1
|
Allegretta C, Montemitro E, Sgobba MN, Capurro V, Pesce E, Ciciriello F, La Bella G, Rossito M, Tuccio V, Arena F, Gunawardena TN, Guerra L, Pedemonte N, Capitanio N, Piccoli C, Laselva O. Deleterious effect of Pseudomonas aeruginosa on F508del-CFTR rescued by elexacaftor/tezacaftor/ivacaftor is clinical strain-dependent in patient-derived nasal cells. ERJ Open Res 2025; 11:00970-2024. [PMID: 40432819 PMCID: PMC12107384 DOI: 10.1183/23120541.00970-2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Accepted: 11/04/2024] [Indexed: 05/29/2025] Open
Abstract
Background The triple cystic fibrosis transmembrane conductance regulator (CFTR) modulators combination elexacaftor/tezacaftor/ivacaftor (ETI) has been approved for people with cystic fibrosis (pwCF) bearing at least one F508del allele. Despite the development of CFTR modulators having dramatically improved respiratory outcomes in pwCF, clinical studies have showed variable responses to this drug formulation. Of note, airway inflammation and bacterial colonisation persist in the upper and lower respiratory tract even in ETI-treated patients. Methods We first tested the clinical exoproducts (EXO) of Pseudomonas aeruginosa isolated from 15 CF patients in wild-type (WT) and F508del-CFTR CF bronchial epithelial (CFBE) cells. We were then prompted to evaluate the effects of EXO in ex-vivo patient-derived tissues. Therefore, we cultured primary nasal epithelial cells (HNECs) with EXO isolated from the corresponding pwCF to mimic the native status of CF airway. Results We found that EXO variably decreased WT-, F508del- and ETI-dependent F508del-CFTR function and increased proinflammatory cytokines and reactive oxygen species (ROS) levels in a clinical strain-specific manner. Similarly, we observed a variable reduction of F508del-CFTR function in presence or absence of ETI and upregulation of proinflammatory cytokines and ROS levels. Interestingly, HNECs treated with EXO isolated from the corresponding donor and three different pwCF showed a variable reduction of ETI-dependent F508del-CFTR function mainly due to clinical strains with limited effect of patient background. Furthermore, we demonstrated that ETI pretreatment decreased the cytokines and ROS levels down to the levels of uninfected cells. Conclusion These preclinical studies suggest that in vitro screening of patient-specific response to CFTR modulators under infection/inflammation conditions could prove to be a valuable tool to enhance the prediction of clinical response.
Collapse
Affiliation(s)
- Caterina Allegretta
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Enza Montemitro
- Cystic Fibrosis Center, Specialistic Pediatrics Department, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Maria Noemi Sgobba
- Department of Biosciences, Biotechnologies and Environment, University of Bari “Aldo Moro”, Bari, Italy
| | - Valeria Capurro
- UOC Genetica Medica, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Emanuela Pesce
- UOC Genetica Medica, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Fabiana Ciciriello
- Cystic Fibrosis Center, Specialistic Pediatrics Department, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Gianfranco La Bella
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Martina Rossito
- Cystic Fibrosis Diagnostic Unit, Laboratory and Specialistic Pediatrics Departments, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Vanessa Tuccio
- Cystic Fibrosis Diagnostic Unit, Laboratory and Specialistic Pediatrics Departments, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Fabio Arena
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | | | - Lorenzo Guerra
- Department of Biosciences, Biotechnologies and Environment, University of Bari “Aldo Moro”, Bari, Italy
| | | | - Nazzareno Capitanio
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Claudia Piccoli
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
- C. Piccoli and O. Laselva contributed equally to this article as lead authors and supervised the work
| | - Onofrio Laselva
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
- C. Piccoli and O. Laselva contributed equally to this article as lead authors and supervised the work
| |
Collapse
|
2
|
Daoud A, Xia S, Laselva O, Jiang J, Bear CE. Testing organ-specific responses to therapies in tissues differentiated from Cystic Fibrosis patient derived iPSCs. Stem Cell Res 2025; 83:103653. [PMID: 39793274 DOI: 10.1016/j.scr.2025.103653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 01/03/2025] [Accepted: 01/04/2025] [Indexed: 01/13/2025] Open
Abstract
Cystic Fibrosis (CF) is a life-shortening disease that is caused by mutations in the CFTR gene, a gene that is expressed in multiple organs. There are several primary tissue models of CF disease, including nasal epithelial cultures and rectal organoids, that are effective in reporting the potential efficacy of mutation-targeted therapies called CFTR modulators. However, there is the well-documented variation in tissue dependent, therapeutic response amongst CF patients, even those with the same CF-causing mutation. Hence, there is an interest in developing strategies for comparing therapeutic efficacy in different organs relative to isogenic controls. In this study, we evaluated the CFTR chloride channel response to the highly effective CFTR modulator: Trikafta, in CF patient specific, iPSC-derived colonic and airway cultures relative to mutation-corrected (non-CF) tissues from that same individual. We measured pharmacological rescue in both tissues. This proof-of-concept study provides a roadmap for future comparisons of patient-specific CF therapeutic responses in both pulmonary and extra-pulmonary systems.
Collapse
Affiliation(s)
- Abdelkader Daoud
- Programme in Molecular Medicine, Research Institute for SickKids Hospital, Toronto, Canada
| | - Sunny Xia
- Programme in Molecular Medicine, Research Institute for SickKids Hospital, Toronto, Canada; Department of Physiology, University of Toronto, ON, Canada
| | - Onofrio Laselva
- Programme in Molecular Medicine, Research Institute for SickKids Hospital, Toronto, Canada; Department of Clinical and Experimental Medicine, University of Foggia, Italy
| | - Janet Jiang
- Programme in Molecular Medicine, Research Institute for SickKids Hospital, Toronto, Canada
| | - Christine E Bear
- Programme in Molecular Medicine, Research Institute for SickKids Hospital, Toronto, Canada; Department of Clinical and Experimental Medicine, University of Foggia, Italy.
| |
Collapse
|
3
|
Wang Z, Ye S, van der Laan LJ, Schneeberger K, Masereeuw R, Spee B. Chemically Defined Organoid Culture System for Cholangiocyte Differentiation. Adv Healthc Mater 2024; 13:e2401511. [PMID: 39044566 PMCID: PMC11616262 DOI: 10.1002/adhm.202401511] [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: 04/24/2024] [Revised: 07/15/2024] [Indexed: 07/25/2024]
Abstract
Cholangiocyte organoids provide a powerful platform for applications ranging from in vitro modeling to tissue engineering for regenerative medicine. However, their expansion and differentiation are typically conducted in animal-derived hydrogels, which impede the full maturation of organoids into functional cholangiocytes. In addition, these hydrogels are poorly defined and complex, limiting the clinical applicability of organoids. In this study, a novel medium composition combined with synthetic polyisocyanopeptide (PIC) hydrogels to enhance the maturation of intrahepatic cholangiocyte organoids (ICOs) into functional cholangiocytes is utilized. ICOs cultured in the presence of sodium butyrate and valproic acid, a histone deacetylase inhibitor, and a Notch signaling activator, respectively, in PIC hydrogel exhibit a more mature phenotype, as evidenced by increased expression of key cholangiocyte markers, crucial for biliary function. Notably, mature cholangiocyte organoids in PIC hydrogel display apical-out polarity, in contrast to the traditional basal-out polarization of ICOs cultured in Matrigel. Moreover, these mature cholangiocyte organoids effectively model the biliary pro-fibrotic response induced by transforming growth factor beta. Taken together, an animal-free, chemically defined culture system that promotes the ICOs into mature cholangiocytes with apical-out polarity, facilitating regenerative medicine applications and in vitro studies that require access to the apical membrane, is developed.
