151
|
Zong Y, Stanger BZ. Molecular mechanisms of liver and bile duct development. WILEY INTERDISCIPLINARY REVIEWS-DEVELOPMENTAL BIOLOGY 2012; 1:643-55. [DOI: 10.1002/wdev.47] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
152
|
Truman LA, Bentley KL, Smith EC, Massaro SA, Gonzalez DG, Haberman AM, Hill M, Jones D, Min W, Krause DS, Ruddle NH. ProxTom lymphatic vessel reporter mice reveal Prox1 expression in the adrenal medulla, megakaryocytes, and platelets. THE AMERICAN JOURNAL OF PATHOLOGY 2012; 180:1715-25. [PMID: 22310467 PMCID: PMC3349900 DOI: 10.1016/j.ajpath.2011.12.026] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/13/2011] [Revised: 11/23/2011] [Accepted: 12/08/2011] [Indexed: 11/29/2022]
Abstract
Lymphatic vessels (LVs) are important structures for antigen presentation, for lipid metabolism, and as conduits for tumor metastases, but they have been difficult to visualize in vivo. Prox1 is a transcription factor that is necessary for lymphangiogenesis in ontogeny and the maintenance of LVs. To visualize LVs in the lymph node of a living mouse in real time, we made the ProxTom transgenic mouse in a C57BL/6 background using red fluorescent LVs that are suitable for in vivo imaging. The ProxTom transgene contained all Prox1 regulatory sequences and was faithfully expressed in LVs coincident with endogenous Prox1 expression. The progenies of a ProxTom × Hec6stGFP cross were imaged using two-photon laser scanning microscopy, allowing the simultaneous visualization of LVs and high endothelial venules in a lymph node of a living mouse for the first time. We confirmed the expression of Prox1 in the adult liver, lens, and dentate gyrus. These intensely fluorescent mice revealed the expression of Prox1 in three novel sites: the neuroendocrine cells of the adrenal medulla, megakaryocytes, and platelets. The novel sites identified herein suggest previously unknown roles for Prox1. The faithful expression of the fluorescent reporter in ProxTom LVs indicates that these mice have potential utility in the study of diseases as diverse as lymphedema, filariasis, transplant rejection, obesity, and tumor metastasis.
Collapse
Affiliation(s)
- Lucy A. Truman
- Department of Epidemiology and Public Health, Yale University School of Medicine, New Haven, Connecticut
| | - Kevin L. Bentley
- Department of Epidemiology and Public Health, Yale University School of Medicine, New Haven, Connecticut
| | - Elenoe C. Smith
- Department of Cell Biology, Yale University School of Medicine, New Haven, Connecticut
| | - Stephanie A. Massaro
- Department of Pediatrics, Yale University School of Medicine, New Haven, Connecticut
| | - David G. Gonzalez
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut
| | - Ann M. Haberman
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, Connecticut
| | - Myriam Hill
- Department of Epidemiology and Public Health, Yale University School of Medicine, New Haven, Connecticut
| | - Dennis Jones
- Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut
| | - Wang Min
- Department of Pathology, Yale University School of Medicine, New Haven, Connecticut
| | - Diane S. Krause
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, Connecticut
| | - Nancy H. Ruddle
- Department of Epidemiology and Public Health, Yale University School of Medicine, New Haven, Connecticut
- Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut
| |
Collapse
|
153
|
WESTMORELAND JOBYJ, KILIC GAMZE, SARTAIN CAROLINE, SIRMA SEMA, BLAIN JENNIFER, REHG JEROLD, HARVEY NATASHA, SOSA–PINEDA BEATRIZ. Pancreas-specific deletion of Prox1 affects development and disrupts homeostasis of the exocrine pancreas. Gastroenterology 2012; 142:999-1009.e6. [PMID: 22178591 PMCID: PMC3398795 DOI: 10.1053/j.gastro.2011.12.007] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2011] [Revised: 11/30/2011] [Accepted: 12/03/2011] [Indexed: 12/02/2022]
Abstract
BACKGROUND & AIMS The exocrine portion of the pancreas functions in digestion and preserves pancreatic homeostasis. Learning how this tissue forms during embryogenesis could improve our understanding of human pancreatic diseases. Expression of the homeobox gene Prox1 in the exocrine pancreas changes throughout development in mice. We investigated the role of Prox1 in development of the exocrine pancreas in mice. METHODS Mice with pancreas-specific deletion of Prox1 (Prox1(ΔPanc)) were generated and their pancreatic tissues were analyzed using immunohistochemistry, transmission electron microscopy, histologic techniques, quantitative real-time polymerase chain reaction, immunoblotting, and morphometric analysis. RESULTS Loss of Prox1 from the pancreas led to multiple exocrine alterations, most notably premature acinar cell differentiation, increased ductal cell proliferation, altered duct morphogenesis, and imbalanced expression of claudin proteins. Prox1(ΔPanc) mice also had some minor alterations in islet cells, but beta-cell development was not affected. The exocrine congenital defects of Prox1(ΔPanc) pancreata appeared to initiate a gradual process of deterioration that resulted in extensive loss of acinar cells, lipomatosis, and damage to ductal tissue in adult mice. CONCLUSIONS Pancreas-specific deletion of Prox1 causes premature differentiation of acinar cells and poor elongation of epithelial branches; these defects indicate that Prox1 controls the expansion of tip progenitors in the early developing pancreas. During later stages of embryogenesis, Prox1 appears to regulate duct cell proliferation and morphogenesis. These findings identify Prox1 as an important regulator of pancreatic exocrine development.
Collapse
Affiliation(s)
- JOBY J. WESTMORELAND
- Department of Genetics, St Jude Children’s Research Hospital, Memphis, Tennessee
| | - GAMZE KILIC
- Department of Genetics, St Jude Children’s Research Hospital, Memphis, Tennessee
| | - CAROLINE SARTAIN
- Department of Genetics, St Jude Children’s Research Hospital, Memphis, Tennessee
| | - SEMA SIRMA
- Department of Genetics, St Jude Children’s Research Hospital, Memphis, Tennessee
| | - JENNIFER BLAIN
- Department of Genetics, St Jude Children’s Research Hospital, Memphis, Tennessee
| | - JEROLD REHG
- Department of Pathology, St Jude Children’s Research Hospital, Memphis, Tennessee
| | - NATASHA HARVEY
- Department of Genetics, St Jude Children’s Research Hospital, Memphis, Tennessee
| | - BEATRIZ SOSA–PINEDA
- Department of Genetics, St Jude Children’s Research Hospital, Memphis, Tennessee
| |
Collapse
|
154
|
Saito Y, Kojima T, Takahashi N. Mab21l2 is essential for embryonic heart and liver development. PLoS One 2012; 7:e32991. [PMID: 22412967 PMCID: PMC3297618 DOI: 10.1371/journal.pone.0032991] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2011] [Accepted: 02/06/2012] [Indexed: 12/01/2022] Open
Abstract
During mouse embryogenesis, proper formation of the heart and liver is especially important and is crucial for embryonic viability. In this study, we showed that Mab21l2 was expressed in the trabecular and compact myocardium, and that deletion of Mab21l2 resulted in a reduction of the trabecular myocardium and thinning of the compact myocardium. Mab21l2-deficient embryonic hearts had decreased expression of genes that regulate cell proliferation and apoptosis of cardiomyocytes. These results show that Mab21l2 functions during heart development by regulating the expression of such genes. Mab21l2 was also expressed in the septum transversum mesenchyme (STM). Epicardial progenitor cells are localized to the anterior surface of the STM (proepicardium), and proepicardial cells migrate onto the surface of the heart and form the epicardium, which plays an important role in heart development. The rest of the STM is essential for the growth and survival of hepatoblasts, which are bipotential progenitors for hepatocytes and cholangiocytes. Proepicardial cells in Mab21l2-deficient embryos had defects in cell proliferation, which led to a small proepicardium, in which α4 integrin expression, which is essential for the migration of proepicardial cells, was down-regulated, suggesting that defects occurred in its migration. In Mab21l2-deficient embryos, epicardial formation was defective, suggesting that Mab21l2 plays important roles in epicardial formation through the regulation of the cell proliferation of proepicardial cells and the migratory process of proepicardial cells. Mab21l2-deficient embryos also exhibited hypoplasia of the STM surrounding hepatoblasts and decreased hepatoblast proliferation with a resultant loss of defective morphogenesis of the liver. These findings demonstrate that Mab21l2 plays a crucial role in both heart and liver development through STM formation.
Collapse
Affiliation(s)
| | | | - Naoki Takahashi
- Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
- * E-mail:
| |
Collapse
|
155
|
Xu CR, Zaret KS. Chromatin "pre-pattern" and epigenetic modulation in the cell fate choice of liver over pancreas in the endoderm. Nucleus 2012; 3:150-4. [PMID: 22555599 DOI: 10.4161/nucl.19321] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Understanding the basis for multipotency, whereby stem cells and other progenitors can differentiate into certain tissues and not others, provides insights into the mechanism of cell programming in development, homeostasis, and disease. We recently reported a screen of diverse chromatin marks to obtain clues about chromatin states in the multipotent embryonic endoderm. Genetic and pharmacologic tests of certain marks' function demonstrated that the relevant chromatin modifying factors modulate the fate choice for liver or pancreas induction in the endoderm. The information about chromatin states from embryonic studies can be used to predict lineage-specific developmental potential and chromatin modifiers to enhance particular cell fate transitions from stem cells.
