Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
EMBO J ; 43(3): 339-361, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38238476

ABSTRACT

Hematopoietic stem cell (HSC) divisional fate and function are determined by cellular metabolism, yet the contribution of specific cellular organelles and metabolic pathways to blood maintenance and stress-induced responses in the bone marrow remains poorly understood. The outer mitochondrial membrane-localized E3 ubiquitin ligase MITOL/MARCHF5 (encoded by the Mitol gene) is known to regulate mitochondrial and endoplasmic reticulum (ER) interaction and to promote cell survival. Here, we investigated the functional involvement of MITOL in HSC maintenance by generating MX1-cre inducible Mitol knockout mice. MITOL deletion in the bone marrow resulted in HSC exhaustion and impairment of bone marrow reconstitution capability in vivo. Interestingly, MITOL loss did not induce major mitochondrial dysfunction in hematopoietic stem and progenitor cells. In contrast, MITOL deletion induced prolonged ER stress in HSCs, which triggered cellular apoptosis regulated by IRE1α. In line, dampening of ER stress signaling by IRE1α inihibitor KIRA6 partially rescued apoptosis of long-term-reconstituting HSC. In summary, our observations indicate that MITOL is a principal regulator of hematopoietic homeostasis and protects blood stem cells from cell death through its function in ER stress signaling.


Subject(s)
Endoribonucleases , Protein Serine-Threonine Kinases , Animals , Mice , Apoptosis , Hematopoietic Stem Cells/metabolism , Protein Serine-Threonine Kinases/genetics , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
2.
J Bone Miner Res ; 33(10): 1785-1798, 2018 10.
Article in English | MEDLINE | ID: mdl-29893999

ABSTRACT

Osteoclast differentiation is a dynamic differentiation process, which is accompanied by dramatic changes in metabolic status as well as in gene expression. Recent findings have revealed an essential connection between metabolic reprogramming and dynamic gene expression changes during osteoclast differentiation. However, the upstream regulatory mechanisms that drive these metabolic changes in osteoclastogenesis remain to be elucidated. Here, we demonstrate that induced deletion of a tumor suppressor gene, Folliculin (Flcn), in mouse osteoclast precursors causes severe osteoporosis in 3 weeks through excess osteoclastogenesis. Flcn-deficient osteoclast precursors reveal cell autonomous accelerated osteoclastogenesis with increased sensitivity to receptor activator of NF-κB ligand (RANKL). We demonstrate that Flcn regulates oxidative phosphorylation and purine metabolism through suppression of nuclear localization of the transcription factor Tfe3, thereby inhibiting expression of its target gene Pgc1. Metabolome studies revealed that Flcn-deficient osteoclast precursors exhibit significant augmentation of oxidative phosphorylation and nucleotide production, resulting in an enhanced purinergic signaling loop that is composed of controlled ATP release and autocrine/paracrine purinergic receptor stimulation. Inhibition of this purinergic signaling loop efficiently blocks accelerated osteoclastogenesis in Flcn-deficient osteoclast precursors. Here, we demonstrate an essential and novel role of the Flcn-Tfe3-Pgc1 axis in osteoclastogenesis through the metabolic reprogramming of oxidative phosphorylation and purine metabolism. © 2018 The Authors Journal of Bone and Mineral Research published by Wiley Periodicals, Inc. on behalf of American Society for Bone and Mineral Research (ASBMR).


Subject(s)
Osteoclasts/metabolism , Osteogenesis , Proto-Oncogene Proteins/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Bone Marrow/pathology , Mice , Mice, Knockout , Organelle Biogenesis , Osteoclasts/pathology , Osteoporosis/metabolism , Osteoporosis/pathology , Oxidative Phosphorylation , Purines/metabolism , RAW 264.7 Cells , Signal Transduction , Transcription Factors/metabolism , Up-Regulation
3.
Lab Invest ; 93(1): 54-71, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23147226

ABSTRACT

We previously produced mice with human hepatocyte (h-hep) chimeric livers by transplanting h-heps into albumin enhancer/promoter-driven urokinase-type plasminogen activator-transgenic severe combined immunodeficient (SCID) mice with liver disease. The chimeric livers were constructed with h-heps, mouse hepatocytes, and mouse hepatic sinusoidal cells (m-HSCs). Here, we investigated the morphological features of the chimeric livers and the h-hep gene expression profiles in the xenogeneic animal body. To do so, we performed immunohistochemistry, morphometric analyses, and electron microscopic observations on chimeric mouse livers, and used microarray analyses to compare gene expression patterns in hepatocytes derived from chimeric mouse hepatocytes (c-heps) and h-heps. Morphometric analysis revealed that the ratio of hepatocytes to m-HSCs in the chimeric mouse livers were twofold higher than those in the SCID mouse livers, corresponding to twin-cell plates in the chimeric mouse liver. The h-heps in the chimeric mouse did not show hypoxia even in the twin-cell plate structure, probably because of low oxygen consumption by the h-heps relative to the mouse hepatocytes (m-heps). Immunohistochemical and electron microscopic examinations revealed that the sinusoids in the chimeric mouse livers were normally constructed with h-heps and m-HSCs. However, a number of microvilli projected into the intercellular clefts on the lateral aspects of the hepatocytes, features typical of a growth phase. Microarray profiles indicated that ∼82% of 16 605 probes were within a twofold range difference between h-heps and c-heps. Cluster and principal component analyses showed that the gene expression patterns of c-heps were extremely similar to those of h-heps. In conclusion, the chimeric mouse livers were normally reconstructed with h-heps and m-HSCs, and expressed most human genes at levels similar to those in human livers, although the chimeric livers showed morphological characteristics typical of growth.


