Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
1.
Semin Liver Dis ; 43(1): 13-23, 2023 02.
Article in English | MEDLINE | ID: mdl-36764306

ABSTRACT

The liver field has been debating for decades the contribution of the plasticity of the two epithelial compartments in the liver, hepatocytes and biliary epithelial cells (BECs), to derive each other as a repair mechanism. The hepatobiliary plasticity has been first observed in diseased human livers by the presence of biphenotypic cells expressing hepatocyte and BEC markers within bile ducts and regenerative nodules or budding from strings of proliferative BECs in septa. These observations are not surprising as hepatocytes and BECs derive from a common fetal progenitor, the hepatoblast, and, as such, they are expected to compensate for each other's loss in adults. To investigate the cell origin of regenerated cell compartments and associated molecular mechanisms, numerous murine and zebrafish models with ability to trace cell fates have been extensively developed. This short review summarizes the clinical and preclinical studies illustrating the hepatobiliary plasticity and its potential therapeutic application.


Subject(s)
Liver , Zebrafish , Animals , Mice , Humans , Hepatocytes , Epithelial Cells
3.
Development ; 142(12): 2094-108, 2015 Jun 15.
Article in English | MEDLINE | ID: mdl-26081571

ABSTRACT

The liver is a central regulator of metabolism, and liver failure thus constitutes a major health burden. Understanding how this complex organ develops during embryogenesis will yield insights into how liver regeneration can be promoted and how functional liver replacement tissue can be engineered. Recent studies of animal models have identified key signaling pathways and complex tissue interactions that progressively generate liver progenitor cells, differentiated lineages and functional tissues. In addition, progress in understanding how these cells interact, and how transcriptional and signaling programs precisely coordinate liver development, has begun to elucidate the molecular mechanisms underlying this complexity. Here, we review the lineage relationships, signaling pathways and transcriptional programs that orchestrate hepatogenesis.


Subject(s)
Hepatocytes/cytology , Liver/embryology , Organogenesis/physiology , Stem Cells/cytology , Animals , Cell Differentiation , Cell Lineage , Humans , Liver/growth & development , Liver/metabolism , Liver Diseases/pathology , Mice , Signal Transduction
4.
Gastroenterology ; 145(5): 966-969.e7, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23891978

ABSTRACT

The narrow species tropism of hepatitis C virus (HCV) limits animal studies. We found that pigtail macaque (Macaca nemestrina) hepatic cells derived from induced pluripotent stem cells support the entire HCV life cycle, although infection efficiency was limited by defects in the HCV cell entry process. This block was overcome by either increasing occludin expression, complementing the cells with human CD81, or infecting them with a strain of HCV with less restricted requirements for CD81. Using this system, we can modify viral and host cell genetics to make pigtail macaques a suitable, clinically relevant model for the study of HCV infection.


Subject(s)
Disease Models, Animal , Hepacivirus/pathogenicity , Hepatitis C/virology , Hepatocytes/virology , Induced Pluripotent Stem Cells/virology , Macaca nemestrina , Animals , Cell Line , Cells, Cultured , Hepatitis C/pathology , Hepatitis C/physiopathology , Hepatocytes/pathology , Host-Pathogen Interactions/genetics , Humans , Induced Pluripotent Stem Cells/pathology , Occludin/physiology , Tetraspanin 28/deficiency , Tetraspanin 28/physiology , Virus Internalization , Virus Replication/physiology
5.
bioRxiv ; 2024 Jan 11.
Article in English | MEDLINE | ID: mdl-38260488

ABSTRACT

Primary human hepatocyte (PHH) transplantation is a promising alternative to liver transplantation, whereby liver function could be restored by partial repopulation of the diseased organ with healthy cells. However, currently PHH engraftment efficiency is low and benefits are not maintained long-term. Here we refine two mouse models of human chronic and acute liver diseases to recapitulate compromised hepatocyte proliferation observed in nearly all human liver diseases by overexpression of p21 in hepatocytes. In these clinically relevant contexts, we demonstrate that transient, yet robust expression of human hepatocyte growth factor and epidermal growth factor in the liver via nucleoside-modified mRNA in lipid nanoparticles, whose safety was validated with mRNA-based COVID-19 vaccines, drastically improves PHH engraftment, reduces disease burden, and improves overall liver function. This novel strategy may overcome the critical barriers to clinical translation of cell therapies with primary or stem cell-derived hepatocytes for the treatment of liver diseases.

6.
Nat Commun ; 15(1): 5010, 2024 Jun 12.
Article in English | MEDLINE | ID: mdl-38866762

ABSTRACT

Primary human hepatocyte (PHH) transplantation is a promising alternative to liver transplantation, whereby liver function could be restored by partial repopulation of the diseased organ with healthy cells. However, currently PHH engraftment efficiency is low and benefits are not maintained long-term. Here we refine two male mouse models of human chronic and acute liver diseases to recapitulate compromised hepatocyte proliferation observed in nearly all human liver diseases by overexpression of p21 in hepatocytes. In these clinically relevant contexts, we demonstrate that transient, yet robust expression of human hepatocyte growth factor and epidermal growth factor in the liver via nucleoside-modified mRNA in lipid nanoparticles, whose safety was validated with mRNA-based COVID-19 vaccines, drastically improves PHH engraftment, reduces disease burden, and improves overall liver function. This strategy may overcome the critical barriers to clinical translation of cell therapies with primary or stem cell-derived hepatocytes for the treatment of liver diseases.


Subject(s)
Hepatocyte Growth Factor , Hepatocytes , Nanoparticles , RNA, Messenger , Animals , Hepatocytes/metabolism , Hepatocytes/transplantation , Humans , Mice , Male , RNA, Messenger/metabolism , RNA, Messenger/genetics , Nanoparticles/chemistry , Hepatocyte Growth Factor/metabolism , Hepatocyte Growth Factor/genetics , Disease Models, Animal , Liver/metabolism , Epidermal Growth Factor/metabolism , Cell- and Tissue-Based Therapy/methods , COVID-19/therapy , Liver Diseases/therapy , Liver Diseases/metabolism , Liver Diseases/genetics , Cell Proliferation , SARS-CoV-2/genetics , Liposomes
7.
bioRxiv ; 2023 Apr 18.
Article in English | MEDLINE | ID: mdl-37131727

ABSTRACT

Background and Aims: Acetaminophen (APAP) overdose is the leading cause of acute liver failure, with one available treatment, N-acetyl cysteine (NAC). Yet, NAC effectiveness diminishes about ten hours after APAP overdose, urging for therapeutic alternatives. This study addresses this need by deciphering a mechanism of sexual dimorphism in APAP-induced liver injury, and leveraging it to accelerate liver recovery via growth hormone (GH) treatment. GH secretory patterns, pulsatile in males and near-continuous in females, determine the sex bias in many liver metabolic functions. Here, we aim to establish GH as a novel therapy to treat APAP hepatotoxicity. Approach and Results: Our results demonstrate sex-dependent APAP toxicity, with females showing reduced liver cell death and faster recovery than males. Single-cell RNA sequencing analyses reveal that female hepatocytes have significantly greater levels of GH receptor expression and GH pathway activation compared to males. In harnessing this female-specific advantage, we demonstrate that a single injection of recombinant human GH protein accelerates liver recovery, promotes survival in males following sub-lethal dose of APAP, and is superior to standard-of-care NAC. Alternatively, slow-release delivery of human GH via the safe nonintegrative lipid nanoparticle-encapsulated nucleoside-modified mRNA (mRNA-LNP), a technology validated by widely used COVID-19 vaccines, rescues males from APAP-induced death that otherwise occurred in control mRNA-LNP-treated mice. Conclusions: Our study demonstrates a sexually dimorphic liver repair advantage in females following APAP overdose, leveraged by establishing GH as an alternative treatment, delivered either as recombinant protein or mRNA-LNP, to potentially prevent liver failure and liver transplant in APAP-overdosed patients.

