Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 63
Filter
1.
Proc Natl Acad Sci U S A ; 120(42): e2307972120, 2023 10 17.
Article in English | MEDLINE | ID: mdl-37812721

ABSTRACT

Although generating new neurons in the ischemic injured brain would be an ideal approach to replenish the lost neurons for repairing the damage, the adult mammalian brain retains only limited neurogenic capability. Here, we show that direct conversion of microglia/macrophages into neurons in the brain has great potential as a therapeutic strategy for ischemic brain injury. After transient middle cerebral artery occlusion in adult mice, microglia/macrophages converge at the lesion core of the striatum, where neuronal loss is prominent. Targeted expression of a neurogenic transcription factor, NeuroD1, in microglia/macrophages in the injured striatum enables their conversion into induced neuronal cells that functionally integrate into the existing neuronal circuits. Furthermore, NeuroD1-mediated induced neuronal cell generation significantly improves neurological function in the mouse stroke model, and ablation of these cells abolishes the gained functional recovery. Our findings thus demonstrate that neuronal conversion contributes directly to functional recovery after stroke.


Subject(s)
Brain Ischemia , Stroke , Mice , Animals , Microglia/metabolism , Stroke/metabolism , Macrophages/metabolism , Brain/metabolism , Neurons/metabolism , Brain Ischemia/metabolism , Infarction, Middle Cerebral Artery/metabolism , Mammals
2.
Prog Mol Biol Transl Sci ; 199: 155-178, 2023.
Article in English | MEDLINE | ID: mdl-37678970

ABSTRACT

As a metabolic organ, the liver plays a variety of roles, including detoxification. It has been difficult to obtain stable supplies of hepatocytes for transplantation and for accurate hepatotoxicity determination in drug discovery research. Human pluripotent stem cells, capable of unlimited self-renewal, may be a promising source of hepatocytes. In order to develop a stable supply of embryonic stem cell (ESC)-derived hepatocytes, we have purified human ESC-derived hepatic progenitor cells with exposure to cytocidal puromycin by using their ability to metabolize drugs. Hepatic progenitor cells stably proliferated at least 220-fold over 120 days, maintaining hepatic progenitor cell-like properties. High drug-metabolizing hepatic progenitor cells can be matured into liver cells by suppressing hepatic proliferative signals. The method we developed enables the isolation and proliferation of functional hepatic progenitors from human ESCs, thereby providing a stable supply of high-quality cell resources at high efficiency. Cells produced by this method may facilitate cell therapy for hepatic diseases and reliable drug discovery research.


Subject(s)
Eye Diseases, Hereditary , Pluripotent Stem Cells , Humans , Embryonic Stem Cells , Liver , Drug Discovery
3.
Cells ; 11(23)2022 Nov 24.
Article in English | MEDLINE | ID: mdl-36497010

ABSTRACT

The liver is a crucial organ for maintaining homeostasis in living organisms and is the center of various metabolic functions. Therefore, abnormal metabolic activity, as in metabolic syndrome, leads to pathological conditions, such as abnormal accumulation of lipids in the liver. Inflammation and cell death are induced by several stresses in the fatty liver, namely steatohepatitis. In recent years, an increase in non-alcoholic steatohepatitis (NASH), which is not dependent on excessive alcohol intake, has become an issue as a major cause of liver cirrhosis and liver cancer. There are several recent findings on functional sex-based differences, NASH, and cell stress and death in the liver. In particular, NASH-induced liver injury and tumorigeneses were suppressed by B cell lymphoma 6, the transcriptional factor regulating sex-based liver functional gene expression. In this review, we discuss cell response to stress and lipotoxicity in NASH and its regulatory mechanisms.