Subject(s)
Hepatocytes/cytology , Liver/cytology , Analysis of Variance , Animals , Cell Adhesion/physiology , Cell Hypoxia/physiology , Female , Gene Expression Profiling , Hepatic Stellate Cells/cytology , Humans , Immunohistochemistry , Kupffer Cells/cytology , Liver/chemistry , Male , Mice , Mice, SCID , Tissue Array Analysis/methods , Transplantation, Heterologous
4.
Cell Transplant ; 21(2-3): 429-36, 2012.
Article in English | MEDLINE | ID: mdl-22793050

ABSTRACT

Recent advances in tissue engineering technologies have highlighted the ability to create functional liver systems using isolated hepatocytes in vivo. Considering the serious shortage of donor livers that can be used for hepatocyte isolation, it has remained imperative to establish a hepatocyte propagation protocol to provide highly efficient cell recovery allowing for subsequent tissue engineering procedures. Donor primary hepatocytes were isolated from human α-1 antitrypsin (hA1AT) transgenic mice and were transplanted into the recipient liver of urokinase-type plasminogen activator-severe combined immunodeficiency (uPA/SCID) mice. Transplanted donor hepatocytes actively proliferated within the recipient liver of the uPA/SCID mice. At week 8 or later, full repopulation of the uPA/SCID livers with the transplanted hA1AT hepatocytes were confirmed by blood examination and histological assessment. Proliferated hA1AT hepatocytes were recovered from the recipient uPA/SCID mice, and we generated hepatocyte sheets using these recovered hepatocytes for subsequent transplantation into the subcutaneous space of mice. Stable persistency of the subcutaneously engineered liver tissues was confirmed for up to 90 days, which was the length of our present study. These new data demonstrate the feasibility in propagating murine hepatocytes prior to the development of hepatic cells and bioengineered liver systems. The ability to regenerate and expand hepatocytes has potential clinical value whereby procurement of small amounts of tissue could be expanded to sufficient quantities prior to their use in hepatocyte transplantation or other hepatocyte-based therapies.


Subject(s)
Hepatocytes/cytology , Liver/pathology , Tissue Engineering , Acrylamides/chemistry , Acrylic Resins , Animals , Cell Culture Techniques/instrumentation , Cell Culture Techniques/methods , Cell Proliferation , Hepatocytes/transplantation , Humans , Liver/metabolism , Mice , Mice, SCID , Mice, Transgenic , Polymers/chemistry , Regenerative Medicine , Transplantation, Heterologous , alpha 1-Antitrypsin/metabolism
5.
Endocrinology ; 152(4): 1479-91, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21303949

ABSTRACT

Clinical studies have shown a close association between nonalcoholic fatty liver disease and adult-onset GH deficiency, but the relevant molecular mechanisms are still unclear. No mouse model has been suitable to study the etiological relationship of human nonalcoholic fatty liver disease and human adult-onset GH deficiency under conditions similar to the human liver in vivo. We generated human (h-)hepatocyte chimeric mice with livers that were predominantly repopulated with h-hepatocytes in a h-GH-deficient state. The chimeric mouse liver was mostly repopulated with h-hepatocytes about 50 d after transplantation and spontaneously became fatty in the h-hepatocyte regions after about 70 d. Infusion of the chimeric mouse with h-GH drastically decreased steatosis, showing the direct cause of h-GH deficiency in the generation of hepatic steatosis. Using microarray profiles aided by real-time quantitative RT-PCR, comparison between h-hepatocytes from h-GH-untreated and -treated mice identified 14 GH-up-regulated and four GH-down-regulated genes, including IGF-I, SOCS2, NNMT, IGFLS, P4AH1, SLC16A1, SRD5A1, FADS1, and AKR1B10, respectively. These GH-up- and -down-regulated genes were expressed in the chimeric mouse liver at lower and higher levels than in human livers, respectively. Treatment of the chimeric mice with h-GH ameliorated their altered expression. h-Hepatocytes were separated from chimeric mouse livers for testing in vitro effects of h-GH or h-IGF-I on gene expression, and results showed that GH directly regulated the expression of IGF-I, SOCS2, NNMT, IGFALS, P4AH1, FADS1, and AKR1B10. In conclusion, the chimeric mouse is a novel h-GH-deficient animal model for studying in vivo h-GH-dependent human liver dysfunctions.


