1
|
Li P, Miyamoto D, Huang Y, Adachi T, Hidaka M, Hara T, Soyama A, Matsushima H, Imamura H, Kanetaka K, Gu W, Eguchi S. Three-dimensional human bile duct formation from chemically induced human liver progenitor cells. Front Bioeng Biotechnol 2023; 11:1249769. [PMID: 37671190 PMCID: PMC10475568 DOI: 10.3389/fbioe.2023.1249769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 08/10/2023] [Indexed: 09/07/2023] Open
Abstract
Background: The intrahepatic bile ducts (BDs) play an important role in the modification and transport of bile, and the integration between the BD and hepatocytes is the basis of the liver function. However, the lack of a source of cholangiocytes limits in vitro research. The aim of the present study was to establish three-dimensional BDs combined with human mature hepatocytes (hMHs) in vitro using chemically induced human liver progenitor cells (hCLiPs) derived from hMHs. Methods: In this study, we formed functional BDs from hCLiPs using hepatocyte growth factor and extracellular matrix. BDs expressed the typical biliary markers CK-7, GGT1, CFTR and EpCAM and were able to transport the bile-like substance rhodamine 123 into the lumen. The established three-dimensional BDs were cocultured with hMHs. These cells were able to bind to the BDs, and the bile acid analog CLF was transported from the culture medium through the hMHs and accumulated in the lumen of the BDs. The BDs generated from the hCLiPs showed a BD function and a physiological system (e.g., the transport of bile within the liver) when they were connected to the hMHs. Conclusion: We present a novel in vitro three-dimensional BD combined with hMHs for study, drug screening and the therapeutic modulation of the cholangiocyte function.
Collapse
Affiliation(s)
- Peilin Li
- Department of Surgery, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
- Department of Surgery, Guangzhou First People’s Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
| | - Daisuke Miyamoto
- Department of Surgery, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Yu Huang
- Department of Surgery, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
- Department of Surgery, Guangzhou First People’s Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
| | - Tomohiko Adachi
- Department of Surgery, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Masaaki Hidaka
- Department of Surgery, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Takanobu Hara
- Department of Surgery, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Akihiko Soyama
- Department of Surgery, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Hajime Matsushima
- Department of Surgery, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Hajime Imamura
- Department of Surgery, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Kengo Kanetaka
- Department of Surgery, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Weili Gu
- Department of Surgery, Guangzhou First People’s Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
| | - Susumu Eguchi
- Department of Surgery, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| |
Collapse
|
2
|
Maekawa K, Natsuda K, Hidaka M, Uematsu M, Soyama A, Hara T, Takatsuki M, Nagai K, Miura K, Eguchi S. Long-term culture of rat hepatocytes using human amniotic membrane as a culture substrate. Regen Ther 2021; 18:384-390. [PMID: 34660855 PMCID: PMC8488178 DOI: 10.1016/j.reth.2021.09.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 08/28/2021] [Accepted: 09/12/2021] [Indexed: 11/17/2022] Open
Abstract
Amniotic membrane is attracting attention as a new material for regenerative medicine. We herein report that the culture of primary rat hepatocytes on human amniotic membrane maintained their morphology and their production of albumin for at least two months. Human amniotic membrane was collected during planned cesarean section and kept frozen until usage. Primary rat hepatocytes were plated on human amniotic membrane. Hepatocytes accumulated as colonies on amniotic membrane, and their rat albumin level was maintained for two months. Their three-dimensional structure on extracellular matrix, which is abundant in amniotic membranes might influence the maintenance of the hepatocyte-specific function. Long-term primary culture of rat hepatocyte on the human amniotic membrane was successful. Albumin production from primary isolated hepatocytes was maintained for the long term. Amniotic membrane provided the situation of 3D structure for isolated rat hepatocyte.