Collapse
Affiliation(s)
- Cheng-Ran Xu
- Institute for Regenerative Medicine, Department of Cell and Developmental Biology, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| | | |
Collapse
|
156
|
Azuma K, Urano T, Watabe T, Ouchi Y, Inoue S. PROX1 suppresses vitamin K-induced transcriptional activity of Steroid and Xenobiotic Receptor. Genes Cells 2012; 16:1063-70. [PMID: 22023334 DOI: 10.1111/j.1365-2443.2011.01551.x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Steroid and Xenobiotic Receptor (SXR) belongs to nuclear receptor superfamily. It was shown that secondary bile acids such as lithocholic acid and several chemical compounds such as rifampicin could be ligands for this receptor. Recently, we have demonstrated that vitamin K2 also serves as a ligand for SXR and activation of SXR by vitamin K2 suppressed proliferation and motility of hepatocellular carcinoma (HCC) cells. To analyze function of SXR in HCC cells, we overexpressed exogenous SXR double-tagged with FLAG and HA in a HCC cell line, HepG2 cells, and purified SXR-binding molecules by immunoprecipitation from the nuclear extracts of these cells. Several binding molecules were identified by TOF-MS analyses. One of the SXR-binding molecules was a transcription factor PROX1. We confirmed the interaction of PROX1 and SXR in HEK293 cells. Then, we have shown that AF2 domain of SXR is necessary for binding with PROX1. We further demonstrated that PROX1 negatively regulated the transcriptional activity of SXR by promoter analyses of SXR target gene. These results suggest that PROX1 could negatively regulate SXR signals in some tumor cells, such as HCC cells, where both SXR and PROX1 are expressed.
Collapse
Affiliation(s)
- Kotaro Azuma
- Department of Geriatric Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | | | | | | | | |
Collapse
|
157
|
|
158
|
|
159
|
Yoshimatsu Y, Yamazaki T, Mihira H, Itoh T, Suehiro J, Yuki K, Harada K, Morikawa M, Iwata C, Minami T, Morishita Y, Kodama T, Miyazono K, Watabe T. Ets family members induce lymphangiogenesis through physical and functional interaction with Prox1. J Cell Sci 2011; 124:2753-62. [PMID: 21807940 DOI: 10.1242/jcs.083998] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Prox1 plays pivotal roles during embryonic lymphatic development and maintenance of adult lymphatic systems by modulating the expression of various lymphatic endothelial cell (LEC) markers, such as vascular endothelial growth factor receptor 3 (VEGFR3). However, the molecular mechanisms by which Prox1 transactivates its target genes remain largely unknown. Here, we identified Ets-2 as a candidate molecule that regulates the functions of Prox1. Whereas Ets-2 has been implicated in angiogenesis, its roles during lymphangiogenesis have not yet been elucidated. We found that endogenous Ets-2 interacts with Prox1 in LECs. Using an in vivo model of chronic aseptic peritonitis, we found that Ets-2 enhanced inflammatory lymphangiogenesis, whereas a dominant-negative mutant of Ets-1 suppressed it. Ets-2 also enhanced endothelial migration towards VEGF-C through induction of expression of VEGFR3 in collaboration with Prox1. Furthermore, we found that both Prox1 and Ets-2 bind to the VEGFR3 promoter in intact chromatin. These findings suggest that Ets family members function as transcriptional cofactors that enhance Prox1-induced lymphangiogenesis.
Collapse
Affiliation(s)
- Yasuhiro Yoshimatsu
- Department of Molecular Pathology, Graduate School of Medicine, University of Tokyo, Tokyo 113-0033, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
160
|
New approaches in the differentiation of human embryonic stem cells and induced pluripotent stem cells toward hepatocytes. Stem Cell Rev Rep 2011; 7:748-59. [PMID: 21336836 PMCID: PMC3137783 DOI: 10.1007/s12015-010-9216-4] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Orthotropic liver transplantation is the only established treatment for end-stage liver diseases. Utilization of hepatocyte transplantation and bio-artificial liver devices as alternative therapeutic approaches requires an unlimited source of hepatocytes. Stem cells, especially embryonic stem cells, possessing the ability to produce functional hepatocytes for clinical applications and drug development, may provide the answer to this problem. New discoveries in the mechanisms of liver development and the emergence of induced pluripotent stem cells in 2006 have provided novel insights into hepatocyte differentiation and the use of stem cells for therapeutic applications. This review is aimed towards providing scientists and physicians with the latest advancements in this rapidly progressing field.
Collapse
|
161
|
Skog M, Bono P, Lundin M, Lundin J, Louhimo J, Linder N, Petrova TV, Andersson LC, Joensuu H, Alitalo K, Haglund CH. Expression and prognostic value of transcription factor PROX1 in colorectal cancer. Br J Cancer 2011; 105:1346-51. [PMID: 21970873 PMCID: PMC3241535 DOI: 10.1038/bjc.2011.297] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Background: PROX1 is a specific target of the β-catenin/TCF pathway in the intestinal epithelium. It acts as a regulator of progression from a benign to a highly dysplastic phenotype in colorectal tumours. However, the clinical significance of PROX1 expression is not known. Methods: We studied the prognostic value of immunohistochemical expression of PROX1 in a series of 517 patients with colorectal cancer (CRC). Results: The majority of the tumour samples expressed PROX1 (91%, 471 out of 517). High PROX1 expression was associated with a poor grade of tumour differentiation (P<0.0001). In the subgroup of patients with colon cancer, high PROX1 expression was associated with unfavourable colorectal cancer-specific survival (CCSS) as compared with low PROX1 expression (CCSS 47% vs 62% P=0.045; RR 1.47). The association between high PROX1 and poor outcome was further strengthened in female colon cancer patients (CCSS 38% vs 63% P=0.007; RR 2.02). Nonetheless, in multivariate survival analysis PROX1 expression was not retained as an independent prognostic factor. Conclusion: High PROX1 expression is associated with a poor grade of tumour differentiation, and, in colon cancer patients, also with less favourable patient outcome. Our results strengthen the previous preclinical observations that PROX1 has a role in tumour progression in CRC.
Collapse
Affiliation(s)
- M Skog
- Department of Oncology, Helsinki University Central Hospital, Helsinki FIN-00029 HUS, Finland
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
162
|
Nagaoka M, Duncan SA. Transcriptional control of hepatocyte differentiation. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2011; 97:79-101. [PMID: 21074730 DOI: 10.1016/b978-0-12-385233-5.00003-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The liver is the largest glandular organ in the body and plays a central role in controlling metabolism. During hepatogenesis, complex developmental processes must generate an array of cell types that are spatially arranged to generate a hepatic architecture that is essential to support liver function. The processes that control the ultimate formation of the liver are diverse and complex and in many cases poorly defined. Much of the focus of research during the past three decades has been on understanding how hepatocytes, which are the predominant liver parenchymal cells, differentiate during embryogenesis. Through a combination of mouse molecular genetics, embryology, and molecular biochemistry, investigators have defined a myriad of transcription factors that combine to control formation and function of hepatocytes. Here, we will review the major discoveries that underlie our current understanding of transcriptional regulation of hepatocyte differentiation.
Collapse
Affiliation(s)
- Masato Nagaoka
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | | |
Collapse
|
163
|
Alexander JS, Ganta VC, Jordan PA, Witte MH. Gastrointestinal lymphatics in health and disease. ACTA ACUST UNITED AC 2011; 17:315-35. [PMID: 20022228 DOI: 10.1016/j.pathophys.2009.09.003] [Citation(s) in RCA: 83] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2009] [Revised: 09/28/2009] [Accepted: 09/28/2009] [Indexed: 12/17/2022]
Abstract
Lymphatics perform essential transport and immune regulatory functions to maintain homeostasis in the gastrointestinal (GI) system. Although blood and lymphatic vessels function as parallel and integrated systems, our understanding of lymphatic structure, regulation and functioning lags far behind that of the blood vascular system. This chapter reviews lymphatic flow, differences in lymphangiogenic and hemangiogenic factors, lymphatic fate determinants and structural features, and examines how altered molecular signaling influences lymphatic function in organs of the GI system. Innate errors in lymphatic development frequently disturb GI functioning and physiology. Expansion of lymphatics, a prominent feature of GI inflammation, may also play an important role in tissue restitution following injury. Destruction or dysregulation of lymphatics, following injury, surgery or chronic inflammation also exacerbates GI disease activity. Understanding the physiological roles played by GI lymphatics is essential to elucidating their underlying contributions to forms of congenital and acquired forms of GI pathology, and will provide novel approaches for therapy.
