endothelial cell heterogeneity in organ maintenance and regeneration. Dev Cell 2013; 26:204-19. [PMID: 23871589 DOI: 10.1016/j.devcel.2013.06.017] [Citation(s) in RCA: 461] [Impact Index Per Article: 41.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2013] [Revised: 04/01/2013] [Accepted: 06/18/2013] [Indexed: 02/08/2023]
Abstract
Microvascular endothelial cells (ECs) within different tissues are endowed with distinct but as yet unrecognized structural, phenotypic, and functional attributes. We devised EC purification, cultivation, profiling, and transplantation models that establish tissue-specific molecular libraries of ECs devoid of lymphatic ECs or parenchymal cells. These libraries identify attributes that confer ECs with their organotypic features. We show that clusters of transcription factors, angiocrine growth factors, adhesion molecules, and chemokines are expressed in unique combinations by ECs of each organ. Furthermore, ECs respond distinctly in tissue regeneration models, hepatectomy, and myeloablation. To test the data set, we developed a transplantation model that employs generic ECs differentiated from embryonic stem cells. Transplanted generic ECs engraft into regenerating tissues and acquire features of organotypic ECs. Collectively, we demonstrate the utility of informational databases of ECs toward uncovering the extravascular and intrinsic signals that define EC heterogeneity. These factors could be exploited therapeutically to engineer tissue-specific ECs for regeneration.
Collapse
Affiliation(s)
- Daniel J Nolan
- Department of Genetic Medicine, Howard Hughes Medical Institute, Weill Cornell Medical College, New York, NY 10065, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
128
|
Lancman JJ, Zvenigorodsky N, Gates KP, Zhang D, Solomon K, Humphrey RK, Kuo T, Setiawan L, Verkade H, Chi YI, Jhala US, Wright CVE, Stainier DYR, Dong PDS. Specification of hepatopancreas progenitors in zebrafish by hnf1ba and wnt2bb. Development 2013; 140:2669-79. [PMID: 23720049 PMCID: PMC3678338 DOI: 10.1242/dev.090993] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/25/2013] [Indexed: 12/16/2022]
Abstract
Although the liver and ventral pancreas are thought to arise from a common multipotent progenitor pool, it is unclear whether these progenitors of the hepatopancreas system are specified by a common genetic mechanism. Efforts to determine the role of Hnf1b and Wnt signaling in this crucial process have been confounded by a combination of factors, including a narrow time frame for hepatopancreas specification, functional redundancy among Wnt ligands, and pleiotropic defects caused by either severe loss of Wnt signaling or Hnf1b function. Using a novel hypomorphic hnf1ba zebrafish mutant that exhibits pancreas hypoplasia, as observed in HNF1B monogenic diabetes, we show that hnf1ba plays essential roles in regulating β-cell number and pancreas specification, distinct from its function in regulating pancreas size and liver specification, respectively. By combining Hnf1ba partial loss of function with conditional loss of Wnt signaling, we uncover a crucial developmental window when these pathways synergize to specify the entire ventrally derived hepatopancreas progenitor population. Furthermore, our in vivo genetic studies demonstrate that hnf1ba generates a permissive domain for Wnt signaling activity in the foregut endoderm. Collectively, our findings provide a new model for HNF1B function, yield insight into pancreas and β-cell development, and suggest a new mechanism for hepatopancreatic specification.
Collapse
Affiliation(s)
- Joseph J. Lancman
- Sanford Children’s Health Research Center, Programs in Genetic Disease, Development and Aging, and Stem Cell and Regenerative Biology, Graduate School of Biomedical Sciences, Sanford-Burnham Medical Research Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Natasha Zvenigorodsky
- Department of Biochemistry and Biophysics, Programs in Developmental Biology, Genetics and Human Genetics, and the Diabetes Center and Liver Center, University of California, San Francisco, 1550 Fourth Street, San Francisco, CA 94158, USA
| | - Keith P. Gates
- Sanford Children’s Health Research Center, Programs in Genetic Disease, Development and Aging, and Stem Cell and Regenerative Biology, Graduate School of Biomedical Sciences, Sanford-Burnham Medical Research Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Danhua Zhang
- Sanford Children’s Health Research Center, Programs in Genetic Disease, Development and Aging, and Stem Cell and Regenerative Biology, Graduate School of Biomedical Sciences, Sanford-Burnham Medical Research Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Keely Solomon
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Rohan K. Humphrey
- Pediatric Diabetes Research Center, UCSD School of Medicine, La Jolla CA 92037, USA
| | - Taiyi Kuo
- Department of Biochemistry and Biophysics, Programs in Developmental Biology, Genetics and Human Genetics, and the Diabetes Center and Liver Center, University of California, San Francisco, 1550 Fourth Street, San Francisco, CA 94158, USA
| | - Linda Setiawan
- Department of Biochemistry and Biophysics, Programs in Developmental Biology, Genetics and Human Genetics, and the Diabetes Center and Liver Center, University of California, San Francisco, 1550 Fourth Street, San Francisco, CA 94158, USA
| | - Heather Verkade
- Department of Biochemistry and Biophysics, Programs in Developmental Biology, Genetics and Human Genetics, and the Diabetes Center and Liver Center, University of California, San Francisco, 1550 Fourth Street, San Francisco, CA 94158, USA
- School of Biological Sciences, Monash University, Clayton, VIC 3800, Australia
| | - Young-In Chi
- Hormel Institute, University of Minnesota, Austin, MN 55912, USA
| | - Ulupi S. Jhala
- Pediatric Diabetes Research Center, UCSD School of Medicine, La Jolla CA 92037, USA
| | - Christopher V. E. Wright
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Didier Y. R. Stainier
- Department of Biochemistry and Biophysics, Programs in Developmental Biology, Genetics and Human Genetics, and the Diabetes Center and Liver Center, University of California, San Francisco, 1550 Fourth Street, San Francisco, CA 94158, USA
| | - P. Duc Si Dong
- Sanford Children’s Health Research Center, Programs in Genetic Disease, Development and Aging, and Stem Cell and Regenerative Biology, Graduate School of Biomedical Sciences, Sanford-Burnham Medical Research Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA
| |
Collapse
|
129
|
Lu H, Ma J, Yang Y, Shi W, Luo L. EpCAM is an endoderm-specific Wnt derepressor that licenses hepatic development. Dev Cell 2013; 24:543-53. [PMID: 23484855 DOI: 10.1016/j.devcel.2013.01.021] [Citation(s) in RCA: 123] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2012] [Revised: 11/26/2012] [Accepted: 01/28/2013] [Indexed: 12/14/2022]
Abstract
Mechanisms underlying cell-type-specific response to morphogens or signaling molecules during embryonic development are poorly understood. To learn how response to the liver-inductive Wnt2bb signal is achieved, we identify an endoderm-enriched, single transmembrane protein, epithelial-cell-adhesion-molecule (EpCAM), as an endoderm-specific Wnt derepressor in zebrafish. hi2151/epcam mutants exhibit defective liver development similar to prt/wnt2bb mutants. EpCAM directly binds to Kremen1 and disrupts the Kremen1-Dickkopf2 (Dkk2) interaction, which prevents Kremen1-Dkk2-mediated removal of Lipoprotein-receptor-related protein 6 (Lrp6) from the cell surface. These data lead to a model in which EpCAM derepresses Lrp6 and cooperates with Wnt ligand to activate Wnt signaling through stabilizing membrane Lrp6 and allowing Lrp6 clustering into active signalosomes. Thus, EpCAM cell autonomously licenses and cooperatively activates Wnt2bb signaling in endodermal cells. Our results identify EpCAM as the key molecule and its functional mechanism to confer endodermal cells the competence to respond to the liver-inductive Wnt2bb signal.
Collapse
Affiliation(s)
- Huiqiang Lu
- Key Laboratory of Freshwater Fish Reproduction and Development, Ministry of Education, Laboratory of Molecular Developmental Biology, School of Life Sciences, Southwest University, Beibei, 400715 Chongqing, China
| | | | | | | | | |
Collapse
|
130
|
Boulter L, Lu WY, Forbes SJ. Differentiation of progenitors in the liver: a matter of local choice. J Clin Invest 2013; 123:1867-73. [PMID: 23635784 PMCID: PMC3635730 DOI: 10.1172/jci66026] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The liver is a complex organ that requires multiple rounds of cell fate decision for development and homeostasis throughout the lifetime. During the earliest phases of organogenesis, the liver acquires a separate lineage from the pancreas and the intestine, and subsequently, the liver bud must appropriately differentiate to form metabolic hepatocytes and cholangiocytes for proper hepatic physiology. In addition, throughout life, the liver is bombarded with chemical and pathological insults, which require the activation and correct differentiation of adult progenitor cells. This Review seeks to provide an overview of the complex signaling relationships that allow these tightly regulated processes to occur.
Collapse
Affiliation(s)
- Luke Boulter
- Medical Research Council Centre for Regenerative Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | | | | |
Collapse
|
131
|
Yamashita T, Wang XW. Cancer stem cells in the development of liver cancer. J Clin Invest 2013; 123:1911-8. [PMID: 23635789 DOI: 10.1172/jci66024] [Citation(s) in RCA: 404] [Impact Index Per Article: 36.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Liver cancer is an aggressive disease with a poor outcome. Several hepatic stem/progenitor markers are useful for isolating a subset of liver cells with stem cell features, known as cancer stem cells (CSCs). These cells are responsible for tumor relapse, metastasis, and chemoresistance. Liver CSCs dictate a hierarchical organization that is shared in both organogenesis and tumorigenesis. An increased understanding of the molecular signaling events that regulate cellular hierarchy and stemness, and success in defining key CSC-specific genes, have opened up new avenues to accelerate the development of novel diagnostic and treatment strategies. This Review highlights recent advances in understanding the pathogenesis of liver CSCs and discusses unanswered questions about the concept of liver CSCs.
Collapse
Affiliation(s)
- Taro Yamashita
- Department of General Medicine, Kanazawa University Hospital, Kanazawa, Ishikawa, Japan.
| | | |
Collapse
|
132
|
Stuckenholz C, Lu L, Thakur PC, Choi TY, Shin D, Bahary N. Sfrp5 modulates both Wnt and BMP signaling and regulates gastrointestinal organogenesis [corrected] in the zebrafish, Danio rerio. PLoS One 2013; 8:e62470. [PMID: 23638093 PMCID: PMC3639276 DOI: 10.1371/journal.pone.0062470] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2012] [Accepted: 03/21/2013] [Indexed: 02/08/2023] Open
Abstract
Sfrp5 belongs to the family of secreted frizzled related proteins (Sfrp), secreted inhibitors of Wingless-MMTV Integration Site (Wnt) signaling, which play an important role in cancer and development. We selected sfrp5 because of its compelling expression profile in the developing endoderm in zebrafish, Danio rerio. In this study, overexpression of sfrp5 in embryos results in defects in both convergent extension (CE) by inhibition of non-canonical Wnt signaling and defects in dorsoventral patterning by inhibition of Tolloid-mediated proteolysis of the BMP inhibitor Chordin. From 25 hours post fertilization (hpf) to 3 days post fertilization (dpf), both overexpression and knockdown of Sfrp5 decrease the size of the endoderm, significantly reducing liver cell number. At 3 dpf, insulin-positive endodermal cells fail to coalesce into a single pancreatic islet. We show that Sfrp5 inhibits both canonical and non-canonical Wnt signaling during embryonic and endodermal development, resulting in endodermal abnormalities.
Collapse
Affiliation(s)
- Carsten Stuckenholz
- Department of Medicine, Division of Hematology/Oncology, University of Pittsburgh Medical Center, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Lili Lu
- Department of Medicine, Division of Hematology/Oncology, University of Pittsburgh Medical Center, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Prakash C. Thakur
- Department of Medicine, Division of Hematology/Oncology, University of Pittsburgh Medical Center, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Tae-Young Choi
- Department of Developmental Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Donghun Shin
- Department of Developmental Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Nathan Bahary
- Department of Medicine, Division of Hematology/Oncology, University of Pittsburgh Medical Center, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
133
|
The zebrafish reference genome sequence and its relationship to the human genome. Nature 2013; 496:498-503. [PMID: 23594743 DOI: 10.1038/nature12111] [Citation(s) in RCA: 3104] [Impact Index Per Article: 282.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2012] [Accepted: 03/21/2013] [Indexed: 02/07/2023]
Abstract
Zebrafish have become a popular organism for the study of vertebrate gene function. The virtually transparent embryos of this species, and the ability to accelerate genetic studies by gene knockdown or overexpression, have led to the widespread use of zebrafish in the detailed investigation of vertebrate gene function and increasingly, the study of human genetic disease. However, for effective modelling of human genetic disease it is important to understand the extent to which zebrafish genes and gene structures are related to orthologous human genes. To examine this, we generated a high-quality sequence assembly of the zebrafish genome, made up of an overlapping set of completely sequenced large-insert clones that were ordered and oriented using a high-resolution high-density meiotic map. Detailed automatic and manual annotation provides evidence of more than 26,000 protein-coding genes, the largest gene set of any vertebrate so far sequenced. Comparison to the human reference genome shows that approximately 70% of human genes have at least one obvious zebrafish orthologue. In addition, the high quality of this genome assembly provides a clearer understanding of key genomic features such as a unique repeat content, a scarcity of pseudogenes, an enrichment of zebrafish-specific genes on chromosome 4 and chromosomal regions that influence sex determination.
Collapse
|
134
|
Tao T, Shi H, Huang D, Peng J. Def functions as a cell autonomous factor in organogenesis of digestive organs in zebrafish. PLoS One 2013; 8:e58858. [PMID: 23593122 PMCID: PMC3625206 DOI: 10.1371/journal.pone.0058858] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2012] [Accepted: 02/08/2013] [Indexed: 01/29/2023] Open
Abstract
Digestive organs originate from the endoderm. Morphogenesis of the digestive system is precisely controlled by multiple factors that dictate the cell fate and behavior so that the specific digestive organs are timely formed in the right place and develop into right size and structure. We showed previously that digestive organ expansion factor (def) is a gene whose expression is enriched in the liver, pancreas and intestine. Loss-of-function of def in the defhi429 mutant confers hypoplastic digestive organs partly due to alteration of expression of genes related to the p53 pathway. However, the molecular mechanism for the involvement of Def in the organogenesis of digestive organs is still largely unknown. For example, it is not known whether Def regulates specific pathways in a specific organ. To address this question, we generated four independent Tg(fabp10a:def) transgenic fish lines which over-expressed Def specifically in the liver. We characterized Tg-I, one of the transgenic lines, in detail with genetic, molecular and histological approaches. We found that Tg-I restored the liver but not exocrine pancreas and intestine development in the defhi429 mutant. However, Tg-I adult fish in the wild type (WT) background exhibits reduced liver-to-body ratio and all four transgenic lines conferred abnormal intrahepatic structure. Microarray data analysis showed that certain specific functional pathways were affected in the liver of Tg-I. These results demonstrate that Def functions in a cell autonomous manner during early liver development and aberrant Def protein expression might lead to disruption of the structural integrity of a normal adult liver.
Collapse
Affiliation(s)
- Ting Tao
- Key Laboratory for Molecular Animal Nutrition, Ministry of Education, College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang Province, China
| | - Hui Shi
- Key Laboratory for Molecular Animal Nutrition, Ministry of Education, College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang Province, China
| | - Delai Huang
- Key Laboratory for Molecular Animal Nutrition, Ministry of Education, College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang Province, China
| | - Jinrong Peng
- Key Laboratory for Molecular Animal Nutrition, Ministry of Education, College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang Province, China
- * E-mail:
| |
Collapse
|
135
|
Zhang Z, Rankin SA, Zorn AM. Different thresholds of Wnt-Frizzled 7 signaling coordinate proliferation, morphogenesis and fate of endoderm progenitor cells. Dev Biol 2013; 378:1-12. [PMID: 23562607 DOI: 10.1016/j.ydbio.2013.02.024] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2012] [Revised: 02/06/2013] [Accepted: 02/22/2013] [Indexed: 11/27/2022]
Abstract
Wnt signaling has multiple dynamic roles during development of the gastrointestinal and respiratory systems. Differential Wnt signaling is thought to be a critical step in Xenopus endoderm patterning such that during late gastrula and early somite stages of embryogenesis, Wnt activity must be suppressed in the anterior to allow the specification of foregut progenitors. However, the foregut endoderm also expresses the Wnt-receptor Frizzled 7 (Fzd7) as well as several Wnt ligands suggesting that the current model may be too simple. In this study, we show that Fzd7 is required to transduce a low level of Wnt signaling that is essential to maintain foregut progenitors. Foregut-specific Fzd7-depletion from the Xenopus foregut resulted in liver and pancreas agenesis. Fzd7-depleted embryos failed to maintain the foregut progenitor marker hhex and exhibited decreased proliferation; in addition the foregut cells were enlarged with a randomized orientation. We show that in the foregut Fzd7 signals via both the Wnt/β-catenin and Wnt/JNK pathways and that different thresholds of Wnt-Fzd7 activity coordinate progenitor cell fate, proliferation and morphogenesis.
Collapse
Affiliation(s)
- Zheng Zhang
- Perinatal Institute, Division of Developmental Biology, Cincinnati Children's Hospital Medical Center and the College of Medicine, University of Cincinnati, Cincinnati OH 45229, USA
| | | | | |
Collapse
|
136
|
Abstract
Liver is a prime organ responsible for synthesis, metabolism, and detoxification. The organ is endodermal in origin and its development is regulated by temporal, complex, and finely balanced cellular and molecular interactions that dictate its origin, growth, and maturation. We discuss the relevance of endoderm patterning, which truly is the first step toward mapping of domains that will give rise to specific organs. Once foregut patterning is completed, certain cells within the foregut endoderm gain competence in the form of expression of certain transcription factors that allow them to respond to certain inductive signals. Hepatic specification is then a result of such inductive signals, which often emanate from the surrounding mesenchyme. During hepatic specification bipotential hepatic stem cells or hepatoblasts become apparent and undergo expansion, which results in a visible liver primordium during the stage of hepatic morphogenesis. Hepatoblasts next differentiate into either hepatocytes or cholangiocytes. The expansion and differentiation is regulated by cellular and molecular interactions between hepatoblasts and mesenchymal cells including sinusoidal endothelial cells, stellate cells, and also innate hematopoietic elements. Further maturation of hepatocytes and cholangiocytes continues during late hepatic development as a function of various growth factors. At this time, liver gains architectural novelty in the form of zonality and at cellular level acquires polarity. A comprehensive elucidation of such finely tuned developmental cues have been the basis of transdifferentiation of various types of stem cells to hepatocyte-like cells for purposes of understanding health and disease and for therapeutic applications.
