Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 19 de 19
1.
Development ; 150(16)2023 08 15.
Article En | MEDLINE | ID: mdl-37497580

Earlier data on liver development demonstrated that morphogenesis of the bile duct, portal mesenchyme and hepatic artery is interdependent, yet how this interdependency is orchestrated remains unknown. Here, using 2D and 3D imaging, we first describe how portal mesenchymal cells become organised to form hepatic arteries. Next, we examined intercellular signalling active during portal area development and found that axon guidance genes are dynamically expressed in developing bile ducts and portal mesenchyme. Using tissue-specific gene inactivation in mice, we show that the repulsive guidance molecule BMP co-receptor A (RGMA)/neogenin (NEO1) receptor/ligand pair is dispensable for portal area development, but that deficient roundabout 2 (ROBO2)/SLIT2 signalling in the portal mesenchyme causes reduced maturation of the vascular smooth muscle cells that form the tunica media of the hepatic artery. This arterial anomaly does not impact liver function in homeostatic conditions, but is associated with significant tissular damage following partial hepatectomy. In conclusion, our work identifies new players in development of the liver vasculature in health and liver regeneration.


Axon Guidance , Hepatic Artery , Animals , Mice , Bile Ducts , Morphogenesis , Gene Silencing
2.
Front Cell Dev Biol ; 10: 995013, 2022.
Article En | MEDLINE | ID: mdl-36238685

KRAS mutants are common in many cancers and wild-type KRAS is essential in development as its absence causes embryonic lethality. Despite this critical role in development and disease, the normal expression pattern of KRAS protein is still largely unknown at the tissue level due to the lack of valid antibodies. To address this issue, we used the citrine-Kras mouse model in which the Citrine-KRAS (Cit-K) fusion protein functions as a validated surrogate of endogenous KRAS protein that can be detected on tissue sections by immunolabeling with a GFP antibody. In the embryo, we found expression of KRAS protein in a wide range of organs and tissues. This expression tends to decrease near birth, mainly in mesenchymal cells. During transition to the adult stage, the dynamics of KRAS protein expression vary among organs and detection of KRAS becomes restricted to specific cell types. Furthermore, we found that steady state KRAS protein expression is detectable at the cell membrane and in the cytoplasm and that this subcellular partitioning differed among cell types. Our results reveal hitherto unanticipated dynamics in developmental, tissular, cell-specific and subcellular expression of KRAS protein. They provide insight into the reason why specific cell-types are sensitive to KRAS mutations during cancer initiation.

3.
PLoS Comput Biol ; 17(4): e1008854, 2021 04.
Article En | MEDLINE | ID: mdl-33819288

Colony Stimulating Factor 1 Receptor (CSF1R) is a potential target for anti-epileptic drugs. However, inhibition of CSF1R is not well tolerated by patients, thereby prompting the need for alternative targets. To develop a framework for identification of such alternatives, we here develop a mathematical model of a pro-inflammatory gene regulatory network (GRN) involved in epilepsy and centered around CSF1R. This GRN comprises validated transcriptional and post-transcriptional regulations involving STAT1, STAT3, NFκB, IL6R, CSF3R, IRF8, PU1, C/EBPα, TNFR1, CSF1 and CSF1R. The model was calibrated on mRNA levels of all GRN components in lipopolysaccharide (LPS)-treated mouse microglial BV-2 cells, and allowed to predict that STAT1 and STAT3 have the strongest impact on the expression of the other GRN components. Microglial BV-2 cells were selected because, the modules from which the GRN was deduced are enriched for microglial marker genes. The function of STAT1 and STAT3 in the GRN was experimentally validated in BV-2 cells. Further, in silico analysis of the GRN dynamics predicted that a pro-inflammatory stimulus can induce irreversible bistability whereby the expression level of GRN components occurs as two distinct states. The irreversibility of the switch may enforce the need for chronic inhibition of the CSF1R GRN in order to achieve therapeutic benefit. The cell-to-cell heterogeneity driven by the bistability may cause variable therapeutic response. In conclusion, our modeling approach uncovered a GRN controlling CSF1R that is predominantly regulated by STAT1 and STAT3. Irreversible inflammation-induced bistability and cell-to-cell heterogeneity of the GRN provide a theoretical foundation to the need for chronic GRN control and the limited potential for disease modification via inhibition of CSF1R.


Epilepsy/genetics , Gene Regulatory Networks , Models, Biological , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Signal Transduction/genetics , Animals , Cell Line , Mice , STAT1 Transcription Factor/metabolism , STAT3 Transcription Factor/metabolism
4.
J Cell Mol Med ; 25(9): 4387-4394, 2021 05.
Article En | MEDLINE | ID: mdl-33830670