Subject(s)
Lymphoma, B-Cell , Non-alcoholic Fatty Liver Disease , Humans , Cell Survival , Non-alcoholic Fatty Liver Disease/metabolism , Liver Cirrhosis
4.
Stem Cell Res Ther ; 13(1): 104, 2022 03 12.
Article in English | MEDLINE | ID: mdl-35279203

ABSTRACT

BACKGROUND: The liver plays an important role in various metabolic processes, including protein synthesis, lipid and drug metabolisms and detoxifications. Primary culture of hepatocytes is used for the understanding of liver physiology as well as for the drug development. Hepatocytes are, however, hardly expandable in vitro making it difficult to secure large numbers of cells from one donor. Alternatively, systems using animal models and hepatocellular carcinoma cells have been established, but interspecies differences, variation between human cell sources and limited hepatic functions are among the challenges faced when using these models. Therefore, there is still a need for a highly stable method to purify human hepatocytes with functional sufficiency. In this study, we aimed to establish an in vitro long-term culture system that enables stable proliferation and maintenance of human hepatocytes to ensure a constant supply. METHODS: We first established a growth culture system for hepatocytes derived from patients with drug-induced liver injury using fetal mouse fibroblasts and EMUKK-05 medium. We then evaluated the morphology, proliferative capacity, chromosome stability, gene and protein expression profiles, and drug metabolic capacity of hepatocytes in early, middle and late passages with and without puromycin. In addition, hepatic maturation in 3D culture was evaluated from morphological and functional aspects. RESULTS: In our culture system, the stable proliferation of human hepatocytes was achieved by co-culturing with mouse fetal fibroblasts, resulting in dedifferentiation into hepatic progenitor-like cells. We purified human hepatocytes by selection with cytocidal puromycin and cultured them for more than 60 population doublings over a span of more than 350 days. Hepatocytes with high expression of cytochrome P450 genes survived after exposure to cytocidal antibiotics because of enhanced drug-metabolizing activity. CONCLUSIONS: These results show that this simple culture system with usage of the cytocidal antibiotics enables efficient hepatocyte proliferation and is an effective method for generating a stable supply of hepatocytes for drug discovery research at a significant cost reduction.


Subject(s)
Chemical and Drug Induced Liver Injury , Hepatocytes , Animals , Cell Proliferation , Cells, Cultured , Chemical and Drug Induced Liver Injury/metabolism , Cytochrome P-450 Enzyme System/genetics , Cytochrome P-450 Enzyme System/metabolism , Hepatocytes/metabolism , Humans , Liver/metabolism , Mice
5.
Sci Rep ; 11(1): 23104, 2021 11 29.
Article in English | MEDLINE | ID: mdl-34845278

ABSTRACT

To fully understand the mechanisms governing learning and memory, animal models with minor interindividual variability and higher cognitive function are required. THA rats established by crossing those with high learning capacity exhibit excellent learning and memory abilities, but the factors underlying their phenotype are completely unknown. In the current study, we compare the hippocampi of parental strain Wistar rats to those of THA rats via metabolomic analysis in order to identify molecules specific to the THA rat hippocampus. Higher branched-chain amino acid (BCAA) levels and enhanced activation of BCAA metabolism-associated enzymes were observed in THA rats, suggesting that acetyl-CoA and acetylcholine are synthesized through BCAA catabolism. THA rats maintained high blood BCAA levels via uptake of BCAAs in the small intestine and suppression of BCAA catabolism in the liver. Feeding THA rats with a BCAA-reduced diet decreased acetylcholine levels and learning ability, thus, maintaining high BCAA levels while their proper metabolism in the hippocampus is the mechanisms underlying the high learning ability in THA rats. Identifying appropriate BCAA nutritional supplements and activation methods may thus hold potential for the prevention and amelioration of higher brain dysfunction, including learning disabilities and dementia.


Subject(s)
Amino Acids, Branched-Chain/chemistry , Animal Feed , Hippocampus/metabolism , Learning , Animals , Behavior , Behavior, Animal , Diet , Hippocampus/pathology , Liver/metabolism , Male , Memory , Metabolome , Models, Animal , Phenotype , Psychophysics , Rats , Rats, Wistar
7.
Sci Rep ; 11(1): 18551, 2021 09 17.
Article in English | MEDLINE | ID: mdl-34535735