Collapse
Affiliation(s)
- J S Alexander
- Louisiana State University Health Sciences Center-Shreveport, Molecular and Cellular Physiology, Shreveport, LA, United States
| | | | | | | |
Collapse
|
164
|
Flister MJ, Volk LD, Ran S. Characterization of Prox1 and VEGFR-3 expression and lymphatic phenotype in normal organs of mice lacking p50 subunit of NF-κB. Microcirculation 2011; 18:85-101. [PMID: 21166921 DOI: 10.1111/j.1549-8719.2010.00057.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
OBJECTIVE Inflammation and NF-κB are highly associated with lymphangiogenesis but the underlying mechanisms remain unclear. We recently established that activated NF-κB p50 subunit increases expression of the main lymphangiogenic mediators, VEGFR-3 and its transcriptional activator, Prox1. To elucidate the role of p50 in lymphatic vasculature, we compared LVD and phenotype in p50 KO and WT mice. METHODS Normal tissues from KO and WT mice were stained for LYVE-1 to calculate LVD. VEGFR-3 and Prox1 expressions were analyzed by immunofluorescence and qRT-PCR. RESULTS Compared with WT, LVD in the liver and lungs of KO mice was reduced by 39% and 13%, respectively. This corresponded to 25-44% decreased VEGFR-3 and Prox1 expression. In the MFP, LVD was decreased by 18% but VEGFR-3 and Prox1 expression was 80-140% higher than in WT. Analysis of p65 and p52 NF-κB subunits and an array of inflammatory mediators showed a significant increase in p50 alternative pathways in the MFP but not in other organs. CONCLUSIONS These findings demonstrate the role of NF-κB p50 in regulating the expression of VEGFR-3, Prox1 and LVD in the mammary tissue, liver, and lung.
Collapse
Affiliation(s)
- Michael J Flister
- Department of Medical Microbiology, Immunology, and Cell Biology, Southern Illinois University School of Medicine, Springfield, Illinois 62794-9626, USA
| | | | | |
Collapse
|
165
|
Klein C, Mikutta J, Krueger J, Scholz K, Brinkmann J, Liu D, Veerkamp J, Siegel D, Abdelilah-Seyfried S, le Noble F. Neuron navigator 3a regulates liver organogenesis during zebrafish embryogenesis. Development 2011; 138:1935-45. [PMID: 21471154 DOI: 10.1242/dev.056861] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Endodermal organogenesis requires a precise orchestration of cell fate specification and cell movements, collectively coordinating organ size and shape. In Caenorhabditis elegans, uncoordinated-53 (unc-53) encodes a neural guidance molecule that directs axonal growth. One of the vertebrate homologs of unc-53 is neuron navigator 3 (Nav3). Here, we identified a novel vertebrate neuron navigator 3 isoform in zebrafish, nav3a, and we provide genetic evidence in loss- and gain-of-function experiments showing its functional role in endodermal organogenesis during zebrafish embryogenesis. In zebrafish embryos, nav3a expression was initiated at 22 hpf in the gut endoderm and at 40 hpf expanded to the newly formed liver bud. Endodermal nav3a expression was controlled by Wnt2bb signaling and was independent of FGF and BMP signaling. Morpholino-mediated knockdown of nav3a resulted in a significantly reduced liver size, and impaired development of pancreas and swim bladder. In vivo time-lapse imaging of liver development in nav3a morphants revealed a failure of hepatoblast movement out from the gut endoderm during the liver budding stage, with hepatoblasts being retained in the intestinal endoderm. In hepatocytes in vitro, nav3a acts as a positive modulator of actin assembly in lamellipodia and filipodia extensions, allowing cellular movement. Knockdown of nav3a in vitro impeded hepatocyte movement. Endodermal-specific overexpression of nav3a in vivo resulted in additional ectopic endodermal budding beyond the normal liver and pancreatic budding sites. We conclude that nav3a is required for directing endodermal organogenesis involving coordination of endodermal cell behavior.
Collapse
Affiliation(s)
- Christian Klein
- Department of Angiogenesis and Cardiovascular Pathology, Max-Delbrueck-Center for Molecular Medicine (MDC), Berlin, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
166
|
Akagami M, Kawada K, Kubo H, Kawada M, Takahashi M, Kaganoi J, Kato S, Itami A, Shimada Y, Watanabe G, Sakai Y. Transcriptional factor Prox1 plays an essential role in the antiproliferative action of interferon-γ in esophageal cancer cells. Ann Surg Oncol 2011; 18:3868-77. [PMID: 21452064 DOI: 10.1245/s10434-011-1683-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2010] [Indexed: 02/03/2023]
Abstract
BACKGROUND We previously reported interferon-γ (IFN-γ)-induced apoptosis in 10 (32%) of 31 esophageal squamous cell carcinoma (ESCC) cell lines. However, the molecular basis of antiproliferative action by IFN-γ remains elusive. Here we demonstrate that IFN-γ induces transcriptional factor Prox1, and we explore the link between Prox1 and the IFN-γ system in ESCC cells. METHODS By using ESCC cell lines, we investigated the relationship between p53 mutations and the responsibility to IFN-γ, and studied the role of Prox1 in the antiproliferative effect of IFN-γ by knockdown and overexpression methods. RESULTS p53 mutations were found in seven of nine ESCC cell lines responsible for IFN-γ. The frequency was not different from that of p53 mutations in total ESCC cell lines (21 of 28 cell lines). Treatment of ESCC cells with IFN-β but not IFN-γ resulted in increase of p53 messenger RNA (mRNA) expression, whereas IFN-γ but not IFN-β induced cell growth inhibition of ESCCs harboring p53 mutations. IFN-γ induced Prox1 expression in ESCC cells but not in those transfected with dominant-negative STAT1. Cell growth inhibition by IFN-γ was significantly suppressed in ESCC cells transfected with Prox1 short interfering RNA (siRNA). In addition, overexpression of Prox1 induced antiproliferative effect in ESCC cells. We also demonstrate that Prox1 is expressed in primary esophageal cancer tissues (five of nine samples treated with neoadjuvant chemotherapy before surgery). CONCLUSIONS Prox1 mediates the antiproliferative effect by IFN-γ in ESCC cells. Prox1 may be a candidate target for novel therapeutic strategies of ESCCs.
Collapse
Affiliation(s)
- Masatoshi Akagami
- Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
167
|
Elkouris M, Balaskas N, Poulou M, Politis PK, Panayiotou E, Malas S, Thomaidou D, Remboutsika E. Sox1 Maintains the Undifferentiated State of Cortical Neural Progenitor Cells via the Suppression of Prox1-Mediated Cell Cycle Exit and Neurogenesis. Stem Cells 2011; 29:89-98. [DOI: 10.1002/stem.554] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
168
|
Tanaka M, Itoh T, Tanimizu N, Miyajima A. Liver stem/progenitor cells: their characteristics and regulatory mechanisms. J Biochem 2011; 149:231-9. [PMID: 21217146 DOI: 10.1093/jb/mvr001] [Citation(s) in RCA: 86] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Liver stem cells give rise to both hepatocytes and bile duct epithelial cells also known as cholangiocytes. During liver development hepatoblasts emerge from the foregut endoderm and give rise to both cell types. Colony-forming cells are present in the liver primordium and clonally expanded cells differentiate into either hepatocytes or cholangiocytes depending on culture conditions, showing stem cell characteristics. The growth and differentiation of hepatoblasts are regulated by various extrinsic signals. For example, periportal mesenchymal cells provide a cue for bipotential hepatoblasts to become cholangiocytes, and mesothelial cells covering the parenchyma support the expansion of foetal hepatocytes by producing growth factors. The adult liver has an extraordinary capacity to regenerate, and after 70% hepatectomy the liver recovers its original mass by replication of the remaining hepatocytes without the activation of liver stem cells. However, in certain types of liver injury models, liver stem/progenitor-like cells, known as oval cells in rodents, proliferate around the portal vein, while the roles of such cells in liver regeneration remain a matter of debate. Clonogenic and bipotential cells are also present in the normal adult liver. In this minireview we describe recent studies on liver stem/progenitor cells by focusing on extracellular signals.
Collapse
Affiliation(s)
- Minoru Tanaka
- Institute of Molecular and Cellular Biosciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo, Japan
| | | | | | | |
Collapse
|
169
|
Abstract
The endoderm is the innermost germ layer that gives rise to the lining of the gut, the gills, liver, pancreas, gallbladder, and derivatives of the pharyngeal pouch. These organs form the gastrointestinal tract and are involved with the absorption, delivery, and metabolism of nutrients. The liver has a central role in regulating these processes because it controls lipid metabolism, protein synthesis, and breakdown of endogenous and xenobiotic products. Liver dysfunction frequently leads to significant morbidity and mortality; however, in most settings of organ injury, the liver exhibits remarkable regenerative capacity.