Myofibroma is a benign pericytic tumour affecting young children. The presence of multicentric myofibromas defines infantile myofibromatosis (IMF), which is a life-threatening condition when associated with visceral involvement. The disease pathophysiology remains poorly characterized. In this study, we performed deep RNA sequencing on eight myofibroma samples, including two from patients with IMF. We identified five different in-frame gene fusions in six patients, including three previously described fusion transcripts, SRF-CITED1, SRF-ICA1L and MTCH2-FNBP4, and a fusion of unknown significance, FN1-TIMP1. We found a novel COL4A1-VEGFD gene fusion in two cases, one of which also carried a PDGFRB mutation. We observed a robust expression of VEGFD by immunofluorescence on the corresponding tumour sections. Finally, we showed that the COL4A1-VEGFD chimeric protein was processed to mature VEGFD growth factor by proteases, such as the FURIN proprotein convertase. In conclusion, our results unravel a new recurrent gene fusion that leads to VEGFD production under the control of the COL4A1 gene promoter in myofibroma. This fusion is highly reminiscent of the COL1A1-PDGFB oncogene associated with dermatofibrosarcoma protuberans. This work has implications for the diagnosis and, possibly, the treatment of a subset of myofibromas.


Biomarkers, Tumor/genetics , Collagen Type IV/genetics , Gene Expression Regulation, Neoplastic , Gene Fusion , Myofibroma/pathology , Vascular Endothelial Growth Factor D/genetics , Humans , Myofibroma/genetics , Prognosis
5.
Hepatology ; 74(3): 1445-1460, 2021 09.
Article En | MEDLINE | ID: mdl-33768568

BACKGROUND AND AIMS: Earlier diagnosis and treatment of intrahepatic cholangiocarcinoma (iCCA) are necessary to improve therapy, yet limited information is available about initiation and evolution of iCCA precursor lesions. Therefore, there is a need to identify mechanisms driving formation of precancerous lesions and their progression toward invasive tumors using experimental models that faithfully recapitulate human tumorigenesis. APPROACH AND RESULTS: To this end, we generated a mouse model which combines cholangiocyte-specific expression of KrasG12D with 3,5-diethoxycarbonyl-1,4-dihydrocollidine (DDC) diet-induced inflammation to mimic iCCA development in patients with cholangitis. Histological and transcriptomic analyses of the mouse precursor lesions and iCCA were performed and compared with human analyses. The function of genes overexpressed during tumorigenesis was investigated in human cell lines. We found that mice expressing KrasG12D in cholangiocytes and fed a DDC diet developed cholangitis, ductular proliferations, intraductal papillary neoplasms of bile ducts (IPNBs), and, eventually, iCCAs. The histology of mouse and human IPNBs was similar, and mouse iCCAs displayed histological characteristics of human mucin-producing, large-duct-type iCCA. Signaling pathways activated in human iCCA were also activated in mice. The identification of transition zones between IPNB and iCCA on tissue sections, combined with RNA-sequencing analyses of the lesions supported that iCCAs derive from IPNBs. We further provide evidence that tensin-4 (TNS4), which is stimulated by KRASG12D and SRY-related HMG box transcription factor 17, promotes tumor progression. CONCLUSIONS: We developed a mouse model that faithfully recapitulates human iCCA tumorigenesis and identified a gene cascade which involves TNS4 and promotes tumor progression.


Bile Duct Neoplasms/genetics , Carcinoma, Ductal/genetics , Cholangiocarcinoma/genetics , Disease Models, Animal , Liver Neoplasms, Experimental/genetics , Mice , Tensins/genetics , Animals , Bile Duct Neoplasms/chemically induced , Bile Duct Neoplasms/metabolism , Bile Duct Neoplasms/pathology , Carcinoma, Ductal/chemically induced , Carcinoma, Ductal/metabolism , Carcinoma, Ductal/pathology , Carcinoma, Papillary/chemically induced , Carcinoma, Papillary/genetics , Carcinoma, Papillary/metabolism , Carcinoma, Papillary/pathology , Cholangiocarcinoma/chemically induced , Cholangiocarcinoma/metabolism , Cholangiocarcinoma/pathology , Cholangitis/chemically induced , Cholangitis/complications , HMGB Proteins/genetics , HMGB Proteins/metabolism , Liver Neoplasms, Experimental/chemically induced , Liver Neoplasms, Experimental/metabolism , Liver Neoplasms, Experimental/pathology , Proto-Oncogene Proteins p21(ras)/genetics , Pyridines/toxicity , SOXF Transcription Factors/genetics , SOXF Transcription Factors/metabolism , Signal Transduction , Tensins/metabolism
6.
J Hepatol ; 71(2): 323-332, 2019 08.
Article En | MEDLINE | ID: mdl-30953666