Subject(s)
Fatty Liver/pathology , Growth Hormone/metabolism , Hepatocytes/transplantation , Liver/cytology , Liver/pathology , Adult , Animals , Delta-5 Fatty Acid Desaturase , Fatty Liver/metabolism , Female , Growth Hormone/deficiency , Growth Hormone/genetics , Growth Hormone/pharmacology , Hepatocytes/drug effects , Humans , Immunohistochemistry , Insulin-Like Growth Factor I/pharmacology , Liver/metabolism , Male , Mice , Middle Aged , Oligonucleotide Array Sequence Analysis , Reverse Transcriptase Polymerase Chain Reaction
6.
Drug Metab Pharmacokinet ; 25(6): 539-50, 2010.
Article in English | MEDLINE | ID: mdl-20930422

ABSTRACT

Cryopreserved human (h-) hepatocytes are currently regarded as the best in vitro model for predicting human intrinsic clearance of xenobiotics. Although fresh h-hepatocytes have greater plating efficiency on dishes and greater metabolic activities than cryopreserved cells, performing reproducible studies using fresh hepatocytes from the same donor and having an "on demand" supply of fresh hepatocytes are not possible. In this study, cryopreserved h-hepatocytes were transplanted into albumin enhancer/promoter-driven, urokinase-type plasminogen activator, transgenic/severe combined immunodeficient (uPA/SCID) mice to produce chimeric mice, the livers of which were largely replaced with h-hepatocytes. We determined whether the chimeric mouse could serve as a novel source of fresh h-hepatocytes for in vitro studies. h-Hepatocytes were isolated from chimeric mice (chimeric hepatocytes), and cytochrome P450 (P450) activities were determined. Compared with cryopreserved cells, the P450 (1A2, 2C9, 2C19, 2D6, 2E1, 3A) activities of fresh chimeric hepatocytes were similar or greater. Moreover, ketoprofen was more actively metabolized through glucuronide conjugates by fresh chimeric hepatocytes than by cryopreserved cells. We conclude that chimeric mice may be a useful tool for supplying fresh h-hepatocytes on demand that provide high and stable phase I enzyme and glucuronidation activities.


Subject(s)
Cytochrome P-450 Enzyme System/metabolism , Glucuronides/metabolism , Hepatocytes/enzymology , Ketoprofen/metabolism , Transplantation Chimera/metabolism , Aged , Animals , Aryl Hydrocarbon Hydroxylases/metabolism , Child , Child, Preschool , Cryopreservation , Cytochrome P-450 CYP2A6 , Female , Glucuronosyltransferase/metabolism , Humans , Liver/cytology , Liver/enzymology , Male , Mice , Mice, SCID , Middle Aged
7.
Zoolog Sci ; 27(8): 638-46, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20695779

ABSTRACT

The large micromeres of the 32-cell stage of sea urchin embryos are autonomously specified and differentiate into primary mesenchyme cells (PMCs), giving rise to the skeletogenic cells. We previously demonstrated that HpEts, an ets-related transcription factor, plays an essential role in the specification of PMCs in sea urchin embryos. In order to clarify the function of HpEts in the gene regulatory network involved in PMC specification, we analyzed the zygotic expression pattern and the cis-regulatory region of HpEts, and examined the activity of the HpEts protein as a transcription factor. Intron-based PCR reveals that zygotic expression of HpEts starts at the cleavage stage, and that the rate of transcription reaches maximum at the unhatched blastula stage. A series of progressive deletions of the fragments from -4.2 kbp to +1206 bp of the HpEts, which directs PMC-specific expression, caused a gradual decrease in the specificity, implying that coordination of several cis-regulatory elements regulates the expression in PMCs. A minimum cis-element required for the temporal expression is located within a 10 bp from -243 bp to -234 bp. The HpEts protein remains in the cytoplasm of entire embryonic cells in the cleavage stage. At the unhatched blastula stage, the HpEts protein translocates into the nucleus in presumptive PMCs. Transactivation assays demonstrate that the HpEts protein activates a promoter of Spicule Matrix Protein 50 (SM50), which is a target of HpEts, which binds to the regulatory region of SM50.


Subject(s)
Gene Expression Regulation, Developmental/physiology , Gene Regulatory Networks/physiology , Hemicentrotus/cytology , Mesenchymal Stem Cells/physiology , Transcription Factors/metabolism , Animals , Base Sequence , Cell Differentiation , Molecular Sequence Data , Protein Transport , RNA, Messenger/genetics , RNA, Messenger/metabolism , Regulatory Sequences, Nucleic Acid , Transcription Factors/genetics , Zygote/physiology
8.
Am J Pathol ; 177(2): 654-65, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20522646