ABSTRACT

The liver is an important metabolic organ that controls homeostasis in the body. Moreover, it functions as a hematopoietic organ, while its metabolic function is low during development. Hepatocytes, which are parenchymal cells of the liver, acquire various metabolic functions by the maturation of hepatic progenitor cells during the fetal period; however, this molecular mechanism is still unclear. In this study, Kruppel-like factor 15 (KLF15) was identified as a new regulator of hepatic maturation through a comprehensive analysis of the expression of transcriptional regulators in mouse fetal and adult hepatocytes. KLF15 is a transcription factor whose expression in the liver increases from the embryonic stage throughout the developmental process. KLF15 induced the overexpression of liver function genes in mouse embryonic hepatocytes. Furthermore, we found that the expression of KLF15 could also induce the expression of liver function genes in hepatoblasts derived from human induced pluripotent stem cells (iPSCs). Moreover, KLF15 increased the promoter activity of tyrosine aminotransferase, a liver function gene. KLF15 also suppressed the proliferation of hepatoblasts. These results suggest that KLF15 induces hepatic maturation through the transcriptional activation of target genes and cell cycle control.


Subject(s)
Cell Differentiation , Hepatocytes/cytology , Kruppel-Like Transcription Factors/metabolism , Animals , Cell Line , Cells, Cultured , Gene Expression Regulation, Developmental , Hepatocytes/metabolism , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Kruppel-Like Transcription Factors/genetics , Liver/cytology , Liver/embryology , Liver/metabolism , Mice, Inbred C57BL
8.
Sci Rep ; 11(1): 13766, 2021 07 02.
Article in English | MEDLINE | ID: mdl-34215813

ABSTRACT

Sexual dimorphism in gene regulation, including DNA methylation, is the main driver of sexual dimorphism in phenotypes. However, the questions of how and when sex shapes DNA methylation remain unresolved. Recently, using mice with different combinations of genetic and phenotypic sex, we identified sex-associated differentially methylated regions (sDMRs) that depended on the sex phenotype. Focusing on a panel of validated sex-phenotype dependent male- and female-biased sDMRs, we tested the developmental dynamics of sex bias in liver methylation and the impacts of mutations in the androgen receptor, estrogen receptor alpha, or the transcriptional repressor Bcl6 gene. True hermaphrodites that carry both unilateral ovaries and contralateral testes were also tested. Our data show that sex bias in methylation either coincides with or follows sex bias in the expression of sDMR-proximal genes, suggesting that sex bias in gene expression may be required for demethylation at certain sDMRs. Global ablation of AR, ESR1, or a liver-specific loss of BCL6, all alter sDMR methylation, whereas presence of both an ovary and a testis delays the establishment of male-type methylation levels in hermaphrodites. Moreover, the Bcl6-LKO shows dissociation between expression and methylation, suggesting a distinct role of BCL6 in demethylation of intragenic sDMRs.


Subject(s)
DNA Methylation/genetics , Estrogen Receptor alpha/genetics , Proto-Oncogene Proteins c-bcl-6/genetics , Receptors, Androgen/genetics , Animals , Disorders of Sex Development/genetics , Epigenesis, Genetic , Female , Gene Expression Regulation/genetics , Gene Expression Regulation, Developmental/genetics , Liver/growth & development , Liver/metabolism , Male , Mice , Ovary/growth & development , Ovary/metabolism , Sex Characteristics , Sexism , Testis/growth & development , Testis/metabolism
9.
Sci Rep ; 11(1): 11381, 2021 05 31.
Article in English | MEDLINE | ID: mdl-34059723

ABSTRACT

Ammonia has a cytotoxic effect and can therefore be used as a selection agent for enrichment of zone I hepatocytes. However, it has not yet been determined whether ammonia-treated hepatocyte-like cells are able to proliferate in vitro. In this study, we employed an ammonia selection strategy to purify hepatocyte-like cells that were differentiated from human embryonic stem cells (ESCs) and from induced pluripotent stem cells (iPSCs). The resistance to cytotoxicity or cell death by ammonia is likely attributable to the metabolism of ammonia in the cells. In addition to ammonia metabolism-related genes, ammonia-selected hepatocytes showed increased expression of the cytochrome P450 genes. Additionally, the ammonia-selected cells achieved immortality or at least an equivalent life span to human pluripotent stem cells without affecting expression of the liver-associated genes. Ammonia treatment in combination with in vitro propagation is useful for obtaining large quantities of hepatocytes.


Subject(s)
Ammonia/pharmacology , Embryonic Stem Cells/drug effects , Hepatocytes/drug effects , Animals , Cell Proliferation , Cells, Cultured , Embryonic Stem Cells/cytology , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/drug effects , Mice
10.
Regen Ther ; 16: 81-89, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33732817

ABSTRACT

INTRODUCTION: Biliary strictures after bile duct injury or duct-to-duct biliary reconstruction are serious complications that markedly reduce patients' quality of life because their treatment involves periodic stent replacements. This study aimed to create a scaffold-free tubular construct as an interposition graft to treat biliary complications. METHODS: Scaffold-free tubular constructs of allogeneic pig fibroblasts, that is, fibroblast tubes, were created using a Bio-3D Printer and implanted into pigs as interposition grafts for duct-to-duct biliary reconstruction. RESULTS: Although the fibroblast tube was weaker than the native bile duct, it was sufficiently strong to enable suturing. The pigs' serum hepatobiliary enzyme levels remained stable during the experimental period. Micro-computed tomography showed no biliary strictures, no biliary leakages, and no intrahepatic bile duct dilations. The tubular structure was retained in all resected specimens, and the fibroblasts persisted at the graft sites. Immunohistochemical analyses revealed angiogenesis in the fibroblast tube and absence of extensions of the biliary epithelium into the fibroblast tube's lumen. CONCLUSIONS: This study's findings demonstrated successful reconstruction of the extrahepatic bile duct with a scaffold-free tubular construct created from pig fibroblasts using a novel Bio-3D Printer. This construct could provide a novel regenerative treatment for patients with hepatobiliary diseases.

11.
Sci Rep ; 10(1): 9704, 2020 06 16.
Article in English | MEDLINE | ID: mdl-32546802

ABSTRACT

The prevalence of non-alcoholic steatohepatitis (NASH) rapidly increases with metabolic disorders such as dyslipidaemia, high blood pressure, and hyperglycaemia. B cell lymphoma 6 (Bcl6), a transcriptional repressor, is essential for the formation of germinal centre B cells. In this study, we analysed the role of Bcl6 in NASH progression-associated pathological changes, such as hepatic lipid accumulation, liver fibrosis, and hepatocarcinogenesis. The roles of Bcl6 in NASH were analysed using liver-specific Bcl6 knockout (Bcl6-LKO) and control wild-type (WT) mice. The murine NASH model was established by feeding the mice with choline-deficient, L-amino-acid-defined, high-fat diet (CDAHFD). Feeding the WT mice with CDAHFD for 7 weeks induced the formation of histopathological features resembling human NASH, such as hepatic lipid accumulation, hepatocellular injury, and fibrosis. These histopathological changes were significantly attenuated in Bcl6-LKO mice. Additionally, feeding the male WT mice with CDAHFD for 38 weeks induced the formation of liver tumours, which was suppressed in Bcl6-LKO mice. These findings indicate that Bcl6 is involved in the progression of NASH and NASH-derived tumours.


Subject(s)
Liver/metabolism , Non-alcoholic Fatty Liver Disease/pathology , Proto-Oncogene Proteins c-bcl-6/metabolism , Animals , Disease Models, Animal , Disease Progression , Female , Lipid Metabolism , Liver/chemistry , Male , Mice , Mice, Knockout , Non-alcoholic Fatty Liver Disease/etiology , Non-alcoholic Fatty Liver Disease/metabolism , Proto-Oncogene Proteins c-bcl-6/physiology , Real-Time Polymerase Chain Reaction , Triglycerides/analysis , Triglycerides/metabolism
12.
Biochem Biophys Res Commun ; 528(3): 420-425, 2020 07 30.
Article in English | MEDLINE | ID: mdl-32505348

ABSTRACT

Parenchymal hepatocytes are responsible for most of the metabolic functions of the liver, but exhibit distinct functional properties depending on their localization within the hepatic lobule. Cytochrome P450 oxidases represent a family of drug-metabolizing enzymes, which are expressed predominantly in hepatocytes localized in the centrilobular area (zone 3). The present study describes a unique transgenic mouse strain that distinguishes zone 3 hepatocytes from periportal zone 1 hepatocytes by the intensity of EGFP fluorescence. Both zone 1 and zone 3 hepatocytes isolated from these mice showed the same zone-specific gene expression patterns as in liver tissue in vivo. Experiments using primary cultures of hepatocytes indicated that a combination of low oxygen concentration and activation of Wnt/ß-catenin signaling maintained the expression of zone 3-specific P450 drug-metabolizing enzymes, which was characterized by their susceptibility to acetaminophen-induced mitochondrial dysfunction. These zone-specific hepatocytes provide a useful system in the research area of liver pathophysiology and drug development.


Subject(s)
Hepatocytes/metabolism , NADPH-Ferrihemoprotein Reductase/metabolism , Acetaminophen/toxicity , Animals , Cell Separation , Cells, Cultured , Flow Cytometry , Gene Expression , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Hepatocytes/cytology , Hepatocytes/drug effects , Liver/anatomy & histology , Liver/cytology , Liver/metabolism , Mice , Mice, Transgenic , Mitochondria, Liver/metabolism , NADPH-Ferrihemoprotein Reductase/genetics , Oxygen/metabolism , Wnt Signaling Pathway/drug effects
13.
Hepatol Commun ; 4(2): 235-254, 2020 Feb.
Article in English | MEDLINE | ID: mdl-32025608

ABSTRACT

Formation of intrahepatic bile ducts (IHBDs) proceeds in accordance with their microenvironment. Particularly, mesenchymal cells around portal veins regulate the differentiation and ductular morphogenesis of cholangiocytes in the developing liver; however, further studies are needed to fully understand the arrangement of IHBDs into a continuous hierarchical network. This study aims to clarify the interaction between biliary and liver mesenchymal cells during IHBD formation. To identify candidate factors contributing to this cell-cell interaction, mesenchymal cells were isolated from embryonic day 16.5 matrix metalloproteinase 14 (MMP14)-deficient (knockout [KO]) mice livers, in which IHBD formation is retarded, and compared with those of the wild type (WT). WT mesenchymal cells significantly facilitated the formation of luminal structures comprised of hepatoblast-derived cholangiocytes (cholangiocytic cysts), whereas MMP14-KO mesenchymal cells failed to promote cyst formation. Comprehensive analysis revealed that expression of vasoactive intestinal peptide (VIP) was significantly suppressed in MMP14-KO mesenchymal cells. VIP and VIP receptor 1 (VIPR1) were mainly expressed in periportal mesenchymal cells and cholangiocytic progenitors during IHBD development, respectively, in vivo. VIP/VIPR1 signaling significantly encouraged cholangiocytic cyst formation and up-regulated tight junction protein 1, cystic fibrosis transmembrane conductance regulator, and aquaporin 1, in vitro. VIP antagonist significantly suppressed the tight junction assembly and the up-regulation of ion/water transporters during IHBD development in vivo. In a cholestatic injury model of adult mice, exogenous VIP administration promoted the restoration of damaged tight junctions in bile ducts and improved hyperbilirubinemia. Conclusion: VIP is produced by periportal mesenchymal cells during the perinatal stage. It supports bile duct development by establishing tight junctions and up-regulating ion/water transporters in cholangiocytes. VIP contributes to prompt recovery from cholestatic damage through the establishment of tight junctions in the bile ducts.