BACKGROUND & AIMS: Alterations of individual genes variably affect the development of hepatocellular carcinoma (HCC). Thus, we aimed to characterize the function of tumor-promoting genes in the context of gene regulatory networks (GRNs). METHODS: Using data from The Cancer Genome Atlas, from the LIRI-JP (Liver Cancer - RIKEN, JP project), and from our transcriptomic, transfection and mouse transgenic experiments, we identify a GRN which functionally links LIN28B-dependent dedifferentiation with dysfunction of ß-catenin (CTNNB1). We further generated and validated a quantitative mathematical model of the GRN using human cell lines and in vivo expression data. RESULTS: We found that LIN28B and CTNNB1 form a GRN with SMARCA4, Let-7b (MIRLET7B), SOX9, TP53 and MYC. GRN functionality is detected in HCC and gastrointestinal cancers, but not in other cancer types. GRN status negatively correlates with HCC prognosis, and positively correlates with hyperproliferation, dedifferentiation and HGF/MET pathway activation, suggesting that it contributes to a transcriptomic profile typical of the proliferative class of HCC. The mathematical model predicts how the expression of GRN components changes when the expression of another GRN member varies or is inhibited by a pharmacological drug. The dynamics of GRN component expression reveal distinct cell states that can switch reversibly in normal conditions, and irreversibly in HCC. The mathematical model is available via a web-based tool which can evaluate the GRN status of HCC samples and predict the impact of therapeutic agents on the GRN. CONCLUSIONS: We conclude that identification and modelling of the GRN provide insights into the prognosis of HCC and the mechanisms by which tumor-promoting genes impact on HCC development. LAY SUMMARY: Hepatocellular carcinoma (HCC) is a heterogeneous disease driven by the concomitant deregulation of several genes functionally organized as networks. Here, we identified a gene regulatory network involved in a subset of HCCs. This subset is characterized by increased proliferation and poor prognosis. We developed a mathematical model which uncovers the dynamics of the network and allows us to predict the impact of a therapeutic agent, not only on its specific target but on all the genes belonging to the network.


Carcinoma, Hepatocellular/genetics , Gene Regulatory Networks/drug effects , Liver Neoplasms/genetics , Models, Theoretical , beta Catenin/genetics , beta Catenin/metabolism , Animals , Carcinoma, Hepatocellular/pathology , Cohort Studies , Hep G2 Cells , Humans , Liver Neoplasms/pathology , Mice , Mice, Transgenic , Prognosis , RNA-Binding Proteins/antagonists & inhibitors , RNA-Binding Proteins/metabolism , Sequence Analysis, RNA , Transcriptome , Transfection
7.
Hepatology ; 67(1): 313-327, 2018 01.
Article En | MEDLINE | ID: mdl-28833283

Transcriptional networks control the differentiation of the hepatocyte and cholangiocyte lineages from embryonic liver progenitor cells and their subsequent maturation to the adult phenotype. However, how relative levels of hepatocyte and cholangiocyte gene expression are determined during differentiation remains poorly understood. Here, we identify microRNA (miR)-337-3p as a regulator of liver development. miR-337-3p stimulates expression of cholangiocyte genes and represses hepatocyte genes in undifferentiated progenitor cells in vitro and in embryonic mouse livers. Beyond the stage of lineage segregation, miR-337-3p controls the transcriptional network dynamics of developing hepatocytes and balances both cholangiocyte populations that constitute the ductal plate. miR-337-3p requires Notch and transforming growth factor-ß signaling and exerts a biphasic control on the hepatocyte transcription factor hepatocyte nuclear factor 4α by modulating its activation and repression. With the help of an experimentally validated mathematical model, we show that this biphasic control results from an incoherent feedforward loop between miR-337-3p and hepatocyte nuclear factor 4α. CONCLUSION: Our results identify miR-337-3p as a regulator of liver development and highlight how tight quantitative control of hepatic cell differentiation is exerted through specific gene regulatory network motifs. (Hepatology 2018;67:313-327).


Cell Differentiation/genetics , Gene Expression Regulation, Developmental , Hepatocyte Nuclear Factor 1-alpha/genetics , Hepatocytes/metabolism , MicroRNAs/genetics , Animals , Blotting, Western , Cells, Cultured , Mice , Signal Transduction/genetics , Statistics, Nonparametric , Transcription Factors
8.
Differentiation ; 91(1-3): 42-9, 2016.
Article En | MEDLINE | ID: mdl-26856660

Beta-catenin is known to play stage- and cell-specific functions during liver development. However, its role in development of bile ducts has not yet been addressed. Here we used stage-specific in vivo gain- and loss-of-function approaches, as well as lineage tracing experiments in the mouse, to first demonstrate that ß-catenin is dispensable for differentiation of liver precursor cells (hepatoblasts) to cholangiocyte precursors. Second, when ß-catenin was depleted in the latter, maturation of cholangiocytes, bile duct morphogenesis and differentiation of periportal hepatocytes from cholangiocyte precursors was normal. In contrast, stabilization of ß-catenin in cholangiocyte precursors perturbed duct development and cholangiocyte differentiation. We conclude that ß-catenin is dispensable for biliary development but that its activity must be kept within tight limits. Our work is expected to significantly impact on in vitro differentiation of stem cells to cholangiocytes for toxicology studies and disease modeling.