Collapse
Affiliation(s)
- Donghun Shin
- Department of Developmental Biology, University of Pittsburgh, School of Medicine, Pittsburgh, Pennsylvania, USA.
| | | |
Collapse
|
137
|
Wntless is required for peripheral lung differentiation and pulmonary vascular development. Dev Biol 2013; 379:38-52. [PMID: 23523683 DOI: 10.1016/j.ydbio.2013.03.010] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2012] [Revised: 02/21/2013] [Accepted: 03/12/2013] [Indexed: 12/12/2022]
Abstract
Wntless (Wls), a gene highly conserved across the animal kingdom, encodes for a transmembrane protein that mediates Wnt ligand secretion. Wls is expressed in developing lung, wherein Wnt signaling is necessary for pulmonary morphogenesis. We hypothesize that Wls plays a critical role in modulating Wnt signaling during lung development and therefore affects processes critical for pulmonary morphogenesis. We generated conditional Wls mutant mice utilizing Shh-Cre and Dermo1-Cre mice to delete Wls in the embryonic respiratory epithelium and mesenchyme, respectively. Epithelial deletion of Wls disrupted lung branching morphogenesis, peripheral lung development and pulmonary endothelial differentiation. Epithelial Wls mutant mice died at birth due to respiratory failure caused by lung hypoplasia and pulmonary hemorrhage. In the lungs of these mice, VEGF and Tie2-angiopoietin signaling pathways, which mediate vascular development, were downregulated from early stages of development. In contrast, deletion of Wls in mesenchymal cells of the developing lung did not alter branching morphogenesis or early mesenchymal differentiation. In vitro assays support the concept that Wls acts in part via Wnt5a to regulate pulmonary vascular development. We conclude that epithelial Wls modulates Wnt ligand activities critical for pulmonary vascular differentiation and peripheral lung morphogenesis. These studies provide a new framework for understanding the molecular mechanisms underlying normal pulmonary vasculature formation and the dysmorphic pulmonary vasculature development associated with congenital lung disease.
Collapse
|
138
|
Koltowska K, Apitz H, Stamataki D, Hirst EMA, Verkade H, Salecker I, Ober EA. Ssrp1a controls organogenesis by promoting cell cycle progression and RNA synthesis. Development 2013; 140:1912-8. [PMID: 23515471 DOI: 10.1242/dev.093583] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Tightly controlled DNA replication and RNA transcription are essential for differentiation and tissue growth in multicellular organisms. Histone chaperones, including the FACT (facilitates chromatin transcription) complex, are central for these processes and act by mediating DNA access through nucleosome reorganisation. However, their roles in vertebrate organogenesis are poorly understood. Here, we report the identification of zebrafish mutants for the gene encoding Structure specific recognition protein 1a (Ssrp1a), which, together with Spt16, forms the FACT heterodimer. Focussing on the liver and eye, we show that zygotic Ssrp1a is essential for proliferation and differentiation during organogenesis. Specifically, gene expression indicative of progressive organ differentiation is disrupted and RNA transcription is globally reduced. Ssrp1a-deficient embryos exhibit DNA synthesis defects and prolonged S phase, uncovering a role distinct from that of Spt16, which promotes G1 phase progression. Gene deletion/replacement experiments in Drosophila show that Ssrp1b, Ssrp1a and N-terminal Ssrp1a, equivalent to the yeast homologue Pob3, can substitute Drosophila Ssrp function. These data suggest that (1) Ssrp1b does not compensate for Ssrp1a loss in the zebrafish embryo, probably owing to insufficient expression levels, and (2) despite fundamental structural differences, the mechanisms mediating DNA accessibility by FACT are conserved between yeast and metazoans. We propose that the essential functions of Ssrp1a in DNA replication and gene transcription, together with its dynamic spatiotemporal expression, ensure organ-specific differentiation and proportional growth, which are crucial for the forming embryo.
Collapse
Affiliation(s)
- Katarzyna Koltowska
- Developmental Biology, MRC National Institute for Medical Research, London, UK
| | | | | | | | | | | | | |
Collapse
|
139
|
Autophagy induction is a Tor- and Tp53-independent cell survival response in a zebrafish model of disrupted ribosome biogenesis. PLoS Genet 2013; 9:e1003279. [PMID: 23408911 PMCID: PMC3567153 DOI: 10.1371/journal.pgen.1003279] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2012] [Accepted: 12/12/2012] [Indexed: 12/27/2022] Open
Abstract
Ribosome biogenesis underpins cell growth and division. Disruptions in ribosome biogenesis and translation initiation are deleterious to development and underlie a spectrum of diseases known collectively as ribosomopathies. Here, we describe a novel zebrafish mutant, titania (ttis450), which harbours a recessive lethal mutation in pwp2h, a gene encoding a protein component of the small subunit processome. The biochemical impacts of this lesion are decreased production of mature 18S rRNA molecules, activation of Tp53, and impaired ribosome biogenesis. In ttis450, the growth of the endodermal organs, eyes, brain, and craniofacial structures is severely arrested and autophagy is up-regulated, allowing intestinal epithelial cells to evade cell death. Inhibiting autophagy in ttis450 larvae markedly reduces their lifespan. Somewhat surprisingly, autophagy induction in ttis450 larvae is independent of the state of the Tor pathway and proceeds unabated in Tp53-mutant larvae. These data demonstrate that autophagy is a survival mechanism invoked in response to ribosomal stress. This response may be of relevance to therapeutic strategies aimed at killing cancer cells by targeting ribosome biogenesis. In certain contexts, these treatments may promote autophagy and contribute to cancer cells evading cell death. Autophagy is an act of self-preservation whereby a cell responds to stressful conditions such as nutrient depletion and intense muscular activity by digesting its own cytoplasmic organelles and proteins to fuel its longer-term survival. An understanding of the wide spectrum of physiological stimuli that can trigger this beneficial cellular mechanism is only just starting to emerge. However, this process also has a negative side, since autophagy is exploited in certain pathological conditions, including cancer, to extend the lifespan of cells that would otherwise die. Our analysis of a new zebrafish mutant, titania (ttis450), with defective digestive organs and abnormal craniofacial structure, sheds further light on the physiological and pathological ramifications of autophagy. In (ttis450), an inherited mutation in a gene required for ribosome production provides a powerful stimulus to autophagy in affected tissues, allowing them to evade cell death. The phenotypic consequences of impaired ribosome biogenesis in our zebrafish model are reminiscent of some of the clinical features associated with a group of human syndromes known as ribosomopathies.
Collapse
|
140
|
Choe CP, Collazo A, Trinh LA, Pan L, Moens CB, Crump JG. Wnt-dependent epithelial transitions drive pharyngeal pouch formation. Dev Cell 2013; 24:296-309. [PMID: 23375584 DOI: 10.1016/j.devcel.2012.12.003] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2011] [Revised: 10/18/2012] [Accepted: 12/03/2012] [Indexed: 01/15/2023]
Abstract
The pharyngeal pouches, which form by budding of the foregut endoderm, are essential for segmentation of the vertebrate face. To date, the cellular mechanism and segmental nature of such budding have remained elusive. Here, we find that Wnt11r and Wnt4a from the head mesoderm and ectoderm, respectively, play distinct roles in the segmental formation of pouches in zebrafish. Time-lapse microscopy, combined with mutant and tissue-specific transgenic experiments, reveal requirements of Wnt signaling in two phases of endodermal epithelial transitions. Initially, Wnt11r and Rac1 destabilize the endodermal epithelium to promote the lateral movement of pouch-forming cells. Next, Wnt4a and Cdc42 signaling induce the rearrangement of maturing pouch cells into bilayers through junctional localization of the Alcama immunoglobulin-domain protein, which functions to restabilize adherens junctions. We propose that this dynamic control of epithelial morphology by Wnt signaling may be a common theme for the budding of organ anlagen from the endoderm.
Collapse
Affiliation(s)
- Chong Pyo Choe
- Broad California Institute of Regenerative Medicine Center, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | | | | | | | | | | |
Collapse
|
141
|
Garnaas MK, Cutting CC, Meyers A, Kelsey PB, Harris JM, North TE, Goessling W. Rargb regulates organ laterality in a zebrafish model of right atrial isomerism. Dev Biol 2012; 372:178-89. [PMID: 22982668 PMCID: PMC3697125 DOI: 10.1016/j.ydbio.2012.09.001] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2012] [Revised: 07/26/2012] [Accepted: 09/06/2012] [Indexed: 02/02/2023]
Abstract
Developmental signals determine organ morphology and position during embryogenesis. To discover novel modifiers of liver development, we performed a chemical genetic screen in zebrafish and identified retinoic acid as a positive regulator of hepatogenesis. Knockdown of the four RA receptors revealed that all receptors affect liver formation, however specific receptors exert differential effects. Rargb knockdown results in bilateral livers but does not impact organ size, revealing a unique role for Rargb in conferring left-right positional information. Bilateral populations of hepatoblasts are detectable in rargb morphants, indicating Rargb acts during hepatic specification to position the liver, and primitive endoderm is competent to form liver on both sides. Hearts remain at the midline and gut looping is perturbed in rargb morphants, suggesting Rargb affects lateral plate mesoderm migration. Overexpression of Bmp during somitogenesis similarly results in bilateral livers and midline hearts, and inhibition of Bmp signaling rescues the rargb morphant phenotype, indicating Rargb functions upstream of Bmp to regulate organ sidedness. Loss of rargb causes biliary and organ laterality defects as well as asplenia, paralleling symptoms of the human condition right atrial isomerism. Our findings uncover a novel role for RA in regulating organ laterality and provide an animal model of one form of human heterotaxia.
Collapse
Affiliation(s)
- Maija K Garnaas
- Genetics Division, Brigham and Women's Hospital, Harvard Medical School, Brigham and Women's Hospital, Boston, MA, USA
| | | | | | | | | | | | | |
Collapse
|
142
|
Abstract
The liver has an enormous potential to restore the parenchymal tissue loss due to injury. This is accomplished by the proliferation of either the hepatocytes or liver progenitor cells in cases where massive damage prohibits hepatocytes from entering the proliferative response. Under debate is still whether hepatic stem cells are involved in liver tissue maintenance and regeneration or even whether they exist at all. The definition of an adult tissue-resident stem cell comprises basic functional stem cell criteria like the potential of self-renewal, multipotent, i.e. at least bipotent differentiation capacity and serial transplantability featuring the ability of functional tissue repopulation. The relationship between a progenitor and its progeny should exemplify the lineage commitment from the putative stem cell to the differentiated cell. This is mainly assessed by lineage tracing and immunohistochemical identification of markers specific to progenitors and their descendants. Flow cytometry approaches revealed that the liver stem cell population in animals is likely to be heterogeneous giving rise to progeny with different molecular signatures, depending on the stimulus to activate the putative stem cell compartment. The stem cell criteria are met by a variety of cells identified in the fetal and adult liver both under normal and injury conditions. It is the purpose of this review to verify hepatic stem cell candidates in the light of the stem cell definition criteria mentioned. Also from this point of view adult stem cells from non-hepatic tissues such as bone marrow, umbilical cord blood or adipose tissue, have the potential to differentiate into cells featuring functional hepatocyte characteristics. This has great impact because it opens the possibility of generating hepatocyte-like cells from adult stem cells in a sufficient amount and quality for their therapeutical application to treat end-stage liver diseases by stem cell-based hepatocytes in place of whole organ transplantation.
Collapse
Affiliation(s)
- Bruno Christ
- Translational Centre for Regenerative Medicine-TRM, University of Leipzig, Philipp-Rosenthal-Straße 55, D-04103 Leipzig, Germany.
| | | |
Collapse
|
143
|
Nejak-Bowen K, Monga SP. Wnt/beta-catenin signaling in hepatic organogenesis. Organogenesis 2012; 4:92-9. [PMID: 19279720 DOI: 10.4161/org.4.2.5855] [Citation(s) in RCA: 82] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2008] [Accepted: 03/06/2008] [Indexed: 02/07/2023] Open
Abstract
Wnt/beta-catenin signaling has come to the forefront of liver biology in recent years. This pathway regulates key pathophysiological events inherent to the liver including development, regeneration and cancer, by dictating several biological processes such as proliferation, apoptosis, differentiation, adhesion, zonation and metabolism in various cells of the liver. This review will examine the studies that have uncovered the relevant roles of Wnt/beta-catenin signaling during the process of liver development. We will discuss the potential roles of Wnt/beta-catenin signaling during the phases of development, including competence, hepatic induction, expansion and morphogenesis. In addition, we will discuss the role of negative and positive regulation of this pathway and how the temporal expression of Wnt/beta-catenin can direct key processes during hepatic development. We will also identify some of the major deficits in the current understanding of the role of Wnt/beta-catenin signaling in liver development in order to provide a perspective for future studies. Thus, this review will provide a contextual overview of the role of Wnt/beta-catenin signaling during hepatic organogenesis.
Collapse
Affiliation(s)
- Kari Nejak-Bowen
- Department of Pathology University of Pittsburgh School of Medcine; Pittsburgh, Pennsylvania USA
| | | |
Collapse
|
144
|
Verzi MP, Shivdasani RA. Wnt signaling in gut organogenesis. Organogenesis 2012; 4:87-91. [PMID: 19279719 DOI: 10.4161/org.4.2.5854] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2008] [Accepted: 03/06/2008] [Indexed: 01/11/2023] Open
Abstract
Wnt signaling regulates some aspect of development of nearly all endoderm-derived organs and Wnts mediate both differentiation and proliferation at different steps during visceral organogenesis. Wnt2b induces liver formation in zebrafish 1 and may combine with other inducers, Fibroblast Growth Factors 1 & 4 and Bone Morphogenetic Protein 4, to specify the mammalian liver.2-5 Later in development, Wnts are critical for liver expansion and, finally, for terminal hepatocyte differentiation,6-12 as reviewed elsewhere in this issue (Monga). Likewise, in the pancreas, Wnts drive proliferation of exocrine and endocrine cells13,14 and promote acinar cell differentiation,13,15 as reviewed in the chapter by Murtaugh. Here we examine the intricate involvement of Wnt signaling in growth and differentiation of the digestive tract.
Collapse
Affiliation(s)
- Michael P Verzi
- Department of Medical Oncology; Dana-Farber Cancer Institute; and Department of Medicine; Harvard Medical School; Boston, Massachusetts, USA
| | | |
Collapse
|
145
|
Lehwald N, Tao GZ, Jang KY, Papandreou I, Liu B, Liu B, Pysz MA, Willmann JK, Knoefel WT, Denko NC, Sylvester KG. β-Catenin regulates hepatic mitochondrial function and energy balance in mice. Gastroenterology 2012; 143:754-764. [PMID: 22684045 DOI: 10.1053/j.gastro.2012.05.048] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2011] [Revised: 05/07/2012] [Accepted: 05/29/2012] [Indexed: 12/22/2022]
Abstract
BACKGROUND & AIMS Wnt signaling regulates hepatic function and nutrient homeostasis. However, little is known about the roles of β-catenin in cellular respiration or mitochondria of hepatocytes. METHODS We investigated β-catenin's role in the metabolic function of hepatocytes under homeostatic conditions and in response to metabolic stress using mice with hepatocyte-specific deletion of β-catenin and their wild-type littermates, given either saline (sham) or ethanol (as a model of binge drinking and acute ethanol intoxication). RESULTS Under homeostatic conditions, β-catenin-deficient hepatocytes demonstrated mitochondrial dysfunctions that included impairments to the tricarboxylic acid cycle and oxidative phosphorylation (OXPHOS) and decreased production of adenosine triphosphate (ATP). There was no evidence for redox imbalance or oxidative cellular injury in the absence of metabolic stress. In mice with β-catenin-deficient hepatocytes, ethanol intoxication led to significant redox imbalance in the hepatocytes and further deterioration in mitochondrial function that included reduced OXPHOS, fatty acid oxidation (FAO), and ATP production. Ethanol feeding significantly increased liver steatosis and oxidative damage, compared with wild-type mice, and disrupted the ratio of nicotinamide adenine dinucleotide. β-catenin-deficient hepatocytes also had showed disrupted signaling of Sirt1/peroxisome proliferator-activated receptor-α signaling. CONCLUSIONS β-catenin has an important role in the maintenance of mitochondrial homeostasis, regulating ATP production via the tricarboxylic acid cycle, OXPHOS, and fatty acid oxidation; β-catenin function in these systems is compromised under conditions of nutrient oxidative stress. Reagents that alter Wnt-β-catenin signaling might be developed as a useful new therapeutic strategy for treatment of liver disease.