Collapse
Affiliation(s)
- Trista E North
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | | |
Collapse
|
170
|
Baxter SA, Cheung DY, Bocangel P, Kim HK, Herbert K, Douville JM, Jangamreddy JR, Zhang S, Eisenstat DD, Wigle JT. Regulation of the lymphatic endothelial cell cycle by the PROX1 homeodomain protein. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2011; 1813:201-12. [DOI: 10.1016/j.bbamcr.2010.10.015] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/16/2010] [Revised: 10/01/2010] [Accepted: 10/25/2010] [Indexed: 11/28/2022]
|
171
|
Locker J. Transcriptional Control of Hepatocyte Differentiation. MOLECULAR PATHOLOGY LIBRARY 2011. [DOI: 10.1007/978-1-4419-7107-4_14] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
|
172
|
Liver Stem Cells. Regen Med 2011. [DOI: 10.1007/978-90-481-9075-1_14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
|
173
|
Kaltezioti V, Kouroupi G, Oikonomaki M, Mantouvalou E, Stergiopoulos A, Charonis A, Rohrer H, Matsas R, Politis PK. Prox1 regulates the notch1-mediated inhibition of neurogenesis. PLoS Biol 2010; 8:e1000565. [PMID: 21203589 PMCID: PMC3006385 DOI: 10.1371/journal.pbio.1000565] [Citation(s) in RCA: 90] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2010] [Accepted: 11/03/2010] [Indexed: 11/18/2022] Open
Abstract
During development of the spinal cord, Prox1 controls the balance between proliferation and differentiation of neural progenitor cells via suppression of Notch1 gene expression. Activation of Notch1 signaling in neural progenitor cells (NPCs) induces self-renewal and inhibits neurogenesis. Upon neuronal differentiation, NPCs overcome this inhibition, express proneural genes to induce Notch ligands, and activate Notch1 in neighboring NPCs. The molecular mechanism that coordinates Notch1 inactivation with initiation of neurogenesis remains elusive. Here, we provide evidence that Prox1, a transcription repressor and downstream target of proneural genes, counteracts Notch1 signaling via direct suppression of Notch1 gene expression. By expression studies in the developing spinal cord of chick and mouse embryo, we showed that Prox1 is limited to neuronal precursors residing between the Notch1+ NPCs and post-mitotic neurons. Physiological levels of Prox1 in this tissue are sufficient to allow binding at Notch1 promoter and they are critical for proper Notch1 transcriptional regulation in vivo. Gain-of-function studies in the chick neural tube and mouse NPCs suggest that Prox1-mediated suppression of Notch1 relieves its inhibition on neurogenesis and allows NPCs to exit the cell cycle and differentiate. Moreover, loss-of-function in the chick neural tube shows that Prox1 is necessary for suppression of Notch1 outside the ventricular zone, inhibition of active Notch signaling, down-regulation of NPC markers, and completion of neuronal differentiation program. Together these data suggest that Prox1 inhibits Notch1 gene expression to control the balance between NPC self-renewal and neuronal differentiation. Early during development, neural progenitor cells (NPCs) can either proliferate or differentiate into neurons. Thus, generation of the correct number of neurons is governed by a tightly regulated balance between proliferation and differentiation, and disruption of this balance can result in severe developmental deficits, malformations, or cancers. Notch1 is a member of the Notch family of receptors, which make up a highly conserved cell signaling system. Notch1 signaling has been shown to inhibit NPC differentiation and to promote self-renewal, thereby allowing NPCs to divide and progressively generate the enormous number of neurons present in the central nervous system. The molecular mechanism by which NPCs overcome Notch1-mediated inhibition in order to differentiate into neurons, however, is not completely understood. In this study, we show that Prox1, a homeobox transcriptional repressor, plays a fundamental role in the switch to differentiation by suppressing the expression of Notch1 receptor, thereby preventing newly produced neuronal precursors from receiving inhibitory signals from Notch ligands present in neighboring cells. This transcriptional repression may regulate cell cycle exit and differentiation of NPCs as they migrate towards different regions and adopt their final cell fates. We suggest that Prox1 may exert its known influence on embryonic development, organ morphogenesis, and cancer through its ability to counteract Notch1 signaling.
Collapse
Affiliation(s)
- Valeria Kaltezioti
- Center for Basic Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Georgia Kouroupi
- Laboratory of Cellular and Molecular Neurobiology, Hellenic Pasteur Institute, Athens, Greece
| | - Maria Oikonomaki
- Center for Basic Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Evangelia Mantouvalou
- Center for Basic Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Athanasios Stergiopoulos
- Center for Basic Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Aristidis Charonis
- Center for Basic Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Hermann Rohrer
- Department of Neurochemistry, Max-Planck Institute for Brain Research, Frankfurt/Main, Germany
| | - Rebecca Matsas
- Laboratory of Cellular and Molecular Neurobiology, Hellenic Pasteur Institute, Athens, Greece
| | - Panagiotis K. Politis
- Center for Basic Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
- * E-mail:
| |
Collapse
|
174
|
Visualization of lymphatic vessels by Prox1-promoter directed GFP reporter in a bacterial artificial chromosome-based transgenic mouse. Blood 2010; 117:362-5. [PMID: 20962325 DOI: 10.1182/blood-2010-07-298562] [Citation(s) in RCA: 207] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Although the blood vessel-specific fluorescent transgenic mouse has been an excellent tool to study vasculogenesis and angiogenesis, a lymphatic-specific fluorescent mouse model has not been established to date. Here we report a transgenic animal model that expresses the green fluorescent protein under the promoter of Prox1, a master control gene in lymphatic development. Generated using an approximately 200-kb-long bacterial artificial chromosome harboring the entire Prox1 gene, this Prox1-green fluorescent protein mouse was found to faithfully recapitulate the expression pattern of the Prox1 gene in lymphatic endothelial cells and other Prox1-expressing organs, and enabled us to conveniently visualize detailed structure and morphology of lymphatic vessels and networks throughout development. Our data demonstrate that this novel transgenic mouse can be extremely useful for detection, imaging, and isolation of lymphatic vessels and monitoring wound-associated lymphangiogenesis. Together, this Prox1-green fluorescent protein transgenic mouse will be a great tool for the lymphatic research.
Collapse
|
175
|
Crawford LW, Foley JF, Elmore SA. Histology atlas of the developing mouse hepatobiliary system with emphasis on embryonic days 9.5-18.5. Toxicol Pathol 2010; 38:872-906. [PMID: 20805319 DOI: 10.1177/0192623310374329] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Animal model phenotyping, in utero exposure toxicity studies, and investigation into causes of embryonic, fetal, or perinatal deaths have required pathologists to recognize and diagnose developmental disorders in spontaneous and engineered mouse models of disease. In mammals, the liver is the main site of hematopoiesis during fetal development, has endocrine and exocrine functions important for maintaining homeostasis in fetal and adult life; and performs other functions including waste detoxification, production and removal of glucose, glycogen storage, triglyceride and fatty acid processing, and serum protein production. Due to its role in many critical functions, alterations in the size, morphology, or function(s) of the liver often lead to embryonic lethality. Many publications and websites describe individual aspects of hepatobiliary development at defined stages. However, no single resource provides a detailed histological evaluation of H&E-stained sections of the developing murine liver and biliary systems using high-magnification and high-resolution color images. The work herein provides a histology atlas of hepatobiliary development between embryonic days 9.5-18.5. Although the focus of this work is normal hepatobiliary development, common defects in liver development are also described as a reference for pathologists who may be asked to phenotype mice with congenital, inherited, or treatment-related hepatobiliary defects. Authors' note: All digital images can be viewed online at https://niehsimagesepl-inc.com with the username "ToxPathLiver" and the password "embryolivers."
Collapse
Affiliation(s)
- Laura Wilding Crawford
- 1Cellular and Molecular Pathology Branch, NIEHS, NIH, Research Triangle Park, NC 27709,USA
| | | | | |
Collapse
|
176
|
Yoo J, Kang J, Lee HN, Aguilar B, Kafka D, Lee S, Choi I, Lee J, Ramu S, Haas J, Koh CJ, Hong YK. Kaposin-B enhances the PROX1 mRNA stability during lymphatic reprogramming of vascular endothelial cells by Kaposi's sarcoma herpes virus. PLoS Pathog 2010; 6:e1001046. [PMID: 20730087 PMCID: PMC2921153 DOI: 10.1371/journal.ppat.1001046] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2009] [Accepted: 07/15/2010] [Indexed: 01/16/2023] Open
Abstract
Kaposi's sarcoma (KS) is the most common cancer among HIV-positive patients. Histogenetic origin of KS has long been elusive due to a mixed expression of both blood and lymphatic endothelial markers in KS tumor cells. However, we and others discovered that Kaposi's sarcoma herpes virus (KSHV) induces lymphatic reprogramming of blood vascular endothelial cells by upregulating PROX1, which functions as the master regulator for lymphatic endothelial differentiation. Here, we demonstrate that the KSHV latent gene kaposin-B enhances the PROX1 mRNA stability and plays an important role in KSHV-mediated PROX1 upregulation. We found that PROX1 mRNA contains a canonical AU-rich element (ARE) in its 3′-untranslated region that promotes PROX1 mRNA turnover and that kaposin-B stimulates cytoplasmic accumulation of the ARE-binding protein HuR through activation of the p38/MK2 pathway. Moreover, HuR binds to and stabilizes PROX1 mRNA through its ARE and is necessary for KSHV-mediated PROX1 mRNA stabilization. Together, our study demonstrates that kaposin-B plays a key role in PROX1 upregulation during lymphatic reprogramming of blood vascular endothelial cells by KSHV. Kaposi's sarcoma (KS) is the most common cancer in HIV-positive patients and KS-associated herpes virus (KSHV) was identified as its causing agent. We and others have discovered that when the virus infects endothelial cells of blood vessels, KSHV reprograms the cell type resembling endothelial cells in lymphatic vessels. Although endothelial cells of the blood vascular system and of the lymphatic system share functional similarities, the cell type-reprogramming does not occur under a normal physiological condition. Therefore, cell-fate reprogramming by the cancer-causing virus KSHV provides an important insight into the molecular mechanism for viral pathogenesis. Our current study investigates the molecular mechanism underlying the KSHV-mediated cell fate reprogramming. We identified that a KSHV latent gene kaposin-B plays an important role in KSHV-mediated regulation of PROX1 to promote PROX1 mRNA stability. This study will provide a better understanding on the tumorigenesis and pathogenesis of KS with a potential implication toward new KS therapy.