Bile Ducts/growth & development , Liver/growth & development , Morphogenesis/genetics , beta Catenin/genetics , Animals , Bile Ducts/cytology , Bile Ducts/metabolism , Cell Differentiation/genetics , Gene Expression Regulation, Developmental , Hepatocytes/cytology , Hepatocytes/metabolism , Liver/metabolism , Mice
9.
PLoS One ; 10(6): e0132295, 2015.
Article En | MEDLINE | ID: mdl-26125584

Biliary cysts in adult patients affected by polycystic liver disease are lined by cholangiocytes that proliferate, suggesting that initiation of cyst formation depends on proliferation. Here, we challenge this view by analyzing cyst-lining cell proliferation and differentiation in Cpk mouse embryos and in livers from human fetuses affected by Autosomal Recessive Polycystic Kidney Disease (ARPKD), at early stages of cyst formation. Proliferation of fetal cholangiocyte precursors, measured by immunostaining in human and mouse livers, was low and did not differ between normal and ARPKD or Cpk livers, excluding excessive proliferation as an initiating cause of liver cysts. Instead, our analyses provide evidence that the polycystic livers exhibit increased and accelerated differentiation of hepatoblasts into cholangiocyte precursors, eventually coalescing into large biliary cysts. Lineage tracing experiments, performed in mouse embryos, indicated that the cholangiocyte precursors in Cpk mice generate cholangiocytes and periportal hepatocytes, like in wild-type animals. Therefore, contrary to current belief, cyst formation in polycystic liver disease does not necessarily depend on overproliferation. Combining our prenatal data with available data from adult livers, we propose that polycystic liver can be initiated by proliferation-independent mechanisms at a fetal stage, followed by postnatal proliferation-dependent cyst expansion.


Biliary Tract/pathology , Cell Proliferation/physiology , Choledochal Cyst/pathology , Cysts/pathology , Liver Diseases/pathology , Polycystic Kidney, Autosomal Recessive/pathology , Animals , Bile Duct Diseases/genetics , Bile Duct Diseases/pathology , Biliary Tract/cytology , Cell Differentiation , Cysts/genetics , Disease Models, Animal , Fetus/pathology , Hepatocytes/cytology , Humans , Liver/pathology , Liver Diseases/genetics , Membrane Proteins/genetics , Mice , Mice, Inbred BALB C , Mice, Transgenic , Polycystic Kidney, Autosomal Recessive/genetics , Tamoxifen/pharmacology
10.
Dev Biol ; 404(2): 136-48, 2015 Aug 15.
Article En | MEDLINE | ID: mdl-26033091

In developing liver, cholangiocytes derive from the hepatoblasts and organize to form the bile ducts. Earlier work has shown that the SRY-related High Mobility Group box transcription factor 9 (SOX9) is transiently required for bile duct development, raising the question of the potential involvement of other SOX family members in biliary morphogenesis. Here we identify SOX4 as a new regulator of cholangiocyte development. Liver-specific inactivation of SOX4, combined or not with inactivation of SOX9, affects cholangiocyte differentiation, apico-basal polarity and bile duct formation. Both factors cooperate to control the expression of mediators of the Transforming Growth Factor-ß, Notch, and Hippo-Yap signaling pathways, which are required for normal development of the bile ducts. In addition, SOX4 and SOX9 control formation of primary cilia, which are known signaling regulators. The two factors also stimulate secretion of laminin α5, an extracellular matrix component promoting bile duct maturation. We conclude that SOX4 is a new regulator of liver development and that it exerts a pleiotropic control on bile duct development in cooperation with SOX9.


Bile Ducts, Intrahepatic/embryology , SOX9 Transcription Factor/genetics , SOXC Transcription Factors/genetics , Adaptor Proteins, Signal Transducing/biosynthesis , Animals , Bile Ducts, Intrahepatic/growth & development , Cell Cycle Proteins , Cell Differentiation/genetics , Cells, Cultured , Gene Expression Regulation, Developmental , Hippo Signaling Pathway , Laminin/metabolism , Mice , Mice, Knockout , Organogenesis/genetics , Phosphoproteins/biosynthesis , Protein Serine-Threonine Kinases/biosynthesis , Receptors, Notch/biosynthesis , SOX9 Transcription Factor/biosynthesis , SOXC Transcription Factors/biosynthesis , Transforming Growth Factor beta/biosynthesis , YAP-Signaling Proteins
11.
Development ; 141(3): 538-47, 2014 Feb.
Article En | MEDLINE | ID: mdl-24449835

The liver has multiple functions that preserve homeostasis. Liver diseases are debilitating, costly and often result in death. Elucidating the developmental mechanisms that establish the liver's architecture or generate the cellular diversity of this organ should help advance the prevention, diagnosis and treatment of hepatic diseases. We previously reported that migration of early hepatic precursors away from the gut epithelium requires the activity of the homeobox gene Prox1. Here, we show that Prox1 is a novel regulator of cell differentiation and morphogenesis during hepatogenesis. Prox1 ablation in bipotent hepatoblasts dramatically reduced the expression of multiple hepatocyte genes and led to very defective hepatocyte morphogenesis. As a result, abnormal epithelial structures expressing hepatocyte and cholangiocyte markers or resembling ectopic bile ducts developed in the Prox1-deficient liver parenchyma. By contrast, excessive commitment of hepatoblasts into cholangiocytes, premature intrahepatic bile duct morphogenesis, and biliary hyperplasia occurred in periportal areas of Prox1-deficient livers. Together, these abnormalities indicate that Prox1 activity is necessary to correctly allocate cell fates in liver precursors. These results increase our understanding of differentiation anomalies in pathological conditions and will contribute to improving stem cell protocols in which differentiation is directed towards hepatocytes and cholangiocytes.