Collapse
Affiliation(s)
- Nadja Lehwald
- Department of Surgery, Divison of Pediatric Surgery, Stanford University School of Medicine, Stanford, California; Department of General, Visceral, and Pediatric Surgery, School of Medicine, Heinrich Heine University, Duesseldorf, Germany
| | - Guo-Zhong Tao
- Department of Surgery, Divison of Pediatric Surgery, Stanford University School of Medicine, Stanford, California
| | - Kyu Yun Jang
- Department of Surgery, Divison of Pediatric Surgery, Stanford University School of Medicine, Stanford, California; Department of Pathology and Research Institute of Clinical Medicine, Chonbuk National University Medical School, South Korea
| | - Ioanna Papandreou
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, California
| | - Bowen Liu
- Department of Surgery, Divison of Pediatric Surgery, Stanford University School of Medicine, Stanford, California
| | - Bo Liu
- Department of Surgery, Divison of Pediatric Surgery, Stanford University School of Medicine, Stanford, California
| | - Marybeth A Pysz
- Department of Radiology, Molecular Imaging Program, Stanford University School of Medicine, Stanford, California
| | - Jürgen K Willmann
- Department of Radiology, Molecular Imaging Program, Stanford University School of Medicine, Stanford, California
| | - Wolfram T Knoefel
- Department of General, Visceral, and Pediatric Surgery, School of Medicine, Heinrich Heine University, Duesseldorf, Germany
| | - Nicholas C Denko
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, California
| | - Karl G Sylvester
- Department of Surgery, Divison of Pediatric Surgery, Stanford University School of Medicine, Stanford, California; The Lucile Packard Children's Hospital, Stanford, California.
| |
Collapse
|
146
|
Ribosome biogenesis factor Bms1-like is essential for liver development in zebrafish. J Genet Genomics 2012; 39:451-62. [PMID: 23021545 DOI: 10.1016/j.jgg.2012.07.007] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2012] [Revised: 07/11/2012] [Accepted: 07/12/2012] [Indexed: 11/20/2022]
Abstract
Ribosome biogenesis in the nucleolus requires numerous nucleolar proteins and small non-coding RNAs. Among them is ribosome biogenesis factor Bms1, which is highly conserved from yeast to human. In yeast, Bms1 initiates ribosome biogenesis through recruiting Rcl1 to pre-ribosomes. However, little is known about the biological function of Bms1 in vertebrates. Here we report that Bms1 plays an essential role in zebrafish liver development. We identified a zebrafish bms1l(sq163) mutant which carries a T to A mutation in the gene bms1-like (bms1l). This mutation results in L(152) to Q(152) substitution in a GTPase motif in Bms1l. Surprisingly, bms1l(sq163) mutation confers hypoplasia specifically in the liver, exocrine pancreas and intestine after 3 days post-fertilization (dpf). Consistent with the bms1l(sq163) mutant phenotypes, whole-mount in situ hybridization (WISH) on wild type embryos showed that bms1l transcripts are abundant in the entire digestive tract and its accessory organs. Immunostaining for phospho-Histone 3 (P-H3) and TUNEL assay revealed that impairment of hepatoblast proliferation rather than cell apoptosis is one of the consequences of bms1l(sq163) giving rise to an under-developed liver. Therefore, our findings demonstrate that Bms1l is necessary for zebrafish liver development.
Collapse
|
147
|
Tsai SM, Liu DW, Wang WP. Fibroblast growth factor (Fgf) signaling pathway regulates liver homeostasis in zebrafish. Transgenic Res 2012; 22:301-14. [DOI: 10.1007/s11248-012-9636-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2012] [Accepted: 07/05/2012] [Indexed: 02/08/2023]
|
148
|
So J, Martin BL, Kimelman D, Shin D. Wnt/β-catenin signaling cell-autonomously converts non-hepatic endodermal cells to a liver fate. Biol Open 2012; 2:30-6. [PMID: 23336074 PMCID: PMC3545266 DOI: 10.1242/bio.20122857] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2012] [Accepted: 09/24/2012] [Indexed: 01/05/2023] Open
Abstract
Wnt/β-catenin signaling plays multiple roles in liver development including hepatoblast proliferation and differentiation, hepatocyte differentiation, and liver zonation. A positive role for Wnt/β-catenin signaling in liver specification was recently identified in zebrafish; however, its underlying cellular mechanisms are unknown. Here, we present two cellular mechanisms by which Wnt/β-catenin signaling regulates liver specification. First, using lineage tracing we show that ectopic hepatoblasts, which form in the endoderm posterior to the liver upon activation of Wnt/β-catenin signaling, are derived from the direct conversion of non-hepatic endodermal cells, but not from the posterior migration of hepatoblasts. We found that endodermal cells at the 4-6(th) somite levels, which normally give rise to the intestinal bulb or intestine, gave rise to hepatoblasts in Wnt8a-overexpressing embryos, and that the distribution of traced endodermal cells in Wnt8a-overexpressing embryos was similar to that in controls. Second, by using an endoderm-restricted cell-transplantation technique and mosaic analysis with transgenic lines that cell-autonomously suppress or activate Wnt/β-catenin signaling upon heat-shock, we show that Wnt/β-catenin signaling acts cell-autonomously in endodermal cells to induce hepatic conversion. Altogether, these data demonstrate that Wnt/β-catenin signaling can induce the fate-change of non-hepatic endodermal cells into a liver fate in a cell-autonomous manner. These findings have potential application to hepatocyte differentiation protocols for the generation of mature hepatocytes from induced pluripotent stem cells, supplying a sufficient amount of hepatocytes for cell-based therapies to treat patients with severe liver diseases.
Collapse
Affiliation(s)
- Juhoon So
- Department of Developmental Biology, University of Pittsburgh , Pittsburgh, PA 15260 , USA
| | | | | | | |
Collapse
|
149
|
Huitema LFA, Renn J, Logister I, Gray JK, Waltz SE, Flik G, Schulte-Merker S. Macrophage-stimulating protein and calcium homeostasis in zebrafish. FASEB J 2012; 26:4092-101. [PMID: 22787265 DOI: 10.1096/fj.11-202663] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
To systematically identify novel gene functions essential for osteogenesis and skeletal mineralization, we performed a forward genetic mutagenesis screen in zebrafish and isolated a mutant that showed delayed skeletal mineralization. Analysis of the mutant phenotype in an osterix:nuclear-GFP transgenic background demonstrated that mutants contain osterix-expressing osteoblasts comparable to wild-type embryos. Positional cloning revealed a premature stop mutation in the macrophage-stimulating protein (msp) gene, predicted to result in a biologically inactive protein. Analysis of the embryonic expression pattern for the receptor for Msp, Ron, shows specific expression in the corpuscles of Stannius, a teleost-specific organ that produces stanniocalcin, a pivotal hormone in fish calcium homeostasis. Knockdown of Ron resulted in identical phenotypes as observed in msp mutants. Msp mutant embryos could be rescued by excess calcium. Consistent with a role for Msp/Ron in calcium homeostasis, calcium-regulating factors, such as pth1, pth2, stc1l, and trpv5/6 were significantly affected in msp mutant larvae. While Msp and Ron have previously been shown to play a critical role in a wide variety of biological processes, we introduce here the Msp/Ron signaling axis as a previously unappreciated player in calcium homeostasis and embryonic skeletal mineralization.