Collapse
Affiliation(s)
- Zhenguo Wang
- Division of PharmacologyUtrecht Institute for Pharmaceutical SciencesFaculty of ScienceUtrecht UniversityUniversiteitsweg 99Utrecht3584 CGThe Netherlands
- Department of Clinical SciencesFaculty of Veterinary MedicineUtrecht UniversityUppsalalaan 8Utrecht3584 CTThe Netherlands
| | - Shicheng Ye
- Department of Clinical SciencesFaculty of Veterinary MedicineUtrecht UniversityUppsalalaan 8Utrecht3584 CTThe Netherlands
| | - Luc J.W. van der Laan
- Department of SurgeryErasmus MC Transplant InstituteUniversity Medical Center RotterdamDoctor Molewaterplein 40Rotterdam3015 GDThe Netherlands
| | - Kerstin Schneeberger
- Department of Clinical SciencesFaculty of Veterinary MedicineUtrecht UniversityUppsalalaan 8Utrecht3584 CTThe Netherlands
| | - Rosalinde Masereeuw
- Division of PharmacologyUtrecht Institute for Pharmaceutical SciencesFaculty of ScienceUtrecht UniversityUniversiteitsweg 99Utrecht3584 CGThe Netherlands
| | - Bart Spee
- Department of Clinical SciencesFaculty of Veterinary MedicineUtrecht UniversityUppsalalaan 8Utrecht3584 CTThe Netherlands
| |
Collapse
|
4
|
Fiorucci S, Urbani G, Di Giorgio C, Biagioli M, Distrutti E. Bile Acids-Based Therapies for Primary Sclerosing Cholangitis: Current Landscape and Future Developments. Cells 2024; 13:1650. [PMID: 39404413 PMCID: PMC11475195 DOI: 10.3390/cells13191650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 09/30/2024] [Accepted: 10/02/2024] [Indexed: 10/19/2024] Open
Abstract
Primary sclerosing cholangitis (PSC) is a rare, chronic liver disease with no approved therapies. The ursodeoxycholic acid (UDCA) has been widely used, although there is no evidence that the use of UDCA delays the time to liver transplant or increases survival. Several candidate drugs are currently being developed. The largest group of these new agents is represented by FXR agonists, including obeticholic acid, cilofexor, and tropifexor. Other agents that target bile acid metabolism are ASTB/IBAP inhibitors and fibroblasts growth factor (FGF)19 analogues. Cholangiocytes, the epithelial bile duct cells, play a role in PSC development. Recent studies have revealed that these cells undergo a downregulation of GPBAR1 (TGR5), a bile acid receptor involved in bicarbonate secretion and immune regulation. Additional agents under evaluation are PPARs (elafibranor and seladelpar), anti-itching agents such as MAS-related G-protein-coupled receptors antagonists, and anti-fibrotic and immunosuppressive agents. Drugs targeting gut bacteria and bile acid pathways are also under investigation, given the strong link between PSC and gut microbiota.
Collapse
Affiliation(s)
- Stefano Fiorucci
- Dipartimento di Medicina e Chirurgia, Università di Perugia, 06123 Perugia, Italy; (G.U.); (C.D.G.); (M.B.)
| | - Ginevra Urbani
- Dipartimento di Medicina e Chirurgia, Università di Perugia, 06123 Perugia, Italy; (G.U.); (C.D.G.); (M.B.)
| | - Cristina Di Giorgio
- Dipartimento di Medicina e Chirurgia, Università di Perugia, 06123 Perugia, Italy; (G.U.); (C.D.G.); (M.B.)
| | - Michele Biagioli
- Dipartimento di Medicina e Chirurgia, Università di Perugia, 06123 Perugia, Italy; (G.U.); (C.D.G.); (M.B.)
| | - Eleonora Distrutti
- SC di Gastroenterologia ed Epatologia, Azienda Ospedaliera di Perugia, 06123 Perugia, Italy;
| |
Collapse
|
5
|
Han YK, Lim HJ, Jang G, Jang SY, Park KM. Kidney ischemia/reperfusion injury causes cholangiocytes primary cilia disruption and abnormal bile secretion. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167225. [PMID: 38749218 DOI: 10.1016/j.bbadis.2024.167225] [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: 02/29/2024] [Revised: 04/29/2024] [Accepted: 05/06/2024] [Indexed: 06/17/2024]
Abstract
BACKGROUND Acute kidney injury (AKI) causes distant liver injury, to date, which causes poor outcomes of patients with AKI. Many studies have been performed to overcome AKI-associated liver injury. However, those studies have mainly focused on hepatocytes, and AKI-induced liver injury still remains a clinical problem. Here, we investigated the implication of cholangiocytes and their primary cilia which are critical in final bile secretion. Cholangiocyte, a lining cell of bile ducts, are the only liver epithelial cell containing primary cilium (a microtubule-based cell surface signal-sensing organelle). METHODS Cystathione γ-lyase (CSE, a transsulfuration enzyme) deficient and wild-type mice were subjected to kidney ischemia followed by reperfusion (KIR). Some mice were administered with N-acetyl-cysteine (NAC). RESULTS KIR damaged hepatocytes and cholagiocytes, disrupted cholangiocytes primary cilia, released the disrupted ciliary fragments into the bile, and caused abnormal bile secretion. Glutathione (GSH) and H2S levels in the livers were significantly reduced by KIR, resulting in increased the ratio oxidized GSH to total GSH, and oxidation of tissue and bile. CSE and cystathione β-synthase (CBS) expression were lowered in the liver after KIR. NAC administration increased total GSH and H2S levels in the liver and attenuated KIR-induced liver injuries. In contrast, Cse deletion caused the reduction of total GSH levels and worsened KIR-induced liver injuries, including primary cilia damage and abnormal bile secretion. CONCLUSIONS These results indicate that KIR causes cholangiocyte damage, cholangiocytes primary cilia disruption, and abnormal bile secretion through reduced antioxidative ability of the liver.