ABSTRACT

Liver mass is optimized in relation to body mass. Rat (r) and human (h) hepatocytes were transplanted into liver-injured immunodeficient mice and allowed to proliferate for 3 or 11 weeks, respectively, when the transplants stopped proliferating. Liver/body weight ratio was normal throughout in r-hepatocyte-bearing mice (r-hep-mice), but increased continuously in h-hepatocyte-bearing mice (h-hep-mice), until reaching approximately three times the normal m-liver size, which was considered to be hyperplasia of h-hepatocytes because there were no significant differences in cell size among host (mouse [m-]) and donor (r- and h-) hepatocytes. Transforming growth factor-beta (TGF-beta) type I receptor, TGF-beta type II receptor, and activin A type IIA receptor mRNAs in proliferating r-hepatocytes of r-hep-mice were lower than in resting r-hepatocytes (normal levels) and increased to normal levels during the termination phase. Concomitantly, m-hepatic stellate cells began to express TGF-beta proteins. In stark contrast, TGF-beta type II receptor and activin A type IIA receptor mRNAs in h-hepatocytes remained low throughout and m-hepatic stellate cells did not express TGF-beta in h-hep-mice. As expected, Smad2 and 3 translocated into nuclei in r-hep-mice but not in h-hep-mice. Histological analysis showed a paucity of m-stellate cells in h-hepatocyte colonies of h-hep-mouse liver. We conclude that m-stellate cells are able to normally interact with concordant r-hepatocytes but not with discordant h-hepatocytes, which seems to be at least partly responsible for the failure of the liver size optimization in h-hep-mice.


Subject(s)
Hepatocytes/metabolism , Hepatocytes/transplantation , Hyperplasia/pathology , Liver/pathology , Activin Receptors, Type II/genetics , Activin Receptors, Type II/metabolism , Adolescent , Adult , Animals , Child , Female , Hepatocytes/cytology , Humans , Hyperplasia/metabolism , Infant , Liver/cytology , Liver/metabolism , Male , Mice , Mice, SCID , Middle Aged , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Rats , Receptor, Transforming Growth Factor-beta Type I , Receptor, Transforming Growth Factor-beta Type II , Receptors, Transforming Growth Factor beta/genetics , Receptors, Transforming Growth Factor beta/metabolism , Transplantation, Heterologous
9.
Hum Gene Ther ; 21(1): 40-50, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19725756

ABSTRACT

We developed hybrid vectors employing high-capacity adenovirus as a first-stage carrier encoding all the components required for in situ production of a second-stage lentivirus, thereby achieving stable transgene expression in secondary target cells. Such vectors have never previously been tested in normal tissues, because of the scarcity of suitable in vivo systems permissive for second-stage lentivirus assembly. Here we employed a novel murine model in which endogenous liver tissue is extensively reconstituted with engrafted human hepatocytes, and successfully achieved stable transduction by the second-stage lentivirus produced in situ from first-stage adenovirus. This represents the first demonstration of the functionality of adenoviral-lentiviral hybrid vectors in a normal parenchymal organ in vivo.


Subject(s)
Adenoviridae/genetics , Chimera/metabolism , Genetic Vectors/genetics , Lentivirus/genetics , Liver/metabolism , Transduction, Genetic/methods , Adenoviridae Infections/metabolism , Animals , Cell Line , Cell Movement , Chimera/virology , Gene Expression , Genetic Vectors/administration & dosage , Humans , Injections, Intravenous , Liver/pathology , Liver/virology , Mice
10.
Xenotransplantation ; 15(4): 235-45, 2008.
Article in English | MEDLINE | ID: mdl-18957046

ABSTRACT

BACKGROUND: Previously, we created, a chimeric mouse (humanized mouse), a severe combined immunodeficiency (SCID) mouse whose liver was >90% repopulated with human (h)-hepatocytes, which are useful for the testing of drug metabolism and toxicity, as well as a hepatitis B virus and hepatitis C virus-susceptible animal model. However, their small body size and small total blood volume limited the utilization for analytical purposes, which led us to develop a method to create a chimeric rat bearing h-hepatocyte-repopulated liver. METHODS: F344 nude rats devoid of T cells were irradiated with X-rays and injected with bone marrow cells (BMCs) from SCID mice (m(SCID)). The rate of replacement with m(SCID)-BMCs was evaluated by two-color flow cytometry analysis of peripheral blood mononuclear cells (PBMCs). After m(SCID)-BMCs repopulated the host bone marrow (BM), the rats were treated with retrorsine, partially hepatectomized (PHx), and transplanted with 5 x 10(6) h-hepatocytes isolated from the chimeric mice. h-Albumin (h-Alb) concentrations in the host blood and the expression levels of protein and mRNA of hepatocyte differentiation markers in the h-hepatocytes were evaluated by ELISA, immunostaining, and reverse transcription-PCR, respectively. RESULTS: The m(SCID)-BMCs successfully repopulated the rats, the percentage of mouse cells reaching 94% among host (r(nudeF344)) PBMCs at 4 weeks after m-BMC transplantation. h-Hepatocytes isolated from the chimeric mice were transplanted to the liver of the m(SCID)-BMC-repopulated rats. The engrafted h-hepatocytes expressed h-Alb and h-cytochrome P450 (CYP) subtypes and survived showing normal phenotypes until at least 3 weeks post-h-hepatocytes transplantation (h-HPCT). However, the blood concentrations of h-Alb declined at 4 weeks post-HPCT, concomitant with the emergence of both r(nudeF344)- and m(SCID)-macrophages, suggesting the rejection of h-hepatocytes due to the activation of macrophages. CONCLUSION: We developed a novel method to create a rat that bears the liver engrafted with h-hepatocytes, utilizing a rat with the BM composed of m(SCID)-BMCs as a host. This h-hepatocyte-bearing rat will be a valuable model for studying the immunologic mechanisms involved in xenogeneic transplantation and for generating rats with higher rates of repopulation with h-hepatocytes.


Subject(s)
Hepatocytes/transplantation , Animals , Base Sequence , Bone Marrow Transplantation/immunology , DNA Primers/genetics , Gene Expression Profiling , Graft Survival , Hepatocytes/immunology , Hepatocytes/metabolism , Humans , Macrophage Activation , Mice , Mice, SCID , RNA/genetics , RNA/metabolism , Rats , Rats, Inbred F344 , Rats, Nude , Transplantation Chimera , Transplantation, Heterologous
11.
Thromb Haemost ; 99(5): 883-91, 2008 May.
Article in English | MEDLINE | ID: mdl-18449417

ABSTRACT

Cell-based therapies using isolated hepatocytes have been proposed to be an attractive application in the treatment of haemophilia B due to the normal production of coagulation factor IX (FIX) in these particular cells. Current cell culture technologies have largely failed to provide adequate isolated hepatocytes, so the present studies were designed to examine a new approach to efficiently proliferate hepatocytes that can retain normal biological function, including the ability to synthesize coagulation factors like FIX. Canine or human primary hepatocytes were transplanted into urokinase-type plasminogen activator-severe combined immunodeficiency (uPA/SCID) transgenic mice. Both donor hepatocytes from canines and humans were found to progressively proliferate in the recipient mouse livers as evidenced by a sharp increase in the circulating blood levels of species-specific albumin, which was correlated with the production and release of canine and human FIX antigen levels into the plasma. Histological examination confirmed that the transplanted canine and human hepatocytes were able to proliferate and occupy >80% of the host livers. In addition, the transplanted hepatocytes demonstrated strong cytoplasmic staining for human FIX, and the secreted coagulation factor IX was found to be haemostatically competent using specific procoagulant assays. In all, the results from the present study indicated that developments based on this technology could provide sufficient FIX-producing hepatocytes for cell-based therapy for haemophilia B.


Subject(s)
Cell Transplantation/methods , Factor IX/metabolism , Hemophilia B/surgery , Hepatocytes/transplantation , Liver/surgery , Animals , Cell Culture Techniques , Cell Proliferation , Cells, Cultured , Child , Dogs , Factor IX/genetics , Female , Hemophilia B/metabolism , Hepatocytes/metabolism , Humans , Infant , Liver/metabolism , Liver/pathology , Male , Mice , Mice, Inbred C57BL , Mice, SCID , Mice, Transgenic , RNA, Messenger/metabolism , Serum Albumin/metabolism , Time Factors , Transplantation, Heterologous , Urokinase-Type Plasminogen Activator/genetics , Urokinase-Type Plasminogen Activator/metabolism
12.
Hepatology ; 47(2): 435-46, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18098326

ABSTRACT

UNLABELLED: We previously identified a small population of replicative hepatocytes in long-term cultures of human adult parenchymal hepatocytes (PHs) at a frequency of 0.01%-0.09%. These hepatocytes were able to grow continuously through serial subcultures as colony-forming parenchymal hepatocytes (CFPHs). In the present study, we generated gene expression profiles for cultured CFPHs and found that they expressed cytokeratin 19, CD90 (Thy-1), and CD44, but not mature hepatocyte markers such as tryptophan-2,3-dioxygenase (TO) and glucose-6-phosphatase (G6P), confirming that these cells are hepatic progenitor-like cells. The cultured CFPHs were resistant to infection with human hepatitis B virus (HBV). To examine the growth and differentiation capacity of the cells in vivo, serially subcultured CFPHs were transplanted into the progeny of a cross between albumin promoter/enhancer-driven urokinase plasminogen activator-transgenic mice and severe combined immunodeficient (SCID) mice. The cells were engrafted into the liver and were able to grow for at least 10 weeks, ultimately reaching a maximum occupancy rate of 27%. The CFPHs in the host liver expressed differentiation markers such as TO, G6P, and cytochrome P450 subtypes and could be infected with HBV. CFPH-chimeric mice with a relatively high replacement rate exhibited viremia and had high serum levels of hepatitis B surface antigen. CONCLUSION: Serially subcultured human hepatic progenitor-like cells from postnatal livers successfully repopulated injured livers and exhibited several phenotypes of mature hepatocytes, including susceptibility to HBV. In vitro-expanded CFPHs can be used to characterize the differentiation state of human hepatic progenitor-like cells.


Subject(s)
Hepatitis B virus , Hepatocytes/physiology , Hepatocytes/transplantation , Transplantation Chimera , Adolescent , Animals , Cells, Cultured , Child , Colony-Forming Units Assay , Cryopreservation , Female , Gene Expression Profiling , Hepatocytes/cytology , Hepatocytes/virology , Humans , Infant , Male , Mice , Mice, SCID , Mice, Transgenic
13.
Histochem Cell Biol ; 125(3): 247-57, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16195892

ABSTRACT

Monkey embryonic stem (ES) cells have characteristics that are similar to human ES cells, and might be useful as a substitute model for preclinical research. When embryoid bodies (EBs) formed from monkey ES cells were cultured, expression of many hepatocyte-related genes including cytochrome P450 (Cyp) 3a and Cyp7a1 was observed. Hepatocytes were immunocytochemically observed using antibodies against albumin (ALB), cytokeratin-8/18, and alpha1-antitrypsin in the developing EBs. The in vitro differentiation potential of monkey ES cells into the hepatic lineage prompted us to examine the transplantability of monkey EB cells. As an initial approach to assess the repopulation potential, we transplanted EB cells into immunodeficient urokinase-type plasminogen activator transgenic mice that undergo liver failure. After transplantation, the hepatocyte colonies expressing monkey ALB were observed in the mouse liver. Fluorescence in-situ hybridization revealed that the repopulating hepatocytes arise from cell fusion between transplanted monkey EB cells and recipient mouse hepatocytes. In contrast, neither cell fusion nor repopulation of hepatocytes was observed in the recipient liver after undifferentiated ES cell transplantation. These results indicate that the differentiated cells in developing monkey EBs, but not contaminating ES cells, generate functional hepatocytes by cell fusion with recipient mouse hepatocytes, and repopulate injured mouse liver.


Subject(s)
Embryonic Stem Cells/cytology , Hepatocytes/cytology , Hybrid Cells/cytology , Liver/injuries , Animals , Base Sequence , Cell Differentiation , Cell Fusion , DNA Primers/genetics , Embryonic Stem Cells/metabolism , Hepatocytes/metabolism , Hybrid Cells/metabolism , Immunohistochemistry , Liver/cytology , Liver/metabolism , Macaca fascicularis , Mice , Mice, SCID , Mice, Transgenic , Stem Cell Transplantation , Transplantation, Heterologous , Urokinase-Type Plasminogen Activator/genetics , Urokinase-Type Plasminogen Activator/metabolism
14.
Drug Metab Pharmacokinet ; 21(6): 465-74, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17220562

ABSTRACT

We investigated and compared the expression of human CYPs mRNA in primary cultures of cryopreserved human hepatocytes and in chimeric mice constructed by transplanting hepatocytes from the same human donors. Analysis was performed by real-time reverse-transcription polymerase chain reaction. Initial expression levels for the 12 human CYPs mRNA in chimeric mouse hepatocytes were higher than those in human hepatocytes, but a low correlation coefficient was observed (r=0.690). After 24 h of culture, the correlation remained low (r=0.699). The medium was replaced with fresh medium without human epidermal growth factor, and after 48 h of culture, expression of the 12 human CYPs mRNA were very similar in human hepatocytes and chimeric mouse hepatocytes, and a higher correlation coefficient was observed (r=0.809). After 72 h of culture, the correlation remained high (r=0.873). The ratio of human CYP1A2 mRNA to beta-actin mRNA in chimeric mouse hepatocytes decreased quickly during the first 24 h of culture, and then remained constant. Expression profiles of human CYP1A2 mRNA in chimeric mouse hepatocytes were similar to those in human hepatocytes after exposure of beta-naphthoflavone. CYP3A4 mRNA expression was increased significantly by rifampicin (Rif) exposure in human hepatocytes, whereas Rif-induced increases in CYP3A4 mRNA expression in chimeric mouse hepatocytes was seen for two of the three donors. In conclusion, we demonstrated that expression and induction of human CYPs in human hepatocytes can be reproduced in chimeric mouse hepatocytes.


Subject(s)
Cytochrome P-450 CYP1A2/genetics , Cytochrome P-450 Enzyme System/genetics , Hepatocytes/enzymology , Transplantation Chimera/genetics , Animals , Cell Transplantation , Cells, Cultured , Cytochrome P-450 CYP3A , Female , Gene Expression/drug effects , Hepatocytes/drug effects , Hepatocytes/transplantation , Humans , Male , Mice , Mice, SCID , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Rifampin/pharmacology , Time Factors , Transplantation, Heterologous , beta-Naphthoflavone/pharmacology
15.
Drug Metab Pharmacokinet ; 20(2): 121-6, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15855724

ABSTRACT

Chimeric mice with near-completely humanized liver were constructed by transplantating hepatocytes from a Japanese and Caucasian donor. In the present study, we investigated the induction of human CYP1A2 and CYP3A4 mRNA in a primary culture of the cryopreserved chimeric mouse hepatocytes. beta-naphthoflavone (beta-NF) and rifampicin (Rif) were used as typical cytochrome P450 (CYP) inducers for CYP1A2 and CYP3A4, respectively. Analysis was performed by the real-time reverse-transcription polymerase chain reaction method. CYP1A2 mRNA in the primary culture of chimeric mouse hepatocytes in mice No. 1, 2, and 3 was significantly increased 3.8-, 6.3-, and 3.3-fold by 5 microM beta-NF exposure, respectively, compared with the 0.1% DMSO treated control (p<0.01). CYP3A4 mRNA in the primary culture of chimeric mouse hepatocytes in mice No. 1, 2, and 3 was significantly increased 8.4-fold (p<0.001), 2.2-fold (p<0.01), and 2.3-fold (p<0.05) by 50 microM Rif exposure, respectively, compared with the 0.1% DMSO treated control. The present study demonstrated that a primary culture of cryopreserved hepatocytes from chimeric mice with humanized liver could be used for evaluating the induction of drug metabolizing enzymes in human. This in vitro method may be a useful method for screening the induction potency of new drug candidates on drug metabolizing enzymes.


Subject(s)
Cytochrome P-450 CYP1A2/biosynthesis , Cytochrome P-450 Enzyme System/biosynthesis , Hepatocytes/enzymology , Adolescent , Animals , Cells, Cultured , Chimera , Cryopreservation , Cytochrome P-450 CYP3A , Enzyme Induction , Humans , Male , Mice , Reverse Transcriptase Polymerase Chain Reaction
16.
Drug Metab Dispos ; 32(12): 1402-10, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15383493

ABSTRACT

Recently, a chimeric mouse line in which the liver could be replaced by more than 80% with human hepatocytes was established in Japan. Because the chimeric mouse produces human albumin (hAlb), replacement by human hepatocytes could be estimated by the hAlb concentration in the blood of chimeric mice. In this study, we investigated human major cytochrome P450 (P450) in the livers of chimeric mice by mRNA, protein, and enzyme activity using real-time polymerase chain reaction, Western blot analysis, and high-performance liquid chromatography, respectively. Chimeric mice with humanized liver generated using hepatocytes from a Japanese and white donor were used. Human P450 mRNAs were expressed in the liver of chimeric mice, and major human P450 proteins such as CYP1A2, CYP2C9, and CYP3A4 were detected. The expression of P450 mRNA and protein was correlated with the hAlb concentration in the blood. The enzyme activities such as diclofenac 4'-hydroxylase activity, dexamethasone 6-hydroxylase activity, and coumarin 7-hydroxylase activity, activities that are specific to human P450 but not to murine P450, were increased in a hAlb concentration-dependent manner. The chimeric mice with nearly 90% replacement by human hepatocytes demonstrated almost the same protein contents of human P450s and drug-metabolizing enzyme activity as those of the donor. It was confirmed that genomic DNA from the livers of the chimeric mice and that from the liver of the donor exhibited the same genotype. In conclusion, the chimeric mice exhibited a similarly efficient capacity of drug metabolism as humans, suggesting that they could be a useful animal model for drug development.


Subject(s)
Cytochrome P-450 Enzyme System/biosynthesis , Liver/enzymology , Alleles , Animals , Cell Transplantation , Child , Cytochrome P-450 Enzyme System/genetics , Electrophoresis, Polyacrylamide Gel , Female , Genotype , Hepatocytes/physiology , Humans , Immunoblotting , Infant , Isoenzymes/biosynthesis , Isoenzymes/genetics , Male , Mice , Mice, Transgenic , Microsomes, Liver/drug effects , Microsomes, Liver/enzymology , RNA/biosynthesis , RNA/isolation & purification , Serum Albumin/biosynthesis , Serum Albumin/genetics
17.
Am J Pathol ; 165(3): 901-12, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15331414

ABSTRACT

Human hepatocytes were transplanted into urokinase-type plasminogen activator-transgenic SCID mice (uPA/SCID mice), which are immunodeficient and undergo liver failure. The transplanted cells were characterized in terms of their in vivo growth potential and functions. The human hepatocytes progressively repopulated the murine host liver. However, the recipients died when the replacement index (RI) of the human hepatocytes exceeded 50%. The hosts (chimeric mice) survived at RI >50% when treated with a drug that has anti-human complement factor activity, and these mice developed livers with RI values as high as 96%. In total, 36 chimeric mice were generated, and the rate of successful engraftment was as high as 92%. The yield of chimeric mice with RI >70% was 32%. The human hepatocytes in the murine host liver expressed mRNAs for a variety of human cytochrome P450 (hCYP) subtypes, in a manner that was similar to the donor liver. The mRNAs for hCYP3A4 and hCYP1A1/2 were induced in the liver in a CYP type-specific manner when the mice were treated with rifampicin and 3-methylcholanthrene, respectively. These results indicate that human hepatocytes that propagate in mice retain their normal pharmacological responses. We conclude that the chimeric mouse developed in the present study is a useful model for assessing the functions and pharmacological responses of human hepatocytes.