Collapse
Key Words
- AM, amniotic membrane
- Albumin synthesis
- DMSO, dimethyl sulfoxide
- EGF, epidermal growth factor
- ELISA, enzyme-linked immunosorbent assay
- FBS, fetal bovine serum
- HBV, hepatitis-B virus
- HCV, hepatitis-C virus
- HGF, hepatocyte growth factor
- HIV, human immunodeficiency virus
- HTLV-1, human T-cell leukemia virus type 1
- Human amniotic membrane
- LT, liver transplantation
- PBS, phosphate-buffered saline
- Rat hepatocyte
Collapse
Affiliation(s)
- Kyoichiro Maekawa
- Department of Surgery, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Koji Natsuda
- Department of Surgery, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Masaaki Hidaka
- Department of Surgery, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Masafumi Uematsu
- Department of Ophthalmology and Visual Sciences, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Akihiko Soyama
- Department of Surgery, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Takanobu Hara
- Department of Surgery, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Mitsuhisa Takatsuki
- Department of Digestive and General Surgery, Graduate School of Medicine, University of the Ryukyus, Okinawa, Japan
| | - Kazuhiro Nagai
- Transfusion and Cell Therapy Unit, Nagasaki University Hospital, Nagasaki, Japan
| | - Kiyonori Miura
- Department of Obstetrics and Gynecology, Nagasaki University Graduate School of BioMedical Sciences, Nagasaki, Japan
| | - Susumu Eguchi
- Department of Surgery, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| |
Collapse
|
3
|
Anzai K, Tsuruya K, Ida K, Kagawa T, Inagaki Y, Kamiya A. Kruppel-like factor 15 induces the development of mature hepatocyte-like cells from hepatoblasts. Sci Rep 2021; 11:18551. [PMID: 34535735 PMCID: PMC8448749 DOI: 10.1038/s41598-021-97937-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Accepted: 09/01/2021] [Indexed: 11/08/2022] Open
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.
Collapse
Affiliation(s)
- Kazuya Anzai
- Department of Molecular Life Sciences, Tokai University School of Medicine, 143 Shimokasuya, Isehara, Kanagawa, 259-1193, Japan
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Tokai University School of Medicine, 143 Shimokasuya, Isehara, Kanagawa, 259-1193, Japan
| | - Kota Tsuruya
- Department of Molecular Life Sciences, Tokai University School of Medicine, 143 Shimokasuya, Isehara, Kanagawa, 259-1193, Japan
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Tokai University School of Medicine, 143 Shimokasuya, Isehara, Kanagawa, 259-1193, Japan
| | - Kinuyo Ida
- Department of Molecular Life Sciences, Tokai University School of Medicine, 143 Shimokasuya, Isehara, Kanagawa, 259-1193, Japan
- Center for Matrix Biology and Medicine, Graduate School of Medicine, Tokai University, 143 Shimokasuya, Isehara, Kanagawa, 259-1193, Japan
| | - Tatehiro Kagawa
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Tokai University School of Medicine, 143 Shimokasuya, Isehara, Kanagawa, 259-1193, Japan
| | - Yutaka Inagaki
- Center for Matrix Biology and Medicine, Graduate School of Medicine, Tokai University, 143 Shimokasuya, Isehara, Kanagawa, 259-1193, Japan
- Department of Innovative Medical Science, Tokai University School of Medicine, 143 Shimokasuya, Isehara, Kanagawa, 259-1193, Japan
| | - Akihide Kamiya
- Department of Molecular Life Sciences, Tokai University School of Medicine, 143 Shimokasuya, Isehara, Kanagawa, 259-1193, Japan.
- Center for Matrix Biology and Medicine, Graduate School of Medicine, Tokai University, 143 Shimokasuya, Isehara, Kanagawa, 259-1193, Japan.
| |
Collapse
|
4
|
Rizki-Safitri A, Shinohara M, Tanaka M, Sakai Y. Tubular bile duct structure mimicking bile duct morphogenesis for prospective in vitro liver metabolite recovery. J Biol Eng 2020; 14:11. [PMID: 32206088 PMCID: PMC7081557 DOI: 10.1186/s13036-020-0230-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Accepted: 02/13/2020] [Indexed: 12/13/2022] Open
Abstract
Background Liver metabolites are used to diagnose disease and examine drugs in clinical pharmacokinetics. Therefore, development of an in vitro assay system that reproduces liver metabolite recovery would provide important benefits to pharmaceutical research. However, liver models have proven challenging to develop because of the lack of an appropriate bile duct structure for the accumulation and transport of metabolites from the liver parenchyma. Currently available bile duct models, such as the bile duct cyst-embedded extracellular matrix (ECM), lack any morphological resemblance to the tubular morphology of the living bile duct. Moreover, these systems cannot overcome metabolite recovery issues because they are established in isolated culture systems. Here, we successfully established a non-continuous tubular bile duct structure model in an open-culture system, which closely resembled an in vivo structure. This system was utilized to effectively collect liver metabolites separately from liver parenchymal cells. Results Triple-cell co-culture of primary rat hepatoblasts, rat biliary epithelial cells, and mouse embryonic fibroblasts was grown to mimic the morphogenesis of the bile duct during liver development. Overlaying the cells with ECM containing a Matrigel and collagen type I gel mixture promoted the development of a tubular bile duct structure. In this culture system, the expression of specific markers and signaling molecules related to biliary epithelial cell differentiation was highly upregulated during the ductal formation process. This bile duct structure also enabled the separate accumulation of metabolite analogs from liver parenchymal cells. Conclusions A morphogenesis-based culture system effectively establishes an advanced bile duct structure and improves the plasticity of liver models feasible for autologous in vitro metabolite-bile collection, which may enhance the performance of high-throughput liver models in cell-based assays.
