1
|
Borisch C, Thum T, Bär C, Hoepfner J. Human in vitro models for Fabry disease: new paths for unravelling disease mechanisms and therapies. J Transl Med 2024; 22:965. [PMID: 39449071 PMCID: PMC11515389 DOI: 10.1186/s12967-024-05756-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Accepted: 10/08/2024] [Indexed: 10/26/2024] Open
Abstract
Fabry disease is a multi-organ disease, caused by mutations in the GLA gene and leading to a progressive accumulation of glycosphingolipids due to enzymatic absence or malfunction of the encoded alpha-galactosidase A. Since pathomechanisms are not yet fully understood and available treatments are not efficient for all mutation types and tissues, further research is highly needed. This research involves many different model types, with significant effort towards the establishment of an in vivo model. However, these models did not replicate the variety of symptoms observed in patients. As an alternative strategy, patient-derived somatic cells as well as patient-independent cell lines were used to model specific aspects of the disease in vitro. Fabry disease patients present different phenotypes according to the mutation and the level of residual enzyme activity, pointing to the necessity of personalized disease modeling. With the advent of induced pluripotent stem cells, the derivation of a multitude of disease-affected cell types became possible, even in a patient-specific and mutation-specific manner. Only recently, three-dimensional Fabry disease models were established that even more closely resemble the native tissue of investigated organs and will bring research closer to the in vivo situation. This review provides an overview of human in vitro models and their achievements in unravelling the Fabry disease pathomechanism as well as in elucidating current and future treatment strategies.
Collapse
Affiliation(s)
- Carla Borisch
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Hannover, Germany
| | - Thomas Thum
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Hannover, Germany
- Center for Translational Regenerative Medicine, Hannover Medical School, Hannover, Germany
| | - Christian Bär
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Hannover, Germany
- Center for Translational Regenerative Medicine, Hannover Medical School, Hannover, Germany
- Fraunhofer Institute for Toxicology and Experimental Medicine ITEM, Hannover, Germany
| | - Jeannine Hoepfner
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Hannover, Germany.
| |
Collapse
|
2
|
Juchem M, Lehmann N, