1
|
Zhu H, Zheng M, He H, Lei H, Tai W, Yang J, Song Z. Development and external validation of an early prediction model to identify irresponsive patients and prognosis of UDCA treatment in primary biliary cholangitis. Sci Rep 2024; 14:31369. [PMID: 39732944 PMCID: PMC11682153 DOI: 10.1038/s41598-024-82854-1] [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: 08/30/2024] [Accepted: 12/09/2024] [Indexed: 12/30/2024] Open
Abstract
Ursodeoxycholic acid (UDCA) is the first-line treatment for primary biliary cholangitis (PBC), but 20-40% of patients do not respond well to UDCA. We aimed to develop and validate a prognostic model for the early prediction of patients who nonresponse to UDCA. This retrospective analysis was conducted among patients with primary biliary cholangitis(N = 257) to develop a predictive model for early-stage nonresponse to ursodeoxycholic acid (UDCA) therapy. The model's reliability was subsequently confirmed through external validation in an independent cohort(N = 71). Multivariate cox regression analysis was used to evaluate variables that were significant in the univariate analysis. Total cholesterol, alkaline phosphatase (ALP), and neutrophil-to-lymphocyte ratio (NLR) were the three independent risk factors associated with early biochemical nonresponse to UDCA treatment. Based on these factors, we established a predictive model that possessed good discriminative ability, as reflected by an AUC of 0.862(95%CI = 0.813-0.911). The ROC curve of the external validation set calculated the AUC of 0.916(95%CI:0.823-1.000). In summary, we developed an early predictive model that could identify potential nonresponse factors to UDCA at baseline, which could facilitate risk evaluation and stratification for PBC patients. The NLR and total cholesterol provided a supplementary means for effectively managing PBC patients.
Collapse
Affiliation(s)
- Huiling Zhu
- Department of Gastroenterology, Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Mengyao Zheng
- Department of Gastroenterology, Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Haiyu He
- Department of Gastroenterology, Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Hongtao Lei
- School of Public Health, Kunming Medical University, Kunming, China
| | - Wenlin Tai
- Clinical Lab, Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Jinhui Yang
- Department of Gastroenterology, Second Affiliated Hospital of Kunming Medical University, Kunming, China.
| | - Zhengji Song
- The First People's Hospital of Yunnan Province, Kunming, China.
| |
Collapse
|
2
|
Carolina E, Kuse Y, Okumura A, Aoshima K, Tadokoro T, Matsumoto S, Kanai E, Okumura T, Kasai T, Yamabe S, Nishikawa Y, Yamaguchi K, Furukawa Y, Tanimizu N, Taniguchi H. Generation of human iPSC-derived 3D bile duct within liver organoid by incorporating human iPSC-derived blood vessel. Nat Commun 2024; 15:7424. [PMID: 39198465 PMCID: PMC11358266 DOI: 10.1038/s41467-024-51487-3] [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] [Accepted: 08/06/2024] [Indexed: 09/01/2024] Open
Abstract
In fetal development, tissue interaction such as the interplay between blood vessel (BV) and epithelial tissue is crucial for organogenesis. Here we recapitulate the spatial arrangement between liver epithelial tissue and the portal vein to observe the formation of intrahepatic bile ducts (BDs) from human induced pluripotent stem cells (hiPSC). We co-culture hiPSC-liver progenitors on the artificial BV consisting of immature smooth muscle cells and endothelial cells, both derived from hiPSCs. After 3 weeks, liver progenitors within hiPSC-BV-incorporated liver organoids (BVLO) differentiate to cholangiocytes and acquire epithelial characteristics, including intercellular junctions, microvilli on the apical membrane, and secretory functions. Furthermore, liver surface transplanted-BVLO temporarily attenuates cholestatic injury symptoms. Single cell RNA sequence analysis suggests that BD interact with the BV in BVLO through TGFβ and Notch pathways. Knocking out JAG1 in hiPSC-BV significantly attenuates bile duct formation, highlighting BVLO potential as a model for Alagille syndrome, a congenital biliary disease. Overall, we develop a novel 3D co-culture method that successfully establishes functional human BDs by emulating liver epithelial-BV interaction.