Subject(s)
Cytochrome P-450 Enzyme System/metabolism , Hepatocytes/transplantation , Liver Failure , Liver/pathology , Transplantation, Heterologous , Urokinase-Type Plasminogen Activator/genetics , Adolescent , Adult , Albumins/metabolism , Animals , Child , Chimera , Complement C3-C5 Convertases/antagonists & inhibitors , Complement C3a/metabolism , Cytochrome P-450 Enzyme System/genetics , Enzyme Induction/drug effects , Enzyme Inhibitors/pharmacology , Hepatocytes/cytology , Hepatocytes/physiology , Heterozygote , Homozygote , Humans , In Situ Hybridization , Liver/metabolism , Liver Failure/metabolism , Liver Failure/pathology , Male , Methylcholanthrene/pharmacology , Mice , Mice, SCID , Mice, Transgenic , Middle Aged , RNA, Messenger/analysis , Rifampin/pharmacology , Urokinase-Type Plasminogen Activator/metabolism
18.
Development ; 129(22): 5205-16, 2002 Nov.
Article in English | MEDLINE | ID: mdl-12399312

ABSTRACT

Signals from micromere descendants play a crucial role in sea urchin development. In this study, we demonstrate that these micromere descendants express HpTb, a T-brain homolog of Hemicentrotus pulcherrimus. HpTb is expressed transiently from the hatched blastula stage through the mesenchyme blastula stage to the gastrula stage. By a combination of embryo microsurgery and antisense morpholino experiments, we show that HpTb is involved in the production of archenteron induction signals. However, HpTb is not involved in the production of signals responsible for the specification of secondary mesenchyme cells, the initial specification of primary mesenchyme cells, or the specification of endoderm. HpTb expression is controlled by nuclear localization of beta-catenin, suggesting that HpTb is in a downstream component of the Wnt signaling cascade. We also propose the possibility that HpTb is involved in the cascade responsible for the production of signals required for the spicule formation as well as signals from the vegetal hemisphere required for the differentiation of aboral ectoderm.


Subject(s)
Sea Urchins/embryology , T-Box Domain Proteins/genetics , T-Box Domain Proteins/metabolism , Amino Acid Sequence , Animals , Arsenite Transporting ATPases , Base Sequence , Blastula/cytology , Blastula/metabolism , Cell Nucleus/metabolism , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/metabolism , Ectoderm , Embryo, Nonmammalian , Embryonic Induction , Gastrula , Gene Expression Regulation, Developmental , Ion Pumps/genetics , Ion Pumps/metabolism , Mice , Molecular Sequence Data , Multienzyme Complexes/genetics , Multienzyme Complexes/metabolism , Protein Biosynthesis , Sea Urchins/genetics , Sequence Homology, Amino Acid , Signal Transduction , Trans-Activators/genetics , Trans-Activators/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , beta Catenin
19.
Am J Pathol ; 160(6): 2191-205, 2002 Jun.
Article in English | MEDLINE | ID: mdl-12057922

ABSTRACT

Previously pleiotrophin (PTN) was identified among proteins secreted by Swiss 3T3 cells as a mitogen for cultured adult rat hepatocytes. The present study showed that the growth of rat hepatocytes was enhanced when cultured with rat hepatic stellate cells (HSCs). HSCs expressed PTN mRNA and secreted its protein in the co-cultures. Recombinant PTN enhanced the growth of hepatocytes in culture, suggesting that HSCs stimulate the growth of hepatocytes through the action of PTN. To know the biological role of PTN in the growth of hepatocytes in vivo, we examined the expression of PTN in four regeneration models of adult liver and embryonic liver of rat. The expression of PTN mRNA in the liver was markedly up-regulated by the treatment with D-galactosamine (GalN) or with acetylaminofluorene followed by partial hepatectomy. HSCs expressed PTN mRNA in response to GalN treatment and its protein was found on hepatocytes. The mRNA expression of N-syndecan, a PTN receptor, was up-regulated in GalN-treated hepatocytes. The mesenchymal cells in the septum transversum enclosing the embryonic liver, but not embryonic HSCs, expressed PTN mRNA. We suggest that PTN is secreted from activated adult HSCs and embryonic mesenchymal cells as a mitogen of parenchymal cells in adult and embryonic liver, respectively.


Subject(s)
Carrier Proteins/physiology , Cytokines/physiology , Liver Regeneration/physiology , Liver/growth & development , 2-Acetylaminofluorene/pharmacology , 3T3 Cells , Animals , Cells, Cultured , Coculture Techniques , Galactosamine/pharmacology , Hepatectomy , Immunohistochemistry , In Situ Hybridization , Liver/drug effects , Liver/embryology , Liver Regeneration/drug effects , Male , Membrane Glycoproteins/biosynthesis , Membrane Glycoproteins/genetics , Mice , Proteoglycans/biosynthesis , Proteoglycans/genetics , RNA, Messenger/metabolism , Rats , Rats, Inbred F344 , Recombinant Proteins/metabolism , Syndecan-3 , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...