Bile Ducts/pathology , Cell Lineage , Gene Deletion , Hepatocytes/metabolism , Hepatocytes/pathology , Stem Cells/metabolism , Tumor Suppressor Proteins/deficiency , Aging/metabolism , Animals , Animals, Newborn , Cell Count , Cell Lineage/genetics , Choristoma/pathology , Epithelial Cells/metabolism , Epithelial Cells/pathology , Fetus/metabolism , Gene Expression Regulation, Developmental , Hepatocyte Nuclear Factor 4/metabolism , Homeodomain Proteins/metabolism , Liver/embryology , Liver/metabolism , Mice , SOX9 Transcription Factor/metabolism , Signal Transduction/genetics , Stem Cells/pathology , Transforming Growth Factor beta/metabolism , Tumor Suppressor Proteins/metabolism
12.
Gastroenterology ; 143(6): 1564-1575.e7, 2012 Dec.
Article En | MEDLINE | ID: mdl-22922013

BACKGROUND & AIMS: Self-renewal of mature hepatocytes promotes homeostasis and regeneration of adult liver. However, recent studies have indicated that liver progenitor cells (LPC) could give rise to hepatic epithelial cells during normal turnover of the liver and after acute injury. We investigated the capacity of LPC to differentiate into hepatocytes in vivo and contribute to liver regeneration. METHODS: We performed lineage tracing experiments, using mice that express tamoxifen-inducible Cre recombinase under control of osteopontin regulatory region crossed with yelow fluorescent protein reporter mice, to follow the fate of LPC and biliary cells. Adult mice received partial (two-thirds) hepatectomy, acute or chronic administration of carbon tetrachloride (CCl(4)), choline-deficient diet supplemented with ethionine, or 3,5-diethoxycarbonyl-1,4-dihydrocollidine diet. RESULTS: LPC and/or biliary cells generated 0.78% and 2.45% of hepatocytes during and upon recovery of mice from liver injury, respectively. Repopulation efficiency by LPC and/or biliary cells increased when extracellular matrix and laminin deposition were reduced. The newly formed hepatocytes integrated into hepatic cords, formed biliary canaliculi, expressed hepato-specific enzymes, accumulated glycogen, and proliferated in response to partial hepatectomy, as neighboring native hepatocytes. By contrast, LPC did not contribute to hepatocyte regeneration during normal liver homeostasis, in response to surgical or toxic loss of liver mass, during chronic liver injury (CCl(4)-induced), or during ductular reactions. CONCLUSIONS: LPC or biliary cells terminally differentiate into functional hepatocytes in mice with liver injury.


Cell Differentiation/physiology , Chemical and Drug Induced Liver Injury/pathology , Hepatocytes/cytology , Liver Regeneration/physiology , Liver/cytology , Stem Cells/cytology , Animals , Carbon Tetrachloride/adverse effects , Chemical and Drug Induced Liver Injury/etiology , Choline Deficiency/complications , Epithelial-Mesenchymal Transition/physiology , Female , Hepatectomy/adverse effects , Homeostasis/physiology , Liver/physiology , Male , Mice , Mice, Inbred Strains , Models, Animal
13.
Gastroenterology ; 142(1): 119-29, 2012 Jan.
Article En | MEDLINE | ID: mdl-21920465

BACKGROUND & AIMS: Hepatocyte differentiation is controlled by liver-enriched transcription factors (LETFs). We investigated whether LETFs control microRNA expression during development and whether this control is required for hepatocyte differentiation. METHODS: Using in vivo DNA binding assays, we identified miR-122 as a direct target of the LETF hepatocyte nuclear factor (HNF) 6. The role and mechanisms of the HNF6-miR-122 gene cascade in hepatocyte differentiation were studied in vivo and in vitro by gain-of-function and loss-of-function experiments, using developing mice and zebrafish as model organisms. RESULTS: HNF6 and its paralog Onecut2 are strong transcriptional stimulators of miR-122 expression. Specific levels of miR-122 were required for proper progression of hepatocyte differentiation; miR-122 stimulated the expression of hepatocyte-specific genes and most LETFs, including HNF6. This indicates that HNF6 and miR-122 form a positive feedback loop. Stimulation of hepatocyte differentiation by miR-122 was lost in HNF6-null mice, revealing that a transcription factor can mediate microRNA function. All hepatocyte-specific genes whose expression was stimulated by miR-122 bound HNF6 in vivo, confirming their direct regulation by this factor. CONCLUSIONS: Hepatocyte differentiation is directed by a positive feedback loop that includes a transcription factor (HNF6) and a microRNA (miR-122) that are specifically expressed in liver. These findings could lead to methods to induce differentiation of hepatocytes in vitro and improve our understanding of liver cell dedifferentiation in pathologic conditions.


