1
|
Ilyas I, Nisa FU, Ali MB, Arshad F, Irfan N, Asif M, Amin I. Unveiling genetic variants: Tetra-primer ARMS-PCR diagnosis and structural insights into BLAD, BC, and DUMPS in Pakistani cattle herds. Mol Biol Rep 2024; 51:963. [PMID: 39235569 DOI: 10.1007/s11033-024-09898-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Accepted: 08/27/2024] [Indexed: 09/06/2024]
Abstract
BACKGROUND Bovine leukocyte adhesion deficiency (BLAD), bovine citrullinemia (BC), and deficiency of Uridine monophosphate synthetase (DUMPS) are the common autosomal recessive disorders affecting the global dairy industry. BLAD leads to poor wound healing and recurrent infections. In BC, ammonia builds up leading to neurological disorders and death. DUMPS results in developmental abnormalities. METHODOLOGY In this study, tetra-primer amplification refractory mutation system polymerase chain reaction (ARMS PCR) based diagnostic tests were optimized for BLAD, BC, and DUMPS. A total of 250 animals (58 indigenous and 192 Holstein Friesian (HF)) were screened from all across Pakistan. In addition to validation of ARMS-PCR results through Sanger sequencing, the protein modeling provided structural insights of the disease-associated reported SNPs. Pathway analysis illustrated gene functions under normal and mutated conditions. Furthermore, haplotype and phylogenetic analysis of ASS1 (Argininosuccinate synthetase) gene were performed on study samples and NCBI retrieved sequences. RESULTS The study's focus was to screen the herds for prevalence of carriers of genetic disorders, as they are the main source of disease dissemination. One animal was found carrier for BC, whereas no carriers were found for BLAD and DUMPS. The protein models corroborated the reported amino acid change in BLAD, and protein truncation in both BC and DUMPS proteins. SNPs found in NCBI retrieved sequences were either silent or missense and had no effect on protein structure. DNA network presented graphical illustration of haplotype interactions and phylogenetic analysis conferred evolutionary landscape of ASS1 gene. The combination of these approaches produced an in-depth genetic picture of BC in Pakistani cattle. CONCLUSION The development of diagnostic tests and identification of the heterozygous BC sample underscores the significance of constant monitoring to avoid the unwanted dissemination of mutant alleles among Pakistani cattle, thereby promoting the general well-being and sustainability of the dairy sector.
Collapse
Affiliation(s)
- Iram Ilyas
- Agricultural Biotechnology Division, National Institute for Biotechnology and Genetic Engineering (NIBGE), Constituent College of Pakistan Institute of Engineering and Applied Sciences, Jhang Road, Faisalabad, 38000, Pakistan
| | - Fakhar Un Nisa
- Agricultural Biotechnology Division, National Institute for Biotechnology and Genetic Engineering (NIBGE), Constituent College of Pakistan Institute of Engineering and Applied Sciences, Jhang Road, Faisalabad, 38000, Pakistan
- Department of Animal Breeding and Genetics, University of Veterinary and Animal Sciences, Lahore, Pakistan
| | - Muhammad Basil Ali
- Agricultural Biotechnology Division, National Institute for Biotechnology and Genetic Engineering (NIBGE), Constituent College of Pakistan Institute of Engineering and Applied Sciences, Jhang Road, Faisalabad, 38000, Pakistan
- Senckenberg Biodiversity and Climate Research Institute (SBiK-F), Georg-Voigt-Strasse 14-16, 60325, Frankfurt am Main, Germany
| | - Fazeela Arshad
- Agricultural Biotechnology Division, National Institute for Biotechnology and Genetic Engineering (NIBGE), Constituent College of Pakistan Institute of Engineering and Applied Sciences, Jhang Road, Faisalabad, 38000, Pakistan
- The Roslin Institute, University of Edinburgh, Easter Bush campus, Edinburgh, Scotland
| | - Najida Irfan
- Agricultural Biotechnology Division, National Institute for Biotechnology and Genetic Engineering (NIBGE), Constituent College of Pakistan Institute of Engineering and Applied Sciences, Jhang Road, Faisalabad, 38000, Pakistan
| | - Muhammad Asif
- Agricultural Biotechnology Division, National Institute for Biotechnology and Genetic Engineering (NIBGE), Constituent College of Pakistan Institute of Engineering and Applied Sciences, Jhang Road, Faisalabad, 38000, Pakistan.
| | - Imran Amin
- Agricultural Biotechnology Division, National Institute for Biotechnology and Genetic Engineering (NIBGE), Constituent College of Pakistan Institute of Engineering and Applied Sciences, Jhang Road, Faisalabad, 38000, Pakistan
| |
Collapse
|
2
|
Gomez N, Hsieh C, Li X, Dykstra M, Waksmacki J, Altheim C, Bechar Y, Klim J, Zaepfel B, Rothstein J, Tank EE, Barmada SJ. Counter-regulation of RNA stability by UPF1 and TDP43. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.31.578310. [PMID: 38352350 PMCID: PMC10862862 DOI: 10.1101/2024.01.31.578310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/22/2024]
Abstract
RNA quality control is crucial for proper regulation of gene expression. Disruption of nonsense mediated mRNA decay (NMD), the primary RNA decay pathway responsible for the degradation of transcripts containing premature termination codons (PTCs), can disrupt development and lead to multiple diseases in humans and other animals. Similarly, therapies targeting NMD may have applications in hematological, neoplastic and neurological disorders. As such, tools capable of accurately quantifying NMD status could be invaluable for investigations of disease pathogenesis and biomarker identification. Toward this end, we assemble, validate, and apply a next-generation sequencing approach (NMDq) for identifying and measuring the abundance of PTC-containing transcripts. After validating NMDq performance and confirming its utility for tracking RNA surveillance, we apply it to determine pathway activity in two neurodegenerative diseases, amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD) characterized by RNA misprocessing and abnormal RNA stability. Despite the genetic and pathologic evidence implicating dysfunctional RNA metabolism, and NMD in particular, in these conditions, we detected no significant differences in PTC-encoding transcripts in ALS models or disease. Contrary to expectations, overexpression of the master NMD regulator UPF1 had little effect on the clearance of transcripts with PTCs, but rather restored RNA homeostasis through differential use and decay of alternatively poly-adenylated isoforms. Together, these data suggest that canonical NMD is not a significant contributor to ALS/FTD pathogenesis, and that UPF1 promotes neuronal survival by regulating transcripts with abnormally long 3'UTRs.