8.
Cell Stem Cell ; 30(12): 1640-1657.e8, 2023 12 07.
Article in English | MEDLINE | ID: mdl-38029740

ABSTRACT

The liver is known for its remarkable regenerative ability through proliferation of hepatocytes. Yet, during chronic injury or severe hepatocyte death, proliferation of hepatocytes is exhausted. To overcome this hurdle, we propose vascular-endothelial-growth-factor A (VEGFA) as a therapeutic means to accelerate biliary epithelial-cell (BEC)-to-hepatocyte conversion. Investigation in zebrafish establishes that blocking VEGF receptors abrogates BEC-driven liver repair, while VEGFA overexpression promotes it. Delivery of VEGFA via nonintegrative and safe nucleoside-modified mRNA encapsulated into lipid nanoparticles (mRNA-LNPs) in acutely or chronically injured mouse livers induces robust BEC-to-hepatocyte conversion and elimination of steatosis and fibrosis. In human and murine diseased livers, we further identified VEGFA-receptor KDR-expressing BECs associated with KDR-expressing cell-derived hepatocytes. This work defines KDR-expressing cells, most likely being BECs, as facultative progenitors. This study reveals unexpected therapeutic benefits of VEGFA delivered via nucleoside-modified mRNA-LNP, whose safety is widely validated with COVID-19 vaccines, for harnessing BEC-driven repair to potentially treat liver diseases.


Subject(s)
Liver Diseases , Zebrafish , Animals , Mice , Humans , RNA, Messenger/genetics , COVID-19 Vaccines , Nucleosides , Hepatocytes , Liver , Epithelial Cells , Liver Diseases/pathology , Fibrosis , Liver Regeneration , Vascular Endothelial Growth Factor A/genetics
9.
bioRxiv ; 2023 Apr 18.
Article in English | MEDLINE | ID: mdl-37131823

ABSTRACT

The liver is known for its remarkable regenerative ability through proliferation of hepatocytes. Yet, during chronic injury or severe hepatocyte death, proliferation of hepatocytes is exhausted. To overcome this hurdle, we propose vascular-endothelial-growth-factor A (VEGFA) as a therapeutic means to accelerate biliary epithelial cell (BEC)-to-hepatocyte conversion. Investigation in zebrafish establishes that blocking VEGF receptors abrogates BEC-driven liver repair, while VEGFA overexpression promotes it. Delivery of VEGFA via non-integrative and safe nucleoside-modified mRNA encapsulated into lipid-nanoparticles (mRNA-LNP) in acutely or chronically injured mouse livers induces robust BEC-to-hepatocyte conversion and reversion of steatosis and fibrosis. In human and murine diseased livers, we further identified VEGFA-receptor KDR-expressing BECs associated with KDR-expressing cell-derived hepatocytes. This defines KDR-expressing cells, most likely being BECs, as facultative progenitors. This study reveals novel therapeutic benefits of VEGFA delivered via nucleoside-modified mRNA-LNP, whose safety is widely validated with COVID-19 vaccines, for harnessing BEC-driven repair to potentially treat liver diseases. Highlights: Complementary mouse and zebrafish models of liver injury demonstrate the therapeutic impact of VEGFA-KDR axis activation to harness BEC-driven liver regeneration.VEGFA mRNA LNPs restore two key features of the chronic liver disease in humans such as steatosis and fibrosis.Identification in human cirrhotic ESLD livers of KDR-expressing BECs adjacent to clusters of KDR+ hepatocytes suggesting their BEC origin.KDR-expressing BECs may represent facultative adult progenitor cells, a unique BEC population that has yet been uncovered.