14.
Hepatology ; 71(4): 1437-1452, 2020 04.
Article in English | MEDLINE | ID: mdl-31549421

ABSTRACT

BACKGROUND AND AIMS: Hepatic stellate cells (HSCs), a key player in the progression of liver fibrosis, are activated by various inflammatory stimuli and converted to myofibroblast-like cells with excessive collagen production. Despite many attempts to suppress activation of HSCs or inhibit collagen production in activated HSCs, their clinical applications have not been established yet. Recently, the deactivation of HSCs has been reported as a mechanism underlying the reversibility of experimental liver fibrosis. In the present study, we sought for deactivation factors of HSCs that induce regression of established liver fibrosis. APPROACH AND RESULTS: We identified transcription factor 21 (Tcf21) as one of the transcription factors whose expression was up-regulated in parallel to the differentiation of fetal HSCs. Expression of Tcf21 in HSCs remarkably decreased during culture-induced activation in vitro and in murine and human fibrotic liver tissue in vivo. This reduced Tcf21 expression was recovered during the spontaneous regression of murine liver fibrosis. Tcf21 was also examined for its effects by adeno-associated virus serotype 6-mediated Tcf21 gene transfer into cultured activated HSCs and mice with carbon tetrachloride- or methionine-choline deficient diet-induced liver fibrosis. Overexpression of Tcf21 in activated HSCs not only suppressed fibrogenic gene expression but also restored cells, at least in part, to a quiescent phenotype both in vitro and in vivo. These phenotypic changes of HSCs were accompanied by the regression of steatohepatitis and fibrosis and improved hepatic architecture and function. CONCLUSIONS: Tcf21 has been identified as a deactivation factor of fibrogenic HSCs, providing insight into a treatment strategy for the otherwise intractable liver fibrosis.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/physiology , Hepatic Stellate Cells/metabolism , Liver Cirrhosis/pathology , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Gene Expression Regulation , Gene Transfer Techniques , Hepatic Stellate Cells/pathology , Liver Cirrhosis/genetics , Mice , Mice, Inbred C57BL
15.
J Hepatol ; 71(1): 143-152, 2019 07.
Article in English | MEDLINE | ID: mdl-30898581

ABSTRACT

BACKGROUND & AIMS: Congenital hepatic fibrosis (CHF) is a genetic liver disease resulting in abnormal proliferation of cholangiocytes and progressive hepatic fibrosis. CHF is caused by mutations in the PKHD1 gene and the subsequent dysfunction of the protein it encodes, fibrocystin. However, the underlying molecular mechanism of CHF, which is quite different from liver cirrhosis, remains unclear. This study investigated the molecular mechanism of CHF pathophysiology using a genetically engineered human induced pluripotent stem (iPS) cell model to aid the discovery of novel therapeutic agents for CHF. METHODS: PKHD1-knockout (PKHD1-KO) and heterozygously mutated PKHD1 iPS clones were established by RNA-guided genome editing using the CRISPR/Cas9 system. The iPS clones were differentiated into cholangiocyte-like cells in cysts (cholangiocytic cysts [CCs]) in a 3D-culture system. RESULTS: The CCs were composed of a monolayer of cholangiocyte-like cells. The proliferation of PKHD1-KO CCs was significantly increased by interleukin-8 (IL-8) secreted in an autocrine manner. IL-8 production was significantly elevated in PKHD1-KO CCs due to mitogen-activated protein kinase pathway activation caused by fibrocystin deficiency. The production of connective tissue growth factor (CTGF) was also increased in PKHD1-KO CCs in an IL-8-dependent manner. Furthermore, validation analysis demonstrated that both the serum IL-8 level and the expression of IL-8 and CTGF in the liver samples were significantly increased in patients with CHF, consistent with our in vitro human iPS-disease model of CHF. CONCLUSIONS: Loss of fibrocystin function promotes IL-8-dependent proliferation of, and CTGF production by, human cholangiocytes, suggesting that IL-8 and CTGF are essential for the pathogenesis of CHF. IL-8 and CTGF are candidate molecular targets for the treatment of CHF. LAY SUMMARY: Congenital hepatic fibrosis (CHF) is a genetic liver disease caused by mutations of the PKHD1 gene. Dysfunction of the protein it encodes, fibrocystin, is closely associated with CHF pathogenesis. Using an in vitro human induced pluripotent stem cell model and patient samples, we showed that the loss of fibrocystin function promotes proliferation of cholangiocytes and the production of connective tissue growth factor (CTGF) in an interleukin 8 (IL-8)-dependent manner. These results suggest that IL-8 and CTGF are essential for the pathogenesis of CHF.


