1
|
Wang WL, Lian H, Liang Y, Ye Y, Tam PKH, Chen Y. Molecular Mechanisms of Fibrosis in Cholestatic Liver Diseases and Regenerative Medicine-Based Therapies. Cells 2024; 13:1997. [PMID: 39682745 DOI: 10.3390/cells13231997] [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: 10/14/2024] [Revised: 11/18/2024] [Accepted: 11/26/2024] [Indexed: 12/18/2024] Open
Abstract
The aim of this review is to explore the potential of new regenerative medicine approaches in the treatment of cholestatic liver fibrosis. Cholestatic liver diseases, such as primary biliary cholangitis (PBC), primary sclerosing cholangitis (PSC), and biliary atresia (BA), due to the accumulation of bile, often progress to liver fibrosis, cirrhosis, and liver failure. When the disease becomes severe enough to require liver transplantation. Deeply understanding the disease's progression and fibrosis formation is crucial for better diagnosis and treatment. Current liver fibrosis treatments mainly target the root causes and no direct treatment method in fibrosis itself. Recent advances in regenerative medicine offer a potential approach that may help find the ways to target fibrosis directly, offering hope for improved outcomes. We also summarize, analyze, and discuss the current state and benefits of regenerative medicine therapies such as mesenchymal stem cell (MSC) therapy, induced pluripotent stem cells (iPSCs), and organoid technology, which may help the treatment of cholestatic liver diseases. Focusing on the latest research may reveal new targets and enhance therapeutic efficacy, potentially leading to more effective management and even curative strategies for cholestatic liver diseases.
Collapse
Affiliation(s)
- Wei-Lu Wang
- School of Pharmacy, Faculty of Medicine, Macau University of Science and Technology, Macao SAR, China
| | - Haoran Lian
- School of Pharmacy, Faculty of Medicine, Macau University of Science and Technology, Macao SAR, China
| | - Yingyu Liang
- School of Pharmacy, Faculty of Medicine, Macau University of Science and Technology, Macao SAR, China
| | - Yongqin Ye
- Faculty of Medicine, Macau University of Science and Technology, Macao SAR, China
| | - Paul Kwong Hang Tam
- School of Pharmacy, Faculty of Medicine, Macau University of Science and Technology, Macao SAR, China
- Faculty of Medicine, Macau University of Science and Technology, Macao SAR, China
- Precision Regenerative Medicine Research Centre, Medical Sciences Division, Macau University of Science and Technology, Macao SAR, China
| | - Yan Chen
- School of Pharmacy, Faculty of Medicine, Macau University of Science and Technology, Macao SAR, China
- Faculty of Medicine, Macau University of Science and Technology, Macao SAR, China
- Precision Regenerative Medicine Research Centre, Medical Sciences Division, Macau University of Science and Technology, Macao SAR, China
| |
Collapse
|
2
|
Alam S, Lal BB, Ravindranath A, Bavdekar A, Dheivamani N, Snehavardhan P, Shah A, Tripathi PR, Nagral A, Srikanth KP, Shah I, Ramakrishna SH, Suchismita A, Waikar Y, Shah V, Nalwalla Z, Kumar K, Maria A, Sibal A, Sivaramakrishnan VM, Wadhwa N, Ashritha A, Sood V, Khanna R. Natural course and outcomes of children with ubiquitin-specific protease 53 (USP53)-related genetic chronic cholestasis. J Pediatr Gastroenterol Nutr 2024. [PMID: 39440620 DOI: 10.1002/jpn3.12392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 07/05/2024] [Accepted: 09/10/2024] [Indexed: 10/25/2024]
Abstract
Ubiquitin-specific protease 53 (USP53) is essential for formation of cellular tight junctions and variations in this gene disrupt the tight junctions, resulting in cholestasis. We describe the clinical manifestations and outcomes of patients with USP53 mutations from the Indian progressive familial intrahepatic cholestasis registry. All 29 patients who harbored mutations in the USP53 gene either in the homozygous, compound heterozygous, or heterozygous state and presented with cholestasis were included. USP53 variants related to cholestasis had good outcomes, with native liver survival in 82.7%, whereas 17.3% required liver transplantation. Jaundice developed in 93% and within 3 months of age in 48.8%. Jaundice resolved in 21 (72.4%). Pruritus 76% at a median age of 7 months (severe in 10/22, 45% and refractory to medical therapy in 4, 18.1%). Majority of them (82.7%) had biallelic mutations. Protein-truncating mutations were present in 20 (69%) and missense mutations in 9 (31%). No correlation was found between the genotype and the outcome.