Collapse
Affiliation(s)
- Yong Kwon Han
- Department of Anatomy, School of Medicine, Kyungpook National University, 680 Gukchaebosang-ro, Junggu, Daegu 41944, Republic of Korea; Cardiovascular Research Institute, Kyungpook National University, 680 Gukchaebosang-ro, Junggu, Daegu 41944, Republic of Korea
| | - Hui Jae Lim
- Department of Anatomy, School of Medicine, Kyungpook National University, 680 Gukchaebosang-ro, Junggu, Daegu 41944, Republic of Korea; Department of Biomedical Science and BK21 Plus, The Graduate School of Medicine, Kyungpook National University, 680 Gukchaebosang-ro, Junggu, Daegu 41944, Republic of Korea
| | - GiBong Jang
- Department of Anatomy, School of Medicine, Kyungpook National University, 680 Gukchaebosang-ro, Junggu, Daegu 41944, Republic of Korea; Department of Biomedical Science and BK21 Plus, The Graduate School of Medicine, Kyungpook National University, 680 Gukchaebosang-ro, Junggu, Daegu 41944, Republic of Korea
| | - Se Young Jang
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, 680 Gukchaebosang-ro, Junggu, Daegu 41944, Republic of Korea
| | - Kwon Moo Park
- Department of Anatomy, School of Medicine, Kyungpook National University, 680 Gukchaebosang-ro, Junggu, Daegu 41944, Republic of Korea; Cardiovascular Research Institute, Kyungpook National University, 680 Gukchaebosang-ro, Junggu, Daegu 41944, Republic of Korea; Department of Biomedical Science and BK21 Plus, The Graduate School of Medicine, Kyungpook National University, 680 Gukchaebosang-ro, Junggu, Daegu 41944, Republic of Korea.
| |
Collapse
|
6
|
Budi NYP, Lai WY, Huang YH, Ho HN. 3D organoid cultivation improves the maturation and functional differentiation of cholangiocytes from human pluripotent stem cells. Front Cell Dev Biol 2024; 12:1361084. [PMID: 39040044 PMCID: PMC11260683 DOI: 10.3389/fcell.2024.1361084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2023] [Accepted: 05/29/2024] [Indexed: 07/24/2024] Open
Abstract
Idiopathic cholangiopathies are diseases that affect cholangiocytes, and they have unknown etiologies. Currently, orthotopic liver transplantation is the only treatment available for end-stage liver disease. Limited access to the bile duct makes it difficult to model cholangiocyte diseases. In this study, by mimicking the embryonic development of cholangiocytes and using a robust, feeder- and serum-free protocol, we first demonstrate the generation of unique functional 3D organoids consisting of small and large cholangiocytes derived from human pluripotent stem cells (PSCs), as opposed to traditional 2D culture systems. At day 28 of differentiation, the human PSC-derived cholangiocytes expressed markers of mature cholangiocytes, such as CK7, CK19, and cystic fibrosis transmembrane conductance regulator (CFTR). Compared with the 2D culture system-generated cholangiocytes, the 3D cholangiocyte organoids (COs) showed higher expression of the region-specific markers of intrahepatic cholangiocytes YAP1 and JAG1 and extrahepatic cholangiocytes AQP1 and MUC1. Furthermore, the COs had small-large tube-like structures and functional assays revealed that they exhibited characteristics of mature cholangiocytes, such as multidrug resistance protein 1 transporter function and CFTR channel activity. In addition to the extracellular matrix supports, the epidermal growth factor receptor (EGFR)-mediated signaling regulation might be involved in this cholangiocyte maturation and differentiation. These results indicated the successful generation of intrahepatic and extrahepatic cholangiocytes by using our 3D organoid protocol. The results highlight the advantages of our 3D culture system over the 2D culture system in promoting the functional differentiation and maturation of cholangiocytes. In summary, in advance of the previous works, our study provides a possible concept of small-large cholangiocyte transdifferentiation of human PSCs under cost-effective 3D culture conditions. The study findings have implications for the development of effective cell-based therapy using COs for patients with cholangiopathies.
Collapse
Affiliation(s)
- Nova Yuli Prasetyo Budi
- International Ph.D. Program in Cell Therapy and Regenerative Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Pediatric Surgery Division, Department of Surgery/Genetics Working Group, Faculty of Medicine, Public Health and Nursing, Universitas Gadjah Mada/Dr. Sardjito Hospital, Yogyakarta, Indonesia
| | - Wei-Yu Lai
- Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Yen-Hua Huang
- International Ph.D. Program in Cell Therapy and Regenerative Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- TMU Research Center for Cell Therapy and Regeneration Medicine, Taipei Medical University, Taipei, Taiwan
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Hong-Nerng Ho
- International Ph.D. Program in Cell Therapy and Regenerative Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- TMU Research Center for Cell Therapy and Regeneration Medicine, Taipei Medical University, Taipei, Taiwan
- Department of Obstetrics and Gynecology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Department of Obstetrics and Gynecology, Taipei Municipal Wanfang Hospital, Taipei Medical University, Taipei, Taiwan
- Taipei Cancer Center, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
7
|
Okumura A, Aoshima K, Tanimizu N. Generation of in vivo-like multicellular liver organoids by mimicking developmental processes: A review. Regen Ther 2024; 26:219-234. [PMID: 38903867 PMCID: PMC11186971 DOI: 10.1016/j.reth.2024.05.020] [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: 05/01/2024] [Revised: 05/24/2024] [Accepted: 05/30/2024] [Indexed: 06/22/2024] Open
Abstract
Liver is involved in metabolic reactions, ammonia detoxification, and immunity. Multicellular liver tissue cultures are more desirable for drug screening, disease modeling, and researching transplantation therapy, than hepatocytes monocultures. Hepatocytes monocultures are not stable for long. Further, hepatocyte-like cells induced from pluripotent stem cells and in vivo hepatocytes are functionally dissimilar. Organoid technology circumvents these issues by generating functional ex vivo liver tissue from intrinsic liver progenitor cells and extrinsic stem cells, including pluripotent stem cells. To function as in vivo liver tissue, the liver organoid cells must be arranged precisely in the 3-dimensional space, closely mimicking in vivo liver tissue. Moreover, for long term functioning, liver organoids must be appropriately vascularized and in contact with neighboring epithelial tissues (e.g., bile canaliculi and intrahepatic bile duct, or intrahepatic and extrahepatic bile ducts). Recent discoveries in liver developmental biology allows one to successfully induce liver component cells and generate organoids. Thus, here, in this review, we summarize the current state of knowledge on liver development with a focus on its application in generating different liver organoids. We also cover the future prospects in creating (functionally and structurally) in vivo-like liver organoids using the current knowledge on liver development.
Collapse
Affiliation(s)
- Ayumu Okumura
- Division of Regenerative Medicine, Center for Stem Cell Biology and Regenerative Medicine, The Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-0071, Japan
| | - Kenji Aoshima
- Division of Regenerative Medicine, Center for Stem Cell Biology and Regenerative Medicine, The Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-0071, Japan
| | - Naoki Tanimizu
- Division of Regenerative Medicine, Center for Stem Cell Biology and Regenerative Medicine, The Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-0071, Japan
| |
Collapse
|
8
|
Afonso MB, Marques V, van Mil SW, Rodrigues CM. Human liver organoids: From generation to applications. Hepatology 2024; 79:1432-1451. [PMID: 36815360 PMCID: PMC11095893 DOI: 10.1097/hep.0000000000000343] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Revised: 12/11/2022] [Accepted: 12/19/2022] [Indexed: 02/24/2023]
Abstract
In the last decade, research into human hepatology has been revolutionized by the development of mini human livers in a dish. These liver organoids are formed by self-organizing stem cells and resemble their native counterparts in cellular content, multicellular architecture, and functional features. Liver organoids can be derived from the liver tissue or pluripotent stem cells generated from a skin biopsy, blood cells, or renal epithelial cells present in urine. With the development of liver organoids, a large part of previous hurdles in modeling the human liver is likely to be solved, enabling possibilities to better model liver disease, improve (personalized) drug testing, and advance bioengineering options. In this review, we address strategies to generate and use organoids in human liver disease modeling, followed by a discussion of their potential application in drug development and therapeutics, as well as their strengths and limitations.