Collapse
Affiliation(s)
- Erica Carolina
- Division of Regenerative Medicine, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa, Chiba, Japan
| | - Yoshiki Kuse
- Division of Regenerative Medicine, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Ayumu Okumura
- Division of Regenerative Medicine, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa, Chiba, Japan
| | - Kenji Aoshima
- Division of Regenerative Medicine, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa, Chiba, Japan
| | - Tomomi Tadokoro
- Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, Kanagawa, Japan
| | - Shinya Matsumoto
- Division of Regenerative Medicine, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Eriko Kanai
- Division of Regenerative Medicine, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Takashi Okumura
- Division of Regenerative Medicine, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Toshiharu Kasai
- Division of Regenerative Medicine, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Souichiro Yamabe
- Division of Regenerative Medicine, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa, Chiba, Japan
| | - Yuji Nishikawa
- Division of Tumor Pathology, Department of Pathology, Asahikawa Medical University, Asahikawa, Hokkaido, Japan
| | - Kiyoshi Yamaguchi
- Division of Clinical Genome Research, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Yoichi Furukawa
- Division of Clinical Genome Research, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Naoki Tanimizu
- Division of Regenerative Medicine, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan.
| | - Hideki Taniguchi
- Division of Regenerative Medicine, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan.
- Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, Kanagawa, Japan.
| |
Collapse
|
3
|
Li CZ, Ogawa H, Ng SS, Chen X, Kishimoto E, Sakabe K, Fukami A, Hu YC, Mayhew CN, Hellmann J, Miethke A, Tasnova NL, Blackford SJ, Tang ZM, Syanda AM, Ma L, Xiao F, Sambrotta M, Tavabie O, Soares F, Baker O, Danovi D, Hayashi H, Thompson RJ, Rashid ST, Asai A. Human iPSC-derived hepatocyte system models cholestasis with tight junction protein 2 deficiency. JHEP Rep 2022; 4:100446. [PMID: 35284810 PMCID: PMC8904612 DOI: 10.1016/j.jhepr.2022.100446] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 01/10/2022] [Accepted: 01/14/2022] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND & AIMS The truncating mutations in tight junction protein 2 (TJP2) cause progressive cholestasis, liver failure, and hepatocyte carcinogenesis. Due to the lack of effective model systems, there are no targeted medications for the liver pathology with TJP2 deficiency. We leveraged the technologies of patient-specific induced pluripotent stem cells (iPSC) and CRISPR genome-editing, and we aim to establish a disease model which recapitulates phenotypes of patients with TJP2 deficiency. METHODS We differentiated iPSC to hepatocyte-like cells (iHep) on the Transwell membrane in a polarized monolayer. Immunofluorescent staining of polarity markers was detected by a confocal microscope. The epithelial barrier function and bile acid transport of bile canaliculi were quantified between the two chambers of Transwell. The morphology of bile canaliculi was measured in iHep cultured in the Matrigel sandwich system using a fluorescent probe and live-confocal imaging. RESULTS The iHep differentiated from iPSC with TJP2 mutations exhibited intracellular inclusions of disrupted apical membrane structures, distorted canalicular networks, altered distribution of apical and basolateral markers/transporters. The directional bile acid transport of bile canaliculi was compromised in the mutant hepatocytes, resembling the disease phenotypes observed in the liver of patients. CONCLUSIONS Our iPSC-derived in vitro hepatocyte system revealed canalicular membrane disruption in TJP2 deficient hepatocytes and demonstrated the ability to model cholestatic disease with TJP2 deficiency to serve as a platform for further pathophysiologic study and drug discovery. LAY SUMMARY We investigated a genetic liver disease, progressive familial intrahepatic cholestasis (PFIC), which causes severe liver disease in newborns and infants due to a lack of gene called TJP2. By using cutting-edge stem cell technology and genome editing methods, we established a novel disease modeling system in cell culture experiments. Our experiments demonstrated that the lack of TJP2 induced abnormal cell polarity and disrupted bile acid transport. These findings will lead to the subsequent investigation to further understand disease mechanisms and develop an effective treatment.