10.
Stem Cells ; 29(2): 217-28, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21732480

ABSTRACT

Complex cross-talk between endoderm and the microenvironment is an absolute requirement to orchestrate hepatic specification and expansion. In the mouse, the septum transversum and cardiac mesoderm, through secreted bone morphogenetic proteins (BMP) and fibroblast growth factors (FGF), respectively, instruct the adjacent ventral endoderm to become hepatic endoderm. Consecutively, endothelial cells promote expansion of the specified hepatic endoderm. By using a mouse reporter embryonic stem cell line, in which hCD4 and hCD25 were targeted to the Foxa2 and Foxa3 loci, we reconstituted an in vitro culture system in which committed endoderm cells coexpressing hCD4-Foxa2 and hCD25-Foxa3 were isolated and cocultured with endothelial cells in the presence of BMP4 and bFGF. In this culture setting, we provide mechanistic evidence that endothelial cells function not only to promote hepatic endoderm expansion but are also required at an earlier step for hepatic specification, at least in part through regulation of the Wnt and Notch pathways. Activation of Wnt and Notch by chemical or genetic approaches increases endoderm cell numbers but inhibits hepatic specification, and conversely, chemical inhibition of both pathways enhances hepatic specification and reduces proliferation. By using identical coculture conditions, we defined a similar dependence of endoderm harvested from embryos on endothelial cells to support their growth and hepatic specification. Our findings (1) confirm a conserved role of Wnt repression for mouse hepatic specification, (2) uncover a novel role for Notch repression in the hepatic fate decision, and (3) demonstrate that repression of Wnt and Notch signaling in hepatic endoderm is controlled by the endothelial cell niche.


Subject(s)
Embryonic Stem Cells/metabolism , Endothelial Cells/metabolism , Hepatocytes/metabolism , Wnt Signaling Pathway , Animals , Bone Morphogenetic Protein 4/metabolism , Bone Morphogenetic Protein 4/pharmacology , CD4 Antigens/biosynthesis , CD4 Antigens/genetics , Cell Differentiation/drug effects , Cell Line , Embryonic Stem Cells/cytology , Endoderm/cytology , Endoderm/metabolism , Fibroblast Growth Factors/pharmacology , Hepatocyte Nuclear Factor 3-beta/biosynthesis , Hepatocyte Nuclear Factor 3-beta/genetics , Hepatocyte Nuclear Factor 3-gamma/biosynthesis , Hepatocyte Nuclear Factor 3-gamma/genetics , Hepatocytes/cytology , Interleukin-2 Receptor alpha Subunit/biosynthesis , Interleukin-2 Receptor alpha Subunit/genetics , Mice , Receptors, Notch/metabolism , Wnt Proteins/metabolism
11.
Cell Stem Cell ; 29(4): 491-493, 2022 04 07.
Article in English | MEDLINE | ID: mdl-35395181

ABSTRACT

In this issue of Cell Stem Cell, Gómez-Salinero et al. (2022) identify c-Maf as a driver for murine liver sinusoidal endothelial cell (LSEC) fate and function during liver development, homeostasis, and repair. Similarly, c-Maf defines human LSECs, and its overexpression specializes generic HUVECs into functional induced-LSECs, potentiating regenerative therapeutics.


Subject(s)
Endothelial Cells , Liver , Proto-Oncogene Proteins c-maf , Animals , Endothelial Cells/cytology , Humans , Liver/cytology , Mice
12.
Dev Biol ; 344(1): 79-93, 2010 Aug 01.
Article in English | MEDLINE | ID: mdl-20430021

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.


Subject(s)
Embryonic Stem Cells/cytology , Gene Expression Regulation, Developmental , Hepatocytes/metabolism , Kruppel-Like Transcription Factors/physiology , Liver/metabolism , Proto-Oncogene Proteins/physiology , Animals , Cell Differentiation , Cell Lineage , Cell Proliferation , Endoderm/metabolism , In Situ Hybridization , Kruppel-Like Factor 6 , Mice , Mice, Transgenic , Models, Biological , Zebrafish
13.
Nat Commun ; 12(1): 613, 2021 01 27.
Article in English | MEDLINE | ID: mdl-33504774

ABSTRACT

Induction of intrinsic liver regeneration is an unmet need that can be achieved by temporally activating key hepatocyte regenerative pathways. Here, we establish an efficient, safe, non-integrative method to transiently express hepatocyte-growth-factor (HGF) and epidermal-growth-factor (EGF) in hepatocytes via nucleoside-modified, lipid-nanoparticle-encapsulated mRNA (mRNA-LNP) delivery in mice. We confirm specific hepatotropism of mRNA-LNP via intravenous injection of firefly luciferase encoding mRNA-LNP, with protein expression lasting about 3 days. In the liver, virtually all hepatocytes are transfected along with a subpopulation of endothelial and Kupffer cells. In homeostasis, HGF mRNA-LNP efficiently induce hepatocyte proliferation. In a chronic liver injury mouse model recapitulating non-alcoholic fatty liver disease, injections of both HGF and EGF mRNA-LNP sharply reverse steatosis and accelerate restoration of liver function. Likewise, HGF and EGF mRNA-LNP accelerate liver regeneration after acetaminophen-induced acute liver injury with rapid return to baseline ALT levels. This study introduces mRNA-LNP as a potentially translatable safe therapeutic intervention to harness liver regeneration via controlled expression of endogenous mitogens in vivo.