Collapse
Affiliation(s)
- Astia Rizki-Safitri
- 1Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, Tokyo, Japan.,2Center for International Research on Integrative Biomedical Systems (CIBiS), Institute of Industrial Science (IIS), The University of Tokyo, Tokyo, Japan
| | - Marie Shinohara
- 2Center for International Research on Integrative Biomedical Systems (CIBiS), Institute of Industrial Science (IIS), The University of Tokyo, Tokyo, Japan.,3Department of Chemical System Engineering, Graduate School of Engineering, The University of Tokyo, Tokyo, Japan
| | - Minoru Tanaka
- 4Laboratory of Stem Cell Regulation, Institute for Quantitative Biosciences (IQB), The University of Tokyo, Tokyo, Japan.,5Department of Regenerative Medicine, Research Institute, National Center for Global Health and Medicine (NCGM), Tokyo, Japan
| | - Yasuyuki Sakai
- 1Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, Tokyo, Japan.,2Center for International Research on Integrative Biomedical Systems (CIBiS), Institute of Industrial Science (IIS), The University of Tokyo, Tokyo, Japan.,3Department of Chemical System Engineering, Graduate School of Engineering, The University of Tokyo, Tokyo, Japan.,6Max Planck-The University of Tokyo, Center for Integrative Inflammology, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
5
|
Sato A, Kakinuma S, Miyoshi M, Kamiya A, Tsunoda T, Kaneko S, Tsuchiya J, Shimizu T, Takeichi E, Nitta S, Kawai-Kitahata F, Murakawa M, Itsui Y, Nakagawa M, Azuma S, Koshikawa N, Seiki M, Nakauchi H, Asahina Y, Watanabe M. Vasoactive Intestinal Peptide Derived From Liver Mesenchymal Cells Mediates Tight Junction Assembly in Mouse Intrahepatic Bile Ducts. Hepatol Commun 2019; 4:235-254. [PMID: 32025608 PMCID: PMC6996346 DOI: 10.1002/hep4.1459] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Accepted: 11/23/2019] [Indexed: 01/20/2023] Open
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.
Collapse
Affiliation(s)
- Ayako Sato
- Department of Gastroenterology and Hepatology Tokyo Medical and Dental University Tokyo Japan
| | - Sei Kakinuma
- Department of Gastroenterology and Hepatology Tokyo Medical and Dental University Tokyo Japan.,Department of Liver Disease Control Tokyo Medical and Dental University (TMDU) Tokyo Japan
| | - Masato Miyoshi
- Department of Gastroenterology and Hepatology Tokyo Medical and Dental University Tokyo Japan
| | - Akihide Kamiya
- Department of Molecular Life Sciences School of Medicine Tokai University Isehara Japan
| | - Tomoyuki Tsunoda
- Department of Gastroenterology and Hepatology Tokyo Medical and Dental University Tokyo Japan
| | - Shun Kaneko
- Department of Gastroenterology and Hepatology Tokyo Medical and Dental University Tokyo Japan
| | - Jun Tsuchiya
- Department of Gastroenterology and Hepatology Tokyo Medical and Dental University Tokyo Japan
| | - Taro Shimizu
- Department of Gastroenterology and Hepatology Tokyo Medical and Dental University Tokyo Japan
| | - Eiko Takeichi
- Department of Gastroenterology and Hepatology Tokyo Medical and Dental University Tokyo Japan
| | - Sayuri Nitta
- Department of Gastroenterology and Hepatology Tokyo Medical and Dental University Tokyo Japan
| | - Fukiko Kawai-Kitahata
- Department of Gastroenterology and Hepatology Tokyo Medical and Dental University Tokyo Japan
| | - Miyako Murakawa
- Department of Gastroenterology and Hepatology Tokyo Medical and Dental University Tokyo Japan
| | - Yasuhiro Itsui
- Department of Gastroenterology and Hepatology Tokyo Medical and Dental University Tokyo Japan
| | - Mina Nakagawa
- Department of Gastroenterology and Hepatology Tokyo Medical and Dental University Tokyo Japan
| | - Seishin Azuma
- Department of Gastroenterology and Hepatology Tokyo Medical and Dental University Tokyo Japan
| | - Naohiko Koshikawa
- Division of Cancer Cell Research Institute of Medical Science University of Tokyo Tokyo Japan
| | - Motoharu Seiki
- Division of Cancer Cell Research Institute of Medical Science University of Tokyo Tokyo Japan
| | - Hiromitsu Nakauchi
- Institute for Stem Cell Biology and Regenerative Medicine Stanford University School of Medicine Stanford CA.,Division of Stem Cell Therapy Institute of Medical Science University of Tokyo Tokyo Japan
| | - Yasuhiro Asahina
- Department of Gastroenterology and Hepatology Tokyo Medical and Dental University Tokyo Japan.,Department of Liver Disease Control Tokyo Medical and Dental University (TMDU) Tokyo Japan
