1
|
Igarashi R, Oda M, Okada R, Yano T, Takahashi S, Pastuhov S, Matano M, Masuda N, Togasaki K, Ohta Y, Sato S, Hishiki T, Suematsu M, Itoh M, Fujii M, Sato T. Generation of human adult hepatocyte organoids with metabolic functions. Nature 2025:10.1038/s41586-025-08861-y. [PMID: 40240606 DOI: 10.1038/s41586-025-08861-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 03/04/2025] [Indexed: 04/18/2025]
Abstract
Proliferating hepatocytes often undergo ductal metaplasia to balance the energy trade-off between cellular functions and replication, hindering the expansion of human adult hepatocytes with functional competency1. Here we demonstrate that the combined activation of Wnt and STAT3 signalling enables long-term self-renewal of human adult hepatocyte organoids. YAP activation facilitates hepatocyte proliferation but commits it towards the biliary duct lineage. By contrast, STAT3 activation by oncostatin M induces hepatocyte proliferation while counteracting ductal metaplasia and maintaining the hepatic identity. Xenotransplanted hepatocyte organoids repopulate the recipient mouse liver and reconstitute the metabolic zonation structure. Upon niche factor removal and hormone supplementation, hepatocyte organoids form cord-like structures with bile canalicular networks and exhibit major liver metabolic functions comparable to those of in vivo hepatocytes. Hepatocyte organoids are amenable to gene editing, prompting functional modelling of inherent metabolic liver diseases. The new culture system offers a promising avenue for developing therapeutic strategies against human liver diseases.
Collapse
Affiliation(s)
- Ryo Igarashi
- Department of Organoid Medicine, Sakaguchi Laboratory, Keio University School of Medicine, Tokyo, Japan
- Department of Integrated Medicine and Biochemistry, Keio University School of Medicine, Tokyo, Japan
| | - Mayumi Oda
- Department of Organoid Medicine, Sakaguchi Laboratory, Keio University School of Medicine, Tokyo, Japan
- Department of Integrated Medicine and Biochemistry, Keio University School of Medicine, Tokyo, Japan
| | - Ryo Okada
- JSR-Keio University Medical and Chemical Innovation Center (JKiC), JSR Corporation, Tokyo, Japan
| | - Tomoki Yano
- Department of Organoid Medicine, Sakaguchi Laboratory, Keio University School of Medicine, Tokyo, Japan
- Department of Integrated Medicine and Biochemistry, Keio University School of Medicine, Tokyo, Japan
| | - Sirirat Takahashi
- Department of Organoid Medicine, Sakaguchi Laboratory, Keio University School of Medicine, Tokyo, Japan
- Department of Integrated Medicine and Biochemistry, Keio University School of Medicine, Tokyo, Japan
| | - Strahil Pastuhov
- Department of Organoid Medicine, Sakaguchi Laboratory, Keio University School of Medicine, Tokyo, Japan
- Department of Integrated Medicine and Biochemistry, Keio University School of Medicine, Tokyo, Japan
| | - Mami Matano
- Department of Organoid Medicine, Sakaguchi Laboratory, Keio University School of Medicine, Tokyo, Japan
- Department of Integrated Medicine and Biochemistry, Keio University School of Medicine, Tokyo, Japan
| | - Norio Masuda
- JSR-Keio University Medical and Chemical Innovation Center (JKiC), JSR Corporation, Tokyo, Japan
| | - Kazuhiro Togasaki
- Department of Organoid Medicine, Sakaguchi Laboratory, Keio University School of Medicine, Tokyo, Japan
- Department of Integrated Medicine and Biochemistry, Keio University School of Medicine, Tokyo, Japan
| | - Yuki Ohta
- Department of Organoid Medicine, Sakaguchi Laboratory, Keio University School of Medicine, Tokyo, Japan
- Department of Integrated Medicine and Biochemistry, Keio University School of Medicine, Tokyo, Japan
| | - Saeko Sato
- Department of Organoid Medicine, Sakaguchi Laboratory, Keio University School of Medicine, Tokyo, Japan
- Department of Integrated Medicine and Biochemistry, Keio University School of Medicine, Tokyo, Japan
| | - Takako Hishiki
- Department of Integrated Medicine and Biochemistry, Keio University School of Medicine, Tokyo, Japan
- Department of Biochemistry, Keio University School of Medicine, Tokyo, Japan
| | - Makoto Suematsu
- Department of Integrated Medicine and Biochemistry, Keio University School of Medicine, Tokyo, Japan
- Department of Biochemistry, Keio University School of Medicine, Tokyo, Japan
| | - Manabu Itoh
- JSR-Keio University Medical and Chemical Innovation Center (JKiC), JSR Corporation, Tokyo, Japan
| | - Masayuki Fujii
- Department of Organoid Medicine, Sakaguchi Laboratory, Keio University School of Medicine, Tokyo, Japan
- Department of Integrated Medicine and Biochemistry, Keio University School of Medicine, Tokyo, Japan
| | - Toshiro Sato
- Department of Organoid Medicine, Sakaguchi Laboratory, Keio University School of Medicine, Tokyo, Japan.
- Department of Integrated Medicine and Biochemistry, Keio University School of Medicine, Tokyo, Japan.
| |
Collapse
|
2
|
Guo XL, Wang YD, Liu YJ, Chu L, Zhu H, Hu Y, Wu RY, Xie HY, Yu J, Li SP, Xiong CY, Li RY, Ke F, Chen L, Chen GQ, Chen L, Bai F, Enver T, Li GH, Li HF, Hong DL. Fetal hepatocytes protect the HSPC genome via fetuin-A. Nature 2025; 637:402-411. [PMID: 39633051 PMCID: PMC11711094 DOI: 10.1038/s41586-024-08307-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 10/30/2024] [Indexed: 12/07/2024]
Abstract
The maintenance of genomic integrity in rapidly proliferating cells is a substantial challenge during embryonic development1-3. Although numerous cell-intrinsic mechanisms have been revealed4-7, little is known about genome-protective effects and influences of developmental tissue microenvironments on tissue-forming cells. Here we show that fetal liver hepatocytes provide protection to haematopoietic stem and progenitor cell (HSPC) genomes. Lineage tracing and depletion in mice demonstrated that delayed hepatocyte development in early fetal livers increased the chromosomal instability of newly colonizing HSPCs. In addition, HSPCs developed tolerance to genotoxins in hepatocyte-conditioned medium, suggesting that hepatocytes protect the HSPC genome in a paracrine manner. Proteomic analyses demonstrated the enrichment of fetuin-A in hepatocyte-conditioned medium but not in early fetal livers. Fetuin-A activates a Toll-like receptor pathway to prevent pathogenic R-loop accumulation in HSPCs undergoing DNA replication and gene transcription in the fetal liver. Numerous haematopoietic regulatory genes frequently involved in leukaemogenic mutations are associated with R-loop-enriched regions. In Fetua-knockout mice, HSPCs showed increased genome instability and susceptibility to malignancy induction. Moreover, low concentrations of fetuin-A correlated with the oncogenesis of childhood leukaemia. Therefore, we uncover a mechanism operating in developmental tissues that offers tissue-forming cell genome protection and is implicated in developmental-related diseases.
Collapse
Affiliation(s)
- Xiao-Lin Guo
- Key Laboratory of Cell Differentiation and Apoptosis of Ministry of Education, Department of Pathophysiology, Shanghai Institute of Haematology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yi-Ding Wang
- Key Laboratory of Cell Differentiation and Apoptosis of Ministry of Education, Department of Pathophysiology, Shanghai Institute of Haematology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yan-Jun Liu
- Key Laboratory of Cell Differentiation and Apoptosis of Ministry of Education, Department of Pathophysiology, Shanghai Institute of Haematology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lei Chu
- Department of Obstetrics and Gynecology, Shanghai Tongji Hospital, Tongji University School of Medicine, Shanghai, China
| | - Hua Zhu
- Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ye Hu
- Key Laboratory of Cell Differentiation and Apoptosis of Ministry of Education, Department of Pathophysiology, Shanghai Institute of Haematology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ren-Yan Wu
- Key Laboratory of Cell Differentiation and Apoptosis of Ministry of Education, Department of Pathophysiology, Shanghai Institute of Haematology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hong-Yu Xie
- Key Laboratory of Cell Differentiation and Apoptosis of Ministry of Education, Department of Pathophysiology, Shanghai Institute of Haematology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Juan Yu
- National laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Shui-Ping Li
- Key Laboratory of Cell Differentiation and Apoptosis of Ministry of Education, Department of Pathophysiology, Shanghai Institute of Haematology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chao-Yang Xiong
- National laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Ruo-Yan Li
- Biomedical Pioneering Innovation Centre (BIOPIC) and Translational Cancer Research Centre, School of Life Sciences, First Hospital, Peking University, Beijing, China
| | - Fang Ke
- Key Laboratory of Cell Differentiation and Apoptosis of Ministry of Education, Department of Pathophysiology, Shanghai Institute of Haematology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lei Chen
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Guo-Qiang Chen
- Key Laboratory of Cell Differentiation and Apoptosis of Ministry of Education, Department of Pathophysiology, Shanghai Institute of Haematology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Research Units of Stress and Tumor, Chinese Academy of Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Hainan Academy of Medical Sciences, School of Basic Medicine and Life Science, Hainan Medical University, Haikou, China
| | - Liang Chen
- RNA Institute, Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, China
| | - Fan Bai
- Biomedical Pioneering Innovation Centre (BIOPIC) and Translational Cancer Research Centre, School of Life Sciences, First Hospital, Peking University, Beijing, China
| | - Tariq Enver
- Department of Cancer Biology, UCL Cancer Institute, University College London, London, UK
| | - Guo-Hong Li
- National laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Huai-Fang Li
- Department of Obstetrics and Gynecology, Shanghai Tongji Hospital, Tongji University School of Medicine, Shanghai, China
| | - Deng-Li Hong
- Key Laboratory of Cell Differentiation and Apoptosis of Ministry of Education, Department of Pathophysiology, Shanghai Institute of Haematology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- Research Units of Stress and Tumor, Chinese Academy of Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- Innovative Research Team of High-level Local Universities in Shanghai, Shanghai, China.
- Shanghai Key Laboratory of Reproductive Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
3
|
Sato K. Suppression of gp130 attenuated insulin-mediated signaling and glucose uptake in skeletal muscle cells. Sci Rep 2024; 14:17496. [PMID: 39080385 PMCID: PMC11289081 DOI: 10.1038/s41598-024-68613-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 07/25/2024] [Indexed: 08/02/2024] Open
Abstract
The aim of the present study was to investigate the effects of Oncostatin M receptor (OSMR) subunit gp130 knockdown on insulin-stimulated glucose metabolism-related signaling pathways and glucose uptake in skeletal muscle cells. siRNA-mediated gp130 knockdown was conducted in C2C12 muscle cells, and insulin was added and incubated for 1 h. The cells were cultivated to analyze the mRNA levels of gp130, phosphorylation of STAT3, and glucose metabolism-regulated signaling pathways, and OSM levels in the culture medium were analyzed. The phosphorylation of STAT 3 was significantly decreased in gp130-/- cell. The insulin stimulation was significantly increased in both gp130-/- and gp130+/+ and the phosphorylation of IRS-1 Ser 1101 was significantly decreased in gp130-/-. PI3-kinase activity and Akt Thr 308 phosphorylation were significantly decreased in gp130-/-. The insulin-stimulated increase in glucose uptake rate was significantly attenuated in gp130-/-. In the culture medium, OSM levels were significantly lower in gp130+/+compared to gp130-/- cell. In conclusion, the knockdown of gp130 caused a decrease in STAT 3 phosphorylation and resulted in the attenuation of insulin-mediated glucose metabolism signaling in skeletal muscle cells. Thus, an excessive increase in extracellular OSM may induce blunted insulin action in skeletal muscle cells.
Collapse
Affiliation(s)
- Koji Sato
- Graduate School of Human Development and Environment, Kobe University, 3-11 Tsurukabuto, Nada-ku, Kobe, Hyogo, 657-8501, Japan.
| |
Collapse
|
4
|
Zhu AZ, Ma Z, Wolff EV, Lin Z, Gao ZJ, Li X, Du W. HES1 is required for mouse fetal hematopoiesis. Stem Cell Res Ther 2024; 15:235. [PMID: 39075526 PMCID: PMC11287931 DOI: 10.1186/s13287-024-03836-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Accepted: 07/06/2024] [Indexed: 07/31/2024] Open
Abstract
BACKGROUND Hematopoiesis in mammal is a complex and highly regulated process in which hematopoietic stem cells (HSCs) give rise to all types of differentiated blood cells. Previous studies have shown that hairy and enhancer of split (HES) repressors are essential regulators of adult HSC development downstream of Notch signaling. METHODS In this study, we investigated the role of HES1, a member of HES family, in fetal hematopoiesis using an embryonic hematopoietic specific Hes1 conditional knockout mouse model by using phenotypic flow cytometry, histopathology analysis, and functional in vitro colony forming unit (CFU) assay and in vivo bone marrow transplant (BMT) assay. RESULTS We found that loss of Hes1 in early embryonic stage leads to smaller embryos and fetal livers, decreases hematopoietic stem progenitor cell (HSPC) pool, results in defective multi-lineage differentiation. Functionally, fetal hematopoietic cells deficient for Hes1 exhibit reduced in vitro progenitor activity and compromised in vivo repopulation capacity in the transplanted recipients. Further analysis shows that fetal hematopoiesis defects in Hes1fl/flFlt3Cre embryos are resulted from decreased proliferation and elevated apoptosis, associated with de-repressed HES1 targets, p27 and PTEN in Hes1-KO fetal HSPCs. Finally, pharmacological inhibition of p27 or PTEN improves fetal HSPCs function both in vitro and in vivo. CONCLUSION Together, our findings reveal a previously unappreciated role for HES1 in regulating fetal hematopoiesis, and provide new insight into the differences between fetal and adult HSC maintenance.
Collapse
Affiliation(s)
- Anthony Z Zhu
- Division of Hematology and Oncology, University of Pittsburgh School of Medicine, 5117 Center Ave, Pittsburgh, PA, 15213, USA
- UPMC Hillman Cancer Center, Pittsburgh, PA, 15213, USA
| | - Zhilin Ma
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN, USA
| | - Emily V Wolff
- Division of Hematology and Oncology, University of Pittsburgh School of Medicine, 5117 Center Ave, Pittsburgh, PA, 15213, USA
- UPMC Hillman Cancer Center, Pittsburgh, PA, 15213, USA
| | - Zichen Lin
- Master of Science in Medical Science, Boston University School of Medicine Graduate Master Program, Boston, MA, USA
| | - Zhenxia J Gao
- Division of Hematology and Oncology, University of Pittsburgh School of Medicine, 5117 Center Ave, Pittsburgh, PA, 15213, USA
- UPMC Hillman Cancer Center, Pittsburgh, PA, 15213, USA
| | - Xue Li
- Institute for Brain Research and Rehabilitation, South China Normal University, Guangzhou, China
| | - Wei Du
- Division of Hematology and Oncology, University of Pittsburgh School of Medicine, 5117 Center Ave, Pittsburgh, PA, 15213, USA.
- UPMC Hillman Cancer Center, Pittsburgh, PA, 15213, USA.
| |
Collapse
|
5
|
Tadokoro T, Murata S, Kato M, Ueno Y, Tsuchida T, Okumura A, Kuse Y, Konno T, Uchida Y, Yamakawa Y, Zushi M, Yajima M, Kobayashi T, Hasegawa S, Kawakatsu-Hatada Y, Hayashi Y, Osakabe S, Maeda T, Kimura K, Mori A, Tanaka M, Kamishibahara Y, Matsuo M, Nie YZ, Okamoto S, Oba T, Tanimizu N, Taniguchi H. Human iPSC-liver organoid transplantation reduces fibrosis through immunomodulation. Sci Transl Med 2024; 16:eadg0338. [PMID: 39047116 DOI: 10.1126/scitranslmed.adg0338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 01/31/2024] [Accepted: 07/01/2024] [Indexed: 07/27/2024]
Abstract
Donor organ shortages for transplantation remain a serious global concern, and alternative treatment is in high demand. Fetal cells and tissues have considerable therapeutic potential as, for example, organoid technology that uses human induced pluripotent stem cells (hiPSCs) to generate unlimited human fetal-like cells and tissues. We previously reported the in vivo vascularization of early fetal liver-like hiPSC-derived liver buds (LBs) and subsquent improved survival of recipient mice with subacute liver failure. Here, we show hiPSC-liver organoids (LOs) that recapitulate midgestational fetal liver promote de novo liver generation when grafted onto the surface of host livers in chemical fibrosis models, thereby recovering liver function. We found that fetal liver, a hematopoietic tissue, highly expressed macrophage-recruiting factors and antifibrotic M2 macrophage polarization factors compared with the adult liver, resulting in fibrosis reduction because of CD163+ M2-macrophage polarization. Next, we created midgestational fetal liver-like hiPSC-LOs by fusion of hiPSC-LBs to induce static cell-cell interactions and found that these contained complex structures such as hepatocytes, vasculature, and bile ducts after transplantation. This fusion allowed the generation of a large human tissue suitable for transplantation into immunodeficient rodent models of liver fibrosis. hiPSC-LOs showed superior liver function compared with hiPSC-LBs and improved survival and liver function upon transplantation. In addition, hiPSC-LO transplantation ameliorated chemically induced liver fibrosis, a symptom of liver cirrhosis that leads to organ dysfunction, through immunomodulatory effects, particularly on CD163+ phagocytic M2-macrophage polarization. Together, our results suggest hiPSC-LO transplantation as a promising therapeutic option for liver fibrosis.