Collapse
Affiliation(s)
- Leonie F A Huitema
- Hubrecht Institute, KNAW and University Medical Centre Utrecht, Uppsalalaan 8, 3584CT Utrecht, The Netherlands
| | | | | | | | | | | | | |
Collapse
|
150
|
Delous M, Yin C, Shin D, Ninov N, Debrito Carten J, Pan L, Ma TP, Farber SA, Moens CB, Stainier DYR. Sox9b is a key regulator of pancreaticobiliary ductal system development. PLoS Genet 2012; 8:e1002754. [PMID: 22719264 PMCID: PMC3375260 DOI: 10.1371/journal.pgen.1002754] [Citation(s) in RCA: 94] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2011] [Accepted: 04/23/2012] [Indexed: 01/19/2023] Open
Abstract
The pancreaticobiliary ductal system connects the liver and pancreas to the intestine. It is composed of the hepatopancreatic ductal (HPD) system as well as the intrahepatic biliary ducts and the intrapancreatic ducts. Despite its physiological importance, the development of the pancreaticobiliary ductal system remains poorly understood. The SRY-related transcription factor SOX9 is expressed in the mammalian pancreaticobiliary ductal system, but the perinatal lethality of Sox9 heterozygous mice makes loss-of-function analyses challenging. We turned to the zebrafish to assess the role of SOX9 in pancreaticobiliary ductal system development. We first show that zebrafish sox9b recapitulates the expression pattern of mouse Sox9 in the pancreaticobiliary ductal system and use a nonsense allele of sox9b, sox9bfh313, to dissect its function in the morphogenesis of this structure. Strikingly, sox9bfh313 homozygous mutants survive to adulthood and exhibit cholestasis associated with hepatic and pancreatic duct proliferation, cyst formation, and fibrosis. Analysis of sox9bfh313 mutant embryos and larvae reveals that the HPD cells appear to mis-differentiate towards hepatic and/or pancreatic fates, resulting in a dysmorphic structure. The intrahepatic biliary cells are specified but fail to assemble into a functional network. Similarly, intrapancreatic duct formation is severely impaired in sox9bfh313 mutants, while the embryonic endocrine and acinar compartments appear unaffected. The defects in the intrahepatic and intrapancreatic ducts of sox9bfh313 mutants worsen during larval and juvenile stages, prompting the adult phenotype. We further show that Sox9b interacts with Notch signaling to regulate intrahepatic biliary network formation: sox9b expression is positively regulated by Notch signaling, while Sox9b function is required to maintain Notch signaling in the intrahepatic biliary cells. Together, these data reveal key roles for SOX9 in the morphogenesis of the pancreaticobiliary ductal system, and they cast human Sox9 as a candidate gene for pancreaticobiliary duct malformation-related pathologies. The liver and pancreas function as exocrine glands that secrete bile and pancreatic juice, respectively, to aid the digestion and absorption of nutrients. These fluids reach the intestine via the pancreaticobiliary ductal system, a complex network of ducts. Despite its pivotal role, the development of this ductal system is poorly understood. We have discovered that the zebrafish transcription factor gene sox9b, like its mammalian ortholog, is specifically expressed in the pancreaticobiliary ductal system. The perinatal lethality of Sox9 heterozygous mice makes the analysis of SOX9 function challenging; thus, we turned to the zebrafish to analyze the role of SOX9 in pancreaticobiliary ductal system development. We found that zebrafish sox9b mutants, which survive to adulthood, display defects in the morphogenesis of this ductal network: the intrahepatic and intrapancreatic ducts fail to form a branched network, whereas the ducts connecting the liver and pancreas to the intestine are malformed. These ductal defects affect bile transport and lead to cholestasis in adult mutant fish. At the molecular level, Sox9b interacts with the Notch signaling pathway to regulate the development of the intrahepatic biliary network. Therefore, our work in zebrafish reveals a broad and complex role for SOX9 in pancreaticobiliary ductal system morphogenesis.
Collapse
Affiliation(s)
- Marion Delous
- Department of Biochemistry and Biophysics, Program in Developmental and Stem Cell Biology, Liver Center and Diabetes Center, University of California San Francisco, San Francisco, California, United States of America
- * E-mail: (MD); (DYRS)
| | - Chunyue Yin
- Department of Biochemistry and Biophysics, Program in Developmental and Stem Cell Biology, Liver Center and Diabetes Center, University of California San Francisco, San Francisco, California, United States of America
| | - Donghun Shin
- Department of Biochemistry and Biophysics, Program in Developmental and Stem Cell Biology, Liver Center and Diabetes Center, University of California San Francisco, San Francisco, California, United States of America
| | - Nikolay Ninov
- Department of Biochemistry and Biophysics, Program in Developmental and Stem Cell Biology, Liver Center and Diabetes Center, University of California San Francisco, San Francisco, California, United States of America
| | - Juliana Debrito Carten
- Department of Biology, Johns Hopkins University, Baltimore, Maryland, United States of America
- Department of Embryology, The Carnegie Institution for Science, Baltimore, Maryland, United States of America
| | - Luyuan Pan
- Howard Hughes Medical Institute and Division of Basic Science, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Taylur P. Ma
- Howard Hughes Medical Institute and Division of Basic Science, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Steven A. Farber
- Department of Biology, Johns Hopkins University, Baltimore, Maryland, United States of America
- Department of Embryology, The Carnegie Institution for Science, Baltimore, Maryland, United States of America
| | - Cecilia B. Moens
- Howard Hughes Medical Institute and Division of Basic Science, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Didier Y. R. Stainier
- Department of Biochemistry and Biophysics, Program in Developmental and Stem Cell Biology, Liver Center and Diabetes Center, University of California San Francisco, San Francisco, California, United States of America
- * E-mail: (MD); (DYRS)
| |
Collapse
|