Collapse
Affiliation(s)
- Marta B. Afonso
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Portugal
| | - Vanda Marques
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Portugal
| | - Saskia W.C. van Mil
- Center for Molecular Medicine, University Medical Center Utrecht and Utrecht University, The Netherlands
| | - Cecilia M.P. Rodrigues
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Portugal
| |
Collapse
|
9
|
Hong R, Tan Y, Tian X, Huang Z, Wang J, Ni H, Yang J, Bu W, Yang S, Li T, Yu F, Zhong W, Sun T, Wang X, Li D, Liu M, Yang Y, Zhou J. XIAP-mediated degradation of IFT88 disrupts HSC cilia to stimulate HSC activation and liver fibrosis. EMBO Rep 2024; 25:1055-1074. [PMID: 38351372 PMCID: PMC10933415 DOI: 10.1038/s44319-024-00092-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Revised: 12/15/2023] [Accepted: 01/25/2024] [Indexed: 02/19/2024] Open
Abstract
Activation of hepatic stellate cells (HSCs) plays a critical role in liver fibrosis. However, the molecular basis for HSC activation remains poorly understood. Herein, we demonstrate that primary cilia are present on quiescent HSCs but exhibit a significant loss upon HSC activation which correlates with decreased levels of the ciliary protein intraflagellar transport 88 (IFT88). Ift88-knockout mice are more susceptible to chronic carbon tetrachloride-induced liver fibrosis. Mechanistic studies show that the X-linked inhibitor of apoptosis (XIAP) functions as an E3 ubiquitin ligase for IFT88. Transforming growth factor-β (TGF-β), a profibrotic factor, enhances XIAP-mediated ubiquitination of IFT88, promoting its proteasomal degradation. Blocking XIAP-mediated IFT88 degradation ablates TGF-β-induced HSC activation and liver fibrosis. These findings reveal a previously unrecognized role for ciliary homeostasis in regulating HSC activation and identify the XIAP-IFT88 axis as a potential therapeutic target for liver fibrosis.
Collapse
Affiliation(s)
- Renjie Hong
- Department of Genetics and Cell Biology, State Key Laboratory of Medicinal Chemical Biology, Haihe Laboratory of Cell Ecosystem, Tianjin Key Laboratory of Protein Science, College of Life Sciences, Nankai University, 300071, Tianjin, China
| | - Yanjie Tan
- Center for Cell Structure and Function, Collaborative Innovation Center of Cell Biology in Universities of Shandong, Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, 250014, Jinan, China
| | - Xiaoyu Tian
- Center for Cell Structure and Function, Collaborative Innovation Center of Cell Biology in Universities of Shandong, Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, 250014, Jinan, China
| | - Zhenzhou Huang
- Center for Cell Structure and Function, Collaborative Innovation Center of Cell Biology in Universities of Shandong, Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, 250014, Jinan, China
| | - Jiaying Wang
- Center for Cell Structure and Function, Collaborative Innovation Center of Cell Biology in Universities of Shandong, Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, 250014, Jinan, China
| | - Hua Ni
- Department of Genetics and Cell Biology, State Key Laboratory of Medicinal Chemical Biology, Haihe Laboratory of Cell Ecosystem, Tianjin Key Laboratory of Protein Science, College of Life Sciences, Nankai University, 300071, Tianjin, China
| | - Jia Yang
- Department of Genetics and Cell Biology, State Key Laboratory of Medicinal Chemical Biology, Haihe Laboratory of Cell Ecosystem, Tianjin Key Laboratory of Protein Science, College of Life Sciences, Nankai University, 300071, Tianjin, China
| | - Weiwen Bu
- Department of Genetics and Cell Biology, State Key Laboratory of Medicinal Chemical Biology, Haihe Laboratory of Cell Ecosystem, Tianjin Key Laboratory of Protein Science, College of Life Sciences, Nankai University, 300071, Tianjin, China
| | - Song Yang
- Department of Genetics and Cell Biology, State Key Laboratory of Medicinal Chemical Biology, Haihe Laboratory of Cell Ecosystem, Tianjin Key Laboratory of Protein Science, College of Life Sciences, Nankai University, 300071, Tianjin, China
| | - Te Li
- Department of Genetics and Cell Biology, State Key Laboratory of Medicinal Chemical Biology, Haihe Laboratory of Cell Ecosystem, Tianjin Key Laboratory of Protein Science, College of Life Sciences, Nankai University, 300071, Tianjin, China
| | - Fan Yu
- Department of Genetics and Cell Biology, State Key Laboratory of Medicinal Chemical Biology, Haihe Laboratory of Cell Ecosystem, Tianjin Key Laboratory of Protein Science, College of Life Sciences, Nankai University, 300071, Tianjin, China
| | - Weilong Zhong
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, 300052, Tianjin, China
| | - Tao Sun
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, Nankai University, 300071, Tianjin, China
| | - Xiaohong Wang
- Department of Pharmacology and Tianjin Key Laboratory of Inflammation Biology, School of Basic Medical Sciences, Tianjin Medical University, 300070, Tianjin, China
| | - Dengwen Li
- Department of Genetics and Cell Biology, State Key Laboratory of Medicinal Chemical Biology, Haihe Laboratory of Cell Ecosystem, Tianjin Key Laboratory of Protein Science, College of Life Sciences, Nankai University, 300071, Tianjin, China
| | - Min Liu
- Department of Genetics and Cell Biology, State Key Laboratory of Medicinal Chemical Biology, Haihe Laboratory of Cell Ecosystem, Tianjin Key Laboratory of Protein Science, College of Life Sciences, Nankai University, 300071, Tianjin, China
| | - Yunfan Yang
- Department of Cell Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, 250012, Jinan, China.
| | - Jun Zhou
- Department of Genetics and Cell Biology, State Key Laboratory of Medicinal Chemical Biology, Haihe Laboratory of Cell Ecosystem, Tianjin Key Laboratory of Protein Science, College of Life Sciences, Nankai University, 300071, Tianjin, China.