Subject(s)
Connective Tissue Growth Factor/metabolism , Epithelial Cells/metabolism , Genetic Diseases, Inborn/metabolism , Liver Cirrhosis/metabolism , Bile Ducts/pathology , Cell Proliferation , Gene Editing/methods , Humans , Induced Pluripotent Stem Cells , Interleukin-8/metabolism , Mutagenesis, Site-Directed/methods , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism
16.
Sci Rep ; 9(1): 2072, 2019 02 14.
Article in English | MEDLINE | ID: mdl-30765795

ABSTRACT

Human induced pluripotent stem (iPS) cells can differentiate into hepatocyte lineages, although the phenotype of the differentiated cells is immature compared to adult hepatocytes. Improvement of cell-cell interactions between epithelium and mesenchyme is a potential approach to address this phenotype issue. In this study, we developed a model system for improving interactions between human iPS-derived hepatic progenitor cells (iPS-HPCs) and human iPS-derived hepatic stellate cell-like cells (iPS-HSCs). The phenotype of iPS-HSCs, including gene and protein expression profiles and vitamin A storage, resembled that of hepatic stellate cells. Direct co-culture of iPS-HSCs with iPS-HPCs significantly improved hepatocytic maturation in iPS-HPCs, such as their capacity for albumin production. Next, we generated iPS cell lines overexpressing LIM homeobox 2 (LHX2), which suppresses myofibroblastic changes in HSCs in mice. Hepatocytic maturation in iPS-HPCs was significantly increased in direct co-culture with iPS-HSCs overexpressing LHX2, but not in co-culture with a human hepatic stellate cell line (LX-2) overexpressing LHX2. LHX2 regulated the expression of extracellular matrices, such as laminin and collagen, in iPS-HSCs. In conclusion, this study provides an evidence that LHX2 upregulation in iPS-HSCs promotes hepatocytic maturation of iPS-HPCs, and indicates that genetically modified iPS-HSCs will be of value for research into cell-cell interactions.


Subject(s)
Hepatic Stellate Cells/metabolism , Induced Pluripotent Stem Cells/metabolism , LIM-Homeodomain Proteins/metabolism , Liver/metabolism , Transcription Factors/metabolism , Animals , Cell Communication/physiology , Cell Culture Techniques/methods , Cell Differentiation/physiology , Cell Line , Coculture Techniques/methods , Hepatocytes/metabolism , Humans , Mice , Up-Regulation/physiology
17.
Stem Cells ; 37(1): 89-101, 2019 01.
Article in English | MEDLINE | ID: mdl-30270488

ABSTRACT

Granulocyte colony stimulating factor (G-CSF) has been reported to ameliorate impaired liver function in patients with advanced liver diseases through mobilization and proliferation of hepatic progenitor cells (HPCs). However, the underlying mechanisms remain unknown. We previously showed that G-CSF treatment increased the number of bone marrow (BM)-derived cells migrating to the fibrotic liver following repeated carbon tetrachloride (CCl4 ) injections into mice. In this study, we identified opioid growth factor receptor-like 1 (OGFRL1) as a novel BM cell-derived accelerator of fibrotic liver regeneration in response to G-CSF treatment. Endogenous Ogfrl1 was highly expressed in the hematopoietic organs such as the BM and spleen, whereas the liver contained a relatively small amount of Ogfrl1 mRNA. Among the peripheral blood cells, monocytes were the major sources of OGFRL1. Endogenous Ogfrl1 expression in both the peripheral blood monocytes and the liver was decreased following repeated CCl4 injections. An intrasplenic injection of cells overexpressing OGFRL1 into CCl4 -treated fibrotic mice increased the number of HPC and stimulated proliferation of hepatic parenchymal cells after partial resection of the fibrotic liver. Furthermore, overexpression of OGFRL1 in cultured HPC accelerated their differentiation as estimated by increased expression of liver-specific genes such as hepatocyte nuclear factor 4α, cytochrome P450, and fatty acid binding protein 1, although it did not affect the colony forming ability of HPC. These results indicate a critical role of OGFRL1 in the mobilization and differentiation of HPC in the fibrotic liver, and administration of OGFRL1-expressing cells may serve as a potential regenerative therapy for advanced liver fibrosis. Stem Cells 2019;37:89-101.