Collapse
Key Words
- ALB, albumin
- ASGR2, asialoglycoprotein receptor 2
- ATP1a1, ATPases subunit alpha-1
- BMP4, bone morphogenetic protein 4
- BSA-FAF, bovine serum albumin fatty acid-free
- BSEP, bile salt export pump
- Bile acid transport
- CDFDA, 5-(and-6)-carboxy-2′,7′-dichlorofluorescein
- Cellular polarity
- DE, definitive endoderm
- DILI, drug-induced liver injury
- FGF2, fibroblast growth factor 2
- GCA, glycocholate
- GCDCA, glycochenodeoxycholate
- HCM, Hepatocyte Culture Medium
- HE, hepatic endodermal
- HGF, hepatocyte growth factor
- HNF4a, hepatic nuclear factor 4a
- MDCKII, Madin–Darby canine kidney II
- MRP2, multidrug resistance-associated protein 2
- NTCP, Na+-TCA cotransporter
- PFIC (progressive familial intrahepatic cholestasis)
- PFIC, progressive familial intrahepatic cholestasis
- PI, propidium iodide
- RT-qPCR, quantitative reverse transcription PCR
- TCA, taurocholic acid
- TCDCA, taurochenodeoxycholate
- TEER, transepithelial electrical resistance
- TEM, transmission electron microscopy
- TJP1, tight junction protein 1
- TJP2, tight junction protein 2
- iHep, iPSC-derived hepatocytes
- iPSC, induced pluripotent stem cell
- sgRNA, single-guide RNA
- ssODN, single-stranded oligonucleotide-DNA
Collapse
Affiliation(s)
- Chao Zheng Li
- Centre for Stem Cells and Regenerative Medicine, King’s College London, London, UK
| | - Hiromi Ogawa
- Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| | - Soon Seng Ng
- Centre for Stem Cells and Regenerative Medicine, King’s College London, London, UK
| | - Xindi Chen
- Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| | - Eriko Kishimoto
- Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| | - Kokoro Sakabe
- Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| | - Aiko Fukami
- Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| | - Yueh-Chiang Hu
- Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| | | | - Jennifer Hellmann
- Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
- Department of Paediatrics, The University of Cincinnati, Cincinnati, OH, USA
| | - Alexander Miethke
- Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
- Department of Paediatrics, The University of Cincinnati, Cincinnati, OH, USA
| | - Nahrin L. Tasnova
- Centre for Stem Cells and Regenerative Medicine, King’s College London, London, UK
| | | | - Zu Ming Tang
- Stem Cell Hotel, King’s College London, London, UK
| | - Adam M. Syanda
- Centre for Stem Cells and Regenerative Medicine, King’s College London, London, UK
| | - Liang Ma
- Centre for Stem Cells and Regenerative Medicine, King’s College London, London, UK
| | - Fang Xiao
- Centre for Stem Cells and Regenerative Medicine, King’s College London, London, UK
| | - Melissa Sambrotta
- Institute of Liver Studies King’s College London, London, United Kingdom
| | - Oliver Tavabie
- Institute of Liver Studies King’s College London, London, United Kingdom
| | | | - Oliver Baker
- Genome Editing and Embryology Core Facility, King’s College London, London, UK
| | - Davide Danovi
- Centre for Stem Cells and Regenerative Medicine, King’s College London, London, UK
| | - Hisamitsu Hayashi
- Graduate School of Pharmaceutical Science, The University of Tokyo, Tokyo, Japan
| | | | - S. Tamir Rashid
- Centre for Stem Cells and Regenerative Medicine, King’s College London, London, UK
| | - Akihiro Asai
- Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
- Department of Paediatrics, The University of Cincinnati, Cincinnati, OH, USA
| |
Collapse
|
4
|
Blaszkiewicz J, Duncan SA. Advancements in Disease Modeling and Drug Discovery Using iPSC-Derived Hepatocyte-like Cells. Genes (Basel) 2022; 13:573. [PMID: 35456379 PMCID: PMC9030659 DOI: 10.3390/genes13040573] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 03/17/2022] [Accepted: 03/21/2022] [Indexed: 02/05/2023] Open