Collapse
|
3
|
Makris G, Veit L, Rüfenacht V, Klassa S, Zürcher N, Matsumoto S, Poms M, Häberle J. Expression and function of the urea cycle in widely-used hepatic cellular models. J Inherit Metab Dis 2024. [PMID: 38192032 DOI: 10.1002/jimd.12701] [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: 05/25/2023] [Revised: 11/30/2023] [Accepted: 12/04/2023] [Indexed: 01/10/2024]
Abstract
The group of rare metabolic defects termed urea cycle disorders (UCDs) occur within the ammonia elimination pathway and lead to significant neurocognitive sequelae for patients surviving decompensation episodes. Besides orthotopic liver transplantation, curative options are lacking for UCDs, with dietary management being the gold clinical standard. Novel therapeutic approaches are essential for UCDs; however, such effort presupposes preclinical testing in cellular models that effectively capture disease manifestation. Several cellular and animal models exist and aim to recapitulate the broad phenotypic spectrum of UCDs; however, the majority of those lack extensive molecular and biochemical characterization. The development of cellular models is emerging since animal models are extremely time and cost consuming, and subject to ethical considerations, including the 3R principle that endorses animal welfare over unchecked preclinical testing. The aim of this study was to compare the extent of expression and functionality of the urea cycle in two commercial hepatoma-derived cell lines, induced pluripotent stem cell hepatocytes (iPSC-Heps), primary human hepatocytes (PHHs) and human liver cell preparations. Using immunoblotting, immunocytochemistry, and stable isotope tracing of the urea cycle metabolites, we identified that the hepatoma-derived, 2-week differentiated HepaRG cells are urea cycle proficient and behave as cellular alternatives to PHHs. Furthermore, HepaRG cells were superior to iPSC-Heps, which are known to exhibit batch-to-batch variabilities in terms of hepatic maturity and enzyme expression. Finally, HepG2 cells lack the urea cycle enzymes ornithine transcarbamylase and arginase 1, the transporter ORNT1, which limits their suitability as model for the study of UCDs.
Collapse
Affiliation(s)
- Georgios Makris
- Division of Metabolism and Children's Research Center, University Children's Hospital Zurich, Zurich, Switzerland
- Center for Integrative Human Physiology, University of Zurich, Zurich, Switzerland
| | - Lara Veit
- Division of Metabolism and Children's Research Center, University Children's Hospital Zurich, Zurich, Switzerland
| | - Véronique Rüfenacht
- Division of Metabolism and Children's Research Center, University Children's Hospital Zurich, Zurich, Switzerland
| | - Sven Klassa
- Division of Metabolism and Children's Research Center, University Children's Hospital Zurich, Zurich, Switzerland
| | - Nadia Zürcher
- Division of Metabolism and Children's Research Center, University Children's Hospital Zurich, Zurich, Switzerland
| | - Shirou Matsumoto
- Department of Pediatrics, Graduate School of Medical Sciences, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Martin Poms
- Division of Clinical Chemistry and Biochemistry, University Children's Hospital Zurich, Zurich, Switzerland
| | - Johannes Häberle
- Division of Metabolism and Children's Research Center, University Children's Hospital Zurich, Zurich, Switzerland
| |
Collapse
|
4
|
Li K, Ren K, Du S, Gao X, Yu J. Development of Liver Cancer Organoids: Reproducing Tumor Microenvironment and Advancing Research for Liver Cancer Treatment. Technol Cancer Res Treat 2024; 23:15330338241285097. [PMID: 39363866 PMCID: PMC11456184 DOI: 10.1177/15330338241285097] [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: 04/26/2024] [Revised: 07/26/2024] [Accepted: 08/30/2024] [Indexed: 10/05/2024] Open
Abstract
Liver cancer a leading cause of cancer-related deaths worldwide, yet understanding of its development mechanism remains limited, and treatment barriers present substantial challenges. Owing to the heterogeneity of tumors, traditional 2D culture models are inadequate for capturing the complexity and diversity of tumor biology and understanding of the disease. Organoids have garnered considerable attention because of their ability to self-renew and develop functional structures in vitro that closely resemble those of human organs. This review explores the history of liver organoids, their cellular origins, techniques of constructing tumor microenvironments that recapitulate liver cancer organoids, and the biological and clinical applications of liver and liver cancer organoids and explores the current challenges related to liver cancer organoid applications and potentially valuable solutions, with the aim of facilitating the construction of in vitro clinical models of liver cancer therapeutic research.
Collapse
Affiliation(s)
- Kangkang Li
- Department of Hepato-Biliary-Pancreatic Surgery, Fuyang Hospital Affiliataed Bengbu Medical College, Fuyang, Anhui province, China, 236000
| | - Kuiwu Ren
- Department of Hepato-Biliary-Pancreatic Surgery, Fuyang Hospital Affiliataed Bengbu Medical College, Fuyang, Anhui province, China, 236000
| | - Sen Du
- Department of Hepato-Biliary-Pancreatic Surgery, Fuyang Hospital Affiliataed Bengbu Medical College, Fuyang, Anhui province, China, 236000
| | - Xiang Gao
- Department of Hepato-Biliary-Pancreatic Surgery, Fuyang People's Hospital of Anhui Medical University, Fuyang, Anhui province, China, 236000
| | - Jiangtao Yu
- Department of Hepato-Biliary-Pancreatic Surgery, Fuyang Hospital Affiliataed Bengbu Medical College, Fuyang, Anhui province, China, 236000
- Department of Hepato-Biliary-Pancreatic Surgery, Fuyang People's Hospital of Anhui Medical University, Fuyang, Anhui province, China, 236000
| |
Collapse
|
5
|
Luo Q, Wang N, Que H, Mai E, Hu Y, Tan R, Gu J, Gong P. Pluripotent Stem Cell-Derived Hepatocyte-like Cells: Induction Methods and Applications. Int J Mol Sci 2023; 24:11592. [PMID: 37511351 PMCID: PMC10380504 DOI: 10.3390/ijms241411592] [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: 04/17/2023] [Revised: 07/09/2023] [Accepted: 07/12/2023] [Indexed: 07/30/2023] Open
Abstract
The development of regenerative medicine provides new options for the treatment of end-stage liver diseases. Stem cells, such as bone marrow mesenchymal stem cells, embryonic stem cells, and induced pluripotent stem cells (iPSCs), are effective tools for tissue repair in regenerative medicine. iPSCs are an appropriate source of hepatocytes for the treatment of liver disease due to their unlimited multiplication capacity, their coverage of the entire range of genetics required to simulate human disease, and their evasion of ethical implications. iPSCs have the ability to gradually produce hepatocyte-like cells (HLCs) with homologous phenotypes and physiological functions. However, how to induce iPSCs to differentiate into HLCs efficiently and accurately is still a hot topic. This review describes the existing approaches for inducing the differentiation of iPSCs into HLCs, as well as some challenges faced, and summarizes various parameters for determining the quality and functionality of HLCs. Furthermore, the application of iPSCs for in vitro hepatoprotective drug screening and modeling of liver disease is discussed. In conclusion, iPSCs will be a dependable source of cells for stem-cell therapy to treat end-stage liver disease and are anticipated to facilitate individualized treatment for liver disease in the future.