Collapse
Affiliation(s)
- Tomomi Tadokoro
- Department of Regenerative Medicine, Yokohama City University School of Medicine, Yokohama, Kanagawa 236-0004, Japan
- Division of Regenerative Medicine, Center for Stem Cell Biology and Regeneration Medicine, Institute of Medical Science, University of Tokyo, Tokyo 108-8639, Japan
| | - Soichiro Murata
- Department of Regenerative Medicine, Yokohama City University School of Medicine, Yokohama, Kanagawa 236-0004, Japan
- Division of Regenerative Medicine, Center for Stem Cell Biology and Regeneration Medicine, Institute of Medical Science, University of Tokyo, Tokyo 108-8639, Japan
| | - Mimoko Kato
- Department of Regenerative Medicine, Yokohama City University School of Medicine, Yokohama, Kanagawa 236-0004, Japan
| | - Yasuharu Ueno
- Department of Regenerative Medicine, Yokohama City University School of Medicine, Yokohama, Kanagawa 236-0004, Japan
- Division of Regenerative Medicine, Center for Stem Cell Biology and Regeneration Medicine, Institute of Medical Science, University of Tokyo, Tokyo 108-8639, Japan
| | - Tomonori Tsuchida
- Division of Regenerative Medicine, Center for Stem Cell Biology and Regeneration Medicine, Institute of Medical Science, University of Tokyo, Tokyo 108-8639, Japan
| | - Ayumu Okumura
- Division of Regenerative Medicine, Center for Stem Cell Biology and Regeneration Medicine, Institute of Medical Science, University of Tokyo, Tokyo 108-8639, Japan
| | - Yoshiki Kuse
- Department of Regenerative Medicine, Yokohama City University School of Medicine, Yokohama, Kanagawa 236-0004, Japan
- Division of Regenerative Medicine, Center for Stem Cell Biology and Regeneration Medicine, Institute of Medical Science, University of Tokyo, Tokyo 108-8639, Japan
| | - Takahiro Konno
- Department of Regenerative Medicine, Yokohama City University School of Medicine, Yokohama, Kanagawa 236-0004, Japan
| | - Yutaro Uchida
- Department of Regenerative Medicine, Yokohama City University School of Medicine, Yokohama, Kanagawa 236-0004, Japan
| | - Yuriko Yamakawa
- Department of Regenerative Medicine, Yokohama City University School of Medicine, Yokohama, Kanagawa 236-0004, Japan
- Division of Regenerative Medicine, Center for Stem Cell Biology and Regeneration Medicine, Institute of Medical Science, University of Tokyo, Tokyo 108-8639, Japan
| | - Marina Zushi
- Division of Regenerative Medicine, Center for Stem Cell Biology and Regeneration Medicine, Institute of Medical Science, University of Tokyo, Tokyo 108-8639, Japan
| | - Megumi Yajima
- Division of Regenerative Medicine, Center for Stem Cell Biology and Regeneration Medicine, Institute of Medical Science, University of Tokyo, Tokyo 108-8639, Japan
| | - Tatsuya Kobayashi
- Department of Regenerative Medicine, Yokohama City University School of Medicine, Yokohama, Kanagawa 236-0004, Japan
- Division of Regenerative Medicine, Center for Stem Cell Biology and Regeneration Medicine, Institute of Medical Science, University of Tokyo, Tokyo 108-8639, Japan
| | - Shunsuke Hasegawa
- Department of Regenerative Medicine, Yokohama City University School of Medicine, Yokohama, Kanagawa 236-0004, Japan
| | - Yumi Kawakatsu-Hatada
- Department of Regenerative Medicine, Yokohama City University School of Medicine, Yokohama, Kanagawa 236-0004, Japan
| | - Yoshihito Hayashi
- Division of Regenerative Medicine, Center for Stem Cell Biology and Regeneration Medicine, Institute of Medical Science, University of Tokyo, Tokyo 108-8639, Japan
| | - Shun Osakabe
- Department of Regenerative Medicine, Yokohama City University School of Medicine, Yokohama, Kanagawa 236-0004, Japan
| | - Takuji Maeda
- Department of Regenerative Medicine, Yokohama City University School of Medicine, Yokohama, Kanagawa 236-0004, Japan
| | - Kodai Kimura
- Department of Regenerative Medicine, Yokohama City University School of Medicine, Yokohama, Kanagawa 236-0004, Japan
| | - Akihiro Mori
- Department of Regenerative Medicine, Yokohama City University School of Medicine, Yokohama, Kanagawa 236-0004, Japan
| | - Maiko Tanaka
- Department of Regenerative Medicine, Yokohama City University School of Medicine, Yokohama, Kanagawa 236-0004, Japan
| | - Yu Kamishibahara
- Department of Regenerative Medicine, Yokohama City University School of Medicine, Yokohama, Kanagawa 236-0004, Japan
- Division of Regenerative Medicine, Center for Stem Cell Biology and Regeneration Medicine, Institute of Medical Science, University of Tokyo, Tokyo 108-8639, Japan
| | - Megumi Matsuo
- Department of Regenerative Medicine, Yokohama City University School of Medicine, Yokohama, Kanagawa 236-0004, Japan
- Division of Regenerative Medicine, Center for Stem Cell Biology and Regeneration Medicine, Institute of Medical Science, University of Tokyo, Tokyo 108-8639, Japan
| | - Yun-Zhong Nie
- Department of Regenerative Medicine, Yokohama City University School of Medicine, Yokohama, Kanagawa 236-0004, Japan
- Division of Regenerative Medicine, Center for Stem Cell Biology and Regeneration Medicine, Institute of Medical Science, University of Tokyo, Tokyo 108-8639, Japan
| | - Satoshi Okamoto
- Department of Regenerative Medicine, Yokohama City University School of Medicine, Yokohama, Kanagawa 236-0004, Japan
- Division of Regenerative Medicine, Center for Stem Cell Biology and Regeneration Medicine, Institute of Medical Science, University of Tokyo, Tokyo 108-8639, Japan
| | - Takayoshi Oba
- Department of Regenerative Medicine, Yokohama City University School of Medicine, Yokohama, Kanagawa 236-0004, Japan
- Division of Regenerative Medicine, Center for Stem Cell Biology and Regeneration Medicine, Institute of Medical Science, University of Tokyo, Tokyo 108-8639, Japan
| | - Naoki Tanimizu
- Division of Regenerative Medicine, Center for Stem Cell Biology and Regeneration Medicine, Institute of Medical Science, University of Tokyo, Tokyo 108-8639, Japan
| | - Hideki Taniguchi
- Department of Regenerative Medicine, Yokohama City University School of Medicine, Yokohama, Kanagawa 236-0004, Japan
- Division of Regenerative Medicine, Center for Stem Cell Biology and Regeneration Medicine, Institute of Medical Science, University of Tokyo, Tokyo 108-8639, Japan
| |
Collapse
|
6
|
Nishikawa Y. Aberrant differentiation and proliferation of hepatocytes in chronic liver injury and liver tumors. Pathol Int 2024; 74:361-378. [PMID: 38837539 PMCID: PMC11551836 DOI: 10.1111/pin.13441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2024] [Revised: 04/29/2024] [Accepted: 05/12/2024] [Indexed: 06/07/2024]
Abstract
Chronic liver injury induces liver cirrhosis and facilitates hepatocarcinogenesis. However, the effects of this condition on hepatocyte proliferation and differentiation are unclear. We showed that rodent hepatocytes display a ductular phenotype when they are cultured within a collagenous matrix. This process involves transdifferentiation without the emergence of hepatoblastic features and is at least partially reversible. During the ductular reaction in chronic liver diseases with progressive fibrosis, some hepatocytes, especially those adjacent to ectopic ductules, demonstrate ductular transdifferentiation, but the majority of increased ductules originate from the existing bile ductular system that undergoes extensive remodeling. In chronic injury, hepatocyte proliferation is weak but sustained, and most regenerative nodules in liver cirrhosis are composed of clonally proliferating hepatocytes, suggesting that a small fraction of hepatocytes maintain their proliferative capacity in chronic injury. In mouse hepatocarcinogenesis models, hepatocytes activate the expression of various fetal/neonatal genes, indicating that these cells undergo dedifferentiation. Hepatocyte-specific somatic integration of various oncogenes in mice demonstrated that hepatocytes may be the cells of origin for a broad spectrum of liver tumors through transdifferentiation and dedifferentiation. In conclusion, the phenotypic plasticity and heterogeneity of mature hepatocytes are important for understanding the pathogenesis of chronic liver diseases and liver tumors.
Collapse
Affiliation(s)
- Yuji Nishikawa
- President's OfficeAsahikawa Medical UniversityAsahikawaHokkaidoJapan
| |
Collapse
|
7
|
Zhang T, Qian C, Song M, Tang Y, Zhou Y, Dong G, Shen Q, Chen W, Wang A, Shen S, Zhao Y, Lu Y. Application Prospect of Induced Pluripotent Stem Cells in Organoids and Cell Therapy. Int J Mol Sci 2024; 25:2680. [PMID: 38473926 DOI: 10.3390/ijms25052680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 02/13/2024] [Accepted: 02/23/2024] [Indexed: 03/14/2024] Open
Abstract
Since its inception, induced pluripotent stem cell (iPSC) technology has been hailed as a powerful tool for comprehending disease etiology and advancing drug screening across various domains. While earlier iPSC-based disease modeling and drug assessment primarily operated at the cellular level, recent years have witnessed a significant shift towards organoid-based investigations. Organoids derived from iPSCs offer distinct advantages, particularly in enabling the observation of disease progression and drug metabolism in an in vivo-like environment, surpassing the capabilities of iPSC-derived cells. Furthermore, iPSC-based cell therapy has emerged as a focal point of clinical interest. In this review, we provide an extensive overview of non-integrative reprogramming methods that have evolved since the inception of iPSC technology. We also deliver a comprehensive examination of iPSC-derived organoids, spanning the realms of the nervous system, cardiovascular system, and oncology, as well as systematically elucidate recent advancements in iPSC-related cell therapies.
Collapse
Affiliation(s)
- Teng Zhang
- Jiangsu Joint International Research Laboratory of Chinese Medicine and Regenerative Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Cheng Qian
- Jiangsu Joint International Research Laboratory of Chinese Medicine and Regenerative Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Mengyao Song
- Jiangsu Joint International Research Laboratory of Chinese Medicine and Regenerative Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Yu Tang
- Jiangsu Joint International Research Laboratory of Chinese Medicine and Regenerative Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Yueke Zhou
- Jiangsu Joint International Research Laboratory of Chinese Medicine and Regenerative Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Guanglu Dong
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Department of Biochemistry and Molecular Biology, School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Qiuhong Shen
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Department of Biochemistry and Molecular Biology, School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Wenxing Chen
- Jiangsu Joint International Research Laboratory of Chinese Medicine and Regenerative Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Aiyun Wang
- Jiangsu Joint International Research Laboratory of Chinese Medicine and Regenerative Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Sanbing Shen
- Regenerative Medicine Institute, School of Medicine, University of Galway, H91 W2TY Galway, Ireland
| | - Yang Zhao
- Jiangsu Joint International Research Laboratory of Chinese Medicine and Regenerative Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Department of Biochemistry and Molecular Biology, School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Yin Lu
- Jiangsu Joint International Research Laboratory of Chinese Medicine and Regenerative Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, China
| |
Collapse
|
8
|
Hu Y, Hu X, Luo J, Huang J, Sun Y, Li H, Qiao Y, Wu H, Li J, Zhou L, Zheng S. Liver organoid culture methods. Cell Biosci 2023; 13:197. [PMID: 37915043 PMCID: PMC10619312 DOI: 10.1186/s13578-023-01136-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 09/20/2023] [Indexed: 11/03/2023] Open
Abstract
Organoids, three-dimensional structures cultured in vitro, can recapitulate the microenvironment, complex architecture, and cellular functions of in vivo organs or tissues. In recent decades, liver organoids have been developed rapidly, and their applications in biomedicine, such as drug screening, disease modeling, and regenerative medicine, have been widely recognized. However, the lack of repeatability and consistency, including the lack of standardized culture conditions, has been a major obstacle to the development and clinical application of liver organoids. It is time-consuming for researchers to identify an appropriate medium component scheme, and the usage of some ingredients remains controversial. In this review, we summarized and compared different methods for liver organoid cultivation that have been published in recent years, focusing on controversial medium components and discussing their advantages and drawbacks. We aimed to provide an effective reference for the development and standardization of liver organoid cultivation.
Collapse
Affiliation(s)
- Yiqing Hu
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
- NHC Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, 310003, China
| | - Xiaoyi Hu
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
- NHC Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, 310003, China
| | - Jia Luo
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
- NHC Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, 310003, China
| | - Jiacheng Huang
- NHC Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, 310003, China
| | - Yaohan Sun
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
- NHC Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, 310003, China
| | - Haoyu Li
- NHC Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, 310003, China
| | - Yinbiao Qiao
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
- NHC Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, 310003, China
| | - Hao Wu
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
- NHC Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, 310003, China
| | - Jianhui Li
- NHC Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, 310003, China
- Department of Hepatobiliary and Pancreatic Surgery, Shulan (Hangzhou) Hospital, Zhejiang Shuren University School of Medicine, Hangzhou, 310015, China
- The Organ Repair and Regeneration Medicine Institute of Hangzhou, Hangzhou, 310003, China
| | - Lin Zhou
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China.
- NHC Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, 310003, China.
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, 250117, China.
| | - Shusen Zheng
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China.
- NHC Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, 310003, China.
- Department of Hepatobiliary and Pancreatic Surgery, Shulan (Hangzhou) Hospital, Zhejiang Shuren University School of Medicine, Hangzhou, 310015, China.
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, 250117, China.
| |
Collapse
|
9
|
Ietto G, Iori V, Gritti M, Inversini D, Costantino A, Izunza Barba S, Jiang ZG, Carcano G, Dalla Gasperina D, Pettinato G. Multicellular Liver Organoids: Generation and Importance of Diverse Specialized Cellular Components. Cells 2023; 12:1429. [PMID: 37408262 PMCID: PMC10217024 DOI: 10.3390/cells12101429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 05/11/2023] [Accepted: 05/17/2023] [Indexed: 07/07/2023] Open
Abstract
Over 40,000 patients in the United States are estimated to suffer from end-stage liver disease and acute hepatic failure, for which liver transplantation is the only available therapy. Human primary hepatocytes (HPH) have not been employed as a therapeutic tool due to the difficulty in growing and expanding them in vitro, their sensitivity to cold temperatures, and tendency to dedifferentiate following two-dimensional culture. The differentiation of human-induced pluripotent stem cells (hiPSCs) into liver organoids (LO) has emerged as a potential alternative to orthotropic liver transplantation (OLT). However, several factors limit the efficiency of liver differentiation from hiPSCs, including a low proportion of differentiated cells capable of reaching a mature phenotype, the poor reproducibility of existing differentiation protocols, and insufficient long-term viability in vitro and in vivo. This review will analyze various methodologies being developed to improve hepatic differentiation from hiPSCs into liver organoids, paying particular attention to the use of endothelial cells as supportive cells for their further maturation. Here, we demonstrate why differentiated liver organoids can be used as a research tool for drug testing and disease modeling, or employed as a bridge for liver transplantation following liver failure.