Collapse
Affiliation(s)
- Seema Alam
- Department of Pediatric Hepatology, Institute of Liver and Biliary Sciences, New Delhi, India
| | - Bikrant Bihari Lal
- Department of Pediatric Hepatology, Institute of Liver and Biliary Sciences, New Delhi, India
| | - Aathira Ravindranath
- Department of Pediatric Gastroenterology, Apollo BGS Hospital, Mysuru, Karnataka, India
| | - Ashish Bavdekar
- Department of Pediatrics, KEM Hospital and Research Centre, Pune, India
| | - Nirmala Dheivamani
- Department of Pediatric Gastroenterology, Institute of Child Health and Hospital for Children, Chennai, India
| | | | | | - Parijat Ram Tripathi
- Department of Pediatric Gastroenterology, Ankura Hospital for Women and Children, Hyderabad, India
| | - Aabha Nagral
- Jaslok Hospital and Research Center, Mumbai, India
- Apollo Hospital, Navi Mumbai, India
| | | | - Ira Shah
- Pediatric Infectious Diseases and Pediatric Gastroenterology, Hepatology B.J. Wadia Hospital for Children, Mumbai, India
| | | | - Arya Suchismita
- Department of Gastroenterology, Indira Gandhi Institute of Medical Sciences, Patna, India
| | | | - Vaibhav Shah
- Gujarat Superspeciality Clinic, Ahmedabad, India
| | - Zahabiya Nalwalla
- Department of Pediatrics, B.J. Wadia Hospital for Children, Mumbai, India
| | - Karunesh Kumar
- Department of Pediatric Gastroenterology & Liver Transplantation, Indraprastha Apollo Hospitals, New Delhi, India
| | - Arjun Maria
- Department of Pediatrics, Sir Gangaram Hospital, New Delhi, India
| | - Anupam Sibal
- Department of Pediatric Gastroenterology & Liver Transplantation, Indraprastha Apollo Hospitals, New Delhi, India
| | | | - Nishant Wadhwa
- Department of Pediatrics, Sir Gangaram Hospital, New Delhi, India
| | - A Ashritha
- Department of Pediatric Hepatology, Institute of Liver and Biliary Sciences, New Delhi, India
| | - Vikrant Sood
- Department of Pediatric Hepatology, Institute of Liver and Biliary Sciences, New Delhi, India
| | - Rajeev Khanna
- Department of Pediatric Hepatology, Institute of Liver and Biliary Sciences, New Delhi, India
| |
Collapse
|
3
|
Lal BB, Alam S, Sibal A, Kumar K, Hosaagrahara Ramakrishna S, Shah V, Dheivamani N, Bavdekar A, Nagral A, Wadhwa N, Maria A, Shah A, Shah I, Nalwalla Z, Snehavardhan P, Srikanth KP, Gupta S, Sivaramakrishnan VM, Waikar Y, Suchismita A, Ashritha A, Sood V, Khanna R. Genotype correlates with clinical course and outcome of children with tight junction protein 2 (TJP2) deficiency-related cholestasis. Hepatology 2024; 80:511-526. [PMID: 38447037 DOI: 10.1097/hep.0000000000000828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Accepted: 02/09/2024] [Indexed: 03/08/2024]
Abstract
BACKGROUND AND AIMS The study aimed to describe the clinical course and outcomes, and analyze the genotype-phenotype correlation in patients with tight junction protein 2 (TJP2) deficiency. APPROACH AND RESULTS Data from all children with chronic cholestasis and either homozygous or compound heterozygous mutations in TJP2 were extracted and analyzed. The patients were categorized into 3 genotypes: TJP2-A (missense mutations on both alleles), TJP2-B (missense mutation on one allele and a predicted protein-truncating mutation [PPTM] on the other), and TJP2-C (PPTMs on both alleles). A total of 278 cases of genetic intrahepatic cholestasis were studied, with TJP2 deficiency accounting for 44 cases (15.8%). Of these, 29 were homozygous and 15 were compound heterozygous variants of TJP2 . TJP2-A genotype was identified in 21 (47.7%), TJP2-B in 7 cases (15.9%), and TJP2-C in 16 cases (36.4%), respectively. Patients with the TJP2-C genotype were more likely to experience early infantile cholestasis (87.5% vs. 53.5%, p =0.033), less likely to clear jaundice (12.5% vs. 52.2%, p =0.037), more likely to develop ascites, and had higher serum bile acids. Patients with the TJP2-C genotype were more likely to die or require liver transplantation (native liver survival: 12.5% vs. 78.6%, p <0.001), with a median age at death/liver transplantation of 2.5 years. Cox regression analysis revealed that TJP2-C mutations ( p =0.003) and failure to resolve jaundice ( p =0.049) were independent predictors of poor outcomes. CONCLUSIONS Patients with the TJP2-C genotype carrying PPTMs in both alleles had a rapidly progressive course, leading to early decompensation and death if they did not receive timely liver transplantation.