Collapse
Affiliation(s)
- Jaehyuk Yoo
- Departments of Surgery and Department of Biochemistry and Molecular Biology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
| | - Jinjoo Kang
- Departments of Surgery and Department of Biochemistry and Molecular Biology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
| | - Ha Neul Lee
- Departments of Surgery and Department of Biochemistry and Molecular Biology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
| | - Berenice Aguilar
- Departments of Surgery and Department of Biochemistry and Molecular Biology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
| | - Darren Kafka
- Departments of Surgery and Department of Biochemistry and Molecular Biology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
| | - Sunju Lee
- Departments of Surgery and Department of Biochemistry and Molecular Biology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
| | - Inho Choi
- Departments of Surgery and Department of Biochemistry and Molecular Biology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
| | - Juneyong Lee
- Departments of Surgery and Department of Biochemistry and Molecular Biology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
| | - Swapnika Ramu
- Departments of Surgery and Department of Biochemistry and Molecular Biology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
| | - Juergen Haas
- Max-von-Pettenkofer Institut, Ludwig-Maximilians-Universität München, München, Germany
| | - Chester J. Koh
- Division of Pediatric Urology, Childrens Hospital Los Angeles and Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
| | - Young-Kwon Hong
- Departments of Surgery and Department of Biochemistry and Molecular Biology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
- * E-mail:
| |
Collapse
|
177
|
Becker J, Wang B, Pavlakovic H, Buttler K, Wilting J. Homeobox transcription factor Prox1 in sympathetic ganglia of vertebrate embryos: correlation with human stage 4s neuroblastoma. Pediatr Res 2010; 68:112-7. [PMID: 20453716 DOI: 10.1203/pdr.0b013e3181e5bc0f] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Previously, we observed expression of the homeobox transcription factor Prox1 in neuroectodermal embryonic tissues. Besides essential functions during embryonic development, Prox1 has been implicated in both progression and suppression of malignancies. Here, we show that Prox1 is expressed in embryonic sympathetic trunk ganglia of avian and murine embryos. Prox1 protein is localized in the nucleus of neurofilament-positive sympathetic neurons. Sympathetic progenitors represent the cell of origin of neuroblastoma (NB), the most frequent solid extracranial malignancy of children. NB may progress to life-threatening stage 4, or regress spontaneously in the special stage 4s. By qRT-PCR, we show that Prox1 is expressed at low levels in 24 human NB cell lines compared with human lymphatic endothelial cells (LECs), whereas equal immunostaining of nuclei can be seen in embryonic LECs and sympathetic neurons. In NB stages 1, 2, 3, and 4, we observed almost equal expression levels, but significantly higher amounts in stage 4s NB. By immunohistochemistry, we found variable amounts of Prox1 protein in nuclei of NB cells, showing intra and interindividual differences. Because stage 4s NB are susceptible to postnatal apoptosis, we assume that high Prox1 levels are critical for their behavior.
Collapse
Affiliation(s)
- Jürgen Becker
- Department of Anatomy and Cell Biology, University Medicine Goettingen, 37075 Goettingen, Germany
| | | | | | | | | |
Collapse
|
178
|
Katsumoto K, Shiraki N, Miki R, Kume S. Embryonic and adult stem cell systems in mammals: ontology and regulation. Dev Growth Differ 2010; 52:115-29. [PMID: 20078654 DOI: 10.1111/j.1440-169x.2009.01160.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Stem cells are defined as having the ability to self-renew and to generate differentiated cells. During embryogenesis, cells are initially proliferative and pluripotent and then they gradually become restricted to different cell fates. In the adult, tissue stem cells are normally quiescent, but become proliferative upon injury. Knowledge from developmental biology and insights into the properties of stem cells are keys to further understanding and successful manipulation. Here, we first focus on ES cells, then on embryonic development, and then on tissue stem cells of endodermally derived tissues, particularly the liver and pancreas.
Collapse
Affiliation(s)
- Keiichi Katsumoto
- Department of Stem Cell Biology, Institute of Molecular Embryology and Genetics (IMEG), Kumamoto University, Honjo 2-2-1, Kumamoto 860-0811, Japan
| | | | | | | |
Collapse
|
179
|
Zhao X, Monson C, Gao C, Gouon-Evans V, Matsumoto N, Sadler KC, Friedman SL. Klf6/copeb is required for hepatic outgrowth in zebrafish and for hepatocyte specification in mouse ES cells. Dev Biol 2010; 344:79-93. [PMID: 20430021 DOI: 10.1016/j.ydbio.2010.04.018] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2009] [Revised: 04/02/2010] [Accepted: 04/19/2010] [Indexed: 01/05/2023]
Abstract
Krüppel-like factor 6 (Klf6; copeb in zebrafish) is a zinc-finger transcription factor and tumor suppressor gene. Klf6(-)(/)(-) mice have defects in hematopoiesis and angiogenesis and do not form a liver. However, the vascular abnormalities in Klf6(-/-) mice obfuscate its role in liver development since these two processes are linked in mammals. We utilized zebrafish and mouse ES cells to investigate the role of copeb in endoderm specification and hepatogenesis separate from its function in angiogenesis. During zebrafish development, copeb expression is enriched in digestive organs. Morpholino knockdown of copeb blocks expansion of the liver, pancreas and intestine, but does not affect their specification, differentiation or the vascularization of the liver. Decreased hepatocyte proliferation in copeb morphants is accompanied by upregulation of the cell cycle inhibitor, cdkn1a, a Copeb transcriptional target. A cell autonomous role for Klf6 in endoderm and hepatic development was investigated by manipulating Klf6 expression in mouse ES cells driven to differentiate along the hepatic lineage. Expression of the endoderm markers Hnf3beta, Gata4, Sox17, and CxCr4 is not induced in Klf6(-/-) cells but is upregulated in ES cells over-expressing Klf6. Collectively, these findings indicate that copeb/Klf6 is essential for the development of endoderm-derived organs.
Collapse
Affiliation(s)
- Xiao Zhao
- Division of Liver Diseases/Department of Medicine, Black Family Stem Cell Institute, Mount Sinai School of Medicine, New York, NY 10029, USA
| | | | | | | | | | | | | |
Collapse
|
180
|
Karunamuni G, Yang K, Doughman YQ, Wikenheiser J, Bader D, Barnett J, Austin A, Parsons-Wingerter P, Watanabe M. Expression of lymphatic markers during avian and mouse cardiogenesis. Anat Rec (Hoboken) 2010; 293:259-70. [PMID: 19938109 DOI: 10.1002/ar.21043] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The adult heart has been reported to have an extensive lymphatic system, yet the development of this important system during cardiogenesis is still largely unexplored. The nuclear-localized transcription factor Prox-1 identified a sheet of Prox-1-positive cells on the developing aorta and pulmonary trunk in avian and murine embryos just before septation of the four heart chambers. The cells coalesced into a branching lymphatic network that spread within the epicardium to cover the heart. These vessels eventually expressed the lymphatic markers LYVE-1, VEGFR-3, and podoplanin. Before the Prox-1-positive cells were detected in the mouse epicardium, LYVE-1, a homologue of the CD44 glycoprotein, was primarily expressed in individual epicardial cells. Similar staining patterns were observed for CD44 in avian embryos. The proximity of these LYVE-1/CD44-positive mesenchymal cells to Prox-1-positive vessels suggests that they may become incorporated into the lymphatics. Unexpectedly, we detected LYVE-1/PECAM/VEGFR-3-positive vessels within the embryonic and adult myocardium, which remained Prox-1/podoplanin-negative. Lymphatic markers were surprisingly found in adult rat and embryonic mouse epicardial cell lines, with Prox-1 also exhibiting nuclear-localized expression in primary cultures of embryonic avian epicardial cells. Our data identified three types of cells in the embryonic heart expressing lymphatic markers: (1) Prox-1-positive cells from an extracardiac source that migrate within the serosa of the outflow tract into the epicardium of the developing heart, (2) individual LYVE-1-positive cells in the epicardium that may be incorporated into the Prox-1-positive lymphatic vasculature, and (3) LYVE-1-positive cells/vessels in the myocardium that do not become Prox-1-positive even in the adult heart.
Collapse
Affiliation(s)
- Ganga Karunamuni
- Department of Pediatrics, Case Western Reserve University, Cleveland, Ohio, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
181
|
Abstract
Embryonic development of the liver has been studied intensely, yielding insights that impact diverse areas of developmental and cell biology. Understanding the fundamental mechanisms that control hepatogenesis has also laid the basis for the rational differentiation of stem cells into cells that display many hepatic functions. Here, we review the basic molecular mechanisms that control the formation of the liver as an organ.