Collapse
Affiliation(s)
- Giuseppe Ietto
- General, Emergency and Transplant Surgery Department, ASST-Sette Laghi, 21100 Varese, Italy
- Department of Medicine and Innovation Technology (DiMIT), University of Insubria, 21100 Varese, Italy
| | - Valentina Iori
- General, Emergency and Transplant Surgery Department, ASST-Sette Laghi, 21100 Varese, Italy
- Department of Medicine and Innovation Technology (DiMIT), University of Insubria, 21100 Varese, Italy
| | - Mattia Gritti
- Department of General Surgery, Humanitas Clinical and Research Center, Rozzano, 20089 Milan, Italy
| | - Davide Inversini
- General, Emergency and Transplant Surgery Department, ASST-Sette Laghi, 21100 Varese, Italy
- Department of Medicine and Innovation Technology (DiMIT), University of Insubria, 21100 Varese, Italy
| | - Angelita Costantino
- Department of Drug and Health Sciences, University of Catania, 95124 Catania, Italy;
| | - Sofia Izunza Barba
- Division of Gastroenterology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Z. Gordon Jiang
- Division of Gastroenterology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Giulio Carcano
- General, Emergency and Transplant Surgery Department, ASST-Sette Laghi, 21100 Varese, Italy
- Department of Medicine and Innovation Technology (DiMIT), University of Insubria, 21100 Varese, Italy
| | - Daniela Dalla Gasperina
- Department of Medicine and Innovation Technology (DiMIT), University of Insubria, 21100 Varese, Italy
- Department of Infectious Diseases, ASST-Sette Laghi, 21100 Varese, Italy
| | - Giuseppe Pettinato
- Division of Gastroenterology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| |
Collapse
|
10
|
Albiero M, Ciciliot S, Rodella A, Migliozzi L, Amendolagine FI, Boscaro C, Zuccolotto G, Rosato A, Fadini GP. Loss of Hematopoietic Cell-Derived Oncostatin M Worsens Diet-Induced Dysmetabolism in Mice. Diabetes 2023; 72:483-495. [PMID: 36657995 DOI: 10.2337/db22-0054] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Accepted: 01/02/2023] [Indexed: 01/21/2023]
Abstract
Innate immune cells infiltrate growing adipose tissue and propagate inflammatory clues to metabolically distant tissues, thereby promoting glucose intolerance and insulin resistance. Cytokines of the IL-6 family and gp130 ligands are among such signals. The role played by oncostatin M (OSM) in the metabolic consequences of overfeeding is debated, at least in part, because prior studies did not distinguish OSM sources and dynamics. Here, we explored the role of OSM in metabolic responses and used bone marrow transplantation to test the hypothesis that hematopoietic cells are major contributors to the metabolic effects of OSM. We show that OSM is required to adapt during the development of obesity because OSM concentrations are dynamically modulated during high-fat diet (HFD) and Osm-/- mice displayed early-onset glucose intolerance, impaired muscle glucose uptake, and worsened liver inflammation and damage. We found that OSM is mostly produced by blood cells and deletion of OSM in hematopoietic cells phenocopied glucose intolerance of whole-body Osm-/- mice fed a HFD and recapitulated liver damage with increased aminotransferase levels. We thus uncovered that modulation of OSM is involved in the metabolic response to overfeeding and that hematopoietic cell-derived OSM can regulate metabolism, likely via multiple effects in different tissues.
Collapse
Affiliation(s)
- Mattia Albiero
- Department of Medicine, University of Padova, Padova, Italy
- Veneto Institute of Molecular Medicine, Padova, Italy
| | - Stefano Ciciliot
- Veneto Institute of Molecular Medicine, Padova, Italy
- Department of Molecular Medicine, University of Pavia, Pavia, Italy
| | - Anna Rodella
- Department of Medicine, University of Padova, Padova, Italy
- Veneto Institute of Molecular Medicine, Padova, Italy
| | - Ludovica Migliozzi
- Department of Medicine, University of Padova, Padova, Italy
- Veneto Institute of Molecular Medicine, Padova, Italy
| | - Francesco Ivan Amendolagine
- Department of Medicine, University of Padova, Padova, Italy
- Veneto Institute of Molecular Medicine, Padova, Italy
| | - Carlotta Boscaro
- Department of Medicine, University of Padova, Padova, Italy
- Veneto Institute of Molecular Medicine, Padova, Italy
| | | | - Antonio Rosato
- Veneto Institute of Oncology IOV - IRCCS, Padova, Italy
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy
| | - Gian Paolo Fadini
- Department of Medicine, University of Padova, Padova, Italy
- Veneto Institute of Molecular Medicine, Padova, Italy
| |
Collapse
|
11
|
Chen H, Li LL, Du Y. Krüppel-like factor 15 in liver diseases: Insights into metabolic reprogramming. Front Pharmacol 2023; 14:1115226. [PMID: 36937859 PMCID: PMC10017497 DOI: 10.3389/fphar.2023.1115226] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2022] [Accepted: 02/14/2023] [Indexed: 03/06/2023] Open
Abstract
Liver diseases, characterized by metabolic disorder, have become a global public health problem with high morbidity and mortality. Krüppel-like factor 15 (KLF15) is a zinc-finger transcription factor mainly enriched in liver. Increasing evidence suggests that hepatic KLF15 is activated rapidly during fasting, and contributes to the regulation of gluconeogenesis, lipid, amino acid catabolism, bile acids, endobiotic and xenobiotic metabolism. This review summarizes the latest advances of KLF15 in metabolic reprogramming, and explore the function of KLF15 in acute liver injury, hepatitis B virus, and autoimmune hepatitis. which aims to evaluate the potential of KLF15 as a therapeutic target and prognostic biomarker for liver diseases.
Collapse
Affiliation(s)
- Hao Chen
- Department of Pharmacy, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- The Grade 3 Pharmaceutical Chemistry Laboratory of State Administration of Traditional Chinese Medicine, Hefei, Anhui, China
| | - Lan-Lan Li
- Department of Pharmacy, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- The Grade 3 Pharmaceutical Chemistry Laboratory of State Administration of Traditional Chinese Medicine, Hefei, Anhui, China
| | - Yan Du
- Department of Pharmacy, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- The Grade 3 Pharmaceutical Chemistry Laboratory of State Administration of Traditional Chinese Medicine, Hefei, Anhui, China
- *Correspondence: Yan Du,
| |
Collapse
|
12
|
Zheng S, Bian H, Li J, Shen Y, Yang Y, Hu W. Differentiation therapy: Unlocking phenotypic plasticity of hepatocellular carcinoma. Crit Rev Oncol Hematol 2022; 180:103854. [PMID: 36257532 DOI: 10.1016/j.critrevonc.2022.103854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 09/12/2022] [Accepted: 10/12/2022] [Indexed: 11/06/2022] Open
|
13
|
Komori T, Morikawa Y. Essential roles of the cytokine oncostatin M in crosstalk between muscle fibers and immune cells in skeletal muscle after aerobic exercise. J Biol Chem 2022; 298:102686. [PMID: 36370846 PMCID: PMC9720348 DOI: 10.1016/j.jbc.2022.102686] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 10/29/2022] [Accepted: 10/31/2022] [Indexed: 11/10/2022] Open
Abstract
Crosstalk between muscle fibers and immune cells is well known in the processes of muscle repair after exercise, especially resistance exercise. In aerobic exercise, however, this crosstalk is not fully understood. In the present study, we found that macrophages, especially anti-inflammatory (M2) macrophages, and neutrophils accumulated in skeletal muscles of mice 24 h after a single bout of an aerobic exercise. The expression of oncostatin M (OSM), a member of the interleukin 6 family of cytokines, was also increased in muscle fibers immediately after the exercise. In addition, we determined that deficiency of OSM in mice inhibited the exercise-induced accumulation of M2 macrophages and neutrophils, whereas intramuscular injection of OSM increased these immune cells in skeletal muscles. Furthermore, the chemokines related to the recruitment of macrophages and neutrophils were induced in skeletal muscles after aerobic exercise, which were attenuated in OSM-deficient mice. Among them, CC chemokine ligand 2, CC chemokine ligand 7, and CXC chemokine ligand 1 were induced by OSM in skeletal muscles. Next, we analyzed the direct effects of OSM on the skeletal muscle macrophages, because the OSM receptor β subunit was expressed predominantly in macrophages in the skeletal muscle. OSM directly induced the expression of these chemokines and anti-inflammatory markers in the skeletal muscle macrophages. From these findings, we conclude that OSM is essential for aerobic exercise-induced accumulation of M2 macrophages and neutrophils in the skeletal muscle partly through the regulation of chemokine expression in macrophages.
Collapse
|
14
|
Rankouhi TR, Keulen DV, Tempel D, Venhorst J. Oncostatin M: Risks and Benefits of a Novel Therapeutic Target for Atherosclerosis. Curr Drug Targets 2022; 23:1345-1369. [PMID: 35959619 DOI: 10.2174/1389450123666220811101032] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 05/30/2022] [Accepted: 06/03/2022] [Indexed: 01/25/2023]
Abstract
BACKGROUND Cardiovascular disease (CVD) is a leading cause of death worldwide. It is predicted that approximately 23.6 million people will die from CVDs annually by 2030. Therefore, there is a great need for an effective therapeutic approach to combat this disease. The European Cardiovascular Target Discovery (CarTarDis) consortium identified Oncostatin M (OSM) as a potential therapeutic target for atherosclerosis. The benefits of modulating OSM - an interleukin (IL)-6 family cytokine - have since been studied for multiple indications. However, as decades of high attrition rates have stressed, the success of a drug target is determined by the fine balance between benefits and the risk of adverse events. Safety issues should therefore not be overlooked. OBJECTIVE In this review, a risk/benefit analysis is performed on OSM inhibition in the context of atherosclerosis treatment. First, OSM signaling characteristics and its role in atherosclerosis are described. Next, an overview of in vitro, in vivo, and clinical findings relating to both the benefits and risks of modulating OSM in major organ systems is provided. Based on OSM's biological function and expression profile as well as drug intervention studies, safety concerns of inhibiting this target have been identified, assessed, and ranked for the target population. CONCLUSION While OSM may be of therapeutic value in atherosclerosis, drug development should also focus on de-risking the herein identified major safety concerns: tissue remodeling, angiogenesis, bleeding, anemia, and NMDA- and glutamate-induced neurotoxicity. Close monitoring and/or exclusion of patients with various comorbidities may be required for optimal therapeutic benefit.
Collapse
Affiliation(s)
- Tanja Rouhani Rankouhi
- Department of Risk Analysis for Products in Development, TNO, Utrechtseweg 48, 3704 HE, Zeist, The Netherlands
| | - Daniëlle van Keulen
- SkylineDx BV, Science and Clinical Development, 3062 ME Rotterdam, The Netherlands
| | - Dennie Tempel
- SkylineDx BV, Science and Clinical Development, 3062 ME Rotterdam, The Netherlands
| | - Jennifer Venhorst
- Department of Risk Analysis for Products in Development, TNO, Utrechtseweg 48, 3704 HE, Zeist, The Netherlands
| |
Collapse
|
15
|
Guo J, Wang S, Gao Q. Can next-generation humanized mice that reconstituted with both functional human immune system and hepatocytes model the progression of viral hepatitis to hepatocarcinogenesis? Front Med (Lausanne) 2022; 9:1002260. [PMID: 36213658 PMCID: PMC9537463 DOI: 10.3389/fmed.2022.1002260] [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: 07/25/2022] [Accepted: 09/08/2022] [Indexed: 11/30/2022] Open
Abstract
Hepatitis B virus (HBV) and Hepatitis C virus (HCV) chronic infections cause liver immunopathological diseases such as hepatitis, fibrosis, cirrhosis, and hepatocellular carcinomas, which are difficult to treat and continue to be major health problems globally. Due to the species-specific hepato-tropism of HBV and HCV, conventional rodent models are limited in their utility for studying the infection and associated liver immunopathogenesis. Humanized mice reconstituted with both functional human immune system and hepatocytes (HIS-HuHEP mice) have been extremely instrumental for in vivo studies of HBV or HCV infection and human-specific aspects of the progression of liver immunopathogenesis. However, none of the current HIS-HuHEP mice can model the progression of viral hepatitis to hepatocarcinogenesis which may be a notorious result of HBV or HCV chronic infection in patients, suggesting that they were functionally compromised and that there is still significant space to improve and establish next-generation of HIS-HuHEP mice with more sophisticated functions. In this review, we first summarize the principal requirements to establish HIS-HuHEP mice. We then discuss the respective protocols for current HIS-HuHEP mice and their applications, as well as their advantages and disadvantages. We also raise perspectives for further improving and establishing next-generation HIS-HuHEP mice.
Collapse
Affiliation(s)
- Jinglong Guo
- Department of Cardiovascular Disease, The First Hospital of Jilin University, Changchun, China
| | - Siyue Wang
- Graduate Program in Immunology and Microbiology, Baylor College of Medicine, One Baylor Plaza, Houston, TX, United States
| | - Qi Gao
- Department of Cardiovascular Disease, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
16
|
Development and characterization of a novel mouse anti-canine oncostatin M receptor beta monoclonal antibody. Biochem Biophys Res Commun 2022; 614:114-119. [DOI: 10.1016/j.bbrc.2022.05.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Accepted: 05/04/2022] [Indexed: 11/15/2022]
|
17
|
Li R, Zhao Y, Yourick JJ, Sprando RL, Gao X. Phenotypical, functional and transcriptomic comparison of two modified methods of hepatocyte differentiation from human induced pluripotent stem cells. Biomed Rep 2022; 16:43. [PMID: 35371477 PMCID: PMC8972237 DOI: 10.3892/br.2022.1526] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Accepted: 02/18/2022] [Indexed: 02/06/2023] Open
Abstract
Directed differentiation of human induced pluripotent stem cells (iPSCs) into hepatocytes could provide an unlimited source of liver cells, and therefore holds great promise for regenerative medicine, disease modeling, drug screening and toxicology studies. Various methods have been established during the past decade to differentiate human iPSCs into hepatocyte-like cells (HLCs) using growth factors and/or small molecules. However, direct comparison of the differentiation efficiency and the quality of the final HLCs between different methods has rarely been reported. In the current study, two hepatocyte differentiation methods were devised, termed Method 1 and 2, through modifying existing well-known hepatocyte differentiation strategies, and the resultant cells were compared phenotypically and functionally at different stages of hepatocyte differentiation. Compared to Method 1, higher differentiation efficiency and reproducibility were observed in Method 2, which generated highly homogeneous functional HLCs at the end of the differentiation process. The cells exhibited morphology closely resembling primary human hepatocytes and expressed high levels of hepatic protein markers. More importantly, these HLCs demonstrated several essential characteristics of mature hepatocytes, including major serum protein (albumin, fibronectin and α-1 antitrypsin) secretion, urea release, glycogen storage and inducible cytochrome P450 activity. Further transcriptomic comparison of the HLCs derived from the two methods identified 1,481 differentially expressed genes (DEGs); 290 Gene Ontology terms in the biological process category were enriched by these genes, which were further categorized into 34 functional classes. Pathway analysis of the DEGs identified several signaling pathways closely involved in hepatocyte differentiation of pluripotent stem cells, including 'signaling pathways regulating pluripotency of stem cells', 'Wnt signaling pathway', 'TGF-beta signaling pathway' and 'PI3K-Akt signaling pathway'. These results may provide a molecular basis for the differences observed between the two differentiation methods and suggest ways to further improve hepatocyte differentiation in order to obtain more mature HLCs for biomedical applications.
Collapse
Affiliation(s)
- Rong Li
- Division of Toxicology, Office of Applied Research and Safety Assessment, Center for Food Safety and Applied Nutrition, U.S. Food and Drug Administration, Laurel, MD 20708, USA
| | - Yang Zhao
- Division of Toxicology, Office of Applied Research and Safety Assessment, Center for Food Safety and Applied Nutrition, U.S. Food and Drug Administration, Laurel, MD 20708, USA
| | - Jeffrey J Yourick
- Division of Toxicology, Office of Applied Research and Safety Assessment, Center for Food Safety and Applied Nutrition, U.S. Food and Drug Administration, Laurel, MD 20708, USA
| | - Robert L Sprando
- Division of Toxicology, Office of Applied Research and Safety Assessment, Center for Food Safety and Applied Nutrition, U.S. Food and Drug Administration, Laurel, MD 20708, USA
| | - Xiugong Gao
- Division of Toxicology, Office of Applied Research and Safety Assessment, Center for Food Safety and Applied Nutrition, U.S. Food and Drug Administration, Laurel, MD 20708, USA
| |
Collapse
|
18
|
Kobayashi T, Takeba Y, Ohta Y, Ootaki M, Kida K, Watanabe M, Iiri T, Matsumoto N. Prenatal glucocorticoid administration accelerates the maturation of fetal rat hepatocytes. Mol Biol Rep 2022; 49:5831-5842. [PMID: 35304682 DOI: 10.1007/s11033-022-07358-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Accepted: 03/10/2022] [Indexed: 11/28/2022]
Abstract
BACKGROUND Prenatal glucocorticoid (GC) is clinically administered to pregnant women who are at risk of preterm birth for the maturation of cardiopulmonary function. Preterm and low-birth-weight infants often experience liver dysfunction after birth because their livers are immature. However, the effects of prenatal GC administration on the liver remain unclear. We aimed to investigate the effects of prenatal GC administration on the maturation of liver hepatocytes in preterm rats. METHODS AND RESULTS Dexamethasone (DEX) was administered to pregnant Wistar rats on gestational days 17 and 19 before cesarean section. Real-time reverse transcription-polymerase chain reaction (RT-PCR) was performed to determine the mRNA levels of albumin, hepatocyte nuclear factor-4 alpha (HNF4α), hepatocyte growth factor (HGF), thymus cell antigen 1 (Thy-1), cyclin B, and Cyclin-dependent kinase 1 (CDK1) in the liver samples. Immunohistochemical staining and enzyme-linked immunosorbent assay were performed to examine protein production. The hepatocytes enlarged because of growth and prenatal DEX administration. Albumin, HNF4α, and HGF levels increased secondary to growth and prenatal DEX administration. The levels of the cell cycle markers cyclin B and CDK1 gradually decreased during growth and with DEX administration. CONCLUSIONS The results suggest that prenatal GC administration leads to hepatocyte maturation via expression of HNF4α and HGF in preterm fetuses.