Abstract
Serving as the metabolic hub of the human body, the liver is a vital organ that performs a variety of important physiological functions. Although known for its regenerative potential, it remains vulnerable to a variety of diseases. Despite decades of research, liver disease remains a leading cause of mortality in the United States with a multibillion-dollar-per-year economic burden. Prior research with model systems, such as primary hepatocytes and murine models, has provided many important discoveries. However, progress has been impaired by numerous obstacles associated with these models. In recent years, induced pluripotent stem cell (iPSC)-based systems have emerged as advantageous platforms for studying liver disease. Benefits, including preserved differentiation and physiological function, amenability to genetic manipulation via tools such as CRISPR/Cas9, and availability for high-throughput screening, make these systems increasingly attractive for both mechanistic studies of disease and the identification of novel therapeutics. Although limitations exist, recent studies have made progress in ameliorating these issues. In this review, we discuss recent advancements in iPSC-based models of liver disease, including improvements in model system construction as well as the use of high-throughput screens for genetic studies and drug discovery.
Collapse
Affiliation(s)
| | - Stephen A. Duncan
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC 29425, USA;
| |
Collapse
|
5
|
Abstract
The possibility of reprogramming human somatic cells to pluripotency has opened unprecedented opportunities for creating genuinely human experimental models of disease. Inborn errors of metabolism (IEMs) constitute a greatly heterogeneous class of diseases that appear, in principle, especially suited to be modeled by iPSC-based technology. Indeed, dozens of IEMs have already been modeled to some extent using patient-specific iPSCs. Here, we review the advantages and disadvantages of iPSC-based disease modeling in the context of IEMs, as well as particular challenges associated to this approach, together with solutions researchers have proposed to tackle them. We have structured this review around six lessons that we have learnt from those previous modeling efforts, and that we believe should be carefully considered by researchers wishing to embark in future iPSC-based models of IEMs.
Collapse
Affiliation(s)
- Rubén Escribá
- Regenerative Medicine Program, Institut d'Investigació Biomèdica de Bellvitge - IDIBELL, L'Hospitalet de Llobregat, Spain
- Program for Clinical Translation of Regenerative Medicine in Catalonia - P-[CMRC], L'Hospitalet de Llobregat, Spain
- Center for Networked Biomedical Research On Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Barcelona, Spain
| | - Raquel Ferrer-Lorente
- Regenerative Medicine Program, Institut d'Investigació Biomèdica de Bellvitge - IDIBELL, L'Hospitalet de Llobregat, Spain
- Program for Clinical Translation of Regenerative Medicine in Catalonia - P-[CMRC], L'Hospitalet de Llobregat, Spain
- Center for Networked Biomedical Research On Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Barcelona, Spain
| | - Ángel Raya
- Regenerative Medicine Program, Institut d'Investigació Biomèdica de Bellvitge - IDIBELL, L'Hospitalet de Llobregat, Spain.
- Program for Clinical Translation of Regenerative Medicine in Catalonia - P-[CMRC], L'Hospitalet de Llobregat, Spain.
- Center for Networked Biomedical Research On Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Barcelona, Spain.
- Institució Catalana de Recerca I Estudis Avançats (ICREA), Barcelona, Spain.
| |
Collapse
|