- Center for Cell Structure and Function, Collaborative Innovation Center of Cell Biology in Universities of Shandong, Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, 250014, Jinan, China.
| |
Collapse
|
10
|
Hong R, Tian X, Ma H, Ni H, Yang J, Bu W, Li T, Yang S, Li D, Liu M, Tan Y. Primary cilium-mediated signaling cascade suppresses age-related biliary fibrosis. J Cell Physiol 2023; 238:2600-2611. [PMID: 37683035 DOI: 10.1002/jcp.31113] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 07/19/2023] [Accepted: 08/17/2023] [Indexed: 09/10/2023]
Abstract
The primary cilium is increasingly recognized as a crucial player in the physiology of biliary epithelial cells (BECs). However, the precise role of primary cilia in the development of age-related biliary fibrosis remains unclear. Herein, using cilium-deficient mice, we demonstrate that disruption of ciliary homeostasis in BECs in aged mice leads to significant bile duct proliferation, augmented biliary fibrosis, and heightened indicators of liver injury. Our RNA-sequencing data revealed a dysregulation in genes associated with various biological processes such as bile secretion, fatty acid metabolism, and inflammation. Loss of primary cilia also significantly enhanced signaling pathways driving the development of biliary fibrosis. Our findings collectively suggest that loss of primary cilia in the BECs of aged mice initiates a cascade of signaling events that contribute to biliary fibrosis, highlighting the primary cilium as a potential therapeutic target in the treatment of fibrosing cholangiopathies.
Collapse
Affiliation(s)
- Renjie Hong
- State Key Laboratory of Medicinal Chemical Biology, Haihe Laboratory of Cell Ecosystem, College of Life Sciences, Nankai University, Tianjin, China
| | - Xiaoyu Tian
- Center for Cell Structure and Function, Collaborative Innovation Center of Cell Biology in Universities of Shandong, Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, Jinan, China
| | - Hongbo Ma
- State Key Laboratory of Medicinal Chemical Biology, Haihe Laboratory of Cell Ecosystem, College of Life Sciences, Nankai University, Tianjin, China
| | - Hua Ni
- State Key Laboratory of Medicinal Chemical Biology, Haihe Laboratory of Cell Ecosystem, College of Life Sciences, Nankai University, Tianjin, China
| | - Jia Yang
- State Key Laboratory of Medicinal Chemical Biology, Haihe Laboratory of Cell Ecosystem, College of Life Sciences, Nankai University, Tianjin, China
| | - Weiwen Bu
- State Key Laboratory of Medicinal Chemical Biology, Haihe Laboratory of Cell Ecosystem, College of Life Sciences, Nankai University, Tianjin, China
| | - Te Li
- State Key Laboratory of Medicinal Chemical Biology, Haihe Laboratory of Cell Ecosystem, College of Life Sciences, Nankai University, Tianjin, China
| | - Song Yang
- State Key Laboratory of Medicinal Chemical Biology, Haihe Laboratory of Cell Ecosystem, College of Life Sciences, Nankai University, Tianjin, China
| | - Dengwen Li
- State Key Laboratory of Medicinal Chemical Biology, Haihe Laboratory of Cell Ecosystem, College of Life Sciences, Nankai University, Tianjin, China
| | - Min Liu
- Laboratory of Tissue Homeostasis, Haihe Laboratory of Cell Ecosystem, Tianjin, China
| | - Yanjie Tan
- Center for Cell Structure and Function, Collaborative Innovation Center of Cell Biology in Universities of Shandong, Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, Jinan, China
| |
Collapse
|
11
|
Li X, Wang Q, Ai L, Cheng K. Unraveling the activation process and core driver genes of HSCs during cirrhosis by single-cell transcriptome. Exp Biol Med (Maywood) 2023; 248:1414-1424. [PMID: 37674431 PMCID: PMC10657590 DOI: 10.1177/15353702231191109] [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: 12/23/2022] [Accepted: 03/11/2023] [Indexed: 09/08/2023] Open
Abstract
Worldwide, cirrhosis is a common cause of death, manifesting itself as fibrosis of the liver tissue. When the liver is damaged, the liver produces fibrotic, proliferative myofibroblasts, which are formed by the differentiation of activated hepatic stellate cells. There are no effective antifibrotic treatment options. To deeply explore the activation process of hepatic stellate cells (HSCs) and to discover better therapeutic target genes, single-cell RNA sequencing data on 13 non-cirrhotic liver tissue samples and 10 cirrhotic liver tissue samples were analyzed. We identified activated HSCs from the mesenchymal cell population with high expression of ACTA2. By pseudo-time analysis, we found that the key genes for the differentiation of HSCs into myofibroblasts were C3, CCDC80, COL1A1, COL3A1, DCN, FBLN1, IGFBP3, MXRA5, SERPINE1, and MYH11. Then, we found that the main regulators of HSCs from inactive to activated state were NTF3, NTRK3, NTRK2, JAG1, NOTCH3, ESAM, and CD46 by cell-cell communication analysis. In addition, we found that the top2 hub genes of activated HSCs were CRIP1 and ACTA2. The experimental results show that the top2 hub genes were significantly overexpressed in cirrhotic samples. Our work dissected key intercellular regulators and core driver genes during hepatic stellate cell activation during cirrhosis through single-cell transcriptome data analysis, providing a research strategy to discover rational therapeutic targets for cirrhosis and some important information for gene targeting therapy.
Collapse
Affiliation(s)
- Xia Li
- Transplantation Center, Engineering & Technology Research Center for Transplantation Medicine of Hunan Province, The Third Xiangya Hospital, Central South University, Changsha 410013, China
| | - Qiang Wang
- Transplantation Center, Engineering & Technology Research Center for Transplantation Medicine of Hunan Province, The Third Xiangya Hospital, Central South University, Changsha 410013, China
| | - Liang Ai
- Transplantation Center, Engineering & Technology Research Center for Transplantation Medicine of Hunan Province, The Third Xiangya Hospital, Central South University, Changsha 410013, China
| | - Ke Cheng
- Transplantation Center, Engineering & Technology Research Center for Transplantation Medicine of Hunan Province, The Third Xiangya Hospital, Central South University, Changsha 410013, China
| |
Collapse
|
12
|
Nishie T, Ohta Y, Shirai E, Higaki S, Shimozawa N, Narita K, Kawaguchi K, Tanaka H, Mori C, Tanaka T, Hirabayashi M, Suemori H, Kurisaki A, Tooyama I, Asano S, Takeda S, Takada T. Identification of TEKTIN1-expressing multiciliated cells during spontaneous differentiation of non-human primate embryonic stem cells. Genes Cells 2023. [PMID: 37186436 DOI: 10.1111/gtc.13031] [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: 10/12/2022] [Revised: 04/10/2023] [Accepted: 04/14/2023] [Indexed: 05/17/2023]
Abstract
Tektins are a group of microtubule-stabilizing proteins necessary for cilia and flagella assembly. TEKTIN1 (TEKT1) is used as a sperm marker for monitoring germ cell differentiation in embryonic stem (ES) and induced pluripotent stem (iPS) cells. Although upregulation of TEKT1 has been reported during spontaneous differentiation of ES and iPS cells, it is unclear which cells express TEKT1. To identify TEKT1-expressing cells, we established an ES cell line derived from cynomolgus monkeys (Macaca fascicularis), which expresses Venus controlled by the TEKT1 promoter. Venus expression was detected at 5 weeks of differentiation on the surface of the embryoid body (EB), and it gradually increased with the concomitant formation of a leash-like structure at the EB periphery. Motile cilia were observed on the surface of the Venus-positive leash-like structure after 8 weeks of differentiation. The expression of cilia markers as well as TEKT1-5 and 9 + 2 microtubule structures, which are characteristic of motile cilia, were detected in Venus-positive cells. These results demonstrated that TEKT1-expressing cells are multiciliated epithelial-like cells that form a leash-like structure during the spontaneous differentiation of ES and iPS cells. These findings will provide a new research strategy for studying cilia biology, including ciliogenesis and ciliopathies.