Collapse
Affiliation(s)
- Tsukasa Kobayashi
- Department of Pharmacology, St. Marianna University School of Medicine, 2-16-1 Sugao, Miyamae-ku, Kawasaki, Kanagawa, 216-8511, Japan
| | - Yuko Takeba
- Department of Pharmacology, St. Marianna University School of Medicine, 2-16-1 Sugao, Miyamae-ku, Kawasaki, Kanagawa, 216-8511, Japan.
| | - Yuki Ohta
- Department of Pharmacology, St. Marianna University School of Medicine, 2-16-1 Sugao, Miyamae-ku, Kawasaki, Kanagawa, 216-8511, Japan
| | - Masanori Ootaki
- Department of Pharmacology, St. Marianna University School of Medicine, 2-16-1 Sugao, Miyamae-ku, Kawasaki, Kanagawa, 216-8511, Japan
| | - Keisuke Kida
- Department of Pharmacology, St. Marianna University School of Medicine, 2-16-1 Sugao, Miyamae-ku, Kawasaki, Kanagawa, 216-8511, Japan
| | - Minoru Watanabe
- Institute for Animal Experimentation, St. Marianna University Graduate School of Medicine, 2-16-1 Sugao, Miyamae-ku, Kawasaki, Kanagawa, 216-8511, Japan
| | - Taroh Iiri
- Department of Pharmacology, St. Marianna University School of Medicine, 2-16-1 Sugao, Miyamae-ku, Kawasaki, Kanagawa, 216-8511, Japan
| | - Naoki Matsumoto
- Department of Pharmacology, St. Marianna University School of Medicine, 2-16-1 Sugao, Miyamae-ku, Kawasaki, Kanagawa, 216-8511, Japan
| |
Collapse
|
19
|
Graffmann N, Scherer B, Adjaye J. In vitro differentiation of pluripotent stem cells into hepatocyte like cells - basic principles and current progress. Stem Cell Res 2022; 61:102763. [DOI: 10.1016/j.scr.2022.102763] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 03/08/2022] [Accepted: 03/22/2022] [Indexed: 12/11/2022] Open
|
20
|
Tanimizu N. The neonatal liver: Normal development and response to injury and disease. Semin Fetal Neonatal Med 2022; 27:101229. [PMID: 33745829 DOI: 10.1016/j.siny.2021.101229] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
The liver emerges from the ventral foregut endoderm around 3 weeks in human and 1 week in mice after fertilization. The fetal liver works as a hematopoietic organ and then develops functions required for performing various metabolic reactions in late fetal and neonatal periods. In parallel with functional differentiation, the liver establishes three dimensional tissue structures. In particular, establishment of the bile excretion system consisting of bile canaliculi of hepatocytes and bile ducts of cholangiocytes is critical to maintain healthy tissue status. This is because hepatocytes produce bile as they functionally mature, and if allowed to remain within the liver tissue can lead to cytotoxicity. In this review, we focus on epithelial tissue morphogenesis in the perinatal period and cholestatic liver diseases caused by abnormal development of the biliary system.
Collapse
Affiliation(s)
- Naoki Tanimizu
- Department of Tissue Development and Regeneration, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, S-1, W-17, Chuo-ku, Sapporo, 060-8556, Japan.
| |
Collapse
|
21
|
Raggi C, M'Callum MA, Pham QT, Gaub P, Selleri S, Baratang NV, Mangahas CL, Cagnone G, Reversade B, Joyal JS, Paganelli M. Leveraging interacting signaling pathways to robustly improve the quality and yield of human pluripotent stem cell-derived hepatoblasts and hepatocytes. Stem Cell Reports 2022; 17:584-598. [PMID: 35120625 PMCID: PMC9039749 DOI: 10.1016/j.stemcr.2022.01.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 01/04/2022] [Accepted: 01/05/2022] [Indexed: 12/24/2022] Open
Abstract
Pluripotent stem cell (PSC)-derived hepatocyte-like cells (HLCs) have shown great potential as an alternative to primary human hepatocytes (PHHs) for in vitro modeling. Several differentiation protocols have been described to direct PSCs toward the hepatic fate. Here, by leveraging recent knowledge of the signaling pathways involved in liver development, we describe a robust, scalable protocol that allowed us to consistently generate high-quality bipotent human hepatoblasts and HLCs from both embryonic stem cells and induced PSC (iPSCs). Although not yet fully mature, such HLCs were more similar to adult PHHs than were cells obtained with previously described protocols, showing good potential as a physiologically representative alternative to PHHs for in vitro modeling. PSC-derived hepatoblasts effectively generated with this protocol could differentiate into mature hepatocytes and cholangiocytes within syngeneic liver organoids, thus opening the way for representative human 3D in vitro modeling of liver development and pathophysiology. We generated human hepatoblasts and hepatocyte-like cells (HLCs) from pluripotent stem cells Timed action on Wnt/β-catenin and TGFβ pathways improved maturity and yield of HLCs Hepatoblasts matured into hepatocytes and bile ducts within complex liver organoids The protocol is robust and showed potential for scalability and drug testing
Collapse
Affiliation(s)
- Claudia Raggi
- Liver Tissue Engineering and Cell Therapy Laboratory, CHU Sainte-Justine, Montreal, QC, Canada; Morphocell Technologies Inc., Montreal, QC, Canada
| | - Marie-Agnès M'Callum
- Liver Tissue Engineering and Cell Therapy Laboratory, CHU Sainte-Justine, Montreal, QC, Canada
| | - Quang Toan Pham
- Liver Tissue Engineering and Cell Therapy Laboratory, CHU Sainte-Justine, Montreal, QC, Canada
| | - Perrine Gaub
- CHU Sainte-Justine Research Center, Montreal, QC, Canada; Morphocell Technologies Inc., Montreal, QC, Canada
| | - Silvia Selleri
- Liver Tissue Engineering and Cell Therapy Laboratory, CHU Sainte-Justine, Montreal, QC, Canada
| | | | - Chenicka Lyn Mangahas
- Liver Tissue Engineering and Cell Therapy Laboratory, CHU Sainte-Justine, Montreal, QC, Canada
| | - Gaël Cagnone
- CHU Sainte-Justine Research Center, Montreal, QC, Canada
| | - Bruno Reversade
- Institute of Molecular and Cell Biology and Institute of Medical Biology, A(∗)STAR, Singapore, Singapore
| | - Jean-Sébastien Joyal
- CHU Sainte-Justine Research Center, Montreal, QC, Canada; Department of Pediatrics, Université de Montréal, Montreal, QC, Canada
| | - Massimiliano Paganelli
- Liver Tissue Engineering and Cell Therapy Laboratory, CHU Sainte-Justine, Montreal, QC, Canada; Department of Pediatrics, Université de Montréal, Montreal, QC, Canada; Morphocell Technologies Inc., Montreal, QC, Canada; Pediatric Hepatology, CHU Sainte-Justine, Montreal, QC, Canada.
| |
Collapse
|
22
|
Ashmore-Harris C, Fruhwirth GO. Generation of In Vivo Traceable Hepatocyte-Like Cells from Human iPSCs. Methods Mol Biol 2022; 2544:15-49. [PMID: 36125708 DOI: 10.1007/978-1-0716-2557-6_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
In this chapter, we describe a protocol for differentiation of human-induced pluripotent stem cells (iPSCs) into hepatocyte-like cells (HLCs) and their transduction with a lentivirus for gene transfer. Here, we engineer them to express the human sodium iodide symporter, which can be exploited as a radionuclide reporter gene, thereby enabling these cells to be tracked in vivo by single-photon emission computed tomography (SPECT) or positron emission tomography (PET) imaging. Differentiation of HLCs from iPSCs involves three steps: induction of iPSCs to definitive endoderm, differentiation to a hepatic progenitor cell population, and maturation of immature HLCs. Once proliferation of hepatic progenitors has ceased and an immature HLC population is generated, lentiviral transduction can be performed. The immature hepatic gene expression profile/morphology at the stage of transduction will be compatible with further maturation following transgene expression either in vitro or in vivo, with expression of the transgene retained. We detail how transgenic cells can be imaged in vivo. While we provide a protocol for the NIS reporter gene, the cell engineering aspects of this protocol are transferable for use with other (reporter) genes if desired.
Collapse
Affiliation(s)
- Candice Ashmore-Harris
- Imaging Therapies and Cancer Group, Comprehensive Cancer Centre, School of Cancer and Pharmaceutical Sciences, King's College London, Guy's Cancer Centre, London, UK
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
| | - Gilbert O Fruhwirth
- Imaging Therapies and Cancer Group, Comprehensive Cancer Centre, School of Cancer and Pharmaceutical Sciences, King's College London, Guy's Cancer Centre, London, UK.
| |
Collapse
|
23
|
Matakovic L, Overeem AW, Klappe K, van IJzendoorn SCD. Induction of Bile Canaliculi-Forming Hepatocytes from Human Pluripotent Stem Cells. Methods Mol Biol 2022; 2544:71-82. [PMID: 36125710 DOI: 10.1007/978-1-0716-2557-6_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Cell polarity and formation of bile canaliculi can be achieved in hepatocytes which are generated from patient-derived induced pluripotent stem cells. This allows for the study of endogenous mutant proteins, patient-specific pathogenesis, and drug responses for diseases where hepatocyte polarity and bile canaliculi play a key role. Here, we describe a step-by-step protocol for the generation of bile canaliculi-forming hepatocytes from induced pluripotent stem cells and their evaluation.
Collapse
Affiliation(s)
- Lavinija Matakovic
- Department of Biomedical Sciences of Cells and Systems, section Molecular Cell Biology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Arend W Overeem
- Department of Biomedical Sciences of Cells and Systems, section Molecular Cell Biology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, the Netherlands
| | - Karin Klappe
- Department of Biomedical Sciences of Cells and Systems, section Molecular Cell Biology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Sven C D van IJzendoorn
- Department of Biomedical Sciences of Cells and Systems, section Molecular Cell Biology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands.
| |
Collapse
|
24
|
Xie Y, Yao J, Jin W, Ren L, Li X. Induction and Maturation of Hepatocyte-Like Cells In Vitro: Focus on Technological Advances and Challenges. Front Cell Dev Biol 2021; 9:765980. [PMID: 34901010 PMCID: PMC8662991 DOI: 10.3389/fcell.2021.765980] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2021] [Accepted: 11/08/2021] [Indexed: 12/17/2022] Open
Abstract
Limited by the poor proliferation and restricted sources of adult hepatocytes, there is an urgent need to find substitutes for proliferation and cultivation of mature hepatocytes in vitro for use in disease treatment, drug approval, and toxicity testing. Hepatocyte-like cells (HLCs), which originate from undifferentiated stem cells or modified adult cells, are considered good candidates because of their advantages in terms of cell source and in vitro expansion ability. However, the majority of induced HLCs are in an immature state, and their degree of differentiation is heterogeneous, diminishing their usability in basic research and limiting their clinical application. Therefore, various methods have been developed to promote the maturation of HLCs, including chemical approaches, alteration of cell culture systems, and genetic manipulation, to meet the needs of in vivo transplantation and in vitro model establishment. This review proposes different cell types for the induction of HLCs, and provide a comprehensive overview of various techniques to promote the generation and maturation of HLCs in vitro.
Collapse
Affiliation(s)
- Ye Xie
- The First Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Jia Yao
- The First Clinical Medical College, Lanzhou University, Lanzhou, China.,Key Laboratory of Biotherapy and Regenerative Medicine of Gansu Province, Lanzhou, China
| | - Weilin Jin
- The First Clinical Medical College, Lanzhou University, Lanzhou, China.,Institute of Cancer Neuroscience, The First Hospital of Lanzhou University, Lanzhou, China.,The Medical Frontier Innovation Research Center, The First Hospital of Lanzhou University, Lanzhou, China
| | - Longfei Ren
- The First Clinical Medical College, Lanzhou University, Lanzhou, China.,The Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, China
| | - Xun Li
- The First Clinical Medical College, Lanzhou University, Lanzhou, China.,Key Laboratory of Biotherapy and Regenerative Medicine of Gansu Province, Lanzhou, China.,The Medical Frontier Innovation Research Center, The First Hospital of Lanzhou University, Lanzhou, China.,The Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, China.,Hepatopancreatobiliary Surgery Institute of Gansu Province, Lanzhou, China
| |
Collapse
|
25
|
Zhang H, Zhang Y, Ma Y, Elsabagh M, Wang H, Wang M. Dietary rumen-protected L-arginine or N-carbamylglutamate attenuated fetal hepatic inflammation in undernourished ewes suffering from intrauterine growth restriction. ACTA ACUST UNITED AC 2021; 7:1095-1104. [PMID: 34738040 PMCID: PMC8545652 DOI: 10.1016/j.aninu.2021.06.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 06/19/2021] [Accepted: 06/28/2021] [Indexed: 11/29/2022]
Abstract
This study aimed to explore whether dietary rumen-protected L-arginine (RP-Arg) or N-carbamylglutamate (NCG) supplementation to feed-restricted pregnant ewes counteracts fetal hepatic inflammation and innate immune dysfunction associated with intrauterine growth retardation (IUGR) in ovine fetuses. On d 35 of pregnancy, twin-bearing Hu ewes (n = 32) were randomly assigned to 4 treatment groups (8 ewes and 16 fetuses per group) and fed diets containing 100% of the NRC requirements (CON), 50% of the NRC requirements (RES), RES + RP-Arg (20 g/d) (RESA), or RES + NCG (5 g/d) (RESN). At 08:00 on d 110 of gestation, fetal blood and liver tissue samples were collected. The levels of triglyceride, free fatty acid, cholesterol and β-hydroxybutyrate in the fetal blood of RESA and RESN groups were lower (P < 0.05) than those of the RES group, but were higher (P < 0.05) than those of the CON group. The interleukin (IL)-6 and IL-1 levels in fetal blood and liver tissue as well as the myeloid differentiation primary response 88 (MyD88), transforming growth factor β (TGFβ), and nuclear factor kappa B (NF-κB) mRNA levels in the fetal liver were decreased (P < 0.05) by the NCG or RP-Arg supplementation compared to the RES treatment. Similarly, the toll-like receptor (TLR)-4, MyD88, TGFβ, and p-c-Jun N-terminal kinase (JNK) protein levels in the fetal liver were reduced (P < 0.05) in the NCG and RP-Arg -supplemented groups compared to the RES group. These results showed that dietary supplementation of RP-Arg or NCG to underfed pregnant ewes could protect against IUGR fetal hepatic inflammation via improving lipid metabolism, down-regulating the TLR-4 and the inflammatory JNK and NF-κB signaling pathways, and decreasing cytokine production in ovine fetal blood and liver tissue.
Collapse
Affiliation(s)
- Hao Zhang
- State Key Laboratory of Sheep Genetic Improvement and Healthy Production, Xinjiang Academy of Agricultural Reclamation Sciences, Shihezi, 832000, China.,Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou, 225009, China.,Laboratory of Metabolic Manipulation of Herbivorous Animal Nutrition, College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, China
| | - Ying Zhang
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou, 225009, China.,Laboratory of Metabolic Manipulation of Herbivorous Animal Nutrition, College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, China
| | - Yi Ma
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou, 225009, China.,Laboratory of Metabolic Manipulation of Herbivorous Animal Nutrition, College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, China
| | - Mabrouk Elsabagh
- Department of Animal Production and Technology, Faculty of Agricultural Sciences and Technologies, Niğde Ömer Halisdemir University, Nigde, 51240, Turkey.,Department of Nutrition and Clinical Nutrition, Faculty of Veterinary Medicine, Kafrelsheikh University, Kafrelsheikh, 33516, Egypt
| | - Hongrong Wang
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou, 225009, China.,Laboratory of Metabolic Manipulation of Herbivorous Animal Nutrition, College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, China
| | - Mengzhi Wang
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou, 225009, China.,Laboratory of Metabolic Manipulation of Herbivorous Animal Nutrition, College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, China
| |
Collapse
|
26
|
Dong C, Zhang B, Ying Y, Hou L, Wu W, Wei H, Luo X. Oestradiol promotes the intrahepatic bile duct development of C57BL/6CrSlc mice during embryonic period via Notch signalling pathway. J Cell Mol Med 2021; 25:9447-9459. [PMID: 34498380 PMCID: PMC8500961 DOI: 10.1111/jcmm.16888] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 07/28/2021] [Accepted: 08/12/2021] [Indexed: 01/23/2023] Open
Abstract
Oestradiol (E2) is a critical factor for multiple systems' development during the embryonic period. Here, we aimed to investigate the effects of oestradiol on intrahepatic bile duct development, which may allow a better understanding of congenital bile duct dysplasia. DLK+ hepatoblasts were extracted from the C57BL/6CrSlc foetal mice and randomly divided into control group, oestradiol groups (1, 10, 100 nM) and oestradiol (10 nM) + DAPT (inhibitor of Notch signalling; 40 µM) group for in vitro experiments. For in vivo analysis, pregnant mice were divided into control group, oestradiol (intraperitoneal injection of 0.6 mg/kg/day) ± DAPT (subcutaneous injection of 10 mg/kg/day) groups and tamoxifen (gavage administration of 0.4 mg/kg/day) group. The results showed that oestradiol promoted hepatoblast differentiation into cholangiocytes and intrahepatic bile duct development during the embryonic period. Tamoxifen, an antioestrogenic drug, inhibited the above processes. Moreover, oestradiol promoted the expression of Notch signalling pathway-associated proteins and genes both in vitro and in vivo. Notably, DAPT addition inhibited the oestradiol-mediated effects. In conclusion, oestradiol can promote hepatoblast differentiation into cholangiocytes and intrahepatic bile duct development of C57BL/6CrSlc mice during embryonic period via the Notch signalling pathway.