| | - Mamoru Watanabe
- Department of Gastroenterology and Hepatology Tokyo Medical and Dental University Tokyo Japan.,Advanced Research Institute Tokyo Medical and Dental University (TMDU) Tokyo Japan
| |
Collapse
|
6
|
Development of Bifunctional Three-Dimensional Cysts from Chemically Induced Liver Progenitors. Stem Cells Int 2019; 2019:3975689. [PMID: 31565060 PMCID: PMC6745155 DOI: 10.1155/2019/3975689] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Revised: 07/20/2019] [Accepted: 07/30/2019] [Indexed: 12/12/2022] Open
Abstract
Chemically induced liver progenitors (CLiPs) have promising applications in liver regenerative medicine. Three-dimensional (3D) structures generated from liver progenitor cells possess wide applications in cell transplantation, disease model, and drug testing. Here, we report on the spontaneous formation of 3D cystic structures comprising maturing rat CLiPs on gelatin-coated dishes. Our 3D cysts contained Alb+/+CK19+/− and Ck19+/+Alb+/− cells. These cell types gradually diverged into specialized mature cells, as demonstrated by the expression of mature biliary markers (Cftr, Ae2, and Aqp1) and hepatic markers (Alb and Mrp2). The 3D cysts also expressed functional multidrug resistance protein 1 (Mdr1), as indicated by epithelial efflux of rhodamine. Furthermore, we observed bile canaliculi functions between hepatocytes and cholyl-lysyl-fluorescein extrusions, indicating that the functional characteristics of 3D cysts and active bile salt export pump (Bsep) transporters were intact. Thus, our study revealed a natural characteristic of rat CLiPs to spontaneously form 3D cystic structures accompanied with cell maturation in vitro, offering a platform for studies of liver development and drug screening.
Collapse
|
7
|
Zhou T, Wang W, Aimaiti Y, Jin X, Chen Z, Chen L, Li D. Direct and indirect coculture of mouse hepatic progenitor cells with mouse embryonic fibroblasts for the generation of hepatocytes and cholangiocytes. Cytotechnology 2019; 71:267-275. [PMID: 30603925 DOI: 10.1007/s10616-018-0282-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Accepted: 11/14/2018] [Indexed: 12/12/2022] Open
Abstract
The widespread use of hepatocytes and cholangiocytes for regenerative medicine and tissue engineering is restricted by the limited number of hepatocytes and cholangiocytes; a simple and effective method for the expansion and differentiation of the hepatic progenitor cells (HPCs) is required. Recent studies demonstrated that mouse embryonic fibroblasts (MEFs) play an important role in supporting the proliferation of the mouse hepatic progenitor cells (mHPCs). However, the effect of direct and indirect coculture of MEFs with mHPCs on the differentiation of mHPCs is poorly studied. Herein, we show that mHPCs rapidly proliferate and form colonies in direct or indirect contact coculture with MEFs in the serum-free medium. Importantly, after direct contact coculture of the mHPCs with MEFs for 6 days, mHPCs expressed the hepatic marker albumin (ALB) and did not express the cholangiocyte marker CK19, indicating their differentiation into hepatocytes. In contrast, after indirect contact coculture of the mHPCs with MEFs for 6 days, mHPCs expressed the cholangiocyte marker CK19 and did not express the hepatic marker ALB, indicating their differentiation into cholangiocytes. These results indicate that direct and indirect contact cocultures of the mHPCs with MEFs are useful for rapidly producing hepatocytes and cholangiocytes.
Collapse
Affiliation(s)
- Tao Zhou
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Wei Wang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Yasen Aimaiti
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Xin Jin
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Zhixin Chen
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Liang Chen
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Dewei Li
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, People's Republic of China.
| |
Collapse
|
8
|
Kamiya A, Chikada H, Ida K, Ando E, Tsuruya K, Kagawa T, Inagaki Y. An in vitro model of polycystic liver disease using genome-edited human inducible pluripotent stem cells. Stem Cell Res 2018; 32:17-24. [PMID: 30172093 DOI: 10.1016/j.scr.2018.08.018] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Revised: 08/14/2018] [Accepted: 08/22/2018] [Indexed: 01/23/2023] Open
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.