Collapse
Affiliation(s)
- Qiulin Luo
- College of Pharmacy, Southwest Minzu University, Chengdu 610225, China
| | - Nan Wang
- College of Pharmacy, Southwest Minzu University, Chengdu 610225, China
| | - Hanyun Que
- College of Pharmacy, Southwest Minzu University, Chengdu 610225, China
| | - Erziya Mai
- College of Pharmacy, Southwest Minzu University, Chengdu 610225, China
| | - Yanting Hu
- College of Pharmacy, Southwest Minzu University, Chengdu 610225, China
| | - Rui Tan
- College of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610032, China
| | - Jian Gu
- College of Pharmacy, Southwest Minzu University, Chengdu 610225, China
| | - Puyang Gong
- College of Pharmacy, Southwest Minzu University, Chengdu 610225, China
| |
Collapse
|
6
|
Duff C, Baruteau J. Modelling urea cycle disorders using iPSCs. NPJ Regen Med 2022; 7:56. [PMID: 36163209 PMCID: PMC9513077 DOI: 10.1038/s41536-022-00252-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 08/10/2022] [Indexed: 11/23/2022] Open
Abstract
The urea cycle is a liver-based pathway enabling disposal of nitrogen waste. Urea cycle disorders (UCDs) are inherited metabolic diseases caused by deficiency of enzymes or transporters involved in the urea cycle and have a prevalence of 1:35,000 live births. Patients present recurrent acute hyperammonaemia, which causes high rate of death and neurological sequelae. Long-term therapy relies on a protein-restricted diet and ammonia scavenger drugs. Currently, liver transplantation is the only cure. Hence, high unmet needs require the identification of effective methods to model these diseases to generate innovative therapeutics. Advances in both induced pluripotent stem cells (iPSCs) and genome editing technologies have provided an invaluable opportunity to model patient-specific phenotypes in vitro by creating patients' avatar models, to investigate the pathophysiology, uncover novel therapeutic targets and provide a platform for drug discovery. This review summarises the progress made thus far in generating 2- and 3-dimensional iPSCs models for UCDs, the challenges encountered and how iPSCs offer future avenues for innovation in developing the next-generation of therapies for UCDs.
Collapse
Affiliation(s)
- Claire Duff
- Genetics and Genomic Medicine Department, Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Julien Baruteau
- Genetics and Genomic Medicine Department, Great Ormond Street Institute of Child Health, University College London, London, UK.
- National Institute of Health Research Great Ormond Street Biomedical Research Centre, London, UK.
- Metabolic Medicine Department, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK.
| |
Collapse
|
7
|
Fátima Lopes F, Sitta A, de Moura Coelho D, Schmitt Ribas G, Lamberty Faverzani J, Gomes Dos Reis B, Wajner M, Vargas CR. Clinical findings of patients with hyperammonemia affected by urea cycle disorders with hepatic encephalopathy. Int J Dev Neurosci 2022; 82:772-788. [PMID: 36129623 DOI: 10.1002/jdn.10229] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 08/21/2022] [Accepted: 09/15/2022] [Indexed: 11/10/2022] Open
Abstract
Urea Cycle Disorders (UCD) are a group of genetic diseases caused by deficiencies in the enzymes and transporters involved in the urea cycle. The impairment of the cycle results in ammonia accumulation, leading to neurological dysfunctions and poor outcomes to affected patients. The aim of this study is to investigate and describe UCD patients principal clinical and biochemical presentations to support professionals on urgent diagnosis and quick management, aiming better outcomes for patients. We explored medical records of thirty patients diagnosed in a referral center from Brazil to delineate UCD clinical and biochemical profile. Patients demonstrated a range of signs and symptoms, such as altered levels of consciousness, acute encephalopathy, seizures, progressive loss of appetite, vomiting, coma, and respiratory distress, in most cases combined with high levels of ammonia, which is an immediate biomarker, leading to an UCD suspicion. The most prevalent UCD detected were ornithine transcarbamylase deficiency (11), followed by citrullinemia type I (10), hyperargininemia (5), carbamoyl phosphate synthase 1 deficiency (2) and argininosuccinic aciduria (2). Clinical symptoms were highly severe, being the majority developmental and neurological disabilities, with 20% of death rate. Laboratory analysis revealed high levels of ammonia (mean ± SD: 860 ± 470 μmol/L; reference value: ≤ 80 μmol/L), hypoglycemia, metabolic acidosis, and high excretion of orotic acid in the urine (except in CPS1 deficiency). We emphasize the need of urgent identification of UCD clinical and biochemical conditions, and immediate measurement of ammonia, to enable the correct diagnosis and increase the chances of patients survival, minimizing neurological and psychomotor damage caused by hepatic encephalopathy.
Collapse
Affiliation(s)
- Franciele Fátima Lopes
- Programa de Pós-Graduação em Ciências Farmacêuticas, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.,Serviço de Genética Médica, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Angela Sitta
- Serviço de Genética Médica, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | | | | | - Jéssica Lamberty Faverzani
- Programa de Pós-Graduação em Ciências Farmacêuticas, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Bianca Gomes Dos Reis
- Serviço de Genética Médica, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Moacir Wajner
- Programa de Pós-Graduação em Ciências Farmacêuticas, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.,Serviço de Genética Médica, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Carmen Regla Vargas
- Programa de Pós-Graduação em Ciências Farmacêuticas, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.,Serviço de Genética Médica, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| |
Collapse
|
8
|
Carberry CK, Ferguson SS, Beltran AS, Fry RC, Rager JE. Using liver models generated from human-induced pluripotent stem cells (iPSCs) for evaluating chemical-induced modifications and disease across liver developmental stages. Toxicol In Vitro 2022; 83:105412. [PMID: 35688329 PMCID: PMC9296547 DOI: 10.1016/j.tiv.2022.105412] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 05/20/2022] [Accepted: 06/03/2022] [Indexed: 01/09/2023]
Abstract
The liver is a pivotal organ regulating critical developmental stages of fetal metabolism and detoxification. Though numerous studies have evaluated links between prenatal/perinatal exposures and adverse health outcomes in the developing fetus, the central role of liver to health disruptions resulting from these exposures remains understudied, especially concerning early development and later-in-life health outcomes. While numerous in vitro methods for evaluating liver toxicity have been established, the use of iPSC-derived hepatocytes appears to be particularly well suited to contribute to this critical research gap due to their potential to model a diverse range of disease phenotypes and different stages of liver development. The following key aspects are reviewed: (1) an introduction to developmental liver toxicity; (2) an introduction to embryonic and induced pluripotent stem cell models; (3) methods and challenges for deriving liver cells from stem cells; and (4) applications for iPSC-derived hepatocytes to evaluate liver developmental stages and their associated responses to insults. We conclude that iPSC-derived hepatocytes have great potential for informing liver toxicity and underlying disease mechanisms via the generation of patient-specific iPSCs; implementing large-scale drug and chemical screening; evaluating general biological responses as a potential surrogate target cell; and evaluating inter-individual disease susceptibility and response variability.