Collapse
Affiliation(s)
- Chen Dong
- Department of PediatricsTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Ben‐ping Zhang
- Department of EndocrinologyTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Yan‐Qin Ying
- Department of PediatricsTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Ling Hou
- Department of PediatricsTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Wei Wu
- Department of PediatricsTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Hong Wei
- Department of PediatricsTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Xiao‐ping Luo
- Department of PediatricsTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| |
Collapse
|
27
|
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
|
28
|
Dinca SC, Greiner D, Weidenfeld K, Bond L, Barkan D, Jorcyk CL. Novel mechanism for OSM-promoted extracellular matrix remodeling in breast cancer: LOXL2 upregulation and subsequent ECM alignment. Breast Cancer Res 2021; 23:56. [PMID: 34011405 PMCID: PMC8132418 DOI: 10.1186/s13058-021-01430-x] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 04/20/2021] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Invasive ductal carcinoma (IDC) is a serious problem for patients as it metastasizes, decreasing 5-year patient survival from > 95 to ~ 27%. The breast tumor microenvironment (TME) is often saturated with proinflammatory cytokines, such as oncostatin M (OSM), which promote epithelial-to-mesenchymal transitions (EMT) in IDC and increased metastasis. The extracellular matrix (ECM) also plays an important role in promoting invasive and metastatic potential of IDC. Specifically, the reorganization and alignment of collagen fibers in stromal ECM leads to directed tumor cell motility, which promotes metastasis. Lysyl oxidase like-2 (LOXL2) catalyzes ECM remodeling by crosslinking of collagen I in the ECM. We propose a novel mechanism whereby OSM induces LOXL2 expression, mediating stromal ECM remodeling of the breast TME. METHODS Bioinformatics was utilized to determine survival and gene correlation in patients. IDC cell lines were treated with OSM (also IL-6, LIF, and IL-1β) and analyzed for LOXL2 expression by qRT-PCR and immunolabelling techniques. Collagen I contraction assays, 3D invasion assays, and confocal microscopy were performed with and without LOXL2 inhibition to determine the impact of OSM-induced LOXL2 on the ECM. RESULTS Our studies demonstrate that IDC patients with high LOXL2 and OSM co-expression had worse rates of metastasis-free survival than those with high levels of either, individually, and LOXL2 expression is positively correlated to OSM/OSM receptor (OSMR) expression in IDC patients. Furthermore, human IDC cells treated with OSM resulted in a significant increase in LOXL2 mRNA, which led to upregulated protein expression of secreted, glycosylated, and enzymatically active LOXL2. The expression of LOXL2 in IDC cells did not affect OSM-promoted EMT, and LOXL2 was localized to the cytoplasm and/or secreted. OSM-induced LOXL2 promoted an increase in ECM collagen I fiber crosslinking, which led to significant fiber alignment between cells and increased IDC cell invasion. CONCLUSIONS Aligned collagen fibers in the ECM provide pathways for tumor cells to migrate more easily through the stroma to nearby vasculature and tissue. These results provide a new paradigm through which proinflammatory cytokine OSM promotes tumor progression. Understanding the nuances in IDC metastasis will lead to better potential therapeutics to combat against the possibility.
Collapse
Affiliation(s)
- Simion C. Dinca
- Biomolecular Sciences Graduate Program, Boise State University, 1910 University Drive, MS1515, Boise, ID 83725 USA
| | - Daniel Greiner
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT 84112 USA
- Department of Biological Sciences, Boise State University, 1910 University Drive, MS1515, Boise, ID 83725 USA
| | - Keren Weidenfeld
- Department of Human Biology and Medical Sciences, University of Haifa, Haifa, Israel
| | - Laura Bond
- Biomolecular Research Center, Boise State University, 1910 University Drive, MS1515, Boise, ID 83725 USA
| | - Dalit Barkan
- Department of Human Biology and Medical Sciences, University of Haifa, Haifa, Israel
| | - Cheryl L. Jorcyk
- Biomolecular Sciences Graduate Program, Boise State University, 1910 University Drive, MS1515, Boise, ID 83725 USA
- Department of Biological Sciences, Boise State University, 1910 University Drive, MS1515, Boise, ID 83725 USA
| |
Collapse
|
29
|
Kim Y, Kim YW, Lee SB, Kang K, Yoon S, Choi D, Park SH, Jeong J. Hepatic patch by stacking patient-specific liver progenitor cell sheets formed on multiscale electrospun fibers promotes regenerative therapy for liver injury. Biomaterials 2021; 274:120899. [PMID: 34034028 DOI: 10.1016/j.biomaterials.2021.120899] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 05/10/2021] [Accepted: 05/15/2021] [Indexed: 12/15/2022]
Abstract
Recently, use of cell sheets with bio-applicable fabrication materials for transplantation has been an attractive approach for the treatment of patients with liver failure. However, renewable and scalable cell sources for engineered tissue patches remain limited. We previously reported a new type of proliferating bipotent human chemically derived hepatic progenitor cells (hCdHs) developed by small molecule-mediated reprogramming. Here, we developed a patient-specific hepatic cell sheet constructed from liver biopsy-derived hCdHs on a multiscale fibrous scaffold by combining electrospinning and three-dimensional printing. Analysis of biomaterial composition revealed that the high-density electrospun sheet was superior in increasing the functional properties of hCdHs. Furthermore, the hepatic patch assembled by multilayer stacking with alternate cell sheets of hCdHs and human umbilical vein endothelial cells (HUVECs) recapitulated a liver tissue-like structure, with histological and morphological shape and size similar to those of primary human hepatocytes, and exhibited a significant increase in hepatic functions such as albumin secretion and activity of cytochrome P450 during in vitro hepatic differentiation compared with that in hCdH cells cultured in a two-dimensional monolayer. Interestingly, in the hepatic patch, the induction of functional hepatocytes was associated with both the electrospun fibrous-facilitated oncostatin M signaling and selective activation of AKT signaling by HUVECs. Notably, upon transplantation into a mouse model of therapeutic liver repopulation, the hepatic patch effectively repopulated the damaged parenchyma and induced the restoration of liver function with healthy morphology in the lobe and an improved survival rate (>70%) in mice. Overall, these results suggested that liver biopsy-derived hCdHs can be an efficient alternative source for developing hepatic cell sheets and patches with potential clinical applications in tissue engineering to advance liver regeneration.
Collapse
Affiliation(s)
- Yohan Kim
- Department of Surgery, Hanyang University College of Medicine, Seoul, 04763, Republic of Korea; HY Indang Center of Regenerative Medicine and Stem Cell Research, Hanyang University, Seoul, 04763, Republic of Korea
| | - Young Won Kim
- Digital Manufacturing Process Group, Korea Institute of Industrial Technology, 113-58 Seohaean-ro, Siheungsi, Gyeonggi-do, 15014, Republic of Korea
| | - Seung Bum Lee
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological & Medical Science, Seoul, 01812, Republic of Korea
| | - Kyojin Kang
- Department of Surgery, Hanyang University College of Medicine, Seoul, 04763, Republic of Korea
| | - Sangtae Yoon
- Department of Surgery, Hanyang University College of Medicine, Seoul, 04763, Republic of Korea; HY Indang Center of Regenerative Medicine and Stem Cell Research, Hanyang University, Seoul, 04763, Republic of Korea
| | - Dongho Choi
- Department of Surgery, Hanyang University College of Medicine, Seoul, 04763, Republic of Korea; HY Indang Center of Regenerative Medicine and Stem Cell Research, Hanyang University, Seoul, 04763, Republic of Korea.
| | - Suk-Hee Park
- School of Mechanical Engineering, Pusan National University, Busan, 46241, Republic of Korea.
| | - Jaemin Jeong
- Department of Surgery, Hanyang University College of Medicine, Seoul, 04763, Republic of Korea; HY Indang Center of Regenerative Medicine and Stem Cell Research, Hanyang University, Seoul, 04763, Republic of Korea.
| |
Collapse
|
30
|
Harrison SP, Baumgarten SF, Verma R, Lunov O, Dejneka A, Sullivan GJ. Liver Organoids: Recent Developments, Limitations and Potential. Front Med (Lausanne) 2021; 8:574047. [PMID: 34026769 PMCID: PMC8131532 DOI: 10.3389/fmed.2021.574047] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Accepted: 03/29/2021] [Indexed: 12/12/2022] Open
Abstract
Liver cell types derived from induced pluripotent stem cells (iPSCs) share the potential to investigate development, toxicity, as well as genetic and infectious disease in ways currently limited by the availability of primary tissue. With the added advantage of patient specificity, which can play a role in all of these areas. Many iPSC differentiation protocols focus on 3 dimensional (3D) or organotypic differentiation, as these offer the advantage of more closely mimicking in vivo systems including; the formation of tissue like architecture and interactions/crosstalk between different cell types. Ultimately such models have the potential to be used clinically and either with or more aptly, in place of animal models. Along with the development of organotypic and micro-tissue models, there will be a need to co-develop imaging technologies to enable their visualization. A variety of liver models termed "organoids" have been reported in the literature ranging from simple spheres or cysts of a single cell type, usually hepatocytes, to those containing multiple cell types combined during the differentiation process such as hepatic stellate cells, endothelial cells, and mesenchymal cells, often leading to an improved hepatic phenotype. These allow specific functions or readouts to be examined such as drug metabolism, protein secretion or an improved phenotype, but because of their relative simplicity they lack the flexibility and general applicability of ex vivo tissue culture. In the liver field these are more often constructed rather than developed together organotypically as seen in other organoid models such as brain, kidney, lung and intestine. Having access to organotypic liver like surrogates containing multiple cell types with in vivo like interactions/architecture, would provide vastly improved models for disease, toxicity and drug development, combining disciplines such as microfluidic chip technology with organoids and ultimately paving the way to new therapies.
Collapse
Affiliation(s)
- Sean Philip Harrison
- Hybrid Technology Hub–Center of Excellence, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
- Department of Pediatric Research, Oslo University Hospital, Oslo, Norway
| | - Saphira Felicitas Baumgarten
- Hybrid Technology Hub–Center of Excellence, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
- Department of Pediatric Research, Oslo University Hospital, Oslo, Norway
| | - Rajneesh Verma
- Hybrid Technology Hub–Center of Excellence, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
- Department of Pediatric Research, Oslo University Hospital, Oslo, Norway
| | - Oleg Lunov
- Institute of Physics of the Czech Academy of Sciences, Prague, Czechia
| | - Alexandr Dejneka
- Institute of Physics of the Czech Academy of Sciences, Prague, Czechia
| | - Gareth John Sullivan
- Hybrid Technology Hub–Center of Excellence, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
- Department of Pediatric Research, Oslo University Hospital, Oslo, Norway
- Norwegian Center for Stem Cell Research, Oslo University Hospital, University of Oslo, Oslo, Norway
- Institute of Immunology, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
31
|
Sanchez-Infantes D, Stephens JM. Adipocyte Oncostatin Receptor Regulates Adipose Tissue Homeostasis and Inflammation. Front Immunol 2021; 11:612013. [PMID: 33854494 PMCID: PMC8039456 DOI: 10.3389/fimmu.2020.612013] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 12/31/2020] [Indexed: 01/05/2023] Open
Abstract
Adipocytes are the largest cell type in terms of volume, but not number, in adipose tissue. Adipocytes are prominent contributors to systemic metabolic health. Obesity, defined by excess adipose tissue (AT), is recognized as a low-grade chronic inflammatory state. Cytokines are inflammatory mediators that are produced in adipose tissue (AT) and function in both AT homeostatic as well as pathological conditions. AT inflammation is associated with systemic metabolic dysfunction and obesity-associated infiltration and proliferation of immune cells occurs in a variety of fat depots in mice and humans. AT immune cells secrete a variety of chemokines and cytokines that act in a paracrine manner on adjacent adipocytes. TNFα, IL-6, and MCP-1, are well studied mediators of AT inflammation. Oncostatin M (OSM) is another proinflammatory cytokine that is elevated in AT in human obesity, and its specific receptor (OSMRβ) is also induced in conditions of obesity and insulin resistance. OSM production and paracrine signaling in AT regulates adipogenesis and the functions of AT. This review summarizes the roles of the oncostatin M receptor (OSMRβ) as a modulator of adipocyte development and function its contributions to immunological adaptations in AT in metabolic disease states.
Collapse
Affiliation(s)
- David Sanchez-Infantes
- Department of Endocrinology and Nutrition, Germans Trias i Pujol Research Institute, Barcelona, Spain
- Department of Basic Sciences of Health, Area of Biochemistry and Molecular Biology, Universidad Rey Juan Carlos, Alcorcon, Spain
| | - Jacqueline M. Stephens
- Department of Biological Sciences and Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, LA, United States
| |
Collapse
|
32
|
Lewis K, Yoshimoto M, Takebe T. Fetal liver hematopoiesis: from development to delivery. Stem Cell Res Ther 2021; 12:139. [PMID: 33597015 PMCID: PMC7890853 DOI: 10.1186/s13287-021-02189-w] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Accepted: 01/25/2021] [Indexed: 12/25/2022] Open
Abstract
Clinical transplants of hematopoietic stem cells (HSC) can provide a lifesaving therapy for many hematological diseases; however, therapeutic applications are hampered by donor availability. In vivo, HSC exist in a specified microenvironment called the niche. While most studies of the niche focus on those residing in the bone marrow (BM), a better understanding of the fetal liver niche during development is vital to design human pluripotent stem cell (PSC) culture and may provide valuable insights with regard to expanding HSCs ex vivo for transplantation. This review will discuss the importance of the fetal liver niche in HSC expansion, a feat that occurs during development and has great clinical potential. We will also discuss emerging approaches to generate expandable HSC in cell culture that attain more complexity in the form of cells or organoid models in combination with engineering and systems biology approaches. Overall, delivering HSC by charting developmental principles will help in the understanding of the molecular and biological interactions between HSCs and fetal liver cells for their controlled maturation and expansion.
Collapse
Affiliation(s)
- Kyle Lewis
- Center for Stem Cell & Organoid Medicine (CuSTOM), Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
- Division of Gastroenterology, Hepatology and Nutrition and Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Momoko Yoshimoto
- Institute of Molecular Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, Texas, 77030, USA.
| | - Takanori Takebe
- Center for Stem Cell & Organoid Medicine (CuSTOM), Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA.
- Division of Gastroenterology, Hepatology and Nutrition and Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA.
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA.
- Institute of Research, Tokyo Medical and Dental University 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan.
- Communication Design Center, Advanced Medical Research Center, Yokohama City University, Kanazawa-ku 3-9, Yokohama, Kanagawa, 236-0004, Japan.
| |
Collapse
|
33
|
Watanabe Y, Tsuchiya A, Terai S. The development of mesenchymal stem cell therapy in the present, and the perspective of cell-free therapy in the future. Clin Mol Hepatol 2020; 27:70-80. [PMID: 33317249 PMCID: PMC7820202 DOI: 10.3350/cmh.2020.0194] [Citation(s) in RCA: 87] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Accepted: 09/21/2020] [Indexed: 02/06/2023] Open
Abstract
Cirrhosis is a chronic condition that can lead to liver failure. Currently, the viable option for decreasing mortality is liver transplantation. However, transplant surgery is highly invasive. Therefore, cell-based therapy has been developed as an alternative. Based on promising findings from preclinical research, some new trials have been registered. One of them was autologous bone marrow cell infusion therapy and found that ameliorating liver fibrosis activated liver regeneration. Now, majority of trials focus on low-immunogenicity mesenchymal stem cells (MSCs) appropriate for allogeneic administration. However, despite about 20 years of research, only a limited number of cell-based therapies have entered routine practice. Furthermore, potential shortcomings of cell-based therapy include a limit on the number of cells, which may be administered, as well as their failure to infiltrate target organs. On the other hand, these research show that MSCs act as "conducting cells" and regulate host cells including macrophages via extracellular vesicles (EVs) or exosome signals, leading to ameliorate liver fibrosis and promote regeneration. Therefore, the concept of cell-free therapy, which makes use of cell-derived EVs or exosomes, is attracting attention. Cell-free therapies may be safely administered in large doses and are able to infiltrate target organs. However, development of cell-free therapy exhibits its own set of challenges and such therapy may not be completely curative in the context of liver disease. This review describes the history of cell-based therapy research and recent advances in cell-free therapy, as well as discussing the need for more effective therapies.