Collapse
Affiliation(s)
- Bikrant Bihari Lal
- Department of Pediatric Hepatology, Institute of Liver and Biliary Sciences, New Delhi, India
| | - Seema Alam
- Department of Pediatric Hepatology, Institute of Liver and Biliary Sciences, New Delhi, India
| | - Anupam Sibal
- Department of Pediatric Gastroenterology & Liver Transplantation, Indraprastha Apollo Hospitals, New Delhi, India
| | - Karunesh Kumar
- Department of Pediatric Gastroenterology & Liver Transplantation, Indraprastha Apollo Hospitals, New Delhi, India
| | | | - Vaibhav Shah
- Department of Pediatric Gastroenterology and Hepatology, Gujarat Superspeciality Clinic, Ahmedabad, Gujarat, India
| | - Nirmala Dheivamani
- Department of Pediatric Gastroenterology, Institute of Child Health and Hospital for Children, Chennai, Tamil Nadu, India
| | - Ashish Bavdekar
- Department of Pediatrics, KEM Hospital and Research Centre, Pune, Maharashtra, India
| | - Aabha Nagral
- Department of Pediatric Hepatology and Liver Transplant, Jaslok Hospital and Research Center, Mumbai, Maharashtra, India
- Department of Pediatric Hepatology and Liver Transplantation, Apollo Hospital, Navi Mumbai, Maharashtra, India
| | - Nishant Wadhwa
- Department of Pediatrics, Sir Gangaram Hospital, New Delhi, India
| | - Arjun Maria
- Department of Pediatrics, Sir Gangaram Hospital, New Delhi, India
| | - Aashay Shah
- Department of Pediatric Gastroenterology, PRISM Pediatric Gastro, Ahmedabad, Gujarat, India
| | - Ira Shah
- Department of Pediatric Infectious Disease, Pediatric Gastroenterology, and Hepatology B.J. Wadia Hospital for Children, Mumbai, Maharashtra, India
| | - Zahabiya Nalwalla
- Department of Pediatrics, B.J. Wadia Hospital for Children, Mumbai, Maharashtra, India
| | - Pandey Snehavardhan
- Department of Pediatric Hepatology and Liver Transplantation, Sahyadri Superspeciality Hospital Pvt Ltd, Pune, Maharashtra, India
| | - K P Srikanth
- Department of Pediatric Gastroenterology and Hepatology, Pediatric Gastroenterology & Hepatology, Manipal Hospitals, Bengaluru, Karnataka, India
| | - Subhash Gupta
- Department of Liver Transplantation, Centre for Liver and Biliary Sciences, Max Superspeciality Hospital, Saket, New Delhi, India
| | | | - Yogesh Waikar
- Department of Pediatric Gastroenterology, Superspeciality GI Kids Clinics, Nagpur, Maharashtra, India
| | - Arya Suchismita
- Department of Gastroenterology, Indira Gandhi Institute of Medical Sciences, Patna, Bihar, India
| | - A Ashritha
- Department of Pediatric Hepatology, Institute of Liver and Biliary Sciences, New Delhi, India
| | - Vikrant Sood
- Department of Pediatric Hepatology, Institute of Liver and Biliary Sciences, New Delhi, India
| | - Rajeev Khanna
- Department of Pediatric Hepatology, Institute of Liver and Biliary Sciences, New Delhi, India
| |
Collapse
|
4
|
Wakasa K, Tamura R, Osaka S, Takei H, Asai A, Nittono H, Kusuhara H, Hayashi H. Rapid in vivo evaluation system for cholestasis-related genes in mice with humanized bile acid profiles. Hepatol Commun 2024; 8:e0382. [PMID: 38517206 PMCID: PMC10962888 DOI: 10.1097/hc9.0000000000000382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Accepted: 12/05/2023] [Indexed: 03/23/2024] Open
Abstract
BACKGROUND Pediatric cholestatic liver diseases (Ped-CLD) comprise many ultrarare disorders with a genetic basis. Pharmacologic therapy for severe cases of Ped-CLD has not been established. Species differences in bile acid (BA) metabolism between humans and rodents contribute to the lack of phenocopy of patients with Ped-CLD in rodents and hinder the development of therapeutic strategies. We aimed to establish an efficient in vivo system to understand BA-related pathogenesis, such as Ped-CLD. METHODS We generated mice that express spCas9 specifically in the liver (L-Cas9Tg/Tg [liver-specific Cas9Tg/Tg] mice) and designed recombinant adeno-associated virus serotype 8 encoding small-guide RNA (AAV8 sgRNA) targeting Abcc2, Abcb11, and Cyp2c70. In