Collapse
Affiliation(s)
- Akihide Kamiya
- Department of Molecular Life Sciences, Tokai University School of Medicine, 143 Shimokasuya, Isehara, Kanagawa, Japan; Center for Matrix Biology and Medicine, Graduate School of Medicine, Tokai University, 143 Shimokasuya, Isehara, Kanagawa 259-1193, Japan.
| | - Hiromi Chikada
- Department of Molecular Life Sciences, Tokai University School of Medicine, 143 Shimokasuya, Isehara, Kanagawa, Japan; Center for Matrix Biology and Medicine, Graduate School of Medicine, Tokai University, 143 Shimokasuya, Isehara, Kanagawa 259-1193, Japan
| | - Kinuyo Ida
- Department of Molecular Life Sciences, Tokai University School of Medicine, 143 Shimokasuya, Isehara, Kanagawa, Japan
| | - Emi Ando
- Department of Molecular Life Sciences, Tokai University School of Medicine, 143 Shimokasuya, Isehara, Kanagawa, Japan
| | - Kota Tsuruya
- Department of Molecular Life Sciences, Tokai University School of Medicine, 143 Shimokasuya, Isehara, Kanagawa, Japan; Division of Gastroenterology, Department of Internal Medicine, Tokai University School of Medicine, 143 Shimokasuya, Isehara, Kanagawa 259-1193, Japan
| | - Tatehiro Kagawa
- Division of Gastroenterology, Department of Internal Medicine, Tokai University School of Medicine, 143 Shimokasuya, Isehara, Kanagawa 259-1193, Japan
| | - Yutaka Inagaki
- Center for Matrix Biology and Medicine, Graduate School of Medicine, Tokai University, 143 Shimokasuya, Isehara, Kanagawa 259-1193, Japan; Department of Regenerative Medicine, Tokai University School of Medicine, 143 Shimokasuya, Isehara, Kanagawa 259-1193, Japan
| |
Collapse
|
9
|
Efficient functional cyst formation of biliary epithelial cells using microwells for potential bile duct organisation in vitro. Sci Rep 2018; 8:11086. [PMID: 30038407 PMCID: PMC6056467 DOI: 10.1038/s41598-018-29464-w] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Accepted: 07/11/2018] [Indexed: 12/11/2022] Open
Abstract
Establishing a bile duct in vitro is valuable to obtain relevant hepatic tissue culture systems for cell-based assays in chemical and drug metabolism analyses. The cyst constitutes the initial morphogenesis for bile duct formation from biliary epithelial cells (BECs) and serves the main building block of bile duct network morphogenesis from the ductal plate during embryogenesis in rodents. Cysts have been commonly cultured via Matrigel-embedded culture, which does not allow structural organisation and restricts the productivity and homogeneity of cysts. In this study, we propose a new method utilising oxygen permeable honeycomb microwells for efficient cyst establishment. Primary mouse BECs were seeded on four sizes of honeycomb microwell (46, 76, 126, and 326 µm-size in diameter). Matrigel in various concentrations was added to assist in cyst formation. The dimension accommodated by microwells was shown to play an important role in effective cyst formation. Cytological morphology, bile acid transportation, and gene expression of the cysts confirmed the favourable basic bile duct function compared to that obtained using Matrigel-embedded culture. Our method is expected to contribute to engineered in vitro liver tissue formation for cell-based assays.