Collapse
Affiliation(s)
- Celeste K Carberry
- Department of Environmental Sciences and Engineering, Gillings School of Global Public Health, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA; The Institute for Environmental Health Solutions, Gillings School of Global Public Health, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Stephen S Ferguson
- Biomolecular Screening Branch, National Toxicology Program, Research Triangle Park, NC, USA
| | - Adriana S Beltran
- Department of Genetics, School of Medicine, University of North Carolina, Chapel Hill, NC, USA
| | - Rebecca C Fry
- Department of Environmental Sciences and Engineering, Gillings School of Global Public Health, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA; The Institute for Environmental Health Solutions, Gillings School of Global Public Health, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA; Curriculum in Toxicology and Environmental Medicine, School of Medicine, University of North Carolina, Chapel Hill, NC, USA
| | - Julia E Rager
- Department of Environmental Sciences and Engineering, Gillings School of Global Public Health, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA; The Institute for Environmental Health Solutions, Gillings School of Global Public Health, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA; Curriculum in Toxicology and Environmental Medicine, School of Medicine, University of North Carolina, Chapel Hill, NC, USA.
| |
Collapse
|
9
|
Laemmle A, Poms M, Hsu B, Borsuk M, Rüfenacht V, Robinson J, Sadowski MC, Nuoffer J, Häberle J, Willenbring H. Aquaporin 9 induction in human iPSC-derived hepatocytes facilitates modeling of ornithine transcarbamylase deficiency. Hepatology 2022; 76:646-659. [PMID: 34786702 PMCID: PMC9295321 DOI: 10.1002/hep.32247] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 10/30/2021] [Accepted: 11/14/2021] [Indexed: 12/16/2022]
Abstract
BACKGROUND AND AIMS Patient-derived human-induced pluripotent stem cells (hiPSCs) differentiated into hepatocytes (hiPSC-Heps) have facilitated the study of rare genetic liver diseases. Here, we aimed to establish an in vitro liver disease model of the urea cycle disorder ornithine transcarbamylase deficiency (OTCD) using patient-derived hiPSC-Heps. APPROACH AND RESULTS Before modeling OTCD, we addressed the question of why hiPSC-Heps generally secrete less urea than adult primary human hepatocytes (PHHs). Because hiPSC-Heps are not completely differentiated and maintain some characteristics of fetal PHHs, we compared gene-expression levels in human fetal and adult liver tissue to identify genes responsible for reduced urea secretion in hiPSC-Heps. We found lack of aquaporin 9 (AQP9) expression in fetal liver tissue as well as in hiPSC-Heps, and showed that forced expression of AQP9 in hiPSC-Heps restores urea secretion and normalizes the response to ammonia challenge by increasing ureagenesis. Furthermore, we proved functional ureagenesis by challenging AQP9-expressing hiPSC-Heps with ammonium chloride labeled with the stable isotope [15 N] (15 NH4 Cl) and by assessing enrichment of [15 N]-labeled urea. Finally, using hiPSC-Heps derived from patients with OTCD, we generated a liver disease model that recapitulates the hepatic manifestation of the human disease. Restoring OTC expression-together with AQP9-was effective in fully correcting OTC activity and normalizing ureagenesis as assessed by 15 NH4 Cl stable-isotope challenge. CONCLUSION Our results identify a critical role for AQP9 in functional urea metabolism and establish the feasibility of in vitro modeling of OTCD with hiPSC-Heps. By facilitating studies of OTCD genotype/phenotype correlation and drug screens, our model has potential for improving the therapy of OTCD.
Collapse
Affiliation(s)
- Alexander Laemmle
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell ResearchUniversity of California San FranciscoSan FranciscoCaliforniaUSA,Department of PediatricsUniversity Children's HospitalBernSwitzerland,University Institute of Clinical ChemistryUniversity of BernBernSwitzerland
| | - Martin Poms
- Division of Clinical Chemistry and BiochemistryUniversity Children’s Hospital ZurichZurichSwitzerland
| | - Bernadette Hsu
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell ResearchUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | - Mariia Borsuk
- University Institute of Clinical ChemistryUniversity of BernBernSwitzerland
| | - Véronique Rüfenacht
- Division of Metabolism and Children`s Research CenterUniversity Children’s HospitalZurichSwitzerland
| | - Joshua Robinson
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell ResearchUniversity of California San FranciscoSan FranciscoCaliforniaUSA,Center for Reproductive SciencesUniversity of California San FranciscoSan FranciscoCaliforniaUSA,Department of Obstetrics, Gynecology, and Reproductive SciencesUniversity of California San FranciscoSan FranciscoCaliforniaUSA,Department of PediatricsMedical GeneticsUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | | | - Jean‐Marc Nuoffer
- Department of PediatricsUniversity Children's HospitalBernSwitzerland,University Institute of Clinical ChemistryUniversity of BernBernSwitzerland
| | - Johannes Häberle
- Division of Metabolism and Children`s Research CenterUniversity Children’s HospitalZurichSwitzerland,Zurich Center for Integrative Human PhysiologyUniversity of ZurichZurichSwitzerland
| | - Holger Willenbring
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell ResearchUniversity of California San FranciscoSan FranciscoCaliforniaUSA,Department of SurgeryDivision of Transplant SurgeryUniversity of California San FranciscoSan FranciscoCaliforniaUSA,Liver CenterUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| |
Collapse
|
10
|
Novel Gene-Correction-Based Therapeutic Modalities for Monogenic Liver Disorders. Bioengineering (Basel) 2022; 9:bioengineering9080392. [PMID: 36004917 PMCID: PMC9404740 DOI: 10.3390/bioengineering9080392] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 08/04/2022] [Accepted: 08/10/2022] [Indexed: 11/17/2022] Open
Abstract
The majority of monogenic liver diseases are autosomal recessive disorders, with few being sex-related or co-dominant. Although orthotopic liver transplantation (LT) is currently the sole therapeutic option for end-stage patients, such an invasive surgical approach is severely restricted by the lack of donors and post-transplant complications, mainly associated with life-long immunosuppressive regimens. Therefore, the last decade has witnessed efforts for innovative cellular or gene-based therapeutic strategies. Gene therapy is a promising approach for treatment of many hereditary disorders, such as monogenic inborn errors. The liver is an organ characterized by unique features, making it an attractive target for in vivo and ex vivo gene transfer. The current genetic approaches for hereditary liver diseases are mediated by viral or non-viral vectors, with promising results generated by gene-editing tools, such as CRISPR-Cas9 technology. Despite massive progress in experimental gene-correction technologies, limitations in validated approaches for monogenic liver disorders have encouraged researchers to refine promising gene therapy protocols. Herein, we highlighted the most common monogenetic liver disorders, followed by proposed genetic engineering approaches, offered as promising therapeutic modalities.
Collapse
|
11
|
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: 1] [Impact Index Per Article: 0.5] [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
|
12
|
Nitzahn M, Truong B, Khoja S, Vega-Crespo A, Le C, Eliav A, Makris G, Pyle AD, Häberle J, Lipshutz GS. CRISPR-Mediated Genomic Addition to CPS1 Deficient iPSCs is Insufficient to Restore Nitrogen Homeostasis. THE YALE JOURNAL OF BIOLOGY AND MEDICINE 2021; 94:545-557. [PMID: 34970092 PMCID: PMC8686786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
CPS1 deficiency is an inborn error of metabolism caused by loss-of-function mutations in the CPS1 gene, catalyzing the initial reaction of the urea cycle. Deficiency typically leads to toxic levels of plasma ammonia, cerebral edema, coma, and death, with the only curative treatment being liver transplantation; due to limited donor availability and the invasiveness and complications of the procedure, however, alternative therapies are needed. Induced pluripotent stem cells offer an alternative cell source to partial or whole liver grafts that theoretically would not require immune suppression regimens and additionally are amenable to genetic modifications. Here, we genetically modified CPS1 deficient patient-derived stem cells to constitutively express human codon optimized CPS1 from the AAVS1 safe harbor site. While edited stem cells efficiently differentiated to hepatocyte-like cells, they failed to metabolize ammonia more efficiently than their unedited counterparts. This unexpected result appears to have arisen in part due to transgene promoter methylation, and thus transcriptional silencing, in undifferentiated cells, impacting their capacity to restore the complete urea cycle function upon differentiation. As pluripotent stem cell strategies are being expanded widely for potential cell therapies, these results highlight the need for strict quality control and functional analysis to ensure the integrity of cell products.