Subject(s)
Hematopoietic Stem Cell Mobilization/methods , Liver Cirrhosis/genetics , Liver Cirrhosis/therapy , Liver Regeneration/genetics , Regenerative Medicine/methods , Stem Cells/metabolism , Animals , Cell Differentiation , Humans , Male , Mice , Transfection
18.
Methods Mol Biol ; 1905: 143-153, 2019.
Article in English | MEDLINE | ID: mdl-30536097

ABSTRACT

Inducible pluripotent stem (iPS) cells are multipotent stem cells that are produced by gene transfer of reprogramming factors to somatic cells. They are thought to be an important source of regenerative medicine because of their pluripotency and self-renewal ability. Although the liver has high regeneration ability, continuous death of hepatocytes due to chronic inflammation leads to liver cirrhosis and liver carcinoma. With regard to such serious liver diseases, liver transplantation is used as a complete cure, but there is a problem of donor shortage. Therefore, transplantation therapy using liver tissue generated from stem cells in vitro is expected.We are developing a system to induce the differentiation of cholangiocytes, one of important non-parenchymal cells in living liver tissue, from human iPS cells. Bile duct-like cystic structures can be induced by purifying human iPS cell-derived hepatoblasts expressing hepatic progenitor cell surface markers and inducing differentiation under appropriate culture conditions. These cells are considered to be useful in constructing a hepatic organoid that reproduces the liver structure of the living body.


Subject(s)
Bile Ducts/cytology , Cell Culture Techniques/methods , Induced Pluripotent Stem Cells/cytology , Bile Ducts/metabolism , Biomarkers/metabolism , Cell Differentiation , Cells, Cultured , Humans , Induced Pluripotent Stem Cells/metabolism , Organoids/cytology , Organoids/metabolism
19.
Methods Mol Biol ; 1905: 211-219, 2019.
Article in English | MEDLINE | ID: mdl-30536103

ABSTRACT

Hepatic progenitor cells are defined as cells exhibiting potency for active proliferation and capacity for bipotential differentiation into hepatocytes and cholangiocytes. To prove the capacity of target cells for terminal differentiation and reconstitution of organs, cell transplantation models have been widely used in previous studies, including those involving the liver. Here we describe a protocol for transplantation of hepatic progenitor cells using retrorsine pretreatment and partial hepatectomy. This transplantation assay reveals the potential for reconstitution of hepatocytes in recipient livers by primary hepatic progenitor cells. Donor cells are detected as a colony composed of 5-10 mature hepatocytes.


Subject(s)
Hepatocytes/cytology , Liver/injuries , Pyrrolizidine Alkaloids/adverse effects , Stem Cell Transplantation/methods , Animals , Cell Differentiation , Cells, Cultured , Female , Hepatectomy , Liver/cytology , Liver/drug effects , Liver/surgery , Liver Regeneration , Male , Mice , Models, Animal , Stem Cells/cytology
20.
Stem Cell Res ; 32: 17-24, 2018 10.
Article in English | MEDLINE | ID: mdl-30172093

ABSTRACT

In the developing liver, bile duct structure is formed through differentiation of hepatic progenitor cells (HPC) into cholangiocytes. A subtype of polycystic liver diseases characterized by uncontrolled expansion of bile ductal cells is caused by genetic abnormalities such as in that of protein kinase C substrate 80 K-H (PRKCSH). In this study, we aimed to mimic the disease process in vitro by genome editing of the PRKCSH locus in human inducible pluripotent stem (iPS) cells. A proportion of cultured human iPS cell-derived CD13+CD133+ HPC differentiated into CD13- cells. During the subsequent gel embedding culture, CD13- cells formed bile ductal marker-positive cystic structures with the polarity of epithelial cells. A deletion of PRKCSH gene increased expression of cholangiocytic transcription factors in CD13- cells and the number of cholangiocytic cyst structure. These results suggest that PRKCSH deficiency promotes the differentiation of HPC-derived cholangiocytes, providing a good in vitro model to analyze the molecular mechanisms underlying polycystic diseases.


Subject(s)
Cysts/metabolism , Cysts/pathology , Gene Editing/methods , Liver Diseases/metabolism , Liver Diseases/pathology , AC133 Antigen/metabolism , CD13 Antigens/metabolism , Calcium-Binding Proteins , Cell Differentiation , Cell Line , Cells, Cultured , Flow Cytometry , Glucosidases/deficiency , Glucosidases/genetics , Humans , Intracellular Signaling Peptides and Proteins/deficiency , Intracellular Signaling Peptides and Proteins/genetics , Liver/cytology
SELECTION OF CITATIONS
SEARCH DETAIL
...