Collapse
|
10
|
Loarca L, De Assuncao TM, Jalan-Sakrikar N, Bronk S, Krishan A, Huang B, Morton L, Trussoni C, Bonilla LM, Krueger E, O’Hara S, Splinter P, Shi G, Pisarello MJL, Gores GJ, Huebert RC, LaRusso NF. Development and characterization of cholangioids from normal and diseased human cholangiocytes as an in vitro model to study primary sclerosing cholangitis. J Transl Med 2017; 97:1385-1396. [PMID: 28892096 PMCID: PMC5664217 DOI: 10.1038/labinvest.2017.63] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Revised: 05/18/2017] [Accepted: 05/23/2017] [Indexed: 12/12/2022] Open
Abstract
Primary sclerosing cholangitis (PSC) is an incurable, fibroinflammatory biliary disease for which there is no effective pharmacotherapy. We recently reported cholangiocyte senescence as an important phenotype in PSC while others showed that portal macrophages accumulate in PSC. Unfortunately, our ability to explore cholangiocyte senescence and macrophage accumulation has been hampered by limited in vitro models. Thus, our aim was to develop and characterize a three-dimensional (3D) model of normal and diseased bile ducts (cholangioids) starting with normal human cholangiocytes (NHC), senescent NHC (NHC-sen), and cholangiocytes from PSC patients. In 3D culture, NHCs formed spheroids of ~5000 cells with a central lumen of ~150 μm. By confocal microscopy and western blot, cholangioids retained expression of cholangiocyte proteins (cytokeratin 7/19) and markers of epithelial polarity (secretin receptor and GM130). Cholangioids are functionally active, and upon secretin stimulation, luminal size increased by ~80%. Cholangioids exposed to hydrogen peroxide exhibited cellular senescence and the senescence-associated secretory phenotype (SASP; increased IL-6, p21, SA-β-Gal, yH2A.x and p16 expression). Furthermore, cholangioids derived from NHC-sen or PSC patients were smaller and had slower growth than the controls. When co-cultured with THP-1 macrophages, the number of macrophages associated with NHC-sen or PSC cholangioids was five- to seven-fold greater compared to co-culture with non-senescent NHC. We observed that NHC-sen and PSC cholangioids release greater number of extracellular vesicles (EVs) compared to controls. Moreover, conditioned media from NHC-sen cholangioids resulted in an ~2-fold increase in macrophage migration. In summary, we developed a method to generate normal and diseased cholangioids, characterized them morphologically and functionally, showed that they can be induced to senescence and SASP, and demonstrated both EV release and macrophage attraction. This novel model mimics several features of PSC, and thus will be useful for studying the pathogenesis of PSC and potentially identifying new therapeutic targets.
Collapse
MESH Headings
- Autoantigens/metabolism
- Bile Ducts/drug effects
- Bile Ducts/metabolism
- Bile Ducts/pathology
- Bile Ducts/ultrastructure
- Biomarkers/metabolism
- Cell Line
- Cells, Cultured
- Cellular Senescence/drug effects
- Cholangitis, Sclerosing/immunology
- Cholangitis, Sclerosing/metabolism
- Cholangitis, Sclerosing/pathology
- Coculture Techniques
- Culture Media, Conditioned
- Extracellular Vesicles/drug effects
- Extracellular Vesicles/metabolism
- Extracellular Vesicles/pathology
- Extracellular Vesicles/ultrastructure
- Gene Expression Regulation/drug effects
- Humans
- Hydrogen Peroxide/toxicity
- Keratin-19/metabolism
- Keratin-7/metabolism
- Macrophage Activation
- Macrophages/cytology
- Macrophages/immunology
- Membrane Proteins/metabolism
- Microscopy, Electron, Transmission
- Multivesicular Bodies/drug effects
- Multivesicular Bodies/metabolism
- Multivesicular Bodies/pathology
- Multivesicular Bodies/ultrastructure
- Oxidants/toxicity
- Receptors, G-Protein-Coupled/metabolism
- Receptors, Gastrointestinal Hormone/metabolism
- Spheroids, Cellular/drug effects
- Spheroids, Cellular/metabolism
- Spheroids, Cellular/pathology
- Spheroids, Cellular/ultrastructure
Collapse
Affiliation(s)
- Lorena Loarca
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN 55905
| | | | | | - Steve Bronk
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN 55905
| | - Anuradha Krishan
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN 55905
| | - Bing Huang
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN 55905
| | | | - Christy Trussoni
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN 55905
| | | | - Eugene Krueger
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN 55905
| | - Steve O’Hara
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN 55905
| | - Patrick Splinter
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN 55905
| | - Guang Shi
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN 55905
| | | | - Gregory J. Gores
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN 55905
| | - Robert C. Huebert
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN 55905
| | - Nicholas F. LaRusso
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN 55905
| |
Collapse
|
11
|