humans, ABCC2 and ABCB11 deficiencies cause constitutional hyperbilirubinemia and most severe Ped-CLD, respectively. Cyp2c70 encodes an enzyme responsible for the rodent-specific BA profile. Six-week-old L-Cas9Tg/Tg mice were injected with this AAV8 sgRNA and subjected to biochemical and histological analysis. RESULTS Fourteen days after the injection with AAV8 sgRNA targeting Abcc2, L-Cas9Tg/Tg mice exhibited jaundice and phenocopied patients with ABCC2 deficiency. L-Cas9Tg/Tg mice injected with AAV8 sgRNA targeting Abcb11 showed hepatomegaly and cholestasis without histological evidence of liver injury. Compared to Abcb11 alone, simultaneous injection of AAV8 sgRNA for Abcb11 and Cyp2c70 humanized the BA profile and caused higher transaminase levels and parenchymal necrosis, resembling phenotypes with ABCB11 deficiency. CONCLUSIONS This study provides proof of concept for efficient in vivo assessment of cholestasis-related genes in humanized bile acid profiles. Our platform offers a more time- and cost-effective alternative to conventional genetically engineered mice, increasing our understanding of BA-related pathogenesis such as Ped-CLD and expanding the potential for translational research.
Collapse
Affiliation(s)
- Kihiro Wakasa
- Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Science, The University of Tokyo, Tokyo, Japan
| | - Ryutaro Tamura
- Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Science, The University of Tokyo, Tokyo, Japan
| | - Shuhei Osaka
- Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Science, The University of Tokyo, Tokyo, Japan
| | - Hajime Takei
- Junshin Clinic Bile Acid Institute, Tokyo, Japan
| | - Akihiro Asai
- Department of Gastroenterology, and Hepatology, and Nutrition, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | | | - Hiroyuki Kusuhara
- Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Science, The University of Tokyo, Tokyo, Japan
| | - Hisamitsu Hayashi
- Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Science, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
5
|
Tavabie OD, Salehi S, Aluvihare VR. The challenges and potential of microRNA-based therapy for patients with liver failure syndromes and hepatocellular carcinoma. Expert Opin Ther Targets 2024; 28:179-191. [PMID: 38487923 DOI: 10.1080/14728222.2024.2331598] [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: 12/21/2023] [Accepted: 03/13/2024] [Indexed: 03/20/2024]
Abstract
INTRODUCTION Morbidity and mortality from liver disease continues to rise worldwide. There are currently limited curative treatments for patients with liver failure syndromes, encompassing acute liver failure and decompensated cirrhosis states, outside of transplantation. Whilst there have been improvements in therapeutic options for patients with hepatocellular carcinoma (HCC), there remain challenges necessitating novel therapeutic agents. microRNA have long been seen as potential therapeutic targets but there has been limited clinical translation. AREAS COVERED We will discuss the limitations of conventional non-transplant management of patients with liver failure syndromes and HCC. We will provide an overview of microRNA and the challenges in developing and delivering microRNA-based therapeutic agents. We will finally provide an overview of microRNA-based therapeutic agents which have progressed to clinical trials. EXPERT OPINION microRNA have great potential to be developed into therapeutic agents due to their association with critical biological processes which govern health and disease. Utilizing microRNA sponges to target multiple microRNA associated with specific biological processes may improve their therapeutic efficacy. However, there needs to be significant improvements in delivery systems to ensure the safe delivery of microRNA to target sites and minimize systemic distribution. This currently significantly impacts the clinical translation of microRNA-based therapeutic agents.