Collapse
Affiliation(s)
- Matthew Nitzahn
- Molecular Biology Institute, David Geffen School of
Medicine at UCLA, Los Angeles, CA, USA,Department of Surgery, David Geffen School of Medicine
at UCLA, Los Angeles, CA, USA
| | - Brian Truong
- Department of Surgery, David Geffen School of Medicine
at UCLA, Los Angeles, CA, USA,Department of Molecular and Medical Pharmacology, David
Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Suhail Khoja
- Department of Surgery, David Geffen School of Medicine
at UCLA, Los Angeles, CA, USA
| | - Agustin Vega-Crespo
- Department of Molecular and Medical Pharmacology, David
Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Colleen Le
- Department of Surgery, David Geffen School of Medicine
at UCLA, Los Angeles, CA, USA
| | - Adam Eliav
- Department of Surgery, David Geffen School of Medicine
at UCLA, Los Angeles, CA, USA
| | - Georgios Makris
- Division of Metabolism and Children’s Research Center,
University Children’s Hospital Zurich, Switzerland
| | - April D. Pyle
- Department of Microbiology, Immunology, and Molecular
Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA,Eli and Edythe Broad Stem Cell Center, David Geffen
School of Medicine at UCLA, Los Angeles, CA, USA
| | - Johannes Häberle
- Division of Metabolism and Children’s Research Center,
University Children’s Hospital Zurich, Switzerland
| | - Gerald S. Lipshutz
- Molecular Biology Institute, David Geffen School of
Medicine at UCLA, Los Angeles, CA, USA,Department of Surgery, David Geffen School of Medicine
at UCLA, Los Angeles, CA, USA,Department of Molecular and Medical Pharmacology, David
Geffen School of Medicine at UCLA, Los Angeles, CA, USA,Department of Psychiatry, David Geffen School of
Medicine at UCLA, Los Angeles, CA, USA,Intellectual and Developmental Disabilities Research
Center, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA,Semel Institute for Neuroscience, David Geffen School
of Medicine at UCLA, Los Angeles, CA, USA,To whom all correspondence should be addressed:
Gerald S. Lipshutz, David Geffen School of Medicine at UCLA, Los Angeles, CA
90095-7054;
| |
Collapse
|
13
|
Yamamura Y, Furuichi K, Murakawa Y, Hirabayashi S, Yoshihara M, Sako K, Kitajima S, Toyama T, Iwata Y, Sakai N, Hosomichi K, Murphy PM, Tajima A, Okita K, Osafune K, Kaneko S, Wada T. Identification of candidate PAX2-regulated genes implicated in human kidney development. Sci Rep 2021; 11:9123. [PMID: 33907292 PMCID: PMC8079710 DOI: 10.1038/s41598-021-88743-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2020] [Accepted: 04/16/2021] [Indexed: 02/02/2023] Open
Abstract
PAX2 is a transcription factor essential for kidney development and the main causative gene for renal coloboma syndrome (RCS). The mechanisms of PAX2 action during kidney development have been evaluated in mice but not in humans. This is a critical gap in knowledge since important differences have been reported in kidney development in the two species. In the present study, we hypothesized that key human PAX2-dependent kidney development genes are differentially expressed in nephron progenitor cells from induced pluripotent stem cells (iPSCs) in patients with RCS relative to healthy individuals. Cap analysis of gene expression revealed 189 candidate promoters and 71 candidate enhancers that were differentially activated by PAX2 in this system in three patients with RCS with PAX2 mutations. By comparing this list with the list of candidate Pax2-regulated mouse kidney development genes obtained from the Functional Annotation of the Mouse/Mammalian (FANTOM) database, we prioritized 17 genes. Furthermore, we ranked three genes-PBX1, POSTN, and ITGA9-as the top candidates based on closely aligned expression kinetics with PAX2 in the iPSC culture system and susceptibility to suppression by a Pax2 inhibitor in cultured mouse embryonic kidney explants. Identification of these genes may provide important information to clarify the pathogenesis of RCS, human kidney development, and kidney regeneration.
Collapse
Affiliation(s)
- Yuta Yamamura
- Department of Nephrology and Laboratory Medicine, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, 13-1 Takara-machi, Kanazawa, Ishikawa, 920-8640, Japan
| | - Kengo Furuichi
- Department of Nephrology, School of Medicine, Kanazawa Medical University, 1-1 Daigaku, Uchinada, Kahoku, Ishikawa, 920-0293, Japan.
| | - Yasuhiro Murakawa
- RIKEN Preventive Medicine and Diagnosis Innovation Program, Yokohama, Kanagawa, Japan
| | - Shigeki Hirabayashi
- RIKEN Preventive Medicine and Diagnosis Innovation Program, Yokohama, Kanagawa, Japan
| | - Masahito Yoshihara
- Division of Genomic Technologies, RIKEN Center for Life Science Technologies, Yokohama, Kanagawa, Japan
| | - Keisuke Sako
- Department of Nephrology and Laboratory Medicine, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, 13-1 Takara-machi, Kanazawa, Ishikawa, 920-8640, Japan
| | - Shinji Kitajima
- Department of Nephrology and Laboratory Medicine, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, 13-1 Takara-machi, Kanazawa, Ishikawa, 920-8640, Japan
| | - Tadashi Toyama
- Department of Nephrology and Laboratory Medicine, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, 13-1 Takara-machi, Kanazawa, Ishikawa, 920-8640, Japan
| | - Yasunori Iwata
- Department of Nephrology and Laboratory Medicine, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, 13-1 Takara-machi, Kanazawa, Ishikawa, 920-8640, Japan
| | - Norihiko Sakai
- Department of Nephrology and Laboratory Medicine, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, 13-1 Takara-machi, Kanazawa, Ishikawa, 920-8640, Japan
| | - Kazuyoshi Hosomichi
- Department of Bioinformatics and Genomics, Graduate School of Advanced Preventive Medical Sciences, Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Philip M Murphy
- Molecular Signaling Section, Laboratory of Molecular Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Atsushi Tajima
- Department of Bioinformatics and Genomics, Graduate School of Advanced Preventive Medical Sciences, Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Keisuke Okita
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Kenji Osafune
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Shuichi Kaneko
- Department of System Biology, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Takashi Wada
- Department of Nephrology and Laboratory Medicine, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, 13-1 Takara-machi, Kanazawa, Ishikawa, 920-8640, Japan.
| |
Collapse
|
14
|
Gene Editing Correction of a Urea Cycle Defect in Organoid Stem Cell Derived Hepatocyte-like Cells. Int J Mol Sci 2021; 22:ijms22031217. [PMID: 33530582 PMCID: PMC7865883 DOI: 10.3390/ijms22031217] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 01/18/2021] [Accepted: 01/20/2021] [Indexed: 12/17/2022] Open
Abstract
Urea cycle disorders are enzymopathies resulting from inherited deficiencies in any genes of the cycle. In severe cases, currently available therapies are marginally effective, with liver transplantation being the only definitive treatment. Donor liver availability can limit even this therapy. Identification of novel therapeutics for genetic-based liver diseases requires models that provide measurable hepatic functions and phenotypes. Advances in stem cell and genome editing technologies could provide models for the investigation of cell-based genetic diseases, as well as the platforms for drug discovery. This report demonstrates a practical, and widely applicable, approach that includes the successful reprogramming of somatic cells from a patient with a urea cycle defect, their genetic correction and differentiation into hepatic organoids, and the subsequent demonstration of genetic and phenotypic change in the edited cells consistent with the correction of the defect. While individually rare, there is a large number of other genetic-based liver diseases. The approach described here could be applied to a broad range and a large number of patients with these hepatic diseases where it could serve as an in vitro model, as well as identify successful strategies for corrective cell-based therapy.