Subject(s)
Hepatocytes/pathology , Lipids/chemistry , Liver Regeneration/physiology , Liver/pathology , Nanoparticles/chemistry , Nucleosides/metabolism , RNA, Messenger/metabolism , Acetaminophen , Animals , Cell Proliferation/drug effects , Chronic Disease , Disease Models, Animal , Epidermal Growth Factor/pharmacology , Female , Green Fluorescent Proteins/metabolism , Hepatocyte Growth Factor/pharmacology , Hepatocytes/drug effects , Homeostasis/drug effects , Injections , Liver/drug effects , Liver/injuries , Liver/physiopathology , Liver Function Tests , Liver Regeneration/drug effects , Mice, Inbred C57BL
14.
Bio Protoc ; 11(19): e4184, 2021 Oct 05.
Article in English | MEDLINE | ID: mdl-34722830

ABSTRACT

With the recent availability of the SARS-CoV-2 mRNA-based vaccines, public attention has been drawn to this new technology and how it may be applied to other indications. Temporal activation of key hepatic regenerative pathways can induce liver regeneration, overcoming the lack of donor organs for liver transplantation and ineffectiveness of alternative treatments. Recombinant protein therapies and genetic therapies that target these pathways require frequent and repeated injections or, when integrated into the genome, may lead to deleterious effects. In contrast, nucleoside-modified mRNA encapsulated in lipid nanoparticles (mRNA-LNP) are non-integrative and induce transient yet robust expression of proteins that could serve as an ideal therapeutic tool to treat specific liver diseases. For instance, our recent publication in Nature Communications used mRNA-LNP to express hepatic mitogens, hepatocyte growth factor, and epidermal growth factor to induce liver regeneration following both acute and chronic liver injuries. Initial testing with firefly luciferase mRNA-LNP transfection and in vivo imaging confirmed specific hepatotropic delivery. In this protocol, we describe in detail the necessary steps to deliver mRNA-LNP to the murine liver and, following intravenous injection of eGFP mRNA-LNP, verify transfection efficiency using flow cytometry and liver cell specificity using immunofluorescence analyses. This procedure presents an unprecedented tool that can be customized with mRNA-LNP encoding any protein of interest to be expressed by virtually all hepatocytes, ~70% endothelial cells, and ~40% Kupffer cells for promoting liver function and/or regeneration. Graphic abstract: Experimental Design of mRNA-LNP IV Injection and Analysis of Liver Cell Specificity and Efficiency of Transfection (Created with BioRender.com).

15.
Stem Cells ; 27(9): 2103-13, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19522011

ABSTRACT

The development of functional cell populations such as hepatocytes and pancreatic beta cells from embryonic stem cell (ESC) is dependent on the efficient induction of definitive endoderm early in the differentiation process. To monitor definitive endoderm formation in mouse ESC differentiation cultures in a quantitative fashion, we generated a reporter cell line that expresses human CD25 from the Foxa3 locus and human CD4 from the Foxa2 locus. Induction of these reporter ESCs with high concentrations of activin A led to the development of a CD25-Foxa3+CD4-Foxa2+ population within 4-5 days of culture. Isolation and characterization of this population showed that it consists predominantly of definitive endoderm that is able to undergo hepatic specification under the appropriate conditions. To develop reagents that can be used for studies on endoderm development from unmanipulated ESCs, from induced pluripotent stem cells, and from the mouse embryo, we generated monoclonal antibodies against the CD25-Foxa3+CD4-Foxa2+ population. With this approach, we identified two antibodies that react specifically with endoderm from ESC cultures and from the early embryo. The specificity of these antibodies enables one to quantitatively monitor endoderm development in ESC differentiation cultures, to study endoderm formation in the embryo, and to isolate pure populations of culture- or embryo-derived endodermal cells.