Collapse
Affiliation(s)
- Yusuke Watanabe
- Department of Preemptive Medicine for Digestive Disease and Healthy Active Life, School of Medicine, Niigata University, Niigata, Japan
| | - Atsunori Tsuchiya
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Shuji Terai
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| |
Collapse
|
34
|
Lucendo-Villarin B, Meseguer-Ripolles J, Drew J, Fischer L, Ma E, Flint O, Simpson KJ, Machesky LM, Mountford JC, Hay DC. Development of a cost-effective automated platform to produce human liver spheroids for basic and applied research. Biofabrication 2020; 13:015009. [PMID: 33007774 DOI: 10.1088/1758-5090/abbdb2] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Accepted: 10/02/2020] [Indexed: 12/14/2022]
Abstract
Liver disease represents an increasing cause of global morbidity and mortality. Currently, liver transplant is the only treatment curative for end-stage liver disease. Donor organs cannot meet the demand and therefore scalable treatments and new disease models are required to improve clinical intervention. Pluripotent stem cells represent a renewable source of human tissue. Recent advances in three-dimensional cell culture have provided the field with more complex systems that better mimic liver physiology and function. Despite these improvements, current cell-based models are variable in performance and expensive to manufacture at scale. This is due, in part, to the use of poorly defined or cross-species materials within the process, severely affecting technology translation. To address this issue, we have developed an automated and economical platform to produce liver tissue at scale for modelling disease and small molecule screening. Stem cell derived liver spheres were formed by combining hepatic progenitors with endothelial cells and stellate cells, in the ratios found within the liver. The resulting tissue permitted the study of human liver biology 'in the dish' and could be scaled for screening. In summary, we have developed an automated differentiation system that permits reliable self-assembly of human liver tissue for biomedical application. Going forward we believe that this technology will not only serve as anin vitroresource, and may have an important role to play in supporting failing liver function in humans.
Collapse
Affiliation(s)
- B Lucendo-Villarin
- Centre for Regenerative Medicine, University of Edinburgh, Edinburgh EH16 4UU, United Kingdom
- Both authors contributed equally to this manuscript
| | - J Meseguer-Ripolles
- Centre for Regenerative Medicine, University of Edinburgh, Edinburgh EH16 4UU, United Kingdom
- Both authors contributed equally to this manuscript
| | - J Drew
- CRUK Beatson Institute, Garscube Estate, Switchback Road, Bearsden, Glasgow G61 1BD, United Kingdom
| | - L Fischer
- Centre for Regenerative Medicine, University of Edinburgh, Edinburgh EH16 4UU, United Kingdom
| | - E Ma
- CRUK Beatson Institute, Garscube Estate, Switchback Road, Bearsden, Glasgow G61 1BD, United Kingdom
- Institute of Cancer Sciences, University of Glasgow, Garscube Campus, G61 1BD, United Kingdom
| | - O Flint
- Centre for Regenerative Medicine, University of Edinburgh, Edinburgh EH16 4UU, United Kingdom
| | - K J Simpson
- Scottish Liver Transplant Unit, Royal Infirmary, Edinburgh EH16 4SA, United Kingdom
| | - L M Machesky
- CRUK Beatson Institute, Garscube Estate, Switchback Road, Bearsden, Glasgow G61 1BD, United Kingdom
- Institute of Cancer Sciences, University of Glasgow, Garscube Campus, G61 1BD, United Kingdom
| | - J C Mountford
- SNBTS, 52 Research Avenue North, Heriot-Watt Research Park, Edinburgh EH14 4BE, United Kingdom
| | - D C Hay
- Centre for Regenerative Medicine, University of Edinburgh, Edinburgh EH16 4UU, United Kingdom
- Author to whom any correspondence should be addressed
| |
Collapse
|
35
|
Ullah I, Seo K, Wi H, Kim Y, Lee S, Ock SA. Induction of the differentiation of porcine bone marrow mesenchymal stem cells into premature hepatocyte-like cells in an indirect coculture system with primary hepatocytes. Anim Cells Syst (Seoul) 2020; 24:289-298. [PMID: 33209203 PMCID: PMC7646558 DOI: 10.1080/19768354.2020.1823473] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Liver transplantation is currently the only option for patients with end-stage liver disease. Thus, other alternate therapeutic strategies are needed. Bone marrow mesenchymal stem cells (BM-MSCs) are nonhematopoietic cells present in the bone marrow stroma that serve as precursors cells for various other cells. In this study, we evaluated the differentiation of porcine BM-MSCs into hepatocyte-like cells using three types of culture systems: hepatic induction medium (HIM), HIM/primary hepatocyte culture supernatant (HCS; 1:1 ratio), and a hepatocyte coculture system (HCCS; primary hepatocytes in the upper chamber, and BM-MSCs in the lower chamber). Primary hepatocytes were isolated from anesthetized healthy 1-month-old pigs by enzymatic digestion. Hepatic-specific marker expression (albumin [ALB], transferrin [TF], α-fetoprotein [AFP]), glycogen storage, low-density lipoprotein, and indocyanine green uptake were evaluated. Upregulation of hepatic-specific markers (ALB, TF, and AFP) was observed by real-time polymerase chain reaction in the HCCS group. Periodic acid-Schiff staining revealed enhanced glycogen storage in hepatocyte-like cells from the HCCS group compared with that from the HIM/HCS group. Furthermore, hepatocyte like-cells in the HCCS group showed improved LDL and ICG uptake than those in the other groups. Overall, our current study revealed that indirect coculture of primary hepatocytes and BM-MSCs enhanced the differentiation efficacy of BM-MSCs into hepatocyte-like cells by unknown useful soluble factors, including paracrine factors.
Collapse
Affiliation(s)
- Imran Ullah
- Animal Biotechnology Division, National Institute of Animal Science, Rural Development Administration, Wanju-gun, Republic of Korea.,Department of Biochemistry, Quaid-i-Azam University, Islamabad, Pakistan
| | - Kangmin Seo
- Animal Biotechnology Division, National Institute of Animal Science, Rural Development Administration, Wanju-gun, Republic of Korea
| | - Hayeon Wi
- Animal Biotechnology Division, National Institute of Animal Science, Rural Development Administration, Wanju-gun, Republic of Korea
| | - Youngim Kim
- Animal Biotechnology Division, National Institute of Animal Science, Rural Development Administration, Wanju-gun, Republic of Korea
| | - Seunghoon Lee
- Animal Biotechnology Division, National Institute of Animal Science, Rural Development Administration, Wanju-gun, Republic of Korea
| | - Sun A Ock
- Animal Biotechnology Division, National Institute of Animal Science, Rural Development Administration, Wanju-gun, Republic of Korea
| |
Collapse
|
36
|
Okuyama S, Kawamura F, Kubiura M, Tsuji S, Osaki M, Kugoh H, Oshimura M, Kazuki Y, Tada M. Real-time fluorometric evaluation of hepatoblast proliferation in vivo and in vitro using the expression of CYP3A7 coding for human fetus-specific P450. Pharmacol Res Perspect 2020; 8:e00642. [PMID: 32886454 PMCID: PMC7507068 DOI: 10.1002/prp2.642] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 07/10/2020] [Accepted: 07/13/2020] [Indexed: 12/18/2022] Open
Abstract
The fields of drug discovery and regenerative medicine require large numbers of adult human primary hepatocytes. For this purpose, it is desirable to use hepatocyte-like cells (HLCs) differentiated from human pluripotent stem cells (PSCs). Premature hepatoblast-like cells (HB-LCs) differentiated from PSCs provide an intermediate source and steady supply of newly mature HLCs. To develop an efficient HB-LC induction method, we constructed a red fluorescent reporter, CYP3A7R, in which DsRed is placed under the transcriptional control of CYP3A7 coding for a human fetus-type P450 enzyme. Before using this reporter in human cells, we created transgenic mice using mouse embryonic stem cells (ESCs) carrying a CYP3A7R transgene and confirmed that CYP3A7R was specifically expressed in fetal and newborn livers and reactivated in the adult liver in response to hepatic regeneration. Moreover, we optimized the induction procedure of HB-LCs from transgenic mouse ESCs using semi-quantitative fluorometric evaluation. Activation of Wnt signaling together with chromatin modulation prior to Activin A treatment greatly improved the induction efficiency of HB-LCs. BMP2 and 1.7% dimethyl sulfoxide induced selective proliferation of HB-LCs, which matured to HLCs. Therefore, CYP3A7R will provide a fluorometric evaluation system for high content screening of chemicals that induce HB-LC differentiation, hepatocyte regeneration, and hepatotoxicity when it is introduced into human PSCs.
Collapse
Affiliation(s)
- Shota Okuyama
- Stem Cells & Reprogramming LaboratoryDepartment of BiologyFaculty of ScienceToho UniversityFunabashiJapan
| | - Fumihiko Kawamura
- Stem Cells & Reprogramming LaboratoryDepartment of BiologyFaculty of ScienceToho UniversityFunabashiJapan
- Institute of Regenerative Medicine and BiofunctionGraduate School of Medical ScienceTottori UniversityYonagoJapan
| | - Musashi Kubiura
- Stem Cells & Reprogramming LaboratoryDepartment of BiologyFaculty of ScienceToho UniversityFunabashiJapan
| | - Saori Tsuji
- Chromosome Engineering Research CenterTottori UniversityYonagoJapan
| | - Mitsuhiko Osaki
- Chromosome Engineering Research CenterTottori UniversityYonagoJapan
| | - Hiroyuki Kugoh
- Institute of Regenerative Medicine and BiofunctionGraduate School of Medical ScienceTottori UniversityYonagoJapan
- Chromosome Engineering Research CenterTottori UniversityYonagoJapan
| | - Mitsuo Oshimura
- Chromosome Engineering Research CenterTottori UniversityYonagoJapan
| | - Yasuhiro Kazuki
- Institute of Regenerative Medicine and BiofunctionGraduate School of Medical ScienceTottori UniversityYonagoJapan
- Chromosome Engineering Research CenterTottori UniversityYonagoJapan
| | - Masako Tada
- Stem Cells & Reprogramming LaboratoryDepartment of BiologyFaculty of ScienceToho UniversityFunabashiJapan
| |
Collapse
|
37
|
iPSC-Derived Liver Organoids: A Journey from Drug Screening, to Disease Modeling, Arriving to Regenerative Medicine. Int J Mol Sci 2020; 21:ijms21176215. [PMID: 32867371 PMCID: PMC7503935 DOI: 10.3390/ijms21176215] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 08/20/2020] [Accepted: 08/23/2020] [Indexed: 12/11/2022] Open
Abstract
Liver transplantation is the most common treatment for patients suffering from liver failure that is caused by congenital diseases, infectious agents, and environmental factors. Despite a high rate of patient survival following transplantation, organ availability remains the key limiting factor. As such, research has focused on the transplantation of different cell types that are capable of repopulating and restoring liver function. The best cellular mix capable of engrafting and proliferating over the long-term, as well as the optimal immunosuppression regimens, remain to be clearly well-defined. Hence, alternative strategies in the field of regenerative medicine have been explored. Since the discovery of induced pluripotent stem cells (iPSC) that have the potential of differentiating into a broad spectrum of cell types, many studies have reported the achievement of iPSCs differentiation into liver cells, such as hepatocytes, cholangiocytes, endothelial cells, and Kupffer cells. In parallel, an increasing interest in the study of self-assemble or matrix-guided three-dimensional (3D) organoids have paved the way for functional bioartificial livers. In this review, we will focus on the recent breakthroughs in the development of iPSCs-based liver organoids and the major drawbacks and challenges that need to be overcome for the development of future applications.
Collapse
|
38
|
Soares-da-Silva F, Peixoto M, Cumano A, Pinto-do-Ó P. Crosstalk Between the Hepatic and Hematopoietic Systems During Embryonic Development. Front Cell Dev Biol 2020; 8:612. [PMID: 32793589 PMCID: PMC7387668 DOI: 10.3389/fcell.2020.00612] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Accepted: 06/19/2020] [Indexed: 12/14/2022] Open
Abstract
Hematopoietic stem cells (HSCs) generated during embryonic development are able to maintain hematopoiesis for the lifetime, producing all mature blood lineages. HSC transplantation is a widely used cell therapy intervention in the treatment of hematologic, autoimmune and genetic disorders. Its use, however, is hampered by the inability to expand HSCs ex vivo, urging for a better understanding of the mechanisms regulating their physiological expansion. In the adult, HSCs reside in the bone marrow, in specific microenvironments that support stem cell maintenance and differentiation. Conversely, while developing, HSCs are transiently present in the fetal liver, the major hematopoietic site in the embryo, where they expand. Deeper insights on the dynamics of fetal liver composition along development, and on how these different cell types impact hematopoiesis, are needed. Both, the hematopoietic and hepatic fetal systems have been extensively studied, albeit independently. This review aims to explore their concurrent establishment and evaluate to what degree they may cross modulate their respective development. As insights on the molecular networks that govern physiological HSC expansion accumulate, it is foreseeable that strategies to enhance HSC proliferation will be improved.
Collapse
Affiliation(s)
- Francisca Soares-da-Silva
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- Instituto Nacional de Engenharia Biomédica, Universidade do Porto, Porto, Portugal
- Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
- Lymphocytes and Immunity Unit, Immunology Department, Pasteur Institute, Paris, France
- INSERM U1223, Paris, France
- Université Paris Diderot, Sorbonne Paris Cité, Paris, France
| | - Márcia Peixoto
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- Instituto Nacional de Engenharia Biomédica, Universidade do Porto, Porto, Portugal
- Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
- Lymphocytes and Immunity Unit, Immunology Department, Pasteur Institute, Paris, France
- INSERM U1223, Paris, France
- Université Paris Diderot, Sorbonne Paris Cité, Paris, France
| | - Ana Cumano
- Lymphocytes and Immunity Unit, Immunology Department, Pasteur Institute, Paris, France
- INSERM U1223, Paris, France
- Université Paris Diderot, Sorbonne Paris Cité, Paris, France
| | - Perpetua Pinto-do-Ó
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- Instituto Nacional de Engenharia Biomédica, Universidade do Porto, Porto, Portugal
- Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
| |
Collapse
|
39
|
Wang X, Yang L, Wang YC, Xu ZR, Feng Y, Zhang J, Wang Y, Xu CR. Comparative analysis of cell lineage differentiation during hepatogenesis in humans and mice at the single-cell transcriptome level. Cell Res 2020; 30:1109-1126. [PMID: 32690901 PMCID: PMC7784864 DOI: 10.1038/s41422-020-0378-6] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Accepted: 06/23/2020] [Indexed: 12/17/2022] Open
Abstract
During embryogenesis, the liver is the site of hepatogenesis and hematopoiesis and contains many cell lineages derived from the endoderm and mesoderm. However, the characteristics and developmental programs of many of these cell lineages remain unclear, especially in humans. Here, we performed single-cell RNA sequencing of whole human and mouse fetal livers throughout development. We identified four cell lineage families of endoderm-derived, erythroid, non-erythroid hematopoietic, and mesoderm-derived non-hematopoietic cells, and defined the developmental pathways of the major cell lineage families. In both humans and mice, we identified novel markers of hepatic lineages and an ID3+ subpopulation of hepatoblasts as well as verified that hepatoblast differentiation follows the “default-directed” model. Additionally, we found that human but not mouse fetal hepatocytes display heterogeneity associated with expression of metabolism-related genes. We described the developmental process of erythroid progenitor cells during human and mouse hematopoiesis. Moreover, despite the general conservation of cell differentiation programs between species, we observed different cell lineage compositions during hematopoiesis in the human and mouse fetal livers. Taken together, these results reveal the dynamic cell landscape of fetal liver development and illustrate the similarities and differences in liver development between species, providing an extensive resource for inducing various liver cell lineages in vitro.