Collapse
Affiliation(s)
| | - Siamak Salehi
- Institute of Liver Studies, King's College Hospital, London, UK
| | | |
Collapse
|
6
|
Ren L, Cheng SG, Kang PC, Li TF, Li X, Xiao JZ, Jiang D. Silenced LASP1 interacts with DNMT1 to promote TJP2 expression and attenuate articular cartilage injury in mice by suppressing TJP2 methylation. Kaohsiung J Med Sci 2023; 39:1096-1105. [PMID: 37578083 DOI: 10.1002/kjm2.12738] [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: 12/21/2022] [Revised: 06/03/2023] [Accepted: 07/06/2023] [Indexed: 08/15/2023] Open
Abstract
To investigate the regulatory mechanisms and effects of LIM and SH3 protein 1 (LASP1) on osteoarthritis (OA). IL-1β was used to induce OA in cell models. Viability and apoptosis of chondrocytes were assessed. The expressions of tumor necrsis factor-α (TNF-α) and IL-6 were measured by ELISA kit, and Quantitative reverse transcription polymerase chain reaction (qRT-PCR) and Western blot were performed to test the expression of related proteins. The STRING database was used to predict the relationship between LASP1 and DNA methyltransferase 1 (DNMT1). The tight junction protein 2 (TJP2) and Gene Expression Omnibus data were analyzed for differential OA genes. Methylation-specific PCR detected methylation of the TJP2 promoter region, and chromatin immunoprecipitation detected the enrichment of DNMT1 in the TJP2 promoter region. Safranin O-Fast Green staining and hematoxylin and eosin staining were used to determine the OARSI score and evaluate the pathological conditions of the joint tissues. LASP1 was highly expressed in IL-1β-induced cell models. Silencing of LASP1 promoted chondrocyte proliferation and expression of Collagen II and Aggrecan and inhibited chondrocyte apoptosis, inflammatory factors, and matrix metalloprotein expression. TJP2 is weakly expressed in OA models, and LASP1 promotes methylation of the TJP2 promoter region by interacting with DNMT1. Silencing of LASP1 attenuated IL-1β-induced chondrocyte degeneration by promoting TJP2 expression. Similarly, silencing LASP1 promotes TJP2 expression to alleviate articular cartilage injury in mice with OA. Silencing of LASP1 inhibited the methylation of the TJP2 promoter region by interacting with DNMT1, thereby alleviating articular cartilage damage in OA mice.
Collapse
Affiliation(s)
- Lian Ren
- Department of Orthopedic Surgery, Loudi Central Hospital, Loudi, China
| | - Shi-Gao Cheng
- Department of Orthopedic Surgery, Loudi Central Hospital, Loudi, China
| | - Peng-Cheng Kang
- Department of Orthopedic Surgery, Loudi Central Hospital, Loudi, China
| | - Teng-Fei Li
- Department of Orthopedic Surgery, Loudi Central Hospital, Loudi, China
| | - Xun Li
- Department of Orthopedic Surgery, Loudi Central Hospital, Loudi, China
| | - Jiong-Zhe Xiao
- Department of Orthopedic Surgery, Loudi Central Hospital, Loudi, China
| | - Dong Jiang
- Department of Orthopedic Surgery, Loudi Central Hospital, Loudi, China
| |
Collapse
|
7
|
Tamargo-Rubio I, Simpson AB, Hoogerland JA, Fu J. Human induced pluripotent stem cell-derived liver-on-a-chip for studying drug metabolism: the challenge of the cytochrome P450 family. Front Pharmacol 2023; 14:1223108. [PMID: 37448965 PMCID: PMC10338083 DOI: 10.3389/fphar.2023.1223108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Accepted: 06/20/2023] [Indexed: 07/18/2023] Open
Abstract
The liver is the primary organ responsible for the detoxification and metabolism of drugs. To date, a lack of preclinical models that accurately emulate drug metabolism by the human liver presents a significant challenge in the drug development pipeline, particularly for predicting drug efficacy and toxicity. In recent years, emerging microfluidic-based organ-on-a-chip (OoC) technologies, combined with human induced pluripotent stem cell (hiPSC) technology, present a promising avenue for the complete recapitulation of human organ biology in a patient-specific manner. However, hiPSC-derived organoids and liver-on-a-chip models have so far failed to sufficiently express cytochrome P450 monooxygenase (CYP450) enzymes, the key enzymes involved in first-pass metabolism, which limits the effectiveness and translatability of these models in drug metabolism studies. This review explores the potential of innovative organoid and OoC technologies for studying drug metabolism and discusses their existing drawbacks, such as low expression of CYP450 genes. Finally, we postulate potential approaches for enhancing CYP450 expression in the hope of paving the way toward developing novel, fully representative liver drug-metabolism models.