Collapse
Affiliation(s)
- Tomomi Nishie
- Laboratory of Cell Engineering, Department of Pharmaceutical Sciences, Ritsumeikan University, Kusatsu, Shiga, Japan
| | - Yoshio Ohta
- Laboratory of Cell Engineering, Department of Pharmaceutical Sciences, Ritsumeikan University, Kusatsu, Shiga, Japan
| | - Emi Shirai
- Laboratory of Cell Engineering, Department of Pharmaceutical Sciences, Ritsumeikan University, Kusatsu, Shiga, Japan
| | - Shogo Higaki
- Laboratory of Cell Engineering, Department of Pharmaceutical Sciences, Ritsumeikan University, Kusatsu, Shiga, Japan
| | - Nobuhiro Shimozawa
- Tsukuba Primate Research Center, National Institutes of Biomedical Innovation, Health and Nutrition, Tsukuba-shi, Ibaraki, Japan
| | - Keishi Narita
- Department of Anatomy and Cell Biology, Interdisciplinary Graduate School, University of Yamanashi, Chuo, Yamanashi, Japan
| | - Kotoku Kawaguchi
- Department of Molecular Physiology, College of Pharmaceutical Sciences, Ritsumeikan University, Kusatsu, Shiga, Japan
| | - Hideyuki Tanaka
- Department of Anatomy, Teikyo University School of Medicine, Itabashi, Tokyo, Japan
| | - Chika Mori
- Laboratory of Cell Engineering, Department of Pharmaceutical Sciences, Ritsumeikan University, Kusatsu, Shiga, Japan
| | - Taiga Tanaka
- Laboratory of Cell Engineering, Department of Pharmaceutical Sciences, Ritsumeikan University, Kusatsu, Shiga, Japan
| | - Masumi Hirabayashi
- Center for Genetic Analysis of Behavior, National Institute for Physiological Sciences, Okazaki, Aichi, Japan
| | - Hirofumi Suemori
- Center for Human ES Cell Research, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Akira Kurisaki
- Graduate School of Biological Sciences, Nara Institute of Science and Technology, Nara, Japan
| | - Ikuo Tooyama
- Molecular Neuroscience Research Center and Medical Innovation Research Center, Shiga University of Medical Science, Shiga, Japan
| | - Shinji Asano
- Department of Molecular Physiology, College of Pharmaceutical Sciences, Ritsumeikan University, Kusatsu, Shiga, Japan
| | - Sén Takeda
- Department of Anatomy and Cell Biology, Interdisciplinary Graduate School, University of Yamanashi, Chuo, Yamanashi, Japan
- Department of Anatomy, Teikyo University School of Medicine, Itabashi, Tokyo, Japan
| | - Tatsuyuki Takada
- Laboratory of Cell Engineering, Department of Pharmaceutical Sciences, Ritsumeikan University, Kusatsu, Shiga, Japan
| |
Collapse
|
13
|
Jalan-Sakrikar N, Brevini T, Huebert RC, Sampaziotis F. Organoids and regenerative hepatology. Hepatology 2023; 77:305-322. [PMID: 35596930 PMCID: PMC9676408 DOI: 10.1002/hep.32583] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 05/13/2022] [Accepted: 05/14/2022] [Indexed: 02/03/2023]
Abstract
The burden of liver diseases is increasing worldwide, with liver transplantation remaining the only treatment option for end-stage liver disease. Regenerative medicine holds great potential as a therapeutic alternative, aiming to repair or replace damaged liver tissue with healthy functional cells. The properties of the cells used are critical for the efficacy of this approach. The advent of liver organoids has not only offered new insights into human physiology and pathophysiology, but also provided an optimal source of cells for regenerative medicine and translational applications. Here, we discuss various historical aspects of 3D organoid culture, how it has been applied to the hepatobiliary system, and how organoid technology intersects with the emerging global field of liver regenerative medicine. We outline the hepatocyte, cholangiocyte, and nonparenchymal organoids systems available and discuss their advantages and limitations for regenerative medicine as well as future directions.
Collapse
Affiliation(s)
- Nidhi Jalan-Sakrikar
- Division of Gastroenterology and Hepatology, Mayo Clinic and Foundation, Rochester, Minnesota, USA
- Gastroenterology Research Unit, Mayo Clinic and Foundation, Rochester, Minnesota, USA
| | - Teresa Brevini
- Wellcome-MRC Cambridge Stem Cell Institute, Cambridge, UK
| | - Robert C. Huebert
- Division of Gastroenterology and Hepatology, Mayo Clinic and Foundation, Rochester, Minnesota, USA
- Gastroenterology Research Unit, Mayo Clinic and Foundation, Rochester, Minnesota, USA
| | - Fotios Sampaziotis
- Wellcome-MRC Cambridge Stem Cell Institute, Cambridge, UK
- Department of Medicine, University of Cambridge, Cambridge, UK
- Cambridge Liver Unit, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| |
Collapse
|
14
|
Bioengineering Liver Organoids for Diseases Modelling and Transplantation. BIOENGINEERING (BASEL, SWITZERLAND) 2022; 9:bioengineering9120796. [PMID: 36551002 PMCID: PMC9774794 DOI: 10.3390/bioengineering9120796] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 12/05/2022] [Accepted: 12/08/2022] [Indexed: 12/15/2022]
Abstract
Organoids as three-dimension (3D) cellular organizations partially mimic the physiological functions and micro-architecture of native tissues and organs, holding great potential for clinical applications. Advances in the identification of essential factors including physical cues and biochemical signals for controlling organoid development have contributed to the success of growing liver organoids from liver tissue and stem/progenitor cells. However, to recapitulate the physiological properties and the architecture of a native liver, one has to generate liver organoids that contain all the major liver cell types in correct proportions and relative 3D locations as found in a native liver. Recent advances in stem-cell-, biomaterial- and engineering-based approaches have been incorporated into conventional organoid culture methods to facilitate the development of a more sophisticated liver organoid culture resembling a near to native mini-liver in a dish. However, a comprehensive review on the recent advancement in the bioengineering liver organoid is still lacking. Here, we review the current liver organoid systems, focusing on the construction of the liver organoid system with various cell sources, the roles of growth factors for engineering liver organoids, as well as the recent advances in the bioengineering liver organoid disease models and their biomedical applications.