Collapse
Affiliation(s)
- Xin Wang
- Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, College of Life Sciences, Department of Human Anatomy, Histology, and Embryology, and School of Basic Medical Sciences, Peking University Health Science Center, Peking University, Beijing, 100871, China
| | - Li Yang
- Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, College of Life Sciences, Department of Human Anatomy, Histology, and Embryology, and School of Basic Medical Sciences, Peking University Health Science Center, Peking University, Beijing, 100871, China
| | - Yan-Chun Wang
- Haidian Maternal & Child Health Hospital, Beijing, 100080, China
| | - Zi-Ran Xu
- Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, College of Life Sciences, Department of Human Anatomy, Histology, and Embryology, and School of Basic Medical Sciences, Peking University Health Science Center, Peking University, Beijing, 100871, China
| | - Ye Feng
- Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, College of Life Sciences, Department of Human Anatomy, Histology, and Embryology, and School of Basic Medical Sciences, Peking University Health Science Center, Peking University, Beijing, 100871, China
| | - Jing Zhang
- Haidian Maternal & Child Health Hospital, Beijing, 100080, China
| | - Yi Wang
- Haidian Maternal & Child Health Hospital, Beijing, 100080, China
| | - Cheng-Ran Xu
- Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, College of Life Sciences, Department of Human Anatomy, Histology, and Embryology, and School of Basic Medical Sciences, Peking University Health Science Center, Peking University, Beijing, 100871, China.
| |
Collapse
|
40
|
Analysis of hiPSCs differentiation toward hepatocyte-like cells upon extended exposition to oncostatin. Differentiation 2020; 114:36-48. [PMID: 32563741 DOI: 10.1016/j.diff.2020.05.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 04/30/2020] [Accepted: 05/11/2020] [Indexed: 12/20/2022]
Abstract
The capability to produce and maintain functional human adult hepatocytes remains one of the major challenges for the use of in-vitro models toward liver cell therapy and industrial drug-screening applications. Among the suggested strategies to solve this issue, the use of human-induced pluripotent stem cells (hiPSCs), differentiated toward hepatocyte-like cells (HLCs) is promising. In this work, we propose a 31-day long protocol, that includes a final 14-day long phase of oncostatin treatment, as opposed to a 7-day treatment which led to the formation of a hepatic tissue functional for CYP1A2, CYP2B6, CYP2C8, CYP2D6, and CYP3A4. The production of albumin, as well as bile acid metabolism and transport, were also detected. Transcriptome profile comparisons and liver transcription factors (TFs) motif dynamics revealed increased expression of typical hepatic markers such as HNF1A and of important metabolic markers like PPARA. The performed analysis has allowed for the extraction of potential targets and pathways which would allow enhanced hepatic maturation in-vitro. From this investigation, NRF1 and SP3 appeared as transcription factors of importance. Complex epithelial-mesenchymal transition (EMT) and mesenchymal-epithelial transition (MET) patterns were also observed during the differentiation process. Moreover, whole transcriptome analysis highlighted a response typical of the one observed in liver regeneration and hepatocyte proliferation. While a complete maturation of hepatocytes was yet to be obtained, the results presented in this work provide new insights into the process of liver development and highlight potential targets aimed to improve in-vitro liver regeneration.
Collapse
|
41
|
Cotovio JP, Fernandes TG. Production of Human Pluripotent Stem Cell-Derived Hepatic Cell Lineages and Liver Organoids: Current Status and Potential Applications. Bioengineering (Basel) 2020; 7:E36. [PMID: 32283585 PMCID: PMC7356351 DOI: 10.3390/bioengineering7020036] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2020] [Revised: 04/03/2020] [Accepted: 04/07/2020] [Indexed: 02/06/2023] Open
Abstract
Liver disease is one of the leading causes of death worldwide, leading to the death of approximately 2 million people per year. Current therapies include orthotopic liver transplantation, however, donor organ shortage remains a great challenge. In addition, the development of novel therapeutics has been limited due to the lack of in vitro models that mimic in vivo liver physiology. Accordingly, hepatic cell lineages derived from human pluripotent stem cells (hPSCs) represent a promising cell source for liver cell therapy, disease modelling, and drug discovery. Moreover, the development of new culture systems bringing together the multiple liver-specific hepatic cell types triggered the development of hPSC-derived liver organoids. Therefore, these human liver-based platforms hold great potential for clinical applications. In this review, the production of the different hepatic cell lineages from hPSCs, including hepatocytes, as well as the emerging strategies to generate hPSC-derived liver organoids will be assessed, while current biomedical applications will be highlighted.
Collapse
Affiliation(s)
| | - Tiago G. Fernandes
- iBB-Institute for Bioengineering and Biosciences and Department of Bioengineering, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal;
| |
Collapse
|
42
|
Piquer-Garcia I, Campderros L, Taxerås SD, Gavaldà-Navarro A, Pardo R, Vila M, Pellitero S, Martínez E, Tarascó J, Moreno P, Villarroya J, Cereijo R, González L, Reyes M, Rodriguez-Fernández S, Vives-Pi M, Lerin C, Elks CM, Stephens JM, Puig-Domingo M, Villarroya F, Villena JA, Sánchez-Infantes D. A Role for Oncostatin M in the Impairment of Glucose Homeostasis in Obesity. J Clin Endocrinol Metab 2020; 105:5586710. [PMID: 31606738 PMCID: PMC7112982 DOI: 10.1210/clinem/dgz090] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Accepted: 10/02/2019] [Indexed: 12/19/2022]
Abstract
CONTEXT Oncostatin M (OSM) plays a key role in inflammation, but its regulation and function during obesity is not fully understood. OBJECTIVE The aim of this study was to evaluate the relationship of OSM with the inflammatory state that leads to impaired glucose homeostasis in obesity. We also assessed whether OSM immunoneutralization could revert metabolic disturbances caused by a high-fat diet (HFD) in mice. DESIGN 28 patients with severe obesity were included and stratified into two groups: (1) glucose levels <100 mg/dL and (2) glucose levels >100 mg/dL. White adipose tissue was obtained to examine OSM gene expression. Human adipocytes were used to evaluate the effect of OSM in the inflammatory response, and HFD-fed C57BL/6J mice were injected with anti-OSM antibody to evaluate its effects. RESULTS OSM expression was elevated in subcutaneous and visceral fat from patients with obesity and hyperglycemia, and correlated with Glut4 mRNA levels, serum insulin, homeostatic model assessment of insulin resistance, and inflammatory markers. OSM inhibited adipogenesis and induced inflammation in human adipocytes. Finally, OSM receptor knockout mice had increased Glut4 mRNA levels in adipose tissue, and OSM immunoneutralization resulted in a reduction of glucose levels and Ccl2 expression in adipose tissue from HFD-fed mice. CONCLUSIONS OSM contributes to the inflammatory state during obesity and may be involved in the development of insulin resistance.
Collapse
Affiliation(s)
- Irene Piquer-Garcia
- Department of Endocrinology and Nutrition, Germans Trias i Pujol Research Institute, Barcelona, Spain
| | - Laura Campderros
- Department of Biochemistry and Molecular Biomedicine, and Institute of Biomedicine, University of Barcelona, Barcelona, Spain
- BiomedicalResearch Center (Red Fisiopatología de la Obesidad y Nutrición) (CIBEROBN), ISCIII, Madrid, Spain
| | - Siri D Taxerås
- Department of Endocrinology and Nutrition, Germans Trias i Pujol Research Institute, Barcelona, Spain
| | - Aleix Gavaldà-Navarro
- Department of Biochemistry and Molecular Biomedicine, and Institute of Biomedicine, University of Barcelona, Barcelona, Spain
- BiomedicalResearch Center (Red Fisiopatología de la Obesidad y Nutrición) (CIBEROBN), ISCIII, Madrid, Spain
| | - Rosario Pardo
- Laboratory of Metabolism and Obesity, Vall d’Hebron Institut de Recerca, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - María Vila
- Laboratory of Metabolism and Obesity, Vall d’Hebron Institut de Recerca, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Silvia Pellitero
- Department of Endocrinology and Nutrition, Germans Trias i Pujol Research Institute, Barcelona, Spain
- BiomedicalResearch Center (Red Fisiopatología de la Diabetes y enfermedades metabólicas) (CIBERDEM), ISCIII, Madrid, Spain
| | - Eva Martínez
- Department of Endocrinology and Nutrition, Germans Trias i Pujol Research Institute, Barcelona, Spain
| | - Jordi Tarascó
- Department of Surgery, Germans Trias i Pujol Research Institute, Barcelona, Spain
| | - Pau Moreno
- Department of Surgery, Germans Trias i Pujol Research Institute, Barcelona, Spain
| | - Joan Villarroya
- Department of Biochemistry and Molecular Biomedicine, and Institute of Biomedicine, University of Barcelona, Barcelona, Spain
- Infectious Diseases Unit, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Rubén Cereijo
- Department of Biochemistry and Molecular Biomedicine, and Institute of Biomedicine, University of Barcelona, Barcelona, Spain
- BiomedicalResearch Center (Red Fisiopatología de la Obesidad y Nutrición) (CIBEROBN), ISCIII, Madrid, Spain
| | - Lorena González
- Department of Endocrinology and Nutrition, Germans Trias i Pujol Research Institute, Barcelona, Spain
| | - Marjorie Reyes
- Department of Endocrinology and Nutrition, Germans Trias i Pujol Research Institute, Barcelona, Spain
| | | | - Marta Vives-Pi
- BiomedicalResearch Center (Red Fisiopatología de la Diabetes y enfermedades metabólicas) (CIBERDEM), ISCIII, Madrid, Spain
- Immunology Section, Germans Trias i Pujol Research Institute, Barcelona, Spain
| | - Carles Lerin
- Endocrinology, Sant Joan de Déu Hospital, Barcelona, Spain
| | - Carrie M Elks
- Matrix Biology Laboratory, Pennington Biomedical Research Center, Baton Rouge, Louisiana
| | - Jacqueline M Stephens
- Adipocyte Biology Laboratory, Pennington Biomedical Research Center, Baton Rouge, Louisiana
| | - Manel Puig-Domingo
- Department of Endocrinology and Nutrition, Germans Trias i Pujol Research Institute, Barcelona, Spain
- BiomedicalResearch Center (Red Fisiopatología de la Diabetes y enfermedades metabólicas) (CIBERDEM), ISCIII, Madrid, Spain
| | - Francesc Villarroya
- Department of Biochemistry and Molecular Biomedicine, and Institute of Biomedicine, University of Barcelona, Barcelona, Spain
- BiomedicalResearch Center (Red Fisiopatología de la Obesidad y Nutrición) (CIBEROBN), ISCIII, Madrid, Spain
| | - Josep A Villena
- Laboratory of Metabolism and Obesity, Vall d’Hebron Institut de Recerca, Universitat Autònoma de Barcelona, Barcelona, Spain
- BiomedicalResearch Center (Red Fisiopatología de la Diabetes y enfermedades metabólicas) (CIBERDEM), ISCIII, Madrid, Spain
| | - David Sánchez-Infantes
- Department of Endocrinology and Nutrition, Germans Trias i Pujol Research Institute, Barcelona, Spain
- BiomedicalResearch Center (Red Fisiopatología de la Obesidad y Nutrición) (CIBEROBN), ISCIII, Madrid, Spain
- Correspondence and Reprint Requests: David Sánchez-Infantes, PhD, Obesity and Type 2 Diabetes: Adipose Tissue Biology Group Leader, Germans Trias i Pujol Research Institute (IGTP), Campus Can Ruti, Carretera de Can Ruti, Camí de les Escoles s/n 08916 Badalona, Barcelona, Spain. E-mail:
| |
Collapse
|
43
|
Arshad S, Naveed M, Ullia M, Javed K, Butt A, Khawar M, Amjad F. Targeting STAT-3 signaling pathway in cancer for development of novel drugs: Advancements and challenges. Genet Mol Biol 2020; 43:e20180160. [PMID: 32167126 PMCID: PMC7198026 DOI: 10.1590/1678-4685-gmb-2018-0160] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2018] [Accepted: 10/20/2018] [Indexed: 12/25/2022] Open
Abstract
Signal transducers and activators of transcription 3 (STAT-3) is a transcription
factor that regulates the gene expression of several target genes. These factors
are activated by the binding of cytokines and growth factors with STAT-3
specific receptors on cell membrane. Few years ago, STAT-3 was considered an
acute phase response element having several cellular functions such as
inflammation, cell survival, invasion, metastasis and proliferation, genetic
alteration, and angiogenesis. STAT-3 is activated by several types of
inflammatory cytokines, carcinogens, viruses, growth factors, and oncogenes.
Thus, the STAT3 pathway is a potential target for cancer therapeutics. Abnormal
STAT-3 activity in tumor development and cellular transformation can be targeted
by several genomic and pharmacological methodologies. An extensive review of the
literature has been conducted to emphasize the role of STAT-3 as a unique cancer
drug target. This review article discusses in detail the wide range of STAT-3
inhibitors that show antitumor effects both in vitro and
in vivo. Thus, targeting constitutive STAT-3 signaling is a
remarkable therapeutic methodology for tumor progression. Finally, current
limitations, trials and future perspectives of STAT-3 inhibitors are also
critically discussed.
Collapse
Affiliation(s)
- Sundas Arshad
- University of Lahore, Department of Allied Health Sciences, Gujrat Campus, Pakistan
| | - Muhammad Naveed
- University of Central Punjab, Faculty of life sciences, Department of Biotechnology, Lahore, Pakistan
| | - Mahad Ullia
- University of Gujrat, Department of Biochemistry and Biotechnology Sialkot sub Campus, Pakistan
| | - Khadija Javed
- University of Gujrat, Department of Biochemistry and Biotechnology Sialkot sub Campus, Pakistan
| | - Ayesha Butt
- University of Gujrat, Department of Biochemistry and Biotechnology Sialkot sub Campus, Pakistan
| | - Masooma Khawar
- University of Gujrat, Department of Biochemistry and Biotechnology Sialkot sub Campus, Pakistan
| | - Fazeeha Amjad
- University of Gujrat, Department of Biochemistry and Biotechnology Sialkot sub Campus, Pakistan
| |
Collapse
|
44
|
Akbari S, Arslan N, Senturk S, Erdal E. Next-Generation Liver Medicine Using Organoid Models. Front Cell Dev Biol 2019; 7:345. [PMID: 31921856 PMCID: PMC6933000 DOI: 10.3389/fcell.2019.00345] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Accepted: 12/03/2019] [Indexed: 12/24/2022] Open
Abstract
"Liver medicine" refers to all diagnostic and treatment strategies of diseases and conditions that cause liver failure directly or indirectly. Despite significant advances in the field of liver medicine in recent years, improved tools are needed to efficiently define the pathophysiology of liver diseases and provide effective therapeutic options to patients. Recently, organoid technology has been established as the state-of-the-art cell culture tool for studying human biology in health and disease. In general, organoids are simplified three-dimensional (3D) mini-organ structures that can be grown in a 3D matrix where the structural and functional aspects of real organs are efficiently recapitulated. The generation of organoids is facilitated by exogenous factors that regulate multiple signaling pathways and promote the self-renewal, proliferation, and differentiation of the cells to promote spontaneous self-organization and tissue-specific organogenesis. Newly established protocols suggest that liver-specific organoids can be derived from either pluripotent stem cells or liver-specific stem/progenitor cells. Today, robust and long-term cultures of organoids with the closest physiology to in vivo liver, in terms of cellular composition and function, open a new era in studying and understanding the disease pathology as well as high-throughput drug screening. Of note, these next-generation cell culture systems have immense potential to be further improved by genome editing and bioengineering technologies to foster the development of patient-specific therapeutic options for clinical applications. Here, we will discuss recent advances and challenges in the generation of human liver organoids and highlight emerging concepts for their potential applications in liver medicine.
Collapse
Affiliation(s)
| | - Nur Arslan
- İzmir Biomedicine and Genome Center, İzmir, Turkey
- Department of Pediatric Gastroenterology and Metabolism, Faculty of Medicine, Dokuz Eylul University, İzmir, Turkey
| | - Serif Senturk
- İzmir Biomedicine and Genome Center, İzmir, Turkey
- Department of Genome Sciences and Molecular Biotechnology, İzmir International Biomedicine and Genome Institute, Dokuz Eylul University, İzmir, Turkey
| | - Esra Erdal
- İzmir Biomedicine and Genome Center, İzmir, Turkey
- Department of Medical Biology and Genetics, Faculty of Medicine, Dokuz Eylul University, İzmir, Turkey
| |
Collapse
|
45
|
Li S, Huang SQ, Zhao YX, Ding YJ, Ma DJ, Ding QR. Derivation and applications of human hepatocyte-like cells. World J Stem Cells 2019; 11:535-547. [PMID: 31523372 PMCID: PMC6716086 DOI: 10.4252/wjsc.v11.i8.535] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Revised: 05/15/2019] [Accepted: 07/30/2019] [Indexed: 02/06/2023] Open
Abstract
Human hepatocyte-like cells (HLCs) derived from human pluripotent stem cells (hPSCs) promise a valuable source of cells with human genetic background, physiologically relevant liver functions, and unlimited supply. With over 10 years’ efforts in this field, great achievements have been made. HLCs have been successfully derived and applied in disease modeling, toxicity testing and drug discovery. Large cohorts of induced pluripotent stem cells-derived HLCs have been recently applied in studying population genetics and functional outputs of common genetic variants in vitro. This has offered a new paradigm for genome-wide association studies and possibly in vitro pharmacogenomics in the nearly future. However, HLCs have not yet been successfully applied in bioartificial liver devices and have only displayed limited success in cell transplantation. HLCs still have an immature hepatocyte phenotype and exist as a population with great heterogeneity, and HLCs derived from different hPSC lines display variable differentiation efficiency. Therefore, continuous improvement to the quality of HLCs, deeper investigation of relevant biological processes, and proper adaptation of recent advances in cell culture platforms, genome editing technology, and bioengineering systems are required before HLCs can fulfill the needs in basic and translational research. In this review, we summarize the discoveries, achievements, and challenges in the derivation and applications of HLCs.