Subject(s)
Antibodies, Monoclonal/immunology , CD4 Antigens/metabolism , Endoderm/immunology , Gene Expression Regulation, Developmental , Hepatocyte Nuclear Factor 3-beta/metabolism , Hepatocyte Nuclear Factor 3-gamma/metabolism , Interleukin-2 Receptor alpha Subunit/metabolism , Activins/pharmacology , Animals , Cell Differentiation/drug effects , Cell Differentiation/genetics , Embryonic Stem Cells/cytology , Embryonic Stem Cells/immunology , Embryonic Stem Cells/metabolism , Endoderm/cytology , Flow Cytometry , Humans , Mice
16.
Nat Biotechnol ; 24(11): 1402-11, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17086172

ABSTRACT

When differentiated in the presence of activin A in serum-free conditions, mouse embryonic stem cells efficiently generate an endoderm progenitor population defined by the coexpression of either Brachyury, Foxa2 and c-Kit, or c-Kit and Cxcr4. Specification of these progenitors with bone morphogenetic protein-4 in combination with basic fibroblast growth factor and activin A results in the development of hepatic populations highly enriched (45-70%) for cells that express the alpha-fetoprotein and albumin proteins. These cells also express transcripts of Afp, Alb1, Tat, Cps1, Cyp7a1 and Cyp3a11; they secrete albumin, store glycogen, show ultrastructural characteristics of mature hepatocytes, and are able to integrate into and proliferate in injured livers in vivo and mature into hepatocytes expressing dipeptidyl peptidase IV or fumarylacetoacetate hydrolase. Together, these findings establish a developmental pathway in embryonic stem cell differentiation cultures that leads to efficient generation of cells with an immature hepatocytic phenotype.


Subject(s)
Bone Morphogenetic Proteins/physiology , Cell Differentiation/physiology , Cell Transplantation/physiology , Embryonic Stem Cells/physiology , Hepatocytes/physiology , Activins/physiology , Albumins/metabolism , Animals , Biomarkers , Bone Morphogenetic Protein 4 , CD4 Antigens/genetics , Cell Line , Cell Transplantation/methods , Dipeptidyl Peptidase 4/metabolism , Endoderm/cytology , Endoderm/metabolism , Glycogen/metabolism , Green Fluorescent Proteins/analysis , Hepatocyte Nuclear Factor 3-beta/metabolism , Hepatocytes/cytology , Hydrolases/metabolism , Mice , Phenotype , Signal Transduction
17.
Stem Cell Res ; 30: 163-170, 2018 07.
Article in English | MEDLINE | ID: mdl-29936335

ABSTRACT

Liver organogenesis requires complex cell-cell interactions between hepatic endoderm cells and adjacent cell niches. Endothelial cells are key players for endoderm hepatic fate decision. We previously demonstrated that the endothelial cell niche promotes hepatic specification of mouse embryonic stem cell(ESC)-derived endoderm through dual repression of Wnt and Notch pathways in endoderm cells. In the present study, we dissected further the mechanisms by which endothelial cells trigger endoderm hepatic specification. Using our previously established in vitro mouse ESC system mimicking the early hepatic specification process, endoderm cells were purified and co-cultured with endothelial cells to induce hepatic specification. The comparison of transcriptome profiles between hepatic endoderm cells isolated from co-cultures and endoderm cells cultured alone revealed that VEGF signaling instructs hepatic specification of endoderm cells through endothelial VEGFR2 activation. Additionally, epigenetic mark inhibition assays upon co-cultures uncovered that histone acetylation and DNA methylation promote hepatic specification while histone methylation inhibits it. This study provides an efficient 2D platform modelling the endothelial cell niche crosstalk with endoderm, and reveals mechanisms by which endothelial cells promote hepatic specification of mouse ESC-derived endoderm cells through endothelial VEGFR2 activation and endoderm epigenetic modifications.