Cell Differentiation , Hepatocytes/metabolism , MicroRNAs/metabolism , Transcription Factors/metabolism , Animals , Base Sequence , Binding Sites , Cells, Cultured , Embryo Culture Techniques , Feedback, Physiological , Gene Expression Regulation, Developmental , Hepatocyte Nuclear Factor 3-beta/genetics , Hepatocyte Nuclear Factor 3-beta/metabolism , Hepatocyte Nuclear Factor 6/genetics , Hepatocyte Nuclear Factor 6/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Mice , Mice, Knockout , Mice, Transgenic , Molecular Sequence Data , Promoter Regions, Genetic , RNA Interference , Signal Transduction , Transcription Factors/deficiency , Transcription Factors/genetics , Transfection , Zebrafish/embryology , Zebrafish/genetics , Zebrafish/metabolism , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
14.
Gastroenterology ; 141(4): 1432-8, 1438.e1-4, 2011 Oct.
Article En | MEDLINE | ID: mdl-21708104

UNLABELLED: BACKGROUND& AIMS: Embryonic biliary precursor cells form a periportal sheet called the ductal plate, which is progressively remodeled to generate intrahepatic bile ducts. A limited number of ductal plate cells participate in duct formation; those not involved in duct development are believed to involute by apoptosis. Moreover, cells that express the SRY-related HMG box transcription factor 9 (SOX9), which include the embryonic ductal plate cells, were proposed to continuously supply the liver with hepatic cells. We investigated the role of the ductal plate in hepatic morphogenesis. METHODS: Apoptosis and proliferation were investigated by immunostaining of mouse and human fetal liver tissue. The postnatal progeny of SOX9-expressing ductal plate cells was analyzed after genetic labeling, at the ductal plate stage, by Cre-mediated recombination of a ROSA26RYFP reporter allele. Inducible Cre expression was induced by SOX9 regulatory regions, inserted in a bacterial artificial chromosome. Livers were studied from mice under normal conditions and during diet-induced regeneration. RESULTS: Ductal plate cells did not undergo apoptosis and showed limited proliferation. They generated cholangiocytes lining interlobular bile ducts, bile ductules, and canals of Hering, as well as periportal hepatocytes. Oval cells that appeared during regeneration also derived from the ductal plate. We did not find that liver homeostasis required a continuous supply of cells from SOX9-expressing progenitors. CONCLUSIONS: The ductal plate gives rise to cholangiocytes lining the intrahepatic bile ducts, including its most proximal segments. It also generates periportal hepatocytes and adult hepatic progenitor cells.


Adult Stem Cells/physiology , Bile Ducts, Intrahepatic/embryology , Cell Differentiation , Cell Lineage , Embryonic Stem Cells/physiology , Hepatocytes/physiology , Liver/embryology , Adult Stem Cells/metabolism , Animals , Apoptosis , Bile Ducts, Intrahepatic/metabolism , Cell Proliferation , Chromosomes, Artificial, Bacterial , Embryonic Stem Cells/metabolism , Fluorescent Antibody Technique , Gestational Age , Hepatocytes/metabolism , Humans , Immunohistochemistry , In Situ Nick-End Labeling , Integrases/genetics , Liver/metabolism , Liver Regeneration , Luminescent Proteins/biosynthesis , Luminescent Proteins/genetics , Mice , Mice, Transgenic , Microscopy, Confocal , Microscopy, Fluorescence , Proteins/genetics , RNA, Untranslated , SOX9 Transcription Factor/biosynthesis , SOX9 Transcription Factor/genetics , SOX9 Transcription Factor/metabolism
15.
Hepatology ; 53(6): 1959-66, 2011 Jun.
Article En | MEDLINE | ID: mdl-21391226

UNLABELLED: Ductal plate malformations (DPMs) are developmental anomalies considered to result from lack of ductal plate remodeling during bile duct morphogenesis. In mice, bile duct development is initiated by the formation of primitive ductal structures lined by two cell types, namely ductal plate cells and hepatoblasts. During ductal plate remodeling, the primitive ductal structures mature to ducts as a result from differentiation of the ductal plate cells and hepatoblasts to cholangiocytes. Here, we report this process is conserved in human fetal liver. These findings prompted us to evaluate how DPMs develop in three mouse models, namely mice with livers deficient in hepatocyte nuclear factor 6 (HNF6), HNF1ß, or cystin-1 (cpk [congenital polycystic kidney] mice). Human liver from a patient with a HNF1B/TCF2 mutation, and from fetuses affected with autosomal recessive polycystic kidney disease (ARPKD) were also analyzed. Despite the epistatic relationship between HNF6, HNF1ß, and cystin-1, the three mouse models displayed distinct morphogenic mechanisms of DPM. They all developed biliary cysts lined by cells with abnormal apicobasal polarity. However, the absence of HNF6 led to an early defect in ductal plate cell differentiation. In HNF1ß-deficient liver, maturation of the primitive ductal structures was impaired. Normal differentiation and maturation but abnormal duct expansion was apparent in cpk mouse livers and in human fetal ARPKD. CONCLUSION: DPM is the common endpoint of distinct defects initiated at distinct stages of bile duct morphogenesis. Our observations provide a new pathogenic classification of DPM.