Collapse
|
15
|
Mamani-Huanca M, Gradillas A, López-Gonzálvez Á, Barbas C. Unraveling the Cyclization of l-Argininosuccinic Acid in Biological Samples: A Study via Mass Spectrometry and NMR Spectroscopy. Anal Chem 2020; 92:12891-12899. [DOI: 10.1021/acs.analchem.0c01420] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Affiliation(s)
- Maricruz Mamani-Huanca
- Centre for Metabolomics and Bioanalysis (CEMBIO), Department of Chemistry and Biochemistry, Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Urbanización Monteprı́ncipe, 28660 Boadilla del Monte, Madrid, Spain
| | - Ana Gradillas
- Centre for Metabolomics and Bioanalysis (CEMBIO), Department of Chemistry and Biochemistry, Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Urbanización Monteprı́ncipe, 28660 Boadilla del Monte, Madrid, Spain
| | - Ángeles López-Gonzálvez
- Centre for Metabolomics and Bioanalysis (CEMBIO), Department of Chemistry and Biochemistry, Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Urbanización Monteprı́ncipe, 28660 Boadilla del Monte, Madrid, Spain
| | - Coral Barbas
- Centre for Metabolomics and Bioanalysis (CEMBIO), Department of Chemistry and Biochemistry, Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Urbanización Monteprı́ncipe, 28660 Boadilla del Monte, Madrid, Spain
| |
Collapse
|
16
|
Yoshida T, Jonouchi T, Osafune K, Takita J, Sakurai H. A Liver Model of Infantile-Onset Pompe Disease Using Patient-Specific Induced Pluripotent Stem Cells. Front Cell Dev Biol 2019; 7:316. [PMID: 31850350 PMCID: PMC6895003 DOI: 10.3389/fcell.2019.00316] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Accepted: 11/18/2019] [Indexed: 11/13/2022] Open
Abstract
Infantile-onset Pompe disease (IOPD) is a life-threatening multi-organ disease caused by an inborn defect of lysosomal acid α-glucosidase (GAA), which can degrade glycogen into glucose. Lack of GAA causes abnormal accumulation of glycogen in the lysosomes, particularly in the skeletal muscle, liver, and heart. Enzyme replacement therapy (ERT) with recombinant human GAA (rhGAA) is the only available treatment; however, its effect varies by organ. Thus, to fully understand the pathomechanism of IOPD, organ-specific disease models are necessary. We previously generated induced pluripotent stem cells (iPSCs) from three unrelated patients with IOPD and establish a skeletal muscle model of IOPD. Here, we used the same iPSC lines as the previous study and differentiated them into hepatocytes. As a result, hepatocytes differentiated from iPSC of IOPD patients showed abnormal accumulation of lysosomal glycogen, the hallmark of Pompe disease. Using this model, we also demonstrated that glycogen accumulation was dose-dependently restored by rhGAA treatment. In conclusion, we have successfully established an in vitro liver model of IOPD using patient-specific iPSCs. This model can be a platform to elucidate the underlying disease mechanism or to be applied to drug-screening. Moreover, our study also suggest that an iPSC-based approach is suitable for modeling of diseases that affect multiple organs like Pompe disease.
Collapse
Affiliation(s)
- Takeshi Yoshida
- Department of Pediatrics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Tatsuya Jonouchi
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Kenji Osafune
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Junko Takita
- Department of Pediatrics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Hidetoshi Sakurai
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| |
Collapse
|
17
|
Corbett JL, Duncan SA. iPSC-Derived Hepatocytes as a Platform for Disease Modeling and Drug Discovery. Front Med (Lausanne) 2019; 6:265. [PMID: 31803747 PMCID: PMC6873655 DOI: 10.3389/fmed.2019.00265] [Citation(s) in RCA: 85] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Accepted: 10/30/2019] [Indexed: 12/15/2022] Open
Abstract
The liver is one of the largest organs in the body and is responsible for a diverse repertoire of metabolic processes. Such processes include the secretion of serum proteins, carbohydrate and lipid metabolism, bile acid and urea synthesis, detoxification of drugs and metabolic waste products, and vitamin and carbohydrate storage. Currently, liver disease is one of the most prevalent causes of mortality in the USA with congenital liver defects contributing to a significant proportion of these deaths. Historically the study of liver disease has been hampered by a shortage of organ donors, the subsequent scarcity of healthy tissue, and the failure of animal models to fully recapitulate human liver function. In vitro culture of hepatocytes has also proven difficult because primary hepatocytes rapidly de-differentiate in culture. Recent advances in stem cell technology have facilitated the generation of induced pluripotent stem cells (iPSCs) from various somatic cell types from patients. Such cells can be differentiated to a liver cell fate, essentially providing a limitless supply of cells with hepatocyte characteristics that can mimic the pathophysiology of liver disease. Furthermore, development of the CRISPR-Cas9 system, as well as advancement of miniaturized differentiation platforms has facilitated the development of high throughput models for the investigation of hepatocyte differentiation and drug discovery. In this review, we will explore the latest advances in iPSC-based disease modeling and drug screening platforms and examine how this technology is being used to identify new pharmacological interventions, and to advance our understanding of liver development and mechanisms of disease. We will cover how iPSC technology is being used to develop predictive models for rare diseases and how information gained from large in vitro screening experiments can be used to directly inform clinical investigation.