Collapse
|
182
|
Oliver G, Srinivasan RS. Endothelial cell plasticity: how to become and remain a lymphatic endothelial cell. Development 2010; 137:363-72. [PMID: 20081185 DOI: 10.1242/dev.035360] [Citation(s) in RCA: 112] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Lineage commitment and differentiation into mature cell types are mostly considered to be unidirectional and irreversible processes. However, recent results have challenged this by showing that terminally differentiated cell types can be reprogrammed into other cell types, an important step towards devising strategies for gene therapy and tissue regeneration. In this Review, we summarize recent data on the earliest steps in the development of the mammalian lymphatic vasculature: the specification of lymphatic endothelial cells (LECs). We elaborate on a developmental model that integrates the different steps leading to LEC differentiation and lymphatic network formation, discuss evidence that suggests that LEC fate is plastic, and consider the potentially far-reaching implications of the ability to convert one cell type into another.
Collapse
Affiliation(s)
- Guillermo Oliver
- Department of Genetics and Tumor Cell Biology, St Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105-3678, USA.
| | | |
Collapse
|
183
|
Charest-Marcotte A, Dufour CR, Wilson BJ, Tremblay AM, Eichner LJ, Arlow DH, Mootha VK, Giguère V. The homeobox protein Prox1 is a negative modulator of ERR{alpha}/PGC-1{alpha} bioenergetic functions. Genes Dev 2010; 24:537-42. [PMID: 20194433 PMCID: PMC2841331 DOI: 10.1101/gad.1871610] [Citation(s) in RCA: 121] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2009] [Accepted: 01/21/2010] [Indexed: 01/20/2023]
Abstract
Estrogen-related receptor alpha (ERRalpha) and proliferator-activated receptor gamma coactivator-1alpha (PGC-1alpha) play central roles in the transcriptional control of energy homeostasis, but little is known about factors regulating their activity. Here we identified the homeobox protein prospero-related homeobox 1 (Prox1) as one such factor. Prox1 interacts with ERRalpha and PGC-1alpha, occupies promoters of metabolic genes on a genome-wide scale, and inhibits the activity of the ERRalpha/PGC-1alpha complex. DNA motif analysis suggests that Prox1 interacts with the genome through tethering to ERRalpha and other factors. Importantly, ablation of Prox1 and ERRalpha have opposite effects on the respiratory capacity of liver cells, revealing an unexpected role for Prox1 in the control of energy homeostasis.
Collapse
Affiliation(s)
- Alexis Charest-Marcotte
- Goodman Cancer Centre, McGill University, Montréal, Québec H3A 1A3, Canada
- Department of Biochemistry, McGill University, Montréal, Québec H3G 1Y6, Canada
| | | | - Brian J. Wilson
- Goodman Cancer Centre, McGill University, Montréal, Québec H3A 1A3, Canada
| | - Annie M. Tremblay
- Goodman Cancer Centre, McGill University, Montréal, Québec H3A 1A3, Canada
- Department of Biochemistry, McGill University, Montréal, Québec H3G 1Y6, Canada
| | - Lillian J. Eichner
- Goodman Cancer Centre, McGill University, Montréal, Québec H3A 1A3, Canada
- Department of Biochemistry, McGill University, Montréal, Québec H3G 1Y6, Canada
| | - Daniel H. Arlow
- Department of Systems Biology, Massachusetts General Hospital, Cambridge, Massachusetts 02142, USA
- Centre for Human Genetic Research, Massachusetts General Hospital, Cambridge, Massachusetts 02142, USA
- Broad Institute of Massachusetts Institute of Technology/Harvard, Cambridge, Massachusetts 02142, USA
| | - Vamsi K. Mootha
- Department of Systems Biology, Massachusetts General Hospital, Cambridge, Massachusetts 02142, USA
- Centre for Human Genetic Research, Massachusetts General Hospital, Cambridge, Massachusetts 02142, USA
- Broad Institute of Massachusetts Institute of Technology/Harvard, Cambridge, Massachusetts 02142, USA
| | - Vincent Giguère
- Goodman Cancer Centre, McGill University, Montréal, Québec H3A 1A3, Canada
- Department of Biochemistry, McGill University, Montréal, Québec H3G 1Y6, Canada
- Department of Medicine, McGill University, Montréal, Québec H3G 1Y6, Canada
- Department of Oncology, McGill University, Montréal, Québec H3G 1Y6, Canada
| |
Collapse
|
184
|
Expression of PROX1 Is a common feature of high-grade malignant astrocytic gliomas. J Neuropathol Exp Neurol 2010; 69:129-38. [PMID: 20084020 DOI: 10.1097/nen.0b013e3181ca4767] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
PROX1 is a prospero-related transcription factor that plays a critical role in the development of various organs including the mammalian lymphatic and central nervous systems; it controls cell proliferation and differentiation through different transcription pathwaysand has both oncogenic and tumor-suppressive functions. We investigated PROX1 expression patterns in 56 human astrocytic gliomas of different grades using immunohistochemistry. An average of 79% of cells in World Health Organization Grade IV (glioblastoma, n = 15) and 57% of cells in World Health Organization Grade III (anaplastic astrocytoma, n = 13) were strongly PROX1 positive; low-grade diffuse astrocytomas (Grade II, n = 13) had 21% of cells that were strongly positive; Grade I tumors (n = 15) had 1.5%; and non-neoplastic brain tissue (n = 15) had 3.7% of cells that were PROX1 positive. Double immunolabeling showed that PROX1+ cells in glioblastomas frequently coexpressed early neuronal proteins MAP2 and betaIII-tubulin but not the mature neuronal marker protein NeuN. Analyses of coexpression with proliferation markers suggest that PROX1+ cells have a marginally lower rate of proliferation than other tumor cells but are still mitotically active. We conclude that PROX1 may constitute a useful tool for the diagnosis and grading ofastrocytic gliomas and for distinguishing Grade III and Grade IV tumors from Grade I and Grade II tumors.
Collapse
|
185
|
Negishi T, Nagai Y, Asaoka Y, Ohno M, Namae M, Mitani H, Sasaki T, Shimizu N, Terai S, Sakaida I, Kondoh H, Katada T, Furutani-Seiki M, Nishina H. Retinoic acid signaling positively regulates liver specification by inducing wnt2bb gene expression in medaka. Hepatology 2010; 51:1037-45. [PMID: 19957374 DOI: 10.1002/hep.23387] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
UNLABELLED During vertebrate embryogenesis, the liver develops at a precise location along the endodermal primitive gut tube because of signaling delivered by adjacent mesodermal tissues. Although several signaling molecules have been associated with liver formation, the molecular mechanism that regulates liver specification is still unclear. We previously performed a screen in medaka to isolate mutants with impaired liver development. The medaka hio mutants exhibit a profound (but transient) defect in liver specification that resembles the liver formation defect found in zebrafish prometheus (prt) mutants, whose mutation occurs in the wnt2bb gene. In addition to their liver abnormality, hio mutants lack pectoral fins and die after hatching. Positional cloning indicated that the hio mutation affects the raldh2 gene encoding retinaldehyde dehydrogenase type2 (RALDH2), the enzyme principally responsible for retinoic acid (RA) biosynthesis. Mutations of raldh2 in zebrafish preclude the development of pectoral fins. Interestingly, in hio mutants, expression of wnt2bb in the lateral plate mesoderm (LPM) directly adjacent to the liver-forming endoderm was completely lost. CONCLUSION Our data reveal the unexpected finding that RA signaling positively regulates the wnt2bb gene expression required for liver specification in medaka. These results suggest that a common molecular mechanism may underlie liver and pectoral fin specification during piscine embryogenesis.
Collapse
Affiliation(s)
- Takahiro Negishi
- Department of Developmental and Regenerative Biology, Medical Research Institute, Tokyo Medical and Dental University, Tokyo, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
186
|
Liver development, regeneration, and carcinogenesis. J Biomed Biotechnol 2010; 2010:984248. [PMID: 20169172 PMCID: PMC2821627 DOI: 10.1155/2010/984248] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2009] [Accepted: 11/12/2009] [Indexed: 02/06/2023] Open
Abstract
The identification of putative liver stem cells has brought closer the previously separate fields of liver development, regeneration, and carcinogenesis. Significant overlaps in the regulation of these processes are now being described. For example, studies in embryonic liver development have already provided the basis for directed differentiation of human embryonic stem cells and induced pluripotent stem cells into hepatocyte-like cells. As a result, the understanding of the cell biology of proliferation and differentiation in the liver has been improved. This knowledge can be used to improve the function of hepatocyte-like cells for drug testing, bioartificial livers, and transplantation. In parallel, the mechanisms regulating cancer cell biology are now clearer, providing fertile soil for novel therapeutic approaches. Recognition of the relationships between development, regeneration, and carcinogenesis, and the increasing evidence for the role of stem cells in all of these areas, has sparked fresh enthusiasm in understanding the underlying molecular mechanisms and has led to new targeted therapies for liver cirrhosis and primary liver cancers.