Nakano Y, Nakao S, Sumiyoshi H, Mikami K, Tanno Y, Sueoka M, Kasahara D, Kimura H, Moro T, Kamiya A, Hozumi K, Inagaki Y. Identification of a novel alpha-fetoprotein-expressing cell population induced by the Jagged1/Notch2 signal in murine fibrotic liver. Hepatol Commun 2017; 1:215-229. [PMID: 29404455 PMCID: PMC5721449 DOI: 10.1002/hep4.1026] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Revised: 02/16/2017] [Accepted: 02/21/2017] [Indexed: 12/17/2022] Open
Abstract
The liver is well known to possess high regenerative capacity in response to partial resection or tissue injury. However, liver regeneration is often impaired in the case of advanced liver fibrosis/cirrhosis when mature hepatocytes can hardly self‐proliferate. Hepatic progenitor cells have been implicated as a source of hepatocytes in regeneration of the fibrotic liver. Although alpha‐fetoprotein (AFP) is known as a clinical marker of progenitor cell induction in injured/fibrotic adult liver, the origin and features of such AFP‐producing cells are not fully understood. Here, we demonstrate a unique and distinct AFP‐expressing cell population that is induced by the Jagged1/Notch2 signal in murine fibrotic liver. Following repeated carbon tetrachloride injections, a significant number of AFP‐positive cells with high proliferative ability were observed along the fibrous septa depending on the extent of liver fibrosis. These AFP‐positive cells exhibited features of immature hepatocytes that were stained positively for hepatocyte‐lineage markers, such as albumin and hepatocyte nuclear factor 4 alpha, and a stem/progenitor cell marker Sox9. A combination of immunohistological examination of fibrotic liver tissues and coculture experiments with primary hepatocytes and hepatic stellate cells indicated that increased Jagged1 expression in activated hepatic stellate cells stimulated Notch2 signaling and up‐regulated AFP expression in adjacent hepatocytes. The mobilization and proliferation of AFP‐positive cells in fibrotic liver were further enhanced after partial hepatectomy, which was significantly suppressed in Jagged1‐conditional knockout mice. Finally, forced expression of the intracellular domain of Notch2 in normal liver induced a small number of AFP‐expressing hepatocytes in vivo. Conclusion: Insight is provided into a novel pathophysiological role of Jagged1/Notch2 signaling in the induction of AFP‐positive cells in fibrotic liver through the interaction between hepatocytes and activated hepatic stellate cells. (Hepatology Communications 2017;1:215‐229)
Collapse
Affiliation(s)
- Yasuhiro Nakano
- Center for Matrix Biology and Medicine Graduate School of Medicine, Tokai University Isehara Japan.,Department of Regenerative Medicine, Tokai University School of Medicine Isehara Japan
| | - Sachie Nakao
- Center for Matrix Biology and Medicine Graduate School of Medicine, Tokai University Isehara Japan.,Department of Regenerative Medicine, Tokai University School of Medicine Isehara Japan
| | - Hideaki Sumiyoshi
- Center for Matrix Biology and Medicine Graduate School of Medicine, Tokai University Isehara Japan.,Department of Regenerative Medicine, Tokai University School of Medicine Isehara Japan
| | - Kenichiro Mikami
- Department of Regenerative Medicine, Tokai University School of Medicine Isehara Japan.,Present address: Present address for Kenichiro Mikami is Department of Gastroenterology and Hematology Hirosaki University Graduate School of Medicine Hirosaki Japan
| | - Yuri Tanno
- Department of Regenerative Medicine, Tokai University School of Medicine Isehara Japan
| | - Minako Sueoka
- Department of Regenerative Medicine, Tokai University School of Medicine Isehara Japan
| | - Daigo Kasahara
- Center for Matrix Biology and Medicine Graduate School of Medicine, Tokai University Isehara Japan.,Department of Mechanical Engineering Tokai University School of Engineering Hiratsuka Japan
| | - Hiroshi Kimura
- Department of Mechanical Engineering Tokai University School of Engineering Hiratsuka Japan
| | - Tadashi Moro
- Center for Matrix Biology and Medicine Graduate School of Medicine, Tokai University Isehara Japan.,Research Laboratory, Minophagen Pharmaceutical Co., Ltd Zama Japan
| | - Akihide Kamiya
- Center for Matrix Biology and Medicine Graduate School of Medicine, Tokai University Isehara Japan.,Department of Molecular Life Sciences, Tokai University School of Medicine Isehara Japan
| | - Katsuto Hozumi
- Center for Matrix Biology and Medicine Graduate School of Medicine, Tokai University Isehara Japan.,Department of Immunology Tokai University School of Medicine Isehara Japan
| | - Yutaka Inagaki
- Center for Matrix Biology and Medicine Graduate School of Medicine, Tokai University Isehara Japan.,Department of Regenerative Medicine, Tokai University School of Medicine Isehara Japan.,Institute of Medical Sciences Tokai University Isehara Japan
| |
Collapse
|
12
|