Collapse
Affiliation(s)
| | - Stephen A. Duncan
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, United States
| |
Collapse
|
18
|
Akbari S, Sevinç GG, Ersoy N, Basak O, Kaplan K, Sevinç K, Ozel E, Sengun B, Enustun E, Ozcimen B, Bagriyanik A, Arslan N, Önder TT, Erdal E. Robust, Long-Term Culture of Endoderm-Derived Hepatic Organoids for Disease Modeling. Stem Cell Reports 2019; 13:627-641. [PMID: 31522975 PMCID: PMC6829764 DOI: 10.1016/j.stemcr.2019.08.007] [Citation(s) in RCA: 81] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Revised: 08/10/2019] [Accepted: 08/16/2019] [Indexed: 12/18/2022] Open
Abstract
Organoid technologies have become a powerful emerging tool to model liver diseases, for drug screening, and for personalized treatments. These applications are, however, limited in their capacity to generate functional hepatocytes in a reproducible and efficient manner. Here, we generated and characterized the hepatic organoid (eHEPO) culture system using human induced pluripotent stem cell (iPSC)-derived EpCAM-positive endodermal cells as an intermediate. eHEPOs can be produced within 2 weeks and expanded long term (>16 months) without any loss of differentiation capacity to mature hepatocytes. Starting from patient-specific iPSCs, we modeled citrullinemia type 1, a urea cycle disorder caused by mutations in the argininosuccinate synthetase (ASS1) enzyme. The disease-related ammonia accumulation phenotype in eHEPOs could be reversed by the overexpression of the wild-type ASS1 gene, which also indicated that this model is amenable to genetic manipulation. Thus, eHEPOs are excellent unlimited cell sources to generate functional hepatic organoids in a fast and efficient manner. iPSC-derived EpCAM+ endodermal cells generate functional hepatic organoids (eHEPOs) eHEPOs can be stably maintained in long-term culture An organoid-based model for a urea cycle disorder (citrullinemia) is generated Genetic manipulation of disease-specific eHEPOs rescues disease phenotype
Collapse
Affiliation(s)
- Soheil Akbari
- Izmir Biomedicine and Genome Center, 35340 Izmir, Turkey; Department of Medical Biology and Genetics, Health Science Institute, Dokuz Eylul University, 35340 Izmir, Turkey
| | | | - Nevin Ersoy
- Department of Histology and Embryology, Health Science Institute, Dokuz Eylul University, 35340 Izmir, Turkey
| | - Onur Basak
- Hubrecht Institute, University Medical Center Utrecht, 3584 CT Utrecht, the Netherlands; Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht, 3584 CG Utrecht, the Netherlands
| | - Kubra Kaplan
- Izmir Biomedicine and Genome Center, 35340 Izmir, Turkey; Department of Genome Sciences and Molecular Biotechnology, Izmir International Biomedicine and Genome Institute, Dokuz Eylul University, 35340 Izmir, Turkey
| | - Kenan Sevinç
- Faculty of Medicine, Koç University, 34450 Istanbul, Turkey
| | - Erkin Ozel
- Faculty of Medicine, Koç University, 34450 Istanbul, Turkey
| | - Berke Sengun
- Faculty of Medicine, Koç University, 34450 Istanbul, Turkey
| | - Eray Enustun
- Faculty of Medicine, Koç University, 34450 Istanbul, Turkey
| | - Burcu Ozcimen
- Faculty of Medicine, Koç University, 34450 Istanbul, Turkey
| | - Alper Bagriyanik
- Izmir Biomedicine and Genome Center, 35340 Izmir, Turkey; Department of Histology and Embryology, Health Science Institute, Dokuz Eylul University, 35340 Izmir, Turkey; Department of Histology and Embryology, Faculty of Medicine, Dokuz Eylul University, 35340 Izmir, Turkey
| | - Nur Arslan
- Izmir Biomedicine and Genome Center, 35340 Izmir, Turkey; Department of Pediatric Gastroenterology and Metabolism, Faculty of Medicine, Dokuz Eylul University, 35340 Izmir, Turkey
| | | | - Esra Erdal
- Izmir Biomedicine and Genome Center, 35340 Izmir, Turkey; Department of Medical Biology and Genetics, Health Science Institute, Dokuz Eylul University, 35340 Izmir, Turkey.
| |
Collapse
|
19
|
Minami T, Ishii T, Yasuchika K, Fukumitsu K, Ogiso S, Miyauchi Y, Kojima H, Kawai T, Yamaoka R, Oshima Y, Kawamoto H, Kotaka M, Yasuda K, Osafune K, Uemoto S. Novel hybrid three-dimensional artificial liver using human induced pluripotent stem cells and a rat decellularized liver scaffold. Regen Ther 2019; 10:127-133. [PMID: 31032388 PMCID: PMC6477477 DOI: 10.1016/j.reth.2019.03.002] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Revised: 03/07/2019] [Accepted: 03/14/2019] [Indexed: 12/14/2022] Open
Abstract
Introduction Liver transplantation is currently the only curative therapy for end-stage liver failure; however, establishment of alternative treatments is required owing to the serious donor organ shortage. Here, we propose a novel model of hybrid three-dimensional artificial livers using both human induced pluripotent stem cells (hiPSCs) and a rat decellularized liver serving as a scaffold. Methods Rat liver harvesting and decellularization were performed as reported in our previous studies. The decellularized liver scaffold was recellularized with hiPSC-derived hepatocyte-like cells (hiPSC-HLCs) through the biliary duct. The recellularized liver graft was continuously perfused with the culture medium using a pump at a flow rate of 0.5 mL/min in a standard CO2 (5%) cell incubator at 37 °C. Results After 48 h of continuous perfusion culture, the hiPSC-HLCs of the recellularized liver distributed into the parenchymal space. Furthermore, the recellularized liver expressed the albumin (ALB) and CYP3A4 genes, and secreted human ALB into the culture medium. Conclusion Novel hybrid artificial livers using hiPSCs and rat decellularized liver scaffolds were successfully generated, which possessed human hepatic functions. Human iPSC-derived hepatocytes were engrafted in a rat decellularized liver scaffold. The recellularized liver expressed human liver-related markers ALB and CYP3A4. The recellularized liver scaffold secreted human albumin. This novel model shows potential for artificial whole liver transplantation.
Collapse
Affiliation(s)
- Takahito Minami
- Department of Surgery, Graduate School of Medicine, Kyoto University, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan.,Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | - Takamichi Ishii
- Department of Surgery, Graduate School of Medicine, Kyoto University, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | - Kentaro Yasuchika
- Department of Surgery, Graduate School of Medicine, Kyoto University, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan.,Japanese Red Cross Wakayama Medical Center, 4-20 Komatsubara-dori, Wakayama City 640-8558, Japan
| | - Ken Fukumitsu
- Department of Surgery, Graduate School of Medicine, Kyoto University, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | - Satoshi Ogiso
- Department of Surgery, Graduate School of Medicine, Kyoto University, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | - Yuya Miyauchi
- Department of Surgery, Graduate School of Medicine, Kyoto University, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | - Hidenobu Kojima
- Department of Surgery, Graduate School of Medicine, Kyoto University, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | - Takayuki Kawai
- Department of Surgery, Graduate School of Medicine, Kyoto University, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | - Ryoya Yamaoka
- Department of Surgery, Graduate School of Medicine, Kyoto University, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | - Yu Oshima
- Department of Surgery, Graduate School of Medicine, Kyoto University, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | - Hiroshi Kawamoto
- Department of Surgery, Graduate School of Medicine, Kyoto University, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | - Maki Kotaka
- Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | - Katsutaro Yasuda
- Department of Surgery, Graduate School of Medicine, Kyoto University, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan.,Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | - Kenji Osafune
- Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | - Shinji Uemoto
- Department of Surgery, Graduate School of Medicine, Kyoto University, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| |
Collapse
|
20
|
Karagiannis P, Takahashi K, Saito M, Yoshida Y, Okita K, Watanabe A, Inoue H, Yamashita JK, Todani M, Nakagawa M, Osawa M, Yashiro Y, Yamanaka S, Osafune K. Induced Pluripotent Stem Cells and Their Use in Human Models of Disease and Development. Physiol Rev 2019; 99:79-114. [PMID: 30328784 DOI: 10.1152/physrev.00039.2017] [Citation(s) in RCA: 214] [Impact Index Per Article: 42.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
The discovery of somatic cell nuclear transfer proved that somatic cells can carry the same genetic code as the zygote, and that activating parts of this code are sufficient to reprogram the cell to an early developmental state. The discovery of induced pluripotent stem cells (iPSCs) nearly half a century later provided a molecular mechanism for the reprogramming. The initial creation of iPSCs was accomplished by the ectopic expression of four specific genes (OCT4, KLF4, SOX2, and c-Myc; OSKM). iPSCs have since been acquired from a wide range of cell types and a wide range of species, suggesting a universal molecular mechanism. Furthermore, cells have been reprogrammed to iPSCs using a myriad of methods, although OSKM remains the gold standard. The sources for iPSCs are abundant compared with those for other pluripotent stem cells; thus the use of iPSCs to model the development of tissues, organs, and other systems of the body is increasing. iPSCs also, through the reprogramming of patient samples, are being used to model diseases. Moreover, in the 10 years since the first report, human iPSCs are already the basis for new cell therapies and drug discovery that have reached clinical application. In this review, we examine the generation of iPSCs and their application to disease and development.