Collapse
Affiliation(s)
- Shuang Li
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Shi-Qian Huang
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Yong-Xu Zhao
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
- College of Mechanical Engineering, Dongguan University of Technology, Dongguan 523808, Guangdong Province, China
| | - Yu-Jie Ding
- Department of Pharmacy, Mudanjiang Kang’an Hospital, Mudanjiang 157011, Heilongjiang Province, China
| | - Dan-Jun Ma
- College of Mechanical Engineering, Dongguan University of Technology, Dongguan 523808, Guangdong Province, China
| | - Qiu-Rong Ding
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China
| |
Collapse
|
46
|
Nakai S, Shibata I, Shitamichi T, Yamaguchi H, Takagi N, Inoue T, Nakagawa T, Kiyokawa J, Wakabayashi S, Miyoshi T, Higashi E, Ishida S, Shiraki N, Kume S. Collagen vitrigel promotes hepatocytic differentiation of induced pluripotent stem cells into functional hepatocyte-like cells. Biol Open 2019; 8:bio.042192. [PMID: 31182631 PMCID: PMC6679405 DOI: 10.1242/bio.042192] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Differentiation of stem cells to hepatocytes provides an unlimited supply of human hepatocytes and therefore has been vigorously studied. However, to date, the stem cell-derived hepatocytes were suggested to be of immature features. To obtain matured hepatocytes from stem cells, we tested the effect of culturing human-induced pluripotent stem (hiPS) cell-derived endoderm cells on collagen vitrigel membrane and compared with our previous reported nanofiber matrix. We cultured hiPS cell-derived endoderm cells on a collagen vitrigel membrane and examined the expression profiles, and tested the activity of metabolic enzymes. Gene expression profile analysis of hepatocytic differentiation markers revealed that upon culture on collagen vitrigel membrane, immature markers of AFP decreased, with a concomitant increase in the expression of mature hepatocyte transcription factors and mature hepatocyte markers such as ALB, ASGR1. Mature markers involved in liver functions, such as transporters, cytochrome P450 enzymes and phase II metabolic enzymes were also upregulated. We observed the upregulation of the liver markers for at least 2 weeks. Gene array profiling analysis revealed that hiPS cell-derived hepatocyte-like cells (hiPS-hep) resemble those of the primary hepatocytes. Functions of the CYP enzyme activities were tested in multi-institution and all revealed high CYP1A, CYP2C19, CYP2D6, CYP3A activity, which could be maintained for at least 2 weeks in culture. Taken together, the present approach identified that collagen vitrigel membrane provides a suitable environment for the generation of hepatocytes from hiPS cells that resemble many characteristics of primary human hepatocytes. Summary: We found that collagen vitrigel membrane used as scaffold potentiates differentiation of human induced pluripotent stem cells to differentiate into mature hepatocyte-like cells that exhibit mature functions of the hepatocytes.
Collapse
Affiliation(s)
- Shun Nakai
- School of Life Science and Technology, Tokyo Institute of Technology, 4259-B-25 Nagatsuta-cho, Midori-ku, Yokohama, Kanagawa 226-8501, Japan
| | - Ima Shibata
- School of Life Science and Technology, Tokyo Institute of Technology, 4259-B-25 Nagatsuta-cho, Midori-ku, Yokohama, Kanagawa 226-8501, Japan
| | - Takahiro Shitamichi
- School of Life Science and Technology, Tokyo Institute of Technology, 4259-B-25 Nagatsuta-cho, Midori-ku, Yokohama, Kanagawa 226-8501, Japan
| | - Hiroyuki Yamaguchi
- Isehara Research Laboratory, Technology and Development Division, Kanto Chemical Co., Inc., 21 Suzukawa, Isehara, Kanagawa 259-1146, Japan
| | - Nobuyuki Takagi
- Technology and Development Division, Kanto Chemical Co., Inc., 2-1, Nihonbashi Muromachi 2-chome, Chuo-ku, Tokyo 103-0022, Japan
| | - Tomoaki Inoue
- Research Division, Chugai Pharmaceutical Co. Ltd, 1-135 Komakado, Gotemba, Shizuoka 412-8513, Japan
| | - Toshito Nakagawa
- Research Division, Chugai Pharmaceutical Co. Ltd, 1-135 Komakado, Gotemba, Shizuoka 412-8513, Japan
| | - Jumpei Kiyokawa
- Research Division, Chugai Pharmaceutical Co. Ltd, 1-135 Komakado, Gotemba, Shizuoka 412-8513, Japan
| | - Satoshi Wakabayashi
- Pharmacokinetics and Metabolism, Drug Safety and Pharmacokinetics Laboratories, Taisho Pharmaceutical Co., Ltd, 1-403 Yoshino-cho, Saitama-shi, Saitama 330-8530, Japan
| | - Tomoya Miyoshi
- Toxicology and Pharmacokinetics Laboratories, Pharmaceutical Research Laboratories, Toray Industries, Inc., 6-10-1 Tebiro, Kamakura, Kanagawa 248-8555, Japan
| | - Eriko Higashi
- Toxicology and Pharmacokinetics Laboratories, Pharmaceutical Research Laboratories, Toray Industries, Inc., 6-10-1 Tebiro, Kamakura, Kanagawa 248-8555, Japan
| | - Seiichi Ishida
- Division of Pharmacology, National Institute of Health Science, 3-25-26 Tonomati, Kawasaki 210-9501, Japan
| | - Nobuaki Shiraki
- School of Life Science and Technology, Tokyo Institute of Technology, 4259-B-25 Nagatsuta-cho, Midori-ku, Yokohama, Kanagawa 226-8501, Japan
| | - Shoen Kume
- School of Life Science and Technology, Tokyo Institute of Technology, 4259-B-25 Nagatsuta-cho, Midori-ku, Yokohama, Kanagawa 226-8501, Japan
| |
Collapse
|
47
|
Ruiz D, Regnier SM, Kirkley AG, Hara M, Haro F, Aldirawi H, Dybala MP, Sargis RM. Developmental exposure to the endocrine disruptor tolylfluanid induces sex-specific later-life metabolic dysfunction. Reprod Toxicol 2019; 89:74-82. [PMID: 31260803 DOI: 10.1016/j.reprotox.2019.06.010] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 06/16/2019] [Accepted: 06/26/2019] [Indexed: 02/06/2023]
Abstract
Endocrine-disrupting chemicals (EDCs) are implicated in the developmental mis-programming of energy metabolism. This study examined the impact of combined gestational and lactational exposure to the fungicide tolylfluanid (TF) on metabolic physiology in adult offspring. C57BL/6 J dams received standard rodent chow or the same diet containing 67 mg/kg TF. Offspring growth and metabolism were assessed up to 22 weeks of age. TF-exposed offspring exhibited reduced weaning weight. Body weight among female offspring remained low throughout the study, while male offspring matched controls by 17 weeks of age. Female offspring exhibited reduced glucose tolerance, markedly enhanced systemic insulin sensitivity, reduced adiposity, and normal gluconeogenic capacity during adulthood. In contrast, male offspring exhibited impaired glucose tolerance with unchanged insulin sensitivity, no differences in adiposity, and increased gluconeogenic capacity. These data indicate that developmental exposure to TF induces sex-specific metabolic disruptions that recapitulate key aspects of other in utero growth restriction models.
Collapse
Affiliation(s)
- Daniel Ruiz
- Committee on Molecular Metabolism and Nutrition, Chicago, IL, United States; University of Chicago, Chicago, IL, United States
| | - Shane M Regnier
- Committee on Molecular Metabolism and Nutrition, Chicago, IL, United States; Pritzker School of Medicine, Chicago, IL, United States; University of Chicago, Chicago, IL, United States
| | - Andrew G Kirkley
- Committee on Molecular Pathogenesis and Molecular Medicine, Chicago, IL, United States; University of Chicago, Chicago, IL, United States
| | - Manami Hara
- Section of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Chicago, IL, United States; University of Chicago, Chicago, IL, United States
| | - Fidel Haro
- University of Chicago, Chicago, IL, United States
| | - Hani Aldirawi
- Department of Mathematics, Statistics, and Computer Science, University of Illinois at Chicago, Chicago, IL, United States
| | - Michael P Dybala
- Section of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Chicago, IL, United States
| | - Robert M Sargis
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Illinois at Chicago, Chicago, IL, United States.
| |
Collapse
|
48
|
Blau BJ, Miki T. The role of cellular interactions in the induction of hepatocyte polarity and functional maturation in stem cell-derived hepatic cells. Differentiation 2019; 106:42-48. [PMID: 30878880 DOI: 10.1016/j.diff.2019.02.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Revised: 02/25/2019] [Accepted: 02/28/2019] [Indexed: 02/08/2023]
Abstract
The unique microenvironment found within the liver in vivo plays a key role in the induction of functional maturation in the developing hepatocyte. During organogenesis, hepatocytes acquire a polar phenotype that allows them to perform their functions of bile production and transport, protein synthesis, metabolism, and detoxification simultaneously, independently, and efficiently. It is thought that the induction of polarity and functional maturation in hepatocytes is dependent on the complex interplay of cell-cell and cell-extracellular matrix (ECM) interactions. While this process is highly efficient in the human liver, it has been shown that hepatocytes rapidly lose their functions when placed in cell culture. This poses a challenge for the development of a bioartificial liver (BAL) support system, which utilizes a live cellular source to perform hepatic functions in the event of acute liver failure or primary nonfunction. However, once the molecular mechanisms underlying the induction of hepatocyte polarity are fully identified, it will be possible to develop highly functional hepatic cells from human pluripotent stem cells (hPSCs). This new cell line would be an ideal cellular source for a BAL system, as it would have both the functionality and longevity to support a patient through the entire clinical course of treatment. In this review, we explore the literature that has examined the potential mechanisms that induce polarity in the developing hepatocyte and discuss the future implications of this knowledge in a clinical setting from a bioengineering perspective.
Collapse
Affiliation(s)
- Brandon J Blau
- Department of Surgery, Keck School of Medicine, University of Southern California, USA
| | - Toshio Miki
- Department of Surgery, Keck School of Medicine, University of Southern California, USA.
| |
Collapse
|
49
|
Watanabe Y, Tsuchiya A, Seino S, Kawata Y, Kojima Y, Ikarashi S, Starkey Lewis PJ, Lu W, Kikuta J, Kawai H, Yamagiwa S, Forbes SJ, Ishii M, Terai S. Mesenchymal Stem Cells and Induced Bone Marrow-Derived Macrophages Synergistically Improve Liver Fibrosis in Mice. Stem Cells Transl Med 2019; 8:271-284. [PMID: 30394698 PMCID: PMC6392382 DOI: 10.1002/sctm.18-0105] [Citation(s) in RCA: 90] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2018] [Accepted: 09/22/2018] [Indexed: 02/06/2023] Open
Abstract
We describe a novel therapeutic approach for cirrhosis using mesenchymal stem cells (MSCs) and colony-stimulating factor-1-induced bone marrow-derived macrophages (id-BMMs) and analyze the mechanisms underlying fibrosis improvement and regeneration. Mouse MSCs and id-BMMs were cultured from mouse bone marrow and their interactions analyzed in vitro. MSCs, id-BMMs, and a combination therapy using MSCs and id-BMMs were administered to mice with CCl4 -induced cirrhosis. Fibrosis regression, liver regeneration, and liver-migrating host cells were evaluated. Administered cell behavior was also tracked by intravital imaging. In coculture, MSCs induced switching of id-BMMs toward the M2 phenotype with high phagocytic activity. In vivo, the combination therapy reduced liver fibrosis (associated with increased matrix metalloproteinases expression), increased hepatocyte proliferation (associated with increased hepatocyte growth factor, vascular endothelial growth factor, and oncostatin M in the liver), and reduced blood levels of liver enzymes, more effectively than MSCs or id-BMMs monotherapy. Intravital imaging showed that after combination cell administration, a large number of id-BMMs, which phagocytosed hepatocyte debris and were retained in the liver for more than 7 days, along with a few MSCs, the majority of which were trapped in the lung, migrated to the fibrotic area in the liver. Host macrophages and neutrophils infiltrated after combination therapy and contributed to liver fibrosis regression and promoted regeneration along with administered cells. Indirect effector MSCs and direct effector id-BMMs synergistically improved cirrhosis along with host cells in mice. These studies pave the way for new treatments for cirrhosis. Stem Cells Translational Medicine 2019;8:271&284.
Collapse
Affiliation(s)
- Yusuke Watanabe
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental SciencesNiigata UniversityNiigataJapan
| | - Atsunori Tsuchiya
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental SciencesNiigata UniversityNiigataJapan
| | - Satoshi Seino
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental SciencesNiigata UniversityNiigataJapan
| | - Yuzo Kawata
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental SciencesNiigata UniversityNiigataJapan
| | - Yuichi Kojima
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental SciencesNiigata UniversityNiigataJapan
| | - Shunzo Ikarashi
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental SciencesNiigata UniversityNiigataJapan
| | - Philip J. Starkey Lewis
- Medical Research Council Centre for Regenerative MedicineThe University of EdinburghEdinburghUnited Kingdom
| | - Wei‐Yu Lu
- Medical Research Council Centre for Regenerative MedicineThe University of EdinburghEdinburghUnited Kingdom
| | - Junichi Kikuta
- Department of Immunology and Cell Biology, Graduate School of Medicine and Frontier BiosciencesOsaka UniversitySuitaJapan
| | - Hirokazu Kawai
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental SciencesNiigata UniversityNiigataJapan
| | - Satoshi Yamagiwa
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental SciencesNiigata UniversityNiigataJapan
| | - Stuart J. Forbes
- Medical Research Council Centre for Regenerative MedicineThe University of EdinburghEdinburghUnited Kingdom
| | - Masaru Ishii
- Department of Immunology and Cell Biology, Graduate School of Medicine and Frontier BiosciencesOsaka UniversitySuitaJapan
| | - Shuji Terai
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental SciencesNiigata UniversityNiigataJapan
| |
Collapse
|
50
|
Tsuchiya A, Suda T, Oda C, Kimura A, Hosaka K, Kimura N, Tominaga K, Hayashi K, Takamura M, Terai S. EpCAM- and/or NCAM-Expressing Hepatocellular Carcinoma in Which Behavior of Hepatic Progenitor Cell Marker-Positive Cells Are Followed. Case Rep Gastroenterol 2019; 13:118-124. [PMID: 31011311 PMCID: PMC6465753 DOI: 10.1159/000498913] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Accepted: 02/14/2019] [Indexed: 02/05/2023] Open
Abstract
Hepatic progenitor cell (HPC) marker-positive hepatocellular carcinomas (HCCs) have recently been extensively analyzed, and their prognosis has been reported as poor compared to HPC marker-negative HCCs. However, previous studies have analyzed the existence of HPC marker-positive cancer cells only in primary lesions, as well as the recurrence rate and prognosis of such tumors. Here, we are the first to report the behavior of HPC marker-positive cancer cells during vascular invasion and metastasis of an HCC. We concurrently analyzed EpCAM- and/or NCAM-expressing cancer cells in the primary, vascular invasion, and metastatic lesions of an HCC. An HCC which includes EpCAM- and/or NCAM-expressing cancer cells has not been previously reported. EpCAM- and/or NCAM-positive cancer cells invaded the vessels and formed heterogeneous populations of these HPC marker-positive cancer cells with HPC marker-negative cancer cells. The frequency of HPC marker-positive cancer colonies and cells in vessels was higher than that in the primary HCC. In the metastatic lesions, EpCAM-positive cancer cells were more frequently detected than NCAM-positive cancer cells, indicating that EpCAM may be more important than NCAM for cancer cell settlement in the metastatic lesions. Furthermore, bigger metastatic tumors tended to include HPC marker-positive cancer cells, suggesting that HPC marker-positive cancer cells have a growth advantage in the metastatic lesions. These results showed that HPC marker-positive cancer cells would be important for vascular invasion and metastasis and suggested that HPC marker-positive cancer cells are an important target in HCC treatment.
Collapse
Affiliation(s)
- Atsunori Tsuchiya
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Science, Niigata University, Niigata, Japan
| | - Takeshi Suda
- Department of Gastroenterology and Hepatology, Uonuma Institute of Community Medicine, Niigata Medical and Dental Hospital, Niigata, Japan
| | - Chiyumi Oda
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Science, Niigata University, Niigata, Japan
| | - Atsushi Kimura
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Science, Niigata University, Niigata, Japan
| | - Kazunori Hosaka
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Science, Niigata University, Niigata, Japan
| | - Naruhiro Kimura
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Science, Niigata University, Niigata, Japan
| | - Kentaro Tominaga
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Science, Niigata University, Niigata, Japan
| | - Kazunao Hayashi
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Science, Niigata University, Niigata, Japan
| | - Masaaki Takamura
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Science, Niigata University, Niigata, Japan
| | - Shuji Terai
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Science, Niigata University, Niigata, Japan
| |
Collapse
|