Collapse
|
15
|
Rizwan M, Ling C, Guo C, Liu T, Jiang JX, Bear CE, Ogawa S, Shoichet MS. Viscoelastic Notch Signaling Hydrogel Induces Liver Bile Duct Organoid Growth and Morphogenesis. Adv Healthc Mater 2022; 11:e2200880. [PMID: 36180392 DOI: 10.1002/adhm.202200880] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 09/09/2022] [Indexed: 01/28/2023]
Abstract
Cholangiocyte organoids can be used to model liver biliary disease; however, both a defined matrix to emulate cholangiocyte self-assembly and the mechano-transduction pathways involved therein remain elusive. A series of defined viscoelastic hyaluronan hydrogels to culture primary cholangiocytes are designed and it is found that by mimicking the stress relaxation rate of liver tissue, cholangiocyte organoid growth can be induced and expression of Yes-associated protein (YAP) target genes could be significantly increased. Strikingly, inhibition of matrix metalloproteinases (MMPs) does not significantly affect organoid growth in 3D culture, suggesting that mechanical remodeling of the viscoelastic microenvironment-and not MMP-mediated degradation-is the key to cholangiocyte organoid growth. By immobilizing Jagged1 to the hyaluronan, stress relaxing hydrogel, self-assembled bile duct structures form in organoid culture, indicating the synergistic effects of Notch signaling and viscoelasticity. By uncovering critical roles of hydrogel viscoelasticity, YAP signaling, and Notch activation, cholangiocyte organogenesis is controlled, thereby paving the way for their use in disease modeling and/or transplantation.
Collapse
Affiliation(s)
- Muhammad Rizwan
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, Ontario, M5S 3E5, Canada.,Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, M5S 3G9, Canada.,Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, M5S 3E1, Canada
| | - Christopher Ling
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, Ontario, M5S 3E5, Canada.,Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, M5S 3E1, Canada
| | - Chengyu Guo
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, Ontario, M5S 3E5, Canada
| | - Tracy Liu
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, Ontario, M5S 3E5, Canada
| | - Jia-Xin Jiang
- Molecular Medicine Programme, Hospital for Sick Children, Toronto, Ontario, M5G 1X8, Canada
| | - Christine E Bear
- Molecular Medicine Programme, Hospital for Sick Children, Toronto, Ontario, M5G 1X8, Canada.,Department of Physiology, University of Toronto, Toronto, Ontario, M5S 1A8, Canada.,Department of Biochemistry, University of Toronto, Toronto, Ontario, M5G 0A4, Canada
| | - Shinichiro Ogawa
- McEwen Stem Cell Institute, University Health Network, Toronto, Ontario, M5G 1L7, Canada.,Soham & Shalia Ajmera Family Transplant Centre, Toronto General Research Institute, University Health Network, Toronto, Ontario, M5G 2C4, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, M5S 1A8, Canada
| | - Molly S Shoichet
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, Ontario, M5S 3E5, Canada.,Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, M5S 3G9, Canada.,Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, M5S 3E1, Canada.,Department of Chemistry, University of Toronto, Toronto, Ontario, M5S 3H6, Canada
| |
Collapse
|
16
|
Advances in Preclinical In Vitro Models for the Translation of Precision Medicine for Cystic Fibrosis. J Pers Med 2022; 12:jpm12081321. [PMID: 36013270 PMCID: PMC9409685 DOI: 10.3390/jpm12081321] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 08/12/2022] [Accepted: 08/16/2022] [Indexed: 11/19/2022] Open
Abstract
The development of preclinical in vitro models has provided significant progress to the studies of cystic fibrosis (CF), a frequently fatal monogenic disease caused by mutations in the gene encoding the CF transmembrane conductance regulator (CFTR) protein. Numerous cell lines were generated over the last 30 years and they have been instrumental not only in enhancing the understanding of CF pathological mechanisms but also in developing therapies targeting the underlying defects in CFTR mutations with further validation in patient-derived samples. Furthermore, recent advances toward precision medicine in CF have been made possible by optimizing protocols and establishing novel assays using human bronchial, nasal and rectal tissues, and by progressing from two-dimensional monocultures to more complex three-dimensional culture platforms. These models also enable to potentially predict clinical efficacy and responsiveness to CFTR modulator therapies at an individual level. In parallel, advanced systems, such as induced pluripotent stem cells and organ-on-a-chip, continue to be developed in order to more closely recapitulate human physiology for disease modeling and drug testing. In this review, we have highlighted novel and optimized cell models that are being used in CF research to develop novel CFTR-directed therapies (or alternative therapeutic interventions) and to expand the usage of existing modulator drugs to common and rare CF-causing mutations.
Collapse
|
17
|
Abstract
The human liver is a complex organ made up of multiple specialized cell types that carry out key physiological functions. An incomplete understanding of liver biology limits our ability to develop therapeutics to prevent chronic liver diseases, liver cancers, and death as a result of organ failure. Recently, single-cell modalities have expanded our understanding of the cellular phenotypic heterogeneity and intercellular cross-talk in liver health and disease. This review summarizes these findings and looks forward to highlighting new avenues for the application of single-cell genomics to unravel unknown pathogenic pathways and disease mechanisms for the development of new therapeutics targeting liver pathology. As these technologies mature, their integration into clinical data analysis will aid in patient stratification and in developing treatment plans for patients suffering from liver disease.
Collapse
Affiliation(s)
- Jawairia Atif
- Ajmera Transplant Centre, Schwartz Reisman Liver Research Centre, Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada
- Department of Immunology, University of Toronto, Medical Sciences Building, Toronto, Ontario, Canada
| | - Cornelia Thoeni
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Gary D. Bader
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
- The Donnelly Centre, University of Toronto, Toronto, Ontario, Canada
| | - Ian D. McGilvray
- Ajmera Transplant Centre, Schwartz Reisman Liver Research Centre, Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Sonya A. MacParland
- Ajmera Transplant Centre, Schwartz Reisman Liver Research Centre, Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada
- Department of Immunology, University of Toronto, Medical Sciences Building, Toronto, Ontario, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
18
|
Wang Z, Faria J, van der Laan LJW, Penning LC, Masereeuw R, Spee B. Human Cholangiocytes Form a Polarized and Functional Bile Duct on Hollow Fiber Membranes. Front Bioeng Biotechnol 2022; 10:868857. [PMID: 35813994 PMCID: PMC9263983 DOI: 10.3389/fbioe.2022.868857] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 05/12/2022] [Indexed: 12/12/2022] Open
Abstract
Liver diseases affect hundreds of millions of people worldwide; most often the hepatocytes or cholangiocytes are damaged. Diseases of the biliary tract cause severe patient burden, and cholangiocytes, the cells lining the biliary tract, are sensitive to numerous drugs. Therefore, investigations into proper cholangiocyte functions are of utmost importance, which is restricted, in vitro, by the lack of primary human cholangiocytes allowing such screening. To investigate biliary function, including transepithelial transport, cholangiocytes must be cultured as three-dimensional (3D) ductular structures. We previously established murine intrahepatic cholangiocyte organoid-derived cholangiocyte-like cells (CLCs) and cultured them onto polyethersulfone hollow fiber membranes (HFMs) to generate 3D duct structures that resemble native bile ducts at the structural and functional level. Here, we established an efficient, stepwise method for directed differentiation of human intrahepatic cholangiocyte organoids (ICOs) into CLCs. Human ICO-derived CLCs showed key characteristics of cholangiocytes, such as the expression of structural and functional markers, formation of primary cilia, and P-glycoprotein-mediated transport in a polarized fashion. The organoid cultures exhibit farnesoid X receptor (FXR)-dependent functions that are vital to liver bile acid homeostasis in vivo. Furthermore, human ICO-derived CLCs cultured on HFMs in a differentiation medium form tubular architecture with some tight, confluent, and polarized monolayers that better mimic native bile duct characteristics than differentiated cultures in standard 2D or Matrigel-based 3D culture plates. Together, our optimized differentiation protocol to obtain CLC organoids, when applied on HFMs to form bioengineered bile ducts, will facilitate studying cholangiopathies and allow developing therapeutic strategies.