Collapse
Affiliation(s)
- Peter Karagiannis
- Center for iPS Cell Research and Application, Kyoto University , Kyoto , Japan
| | - Kazutoshi Takahashi
- Center for iPS Cell Research and Application, Kyoto University , Kyoto , Japan
| | - Megumu Saito
- Center for iPS Cell Research and Application, Kyoto University , Kyoto , Japan
| | - Yoshinori Yoshida
- Center for iPS Cell Research and Application, Kyoto University , Kyoto , Japan
| | - Keisuke Okita
- Center for iPS Cell Research and Application, Kyoto University , Kyoto , Japan
| | - Akira Watanabe
- Center for iPS Cell Research and Application, Kyoto University , Kyoto , Japan
| | - Haruhisa Inoue
- Center for iPS Cell Research and Application, Kyoto University , Kyoto , Japan
| | - Jun K Yamashita
- Center for iPS Cell Research and Application, Kyoto University , Kyoto , Japan
| | - Masaya Todani
- Center for iPS Cell Research and Application, Kyoto University , Kyoto , Japan
| | - Masato Nakagawa
- Center for iPS Cell Research and Application, Kyoto University , Kyoto , Japan
| | - Mitsujiro Osawa
- Center for iPS Cell Research and Application, Kyoto University , Kyoto , Japan
| | - Yoshimi Yashiro
- Center for iPS Cell Research and Application, Kyoto University , Kyoto , Japan
| | - Shinya Yamanaka
- Center for iPS Cell Research and Application, Kyoto University , Kyoto , Japan
| | - Kenji Osafune
- Center for iPS Cell Research and Application, Kyoto University , Kyoto , Japan
| |
Collapse
|
21
|
Induced Pluripotent Stem Cell-Derived Hepatocytes and Precision Medicine in Human Liver Disease. J Pediatr Gastroenterol Nutr 2018; 66:716-719. [PMID: 29509632 DOI: 10.1097/mpg.0000000000001948] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Liver-like human cells can be generated from human skin by converting fibroblasts to "induced pluripotent stem cells" (iPSCs), then differentiating the iPSCs into "induced hepatocytes". Although still primarily used as a research tool, emerging applications involving iPSC-derived induced hepatocytes have exciting and provocative clinical and translational potential. This review provides a brief summary of the current status of this field and obstacles that must be overcome before this novel tool will enable precision medicine-based approaches to human liver disease.
Collapse
|
22
|
Wasim M, Awan FR, Khan HN, Tawab A, Iqbal M, Ayesha H. Aminoacidopathies: Prevalence, Etiology, Screening, and Treatment Options. Biochem Genet 2017; 56:7-21. [PMID: 29094226 DOI: 10.1007/s10528-017-9825-6] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Accepted: 09/18/2017] [Indexed: 12/26/2022]
Abstract
Inborn errors of metabolism (IEMs) are a group of inherited metabolic disorders which are caused by mutations in the specific genes that lead to impaired proteins or enzymes production. Different metabolic pathways are perturbed due to the deficiency or lack of enzymes. To date, more than 500 IEMs have been reported with most of them being untreatable. However, fortunately 91 such disorders are potentially treatable, if diagnosed at an earlier stage of life. IEMs have been classified into different categories and one class of IEMs, characterized by the physiological disturbances of amino acids is called as aminoacidopathies. Out of 91 treatable IEM, thirteen disorders are amino acid related. Aminoacidopathies can be detected by chromatography and mass spectrometry based analytical techniques (e.g., HPLC, GC-MS, LC-MS/MS) for amino acid level changes, and through genetic assays (e.g., PCR, TaqMan Genotyping, DNA sequencing) at the mutation level in the corresponding genes. Hence, this review is focused to describe thirteen common aminoacidopathies namely: Phenylketonuria (PKU), Maple Syrup Urine Disease (MSUD), Homocystinuria/Methylene Tetrahydrofolate Reductase (MTHFR) deficiency, Tyrosinemia type II, Citrullinemia type I and type II, Argininosuccinic aciduria, Carbamoyl Phosphate Synthetase I (CPS) deficiency, Argininemia (arginase deficiency), Hyperornithinemia-Hyperammonemia-Homocitrullinuria (HHH) syndrome, N-Acetylglutamate Synthase (NAGS) deficiency, Ornithine Transcarbamylase (OTC) deficiency, and Pyruvate Dehydrogenase (PDH) complex deficiency. Furthermore, the etiology, prevalence and commonly used analytical techniques for screening of aminoacidopathies are briefly described. This information would be helpful to researchers and clinicians especially from developing countries to initiate newborn screening programs for aminoacidopathies.
Collapse
Affiliation(s)
- Muhammad Wasim
- Health Biotechnology Division, National Institute for Biotechnology and Genetic Engineering (NIBGE) / [Pakistan Institute of Engineering and Applied Sciences (PIEAS), Islamabad], Jhang Road, P.O. Box. 577, Faisalabad, 38000, Pakistan
| | - Fazli Rabbi Awan
- Health Biotechnology Division, National Institute for Biotechnology and Genetic Engineering (NIBGE) / [Pakistan Institute of Engineering and Applied Sciences (PIEAS), Islamabad], Jhang Road, P.O. Box. 577, Faisalabad, 38000, Pakistan.
| | - Haq Nawaz Khan
- Health Biotechnology Division, National Institute for Biotechnology and Genetic Engineering (NIBGE) / [Pakistan Institute of Engineering and Applied Sciences (PIEAS), Islamabad], Jhang Road, P.O. Box. 577, Faisalabad, 38000, Pakistan
| | - Abdul Tawab
- Health Biotechnology Division, National Institute for Biotechnology and Genetic Engineering (NIBGE) / [Pakistan Institute of Engineering and Applied Sciences (PIEAS), Islamabad], Jhang Road, P.O. Box. 577, Faisalabad, 38000, Pakistan
| | - Mazhar Iqbal
- Health Biotechnology Division, National Institute for Biotechnology and Genetic Engineering (NIBGE) / [Pakistan Institute of Engineering and Applied Sciences (PIEAS), Islamabad], Jhang Road, P.O. Box. 577, Faisalabad, 38000, Pakistan
| | - Hina Ayesha
- DHQ Hospital, Faisalabad Medical University, Faisalabad, Pakistan
| |
Collapse
|