Collapse
Affiliation(s)
- Isabel Tamargo-Rubio
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Anna Bella Simpson
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Joanne A. Hoogerland
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Jingyuan Fu
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| |
Collapse
|
8
|
Xu J, Kausalya PJ, Ong AGM, Goh CMF, Mohamed Ali S, Hunziker W. ZO-2/Tjp2 suppresses Yap and Wwtr1/Taz-mediated hepatocyte to cholangiocyte transdifferentiation in the mouse liver. NPJ Regen Med 2022; 7:55. [PMID: 36151109 PMCID: PMC9508083 DOI: 10.1038/s41536-022-00251-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 09/02/2022] [Indexed: 01/11/2023] Open
Abstract
TJP2/ZO-2-inactivating mutations in humans cause progressive cholestatic liver disease. Liver-specific deletion of Tjp2 in the mouse (Tjp2 cKO mice) leads to mild progressive cholestasis without an overt degradation of the bile-blood barrier (BBB). These mice are more susceptible to cholic acid (CA) induced liver injury. Interestingly, while initially also more susceptible, Tjp2 cKO mice develop tolerance to a DDC-supplemented diet. The DDC diet induces an exacerbated ductular reaction in Tjp2 cKO mice, which arises from the transdifferentiation of hepatocytes to cholangiocytes. Consequently, this transdifferentiation is only observed if Tjp2 is inactivated in hepatocytes, but not if deleted in cholangiocytes. The DDC-diet-induced hepatocyte transdifferentiation in Tjp2 cKO mice requires Yap and Wwtr1/Taz, whose protein expression is upregulated in hepatocytes lacking Tjp2, but is independent of Notch2. Although inactivating Tjp2 is sufficient for the upregulation of Yap and Wwtr1/Taz protein, efficient transdifferentiation requires the DDC-diet insult. Thus, Tjp2 negatively regulates Yap/Taz-mediated transdifferentiation of hepatocytes to cholangiocytes in response to DDC-diet-induced liver injury. Furthermore, transdifferentiation is regulated at multiple levels and the type of injury inflicted on the Tjp2 deficient liver plays an important role in the resulting pathophysiology.
Collapse
Affiliation(s)
- Jianliang Xu
- Epithelial Polarity in Disease and Tissue Regeneration Laboratory, Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive Proteos, Singapore, 138673, Singapore.
| | - P Jaya Kausalya
- Epithelial Polarity in Disease and Tissue Regeneration Laboratory, Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive Proteos, Singapore, 138673, Singapore.,M Diagnostics Pte. Ltd. (MiRXES), 30 Biopolis Road, #09-05/06 Matrix, Singapore, 138671, Singapore
| | - Alicia Ghia Min Ong
- Epithelial Polarity in Disease and Tissue Regeneration Laboratory, Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive Proteos, Singapore, 138673, Singapore
| | - Christine Meng Fan Goh
- Epithelial Polarity in Disease and Tissue Regeneration Laboratory, Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive Proteos, Singapore, 138673, Singapore
| | - Safiah Mohamed Ali
- Epithelial Polarity in Disease and Tissue Regeneration Laboratory, Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive Proteos, Singapore, 138673, Singapore
| | - Walter Hunziker
- Epithelial Polarity in Disease and Tissue Regeneration Laboratory, Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive Proteos, Singapore, 138673, Singapore. .,Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, 2 Medical Drive MD9, Singapore, 117593, Singapore.
| |
Collapse
|