Bile Ducts, Intrahepatic/abnormalities , Bile Ducts, Intrahepatic/embryology , Congenital Abnormalities/classification , Congenital Abnormalities/etiology , Morphogenesis/physiology , Animals , Bile Ducts, Intrahepatic/pathology , Biomarkers/metabolism , Cell Differentiation/physiology , Congenital Abnormalities/physiopathology , Disease Models, Animal , Hepatocyte Nuclear Factor 1-beta/metabolism , Hepatocyte Nuclear Factor 6/metabolism , Humans , Liver/embryology , Liver/metabolism , Liver/pathology , Membrane Proteins/metabolism , Mice , Mice, Mutant Strains , Polycystic Kidney Diseases/congenital , Polycystic Kidney Diseases/metabolism , Polycystic Kidney Diseases/physiopathology , Polycystic Kidney, Autosomal Recessive/metabolism , Polycystic Kidney, Autosomal Recessive/physiopathology
16.
Dev Biol ; 347(1): 216-27, 2010 Nov 01.
Article En | MEDLINE | ID: mdl-20807526

Endothelial cells are required to initiate pancreas development from the endoderm. They also control the function of endocrine islets after birth. Here we investigate in developing pancreas how the endothelial cells become organized during branching morphogenesis and how their development affects pancreatic cell differentiation. We show that endothelial cells closely surround the epithelial bud at the onset of pancreas morphogenesis. During branching morphogenesis, the endothelial cells become preferentially located near the central (trunk) epithelial cells and remain at a distance from the branch tips where acinar cells differentiate. This correlates with predominant expression of the angiogenic factor vascular endothelial growth factor-A (VEGF-A) in trunk cells. In vivo ablation of VEGF-A expression by pancreas-specific inactivation of floxed Vegfa alleles results in reduced endothelial development and in excessive acinar differentiation. On the contrary, acinar differentiation is repressed when endothelial cells are recruited around tip cells that overexpress VEGF-A. Treatment of embryonic day 12.5 explants with VEGF-A or with VEGF receptor antagonists confirms that acinar development is tightly controlled by endothelial cells. We also provide evidence that endothelial cells repress the expression of Ptf1a, a transcription factor essential for acinar differentiation, and stimulate the expression of Hey-1 and Hey-2, two repressors of Ptf1a activity. In explants, we provide evidence that VEGF-A signaling is required, but not sufficient, to induce endocrine differentiation. In conclusion, our data suggest that, in developing pancreas, epithelial production of VEGF-A determines the spatial organization of endothelial cells which, in turn, limit acinar differentiation of the epithelium.


Cell Differentiation , Endothelium/metabolism , Epithelium/metabolism , Morphogenesis , Pancreas, Exocrine/cytology , Pancreas, Exocrine/embryology , Animals , Cell Count , Endothelial Cells/cytology , Endothelial Cells/metabolism , Endothelium/cytology , Epithelial Cells/cytology , Epithelial Cells/metabolism , Epithelium/blood supply , Mice , Neovascularization, Physiologic , Pancreas, Exocrine/blood supply , Pancreas, Exocrine/metabolism , Signal Transduction , Tissue Culture Techniques , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism
17.
BMC Dev Biol ; 9: 66, 2009 Dec 14.
Article En | MEDLINE | ID: mdl-20003423

BACKGROUND: The exocrine pancreas is composed of a branched network of ducts connected to acini. They are lined by a monolayered epithelium that derives from the endoderm and is surrounded by mesoderm-derived mesenchyme. The morphogenic mechanisms by which the ductal network is established as well as the signaling pathways involved in this process are poorly understood. RESULTS: By morphological analyzis of wild-type and mutant mouse embryos and using cultured embryonic explants we investigated how epithelial morphogenesis takes place and is regulated by chemokine signaling. Pancreas ontogenesis displayed a sequence of two opposite epithelial transitions. During the first transition, the monolayered and polarized endodermal cells give rise to tissue buds composed of a mass of non polarized epithelial cells. During the second transition the buds reorganize into branched and polarized epithelial monolayers that further differentiate into tubulo-acinar glands. We found that the second epithelial transition is controlled by the chemokine Stromal cell-Derived Factor (SDF)-1. The latter is expressed by the mesenchyme, whereas its receptor CXCR4 is expressed by the epithelium. Reorganization of cultured pancreatic buds into monolayered epithelia was blocked in the presence of AMD3100, a SDF-1 antagonist. Analyzis of sdf1 and cxcr4 knockout embryos at the stage of the second epithelial transition revealed transient defective morphogenesis of the ventral and dorsal pancreas. Reorganization of a globular mass of epithelial cells in polarized monolayers is also observed during submandibular glands development. We found that SDF-1 and CXCR4 are expressed in this organ and that AMD3100 treatment of submandibular gland explants blocks its branching morphogenesis. CONCLUSION: In conclusion, our data show that the primitive pancreatic ductal network, which is lined by a monolayered and polarized epithelium, forms by remodeling of a globular mass of non polarized epithelial cells. Our data also suggest that SDF-1 controls the branching morphogenesis of several exocrine tissues.