Subject(s)
Embryonic Stem Cells/metabolism , Endoderm/metabolism , Endothelial Cells/metabolism , Epigenesis, Genetic/genetics , Liver/physiopathology , Vascular Endothelial Growth Factor Receptor-2/metabolism , Animals , Cell Differentiation , Mice , Signal Transduction
18.
Nat Commun ; 8: 14428, 2017 02 14.
Article in English | MEDLINE | ID: mdl-28195173

ABSTRACT

The recent identification of progenitor populations that contribute to the developing heart in a distinct spatial and temporal manner has fundamentally improved our understanding of cardiac development. However, the mechanisms that direct atrial versus ventricular specification remain largely unknown. Here we report the identification of a progenitor population that gives rise primarily to cardiovascular cells of the ventricles and only to few atrial cells (<5%) of the differentiated heart. These progenitors are specified during gastrulation, when they transiently express Foxa2, a gene not previously implicated in cardiac development. Importantly, Foxa2+ cells contribute to previously identified progenitor populations in a defined pattern and ratio. Lastly, we describe an analogous Foxa2+ population during differentiation of embryonic stem cells. Together, these findings provide insight into the developmental origin of ventricular and atrial cells, and may lead to the establishment of new strategies for generating chamber-specific cell types from pluripotent stem cells.


Subject(s)
Cell Differentiation/physiology , Heart Ventricles/cytology , Heart Ventricles/growth & development , Hepatocyte Nuclear Factor 3-beta/metabolism , Animals , Cell Line , Embryonic Development/physiology , Female , Gastrulation/physiology , Gene Expression Regulation, Developmental , Heart Atria/cytology , Heart Atria/diagnostic imaging , Heart Atria/growth & development , Heart Atria/metabolism , Heart Ventricles/diagnostic imaging , Hepatocyte Nuclear Factor 3-beta/genetics , Mesoderm/cytology , Mesoderm/growth & development , Mesoderm/metabolism , Mice , Mice, Inbred C57BL , Mouse Embryonic Stem Cells/cytology , Mouse Embryonic Stem Cells/metabolism , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism
19.
Cell Stem Cell ; 18(6): 703-706, 2016 06 02.
Article in English | MEDLINE | ID: mdl-27257759

ABSTRACT

The severe shortage of organ donors for treating patients with liver disease has prompted in vitro efforts to produce the main functional cells of the liver: hepatocyte-like cells (Hep cells). We consider the key challenges posed by various stem cell technologies and liver pathologies for developing clinically useful Hep cells.


Subject(s)
Cell- and Tissue-Based Therapy , Liver/cytology , Pluripotent Stem Cells/cytology , Hepatocytes/cytology , Humans , Liver Diseases/therapy
20.
Stem Cell Reports ; 7(2): 158-66, 2016 08 09.
Article in English | MEDLINE | ID: mdl-27509132

ABSTRACT

In the early fetal liver, hematopoietic progenitors expand and mature together with hepatoblasts, the liver progenitors of hepatocytes and cholangiocytes. Previous analyses of human fetal livers indicated that both progenitors support each other's lineage maturation and curiously share some cell surface markers including CD34 and CD133. Using the human embryonic stem cell (hESC) system, we demonstrate that virtually all hESC-derived hepatoblast-like cells (Hep cells) transition through a progenitor stage expressing CD34 and CD133 as well as GATA2, an additional hematopoietic marker that has not previously been associated with human hepatoblast development. Dynamic expression patterns for CD34, CD133, and GATA2 in hepatoblasts were validated in human fetal livers collected from the first and second trimesters of gestation. Knockdown experiments demonstrate that each gene also functions to regulate hepatic fate mostly in a cell-autonomous fashion, revealing unprecedented roles of fetal hematopoietic progenitor markers in human liver progenitors.


Subject(s)
Biomarkers/metabolism , Hematopoietic Stem Cells/cytology , Liver/cytology , AC133 Antigen/metabolism , Animals , Antigens, CD34/metabolism , Cell Differentiation , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , GATA2 Transcription Factor/metabolism , Hematopoietic Stem Cells/metabolism , Hepatocytes/cytology , Hepatocytes/metabolism , Liver/embryology , Mice , Reproducibility of Results
SELECTION OF CITATIONS
SEARCH DETAIL