Collapse
Affiliation(s)
- Zhenguo Wang
- Division of Pharmacology, Department of Pharmaceutical Sciences, Faculty of Sciences, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands
| | - João Faria
- Division of Pharmacology, Department of Pharmaceutical Sciences, Faculty of Sciences, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands
| | | | - Louis C. Penning
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands
| | - Rosalinde Masereeuw
- Division of Pharmacology, Department of Pharmaceutical Sciences, Faculty of Sciences, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands
- *Correspondence: Rosalinde Masereeuw, ; Bart Spee,
| | - Bart Spee
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands
- *Correspondence: Rosalinde Masereeuw, ; Bart Spee,
| |
Collapse
|
19
|
Sharbidre K, Zahid M, Venkatesh SK, Bhati C, Lalwani N. Imaging of fibropolycystic liver disease. Abdom Radiol (NY) 2022; 47:2356-2370. [PMID: 35670875 DOI: 10.1007/s00261-022-03565-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 05/14/2022] [Accepted: 05/17/2022] [Indexed: 11/01/2022]
Abstract
Fibropolycystic liver diseases (FLDs) make up a rare spectrum of heritable hepatobiliary diseases resulting from congenital ductal plate malformations (DPMs) due to the dysfunction of proteins expressed on the primary cilia of cholangiocytes. The embryonic development of the ductal plate is key to understanding this spectrum of diseases. In particular, DPMs can result in various degrees of intrahepatic duct involvement and a wide spectrum of cholangiopathies, including congenital hepatic fibrosis, Caroli disease, polycystic liver disease, and Von Meyenberg complexes. The most common clinical manifestations of FLDs are portal hypertension, cholestasis, cholangitis, and (in rare cases) cholangiocarcinoma. This article reviews recent updates in the pathophysiology, imaging, and clinical management of FLDs.
Collapse
Affiliation(s)
- Kedar Sharbidre
- Department of Abdominal Imaging, University of Alabama at Birmingham, Birmingham, AB, USA.
| | - Mohd Zahid
- Department of Abdominal Imaging, University of Alabama at Birmingham, Birmingham, AB, USA
| | | | - Chandra Bhati
- Department of Transplant Surgery, University of Maryland Medical Center, Baltimore, ML, USA
| | - Neeraj Lalwani
- Department of Abdominal Imaging, Virginia Commonwealth University, Richmond, VA, USA
| |
Collapse
|
20
|
Ramalho AS, Boon M, Proesmans M, Vermeulen F, Carlon MS, De Boeck K. Assays of CFTR Function In Vitro, Ex Vivo and In Vivo. Int J Mol Sci 2022; 23:1437. [PMID: 35163362 PMCID: PMC8836180 DOI: 10.3390/ijms23031437] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 01/12/2022] [Accepted: 01/21/2022] [Indexed: 12/22/2022] Open
Abstract
Cystic fibrosis, a multi-organ genetic disease, is characterized by abnormal function of the cystic fibrosis transmembrane conductance regulator (CFTR) protein, a chloride channel at the apical membrane of several epithelia. In recent years, therapeutic strategies have been developed to correct the CFTR defect. To evaluate CFTR function at baseline for diagnosis, or the efficacy of CFTR-restoring therapy, reliable tests are needed to measure CFTR function, in vitro, ex vivo and in vivo. In vitro techniques either directly or indirectly measure ion fluxes; direct measurement of ion fluxes and quenching of fluorescence in cell-based assays, change in transmembrane voltage or current in patch clamp or Ussing chamber, swelling of CFTR-containing organoids by secondary water influx upon CFTR activation. Several cell or tissue types can be used. Ex vivo and in vivo assays similarly evaluate current (intestinal current measurement) and membrane potential differences (nasal potential difference), on tissues from individual patients. In the sweat test, the most frequently used in vivo evaluation of CFTR function, chloride concentration or stimulated sweat rate can be directly measured. Here, we will describe the currently available bio-assays for quantitative evaluation of CFTR function, their indications, advantages and disadvantages, and correlation with clinical outcome measures.
Collapse
Affiliation(s)
- Anabela S. Ramalho
- CF Research Lab, Woman and Child Unit, Department of Development and Regeneration, KU Leuven (Catholic University of Leuven), B-3000 Leuven, Belgium; (M.B.); (M.P.); (F.V.); (K.D.B.)
| | - Mieke Boon
- CF Research Lab, Woman and Child Unit, Department of Development and Regeneration, KU Leuven (Catholic University of Leuven), B-3000 Leuven, Belgium; (M.B.); (M.P.); (F.V.); (K.D.B.)
- Department of Pediatrics, Pediatric Pulmonology, University Hospital of Leuven, B-3000 Leuven, Belgium
| | - Marijke Proesmans
- CF Research Lab, Woman and Child Unit, Department of Development and Regeneration, KU Leuven (Catholic University of Leuven), B-3000 Leuven, Belgium; (M.B.); (M.P.); (F.V.); (K.D.B.)
- Department of Pediatrics, Pediatric Pulmonology, University Hospital of Leuven, B-3000 Leuven, Belgium
| | - François Vermeulen
- CF Research Lab, Woman and Child Unit, Department of Development and Regeneration, KU Leuven (Catholic University of Leuven), B-3000 Leuven, Belgium; (M.B.); (M.P.); (F.V.); (K.D.B.)
- Department of Pediatrics, Pediatric Pulmonology, University Hospital of Leuven, B-3000 Leuven, Belgium
| | - Marianne S. Carlon
- Molecular Virology and Gene Therapy, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven (Catholic University of Leuven), B-3000 Leuven, Belgium;
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department CHROMETA, KU Leuven (Catholic University of Leuven), B-3000 Leuven, Belgium
| | - Kris De Boeck
- CF Research Lab, Woman and Child Unit, Department of Development and Regeneration, KU Leuven (Catholic University of Leuven), B-3000 Leuven, Belgium; (M.B.); (M.P.); (F.V.); (K.D.B.)
- Department of Pediatrics, Pediatric Pulmonology, University Hospital of Leuven, B-3000 Leuven, Belgium
| |
Collapse
|