Xu W, Wang NR, Wang HF, Feng Q, Deng J, Gong ZQ, Sun J, Lou XL, Yu XF, Zhou L, Hu JP, Huang XF, Qi XQ, Deng YJ, Gong R, Guo Y, Wang MM, Xiao JC, Deng H. Analysis of epithelial-mesenchymal transition markers in the histogenesis of hepatic progenitor cell in HBV-related liver diseases. Diagn Pathol 2016; 11:136. [PMID: 27881141 PMCID: PMC5121942 DOI: 10.1186/s13000-016-0587-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Accepted: 11/16/2016] [Indexed: 12/27/2022] Open
Abstract
Background The origin and heterogeneity of hepatic progenitor cells (HPCs) remain unclear. This study aimed to investigate the involvement of epithelial-mesenchymal transition (EMT) in the histogenesis of HPCs. Methods Surgical liver specimens from patients with HBV-related hepatitis and cirrhosis were investigated with double immunofluorescence labeling to detect antigens associated with HPCs and EMT. Ductular reactions were subjected to quantitative reverse transcription PCR following isolation by laser capture microdissection. Electron microscopic examination was performed to find an ultrastructural evidence of EMT. Results The number of EpCAM-positive HPCs was proportional to the disease severity. The S100A4 expression of HPCs was firstly observed in mild hepatitis and increased significantly in moderate hepatitis, but decreased in severe hepatitis and cirrhosis. The levels of MMP-2, Twist, and Snail increased in direct proportion to the number of HPCs. Some hepatocytes adjacent to portal tracts in cirrhosis showed positivity for MMP-2. Although CK7 and E-cadherin levels decreased in mild and moderate hepatitis, HPCs re-expressed both of them in severe hepatitis and cirrhosis. However, HPCs expressed neither vimentin nor αSMA. The relative mRNA expression levels of EpCAM and EMT-associated markers supported immunohistochemical results. Electron microscopic examination demonstrated the existence of intercellular junctions among HPCs, cholangiocytes, and intermediate hepatocyte-like cells. Conclusion We provided preliminary evidence for the involvement of EMT in the histogenesis of HPCs from cholangiocytes in HBV-related liver diseases. HPCs may re-transdifferentiate into hepatocytes, and the differentiation direction depends, at least in part, on interactions between HPCs and the surrounding microenvironment, especially the non-resolving inflammation caused by HBV infection.
Collapse
Affiliation(s)
- Wei Xu
- Department of General Surgery, Jiangxi Children's Hospital, Nanchang, China
| | - Nong-Rong Wang
- Molecular Medicine and Genetics Center, The Fourth Affiliated Hospital of Nanchang University, Nanchang, China.,Department of Gastroenterology, The Fourth Affiliated Hospital of Nanchang University, Nanchang, China
| | - Hua-Feng Wang
- Department of Pathology, Ruijin Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Qiong Feng
- Department of Pathology, The Second Affiliated Hospital of Nanchang University, Nanchang, China.,Renmin Institute of Forensic Medicine in Jiangxi, Nanchang, China
| | - Jun Deng
- Emergency Department, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Zhi-Qiang Gong
- Renmin Institute of Forensic Medicine in Jiangxi, Nanchang, China
| | - Jian Sun
- Molecular Medicine and Genetics Center, The Fourth Affiliated Hospital of Nanchang University, Nanchang, China
| | - Xiao-Liang Lou
- Molecular Medicine and Genetics Center, The Fourth Affiliated Hospital of Nanchang University, Nanchang, China
| | - Xue-Feng Yu
- Molecular Medicine and Genetics Center, The Fourth Affiliated Hospital of Nanchang University, Nanchang, China
| | - Lv Zhou
- Department of Pathology, The Fourth Affiliated Hospital of Nanchang University, Nanchang, China
| | - Jin-Ping Hu
- Department of Pathology, The Fourth Affiliated Hospital of Nanchang University, Nanchang, China
| | - Xiao-Feng Huang
- Department of Pathology, The Fourth Affiliated Hospital of Nanchang University, Nanchang, China
| | - Xiao-Qing Qi
- Department of Pathology, The Fourth Affiliated Hospital of Nanchang University, Nanchang, China
| | - Yan-Juan Deng
- Department of Pathology, The Fourth Affiliated Hospital of Nanchang University, Nanchang, China
| | - Rui Gong
- Molecular Medicine and Genetics Center, The Fourth Affiliated Hospital of Nanchang University, Nanchang, China
| | - Yan Guo
- Department of Pathology, Ruijin Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Meng-Meng Wang
- Molecular Medicine and Genetics Center, The Fourth Affiliated Hospital of Nanchang University, Nanchang, China
| | - Jia-Cheng Xiao
- Department of Pathology, Ruijin Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China.,Department of Pathology, Tenth People's Hospital of Tongji University, Shanghai, China
| | - Huan Deng
- Molecular Medicine and Genetics Center, The Fourth Affiliated Hospital of Nanchang University, Nanchang, China. .,Renmin Institute of Forensic Medicine in Jiangxi, Nanchang, China. .,Department of Pathology, The Fourth Affiliated Hospital of Nanchang University, Nanchang, China. .,Department of Pathology, The Fourth Affiliated Hospital of Nanchang University, 133 South Guangchang Road, Nanchang, 330006, Jiangxi, China.
| |
Collapse
|