Collapse
|
187
|
Navarro-Alvarez N, Soto-Gutierrez A, Kobayashi N. Hepatic stem cells and liver development. Methods Mol Biol 2010; 640:181-236. [PMID: 20645053 DOI: 10.1007/978-1-60761-688-7_10] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The liver consists of many cell types with specialized functions. Hepatocytes are one of the main players in the organ and therefore are the most vulnerable cells to damage. Since they are not everlasting cells, they need to be replenished throughout life. Although the capacity of hepatocytes to contribute to their own maintenance has long been recognized, recent studies have indicated the presence of both intrahepatic and extrahepatic stem/progenitor cell populations that serve to maintain the normal organ and to regenerate damaged parenchyma in response to a variety of insults.The intrahepatic compartment most likely derives primarily from the biliary tree, particularly the most proximal branches, i.e. the canals of Hering and smallest ductules. The extrahepatic compartment is at least in part derived from diverse populations of cells from the bone marrow. Embryonic stem cells (ES's) are considered as a part of the extrahepatic compartment. Due to their pluripotent capabilities, ES cell-derived cells form a potential future source of hepatocytes, to replace or restore hepatic tissues that have been damaged by disease or injury. Progressing knowledge about stem cells in the liver would allow a better understanding of the mechanisms of hepatic homeostasis and regeneration. Although a human stem cell-derived cell type equivalent to primary hepatocytes does not yet exist, the promising results obtained with extrahepatic stem cells would open the way to cell-based therapy for liver diseases.
Collapse
Affiliation(s)
- Nalu Navarro-Alvarez
- Department of Surgery, Okayama University Graduate School of Medicine and Dentistry, Okayama, Japan
| | | | | |
Collapse
|
188
|
Lemaigre F. Markers and signaling factors for stem cell differentiation to hepatocytes: lessons from developmental studies. Methods Mol Biol 2010; 640:157-66. [PMID: 20645051 DOI: 10.1007/978-1-60761-688-7_8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Liver transplantation is the preferred option to treat a number of hepatic diseases in adults and children, but the number of patients on the waiting list is exceeding the number of available livers for transplantation. Hepatocytes differentiated in vitro from stem cells are a promising and renewable source of tissue for transplantation. The principles guiding programmed differentiation of stem cells to hepatocytes are largely based on knowledge gained from studies on embryonic development of the liver. How key findings in developmental biology are translated into cell culture protocols driving stepwise differentiation of hepatocytes is illustrated in this chapter.
Collapse
Affiliation(s)
- Frédéric Lemaigre
- de Duve Institute, Université Catholique de Louvain, Brussels, Belgium
| |
Collapse
|
189
|
Pistocchi A, Feijóo CG, Cabrera P, Villablanca EJ, Allende ML, Cotelli F. The zebrafish prospero homolog prox1 is required for mechanosensory hair cell differentiation and functionality in the lateral line. BMC DEVELOPMENTAL BIOLOGY 2009; 9:58. [PMID: 19948062 PMCID: PMC2794270 DOI: 10.1186/1471-213x-9-58] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/14/2009] [Accepted: 11/30/2009] [Indexed: 11/20/2022]
Abstract
Background The lateral line system in zebrafish is composed of a series of organs called neuromasts, which are distributed over the body surface. Neuromasts contain clusters of hair cells, surrounded by accessory cells. Results In this report we describe zebrafish prox1 mRNA expression in the migrating primordium and in the neuromasts of the posterior lateral line. Furthermore, using an antibody against Prox1 we characterize expression of the protein in different cell types within neuromasts, and we show distribution among the supporting cells and hair cells. Conclusion Functional analysis using antisense morpholinos indicates that prox1 activity is crucial for the hair cells to differentiate properly and acquire functionality, while having no role in development of other cell types in neuromasts.
Collapse
Affiliation(s)
- Anna Pistocchi
- Department of Biology, Università degli Studi di Milano, Milan, Italy.
| | | | | | | | | | | |
Collapse
|
190
|
Prox1 expression in rod precursors and Müller cells. Exp Eye Res 2009; 90:267-76. [PMID: 19895810 DOI: 10.1016/j.exer.2009.10.015] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2009] [Revised: 10/07/2009] [Accepted: 10/29/2009] [Indexed: 01/08/2023]
Abstract
The transcription factor Prox1 acts in rodent retinogenesis, at least in promoting cell cycle withdrawal and horizontal cell production. In the mature retina, this protein is detected at the inner nuclear layer of all vertebrate groups. We have made a neurochemical characterisation of Prox1(+) cell types in two different vertebrate groups: mammals and fish. As well as Prox1(+) horizontal cells, we have observed Prox1(+)/PKC-alpha(+) rod bipolar cells in mouse and cone ON and mixed b bipolar cells in goldfish. In mouse, only some CB(+) and CR(+) amacrine cells are Prox1(+) and the TH(+) and CR(+) amacrine cells are Prox1(-). However, in goldfish all CR(+) amacrine cells and TH(+) interplexiform cells are Prox1(+) and in the GCL displaced amacrine cells are also Prox1(+). Besides its expression in different interneuron subpopulations, we demonstrate, for the first time, the presence of Prox1 in the GS(+) and CRALBP(+) Müller cells in the retina of adult mammals and in developing and mature retina of fish. The presence of Prox1 in these cells appears to be related to survival or maintenance of their phenotype. We also demonstrate that in fish, where retinal formation persists into adulthood, Prox1 is expressed in dividing PCNA(+) cells at the peripheral growing zone, in rod progenitors at the inner and outer nuclear layers as well as in early progenitors during a retinal regeneration process after cryo-lesion of the peripheral growing zone. Therefore, Prox1 functions in vertebrate retinogenesis may be more complex than previously expected.
Collapse
|
191
|
Horra A, Salazar J, Ferré R, Vallvé JC, Guardiola M, Rosales R, Masana L, Ribalta J. Prox-1 and FOXC2 gene expression in adipose tissue: A potential contributory role of the lymphatic system to familial combined hyperlipidaemia. Atherosclerosis 2009; 206:343-5. [DOI: 10.1016/j.atherosclerosis.2009.02.026] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2009] [Revised: 02/17/2009] [Accepted: 02/18/2009] [Indexed: 11/25/2022]
|
192
|
Liver development in zebrafish (Danio rerio). J Genet Genomics 2009; 36:325-34. [PMID: 19539242 DOI: 10.1016/s1673-8527(08)60121-6] [Citation(s) in RCA: 113] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2009] [Revised: 04/17/2009] [Accepted: 04/17/2009] [Indexed: 12/17/2022]
Abstract
Liver is one of the largest internal organs in the body and its importance for metabolism, detoxification and homeostasis has been well established. In this review, we summarized recent progresses in studying liver initiation and development during embryogenesis using zebrafish as a model system. We mainly focused on topics related to the specification of hepatoblasts from endoderm, the formation and growth of liver bud, the differentiation of hepatocytes and bile duct cells from hepatoblasts, and finally the role of mesodermal signals in controlling liver development in zebrafish.
Collapse
|
193
|
Lemaigre FP. Mechanisms of liver development: concepts for understanding liver disorders and design of novel therapies. Gastroenterology 2009; 137:62-79. [PMID: 19328801 DOI: 10.1053/j.gastro.2009.03.035] [Citation(s) in RCA: 171] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2009] [Revised: 03/15/2009] [Accepted: 03/18/2009] [Indexed: 12/12/2022]
Abstract
The study of liver development has significantly contributed to developmental concepts about morphogenesis and differentiation of other organs. Knowledge of the mechanisms that regulate hepatic epithelial cell differentiation has been essential in creating efficient cell culture protocols for programmed differentiation of stem cells to hepatocytes as well as developing cell transplantation therapies. Such knowledge also provides a basis for the understanding of human congenital diseases. Importantly, much of our understanding of organ development has arisen from analyses of patients with liver deficiencies. We review how the liver develops in the embryo and discuss the concepts that operate during this process. We focus on the mechanisms that control the differentiation and organization of the hepatocytes and cholangiocytes and refer to other reviews for the development of nonepithelial tissue in the liver. Much progress in the characterization of liver development has been the result of genetic studies of human diseases; gaining a better understanding of these mechanisms could lead to new therapeutic approaches for patients with liver disorders.
Collapse
|
194
|
Wandzioch E, Zaret KS. Dynamic signaling network for the specification of embryonic pancreas and liver progenitors. Science 2009; 324:1707-10. [PMID: 19556507 PMCID: PMC2771431 DOI: 10.1126/science.1174497] [Citation(s) in RCA: 200] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Studies of the formation of pancreas and liver progenitors have focused on individual inductive signals and cellular responses. Here, we investigated how bone morphogenetic protein, transforming growth factor-beta (TGFbeta), and fibroblast growth factor signaling pathways converge on the earliest genes that elicit pancreas and liver induction in mouse embryos. The inductive network was found to be dynamic; it changed within hours. Different signals functioned in parallel to induce different early genes, and two permutations of signals induced liver progenitor domains, which revealed flexibility in cell programming. Also, the specification of pancreas and liver progenitors was restricted by the TGFbeta pathway. These findings may enhance progenitor cell specification from stem cells for biomedical purposes and can help explain incomplete programming in stem cell differentiation protocols.