Chemokine CXCL12/metabolism , Morphogenesis , Pancreas/embryology , Submandibular Gland/embryology , Animals , Benzylamines , Chemokine CXCL12/antagonists & inhibitors , Chemokine CXCL12/genetics , Cyclams , Epithelium/embryology , Heterocyclic Compounds/pharmacology , In Vitro Techniques , Mice , Mice, Knockout , Pancreas/metabolism , Receptors, CXCR4/genetics , Receptors, CXCR4/metabolism , Submandibular Gland/metabolism
18.
Gastroenterology ; 136(7): 2325-33, 2009 Jun.
Article En | MEDLINE | ID: mdl-19403103

BACKGROUND & AIMS: A number of diseases are characterized by defective formation of the intrahepatic bile ducts. In the embryo, hepatoblasts differentiate to cholangiocytes, which give rise to the bile ducts. Here, we investigated duct development in mouse liver and characterized the role of the SRY-related HMG box transcription factor 9 (SOX9). METHODS: We identified SOX9 as a new biliary marker and used it in immunostaining experiments to characterize bile duct morphogenesis. The expression of growth factors was determined by in situ hybridization and immunostaining, and their role was studied on cultured hepatoblasts. SOX9 function was investigated by phenotyping mice with a liver-specific inactivation of Sox9. RESULTS: Biliary tubulogenesis started with formation of asymmetrical ductal structures, lined on the portal side by cholangiocytes and on the parenchymal side by hepatoblasts. When the ducts grew from the hilum to the periphery, the hepatoblasts lining the asymmetrical structures differentiated to cholangiocytes, thereby allowing formation of symmetrical ducts lined only by cholangiocytes. We also provide evidence that transforming growth factor-beta promotes differentiation of the hepatoblasts lining the asymmetrical structures. In the absence of SOX9, the maturation of asymmetrical structures into symmetrical ducts was delayed. This was associated with abnormal expression of CCAAT/Enhancer Binding Protein alpha and Homolog of Hairy/Enhancer of Split-1, as well as of the transforming growth factor-beta receptor type II, which are regulators of biliary development. CONCLUSIONS: Our results suggest that biliary development proceeds according to a new mode of tubulogenesis characterized by transient asymmetry and whose timing is controlled by SOX9.


Bile Ducts, Intrahepatic/embryology , Gene Expression Regulation, Developmental , SOX9 Transcription Factor/genetics , Signal Transduction/genetics , Animals , Bile Ducts, Intrahepatic/growth & development , Cell Differentiation , Epidermal Growth Factor/genetics , Epidermal Growth Factor/metabolism , Female , In Situ Hybridization , Liver/embryology , Liver/growth & development , Mice , Mice, Knockout , Models, Animal , Morphogenesis/genetics , Pregnancy , Probability , Reverse Transcriptase Polymerase Chain Reaction , SOX9 Transcription Factor/metabolism , Sensitivity and Specificity , Signal Transduction/physiology
19.
Gastroenterology ; 130(2): 532-41, 2006 Feb.
Article En | MEDLINE | ID: mdl-16472605

BACKGROUND & AIMS: A number of hereditary polycystic diseases are associated with formation of cysts within the pancreatic ducts. The cysts result from abnormal tubulogenesis, but how normal pancreatic duct development is controlled remains poorly understood. Here, we investigate the transcriptional mechanisms that control pancreatic duct development by addressing the role of the transcription factor hepatocyte nuclear factor (HNF)-6. METHODS: Using immunostaining, we have determined the expression pattern of HNF-6 in pancreatic ducts during mouse development. Hnf6 null mice at various stages of development were studied by immunolocalization methods to assess the morphology, differentiation, and proliferation status of ductal cells. The expression of genes involved in hereditary polycystic diseases was determined by real-time, reverse-transcription polymerase chain reaction (RT-PCR). RESULTS: We show that HNF-6 is expressed in the pancreatic duct epithelium throughout development and that, in the absence of HNF-6, duct morphogenesis is perturbed. Although development of the intercalated ducts is normal, cysts appear within the interlobular and intralobular ducts. This is associated with abnormal development of primary cilia at the apical pole of the duct cells and with reduced expression of a set of genes involved in polycystic diseases, namely those coding for HNF-1beta and for the cilium-associated proteins polyductin/fibrocystin and cystin. CONCLUSIONS: We identify HNF-6 as the first transcriptional regulator of pancreatic duct development and reveal the existence of different regulatory mechanisms in distinct duct compartments. HNF-6 controls a network of genes involved in cilium formation and in hereditary polycystic diseases. Finally, HNF-6 deficiency represents a genetically defined model of pancreatic cystic disease.


Gene Expression Regulation, Developmental , Hepatocyte Nuclear Factor 6/genetics , Pancreatic Ducts/growth & development , Animals , Base Sequence , DNA Primers , Embryonic Development , Hepatocyte Nuclear Factor 6/deficiency , Mice , Mice, Knockout , Morphogenesis , Pancreatic Diseases/genetics , Pancreatic Ducts/embryology , Reverse Transcriptase Polymerase Chain Reaction
...