Collapse
Affiliation(s)
- Ewa Wandzioch
- Cell and Developmental Biology Program, Fox Chase Cancer Center, 333 Cottman Avenue, Philadelphia, PA 19111, USA
| | | |
Collapse
|
195
|
Kan NG, Junghans D, Izpisua Belmonte JCI. Compensatory growth mechanisms regulated by BMP and FGF signaling mediate liver regeneration in zebrafish after partial hepatectomy. FASEB J 2009; 23:3516-25. [PMID: 19546304 DOI: 10.1096/fj.09-131730] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Here, we describe the zebrafish (Danio rerio) as a vertebrate model system to study liver regeneration with the added benefit of its powerful genetics and screening possibilities to uncover the molecular pathways underlying liver regeneration. We developed a partial hepatectomy (PH) protocol in zebrafish and investigated in detail the cellular and morphological changes during the process of liver regeneration. We show that the type of regenerative response is dependent on the size of the injury sustained by the zebrafish liver. Furthermore, we demonstrate for the first time that the mechanisms of liver regeneration in zebrafish after PH are strikingly similar to those of rodents and humans, with 100% recovery of the liver mass after 6-7 d postsurgery. This occurs via compensatory growth mediated by proliferation of hepatocytes throughout the entire liver remnant. By analyzing transgenic fish expressing dominant-negative forms of either bone morphogenetic protein (BMP) receptor or fibroblast growth factor (FGF) receptor 1, we demonstrate that the BMP and FGF signaling pathways are crucial regulators of the early events during liver regeneration after PH. Our study demonstrates that the mechanisms of liver regeneration in zebrafish are highly similar to the processes ongoing during mammalian liver regeneration and make the adult zebrafish a suitable model system to study the mechanisms of liver regeneration.
Collapse
Affiliation(s)
- Natalia G Kan
- Gene Expression Laboratory, Salk Institute for Biological Studies, 10010 N. Torrey Pines Rd., La Jolla, CA 92037, USA
| | | | | |
Collapse
|
196
|
Tanaka M, Okabe M, Suzuki K, Kamiya Y, Tsukahara Y, Saito S, Miyajima A. Mouse hepatoblasts at distinct developmental stages are characterized by expression of EpCAM and DLK1: drastic change of EpCAM expression during liver development. Mech Dev 2009; 126:665-76. [PMID: 19527784 DOI: 10.1016/j.mod.2009.06.939] [Citation(s) in RCA: 100] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2008] [Revised: 05/29/2009] [Accepted: 06/06/2009] [Indexed: 12/15/2022]
Abstract
Hepatoblasts are hepatic progenitor cells that expand and give rise to either hepatocyte or cholangiocytes during liver development. We previously reported that delta-like 1 homolog (DLK1) is expressed in the mouse liver primordium at embryonic day (E) 10.5 and that DLK1(+) cells in E14.5 liver contain high proliferative and bipotential hepatoblasts. While the expression of epithelial cell adhesion molecule (EpCAM) in hepatic stem/progenitor cells has been reported, its expression profile at an early stage of liver development remains unknown. In this study, we show that EpCAM is expressed in mouse liver bud at E9.5 and that EpCAM(+)DLK1(+) hepatoblasts form hepatic cords at the early stage of hepatogenesis. DLK1(+) cells of E11.5 liver were fractionated into EpCAM(+) and EpCAM(-) cells; one forth of EpCAM(+)DLK1(+) cells formed a colony in vitro whereas EpCAM(-)DLK1(+) cells rarely did it. Moreover, EpCAM(+)DLK1(+) cells contained cells capable of forming a large colony, indicating that EpCAM(+)DLK1(+) cells in E11.5 liver contain early hepatoblasts with high proliferation potential. Interestingly, EpCAM expression in hepatoblasts was dramatically reduced along with liver development and the colony-forming capacities of both EpCAM(+)DLK1(+) and EpCAM(-)DLK1(+) cells were comparable in E14.5 liver. It strongly suggested that most of mouse hepatoblasts are losing EpCAM expression at this stage. Moreover, we provide evidence that EpCAM(+)DLK1(+) cells in E11.5 liver contain extrahepatic bile duct cells as well as hepatoblasts, while EpCAM(-)DLK1(+) cells contain mesothelial cell precursors. Thus, the expression of EpCAM and DLK1 suggests the developmental pathways of mouse liver progenitors.
Collapse
Affiliation(s)
- Minoru Tanaka
- Institute of Molecular and Cellular Biosciences, The University of Tokyo, 1-1-1 Yayoi, Tokyo 113-0032, Japan.
| | | | | | | | | | | | | |
Collapse
|
197
|
Molecular cloning and gene expression of the prox1a and prox1b genes in the medaka, Oryzias latipes. Gene Expr Patterns 2009; 9:341-7. [DOI: 10.1016/j.gep.2009.02.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2009] [Revised: 01/29/2009] [Accepted: 02/04/2009] [Indexed: 11/18/2022]
|
198
|
Lüdtke THW, Christoffels VM, Petry M, Kispert A. Tbx3 promotes liver bud expansion during mouse development by suppression of cholangiocyte differentiation. Hepatology 2009; 49:969-78. [PMID: 19140222 DOI: 10.1002/hep.22700] [Citation(s) in RCA: 97] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
UNLABELLED After specification of the hepatic endoderm, mammalian liver organogenesis progresses through a series of morphological stages that culminate in the migration of hepatocytes into the underlying mesenchyme to populate the hepatic lobes. Here, we show that in the mouse the transcriptional repressor Tbx3, a member of the T-box protein family, is required for the transition from a hepatic diverticulum with a pseudo-stratified epithelium to a cell-emergent liver bud. In Tbx3-deficient embryos, proliferation in the hepatic epithelium is severely reduced, hepatoblasts fail to delaminate, and cholangiocyte rather than hepatocyte differentiation occurs. Molecular analyses suggest that the primary function of Tbx3 is to maintain expression of hepatocyte transcription factors, including hepatic nuclear factor 4a (Hnf4a) and CCAAT/enhancer binding protein (C/EBP), alpha (Cebpa), and to repress expression of cholangiocyte transcription factors such as Onecut1 (Hnf6) and Hnf1b. CONCLUSION Tbx3 controls liver bud expansion by suppressing cholangiocyte and favoring hepatocyte differentiation in the liver bud.
Collapse
Affiliation(s)
- Timo H-W Lüdtke
- Institut für Molekularbiologie, Medizinische Hochschule Hannover, Hannover, Germany
| | | | | | | |
Collapse
|
199
|
Repression of interferon-gamma expression in T cells by Prospero-related homeobox protein. Cell Res 2009; 18:911-20. [PMID: 19160541 DOI: 10.1038/cr.2008.275] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Interferon-gamma (IFN-gamma) is a major proinflammatory effector and regulatory cytokine produced by activated T cells and NK cells. IFN-gamma has been shown to play pivotal roles in fundamental immunological processes such as inflammatory reactions, cell-mediated immunity and autoimmunity. A variety of human disorders have now been linked to irregular IFN-gamma expression. In order to achieve proper IFN-gamma-mediated immunological effects, IFN-gamma expression in T cells is subject to both positive and negative regulation. In this study, we report for the first time the negative regulation of IFN-gamma expression by Prospero-related Homeobox (Prox1). In Jurkat T cells and primary human CD4+ T cells, Prox1 expression decreases quickly upon T cell activation, concurrent with a dramatic increase in IFN-gamma expression. Reporter analysis and chromatin immunoprecipitation (ChIP) revealed that Prox1 associates with and inhibits the transcription activity of IFN-,gammapromoter in activated Jurkat T cells. Co-immunoprecipitation and GST pull-down assay demonstrated a direct binding between Prox1 and the nuclear receptor peroxisome proliferator-activated receptor gamma (PPPARgamma, which is also an IFN-gamma repressor in T cells. By introducing deletions and mutations into Prox1, we show that the repression of IFN-gamma promoter by Prox1 is largely dependent upon the physical interaction between Prox1 and PPPARgamma Furthermore, PPPARgammaantagonist treatment removes Prox1 from IFN-gamma promoter and attenuates repression of IFN-gamma expression by Prox1. These findings establish Prox1 as a new negative regulator of IFN-gamma expression in T cells and will aid in the understanding of IFN-gamma transcription regulation mechanisms.
Collapse
|
200
|
Pistocchi A, Bartesaghi S, Cotelli F, Del Giacco L. Identification and expression pattern of zebrafish prox2 during embryonic development. Dev Dyn 2009; 237:3916-20. [PMID: 19035352 DOI: 10.1002/dvdy.21798] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
Prox2, together with the previously isolated Prox1, is the vertebrate homolog of the Drosophila homeobox-containing gene prospero, the founder member of a family of transcription factors which have been shown to play critical roles in many developmental events. We have isolated a cDNA which encodes a putative protein that shares a high degree of homology with mammalian Prox1, Prox2, and zebrafish Prox1. Comparative genomic analysis revealed that this protein corresponds to the zebrafish Prox2 homolog being the gene syntenic with the chromosome region hosting mouse Prox2. Whole-mount in situ experiments demonstrated that prox2 is expressed, during zebrafish embryonic development, in defined structures of the central nervous system and the eye, as previously reported in mouse. Additionally, reverse transcriptase-polymerase chain reaction analysis disclosed prox2 expression in several adult organs. Finally, prox1 loss- and gain-of-function assays have been carried out to search for regulative effects on prox2 expression.
Collapse
Affiliation(s)
- Anna Pistocchi
- Department of Biology, Università degli Studi di Milano, Milano, Italy
| | | | | | | |
Collapse
|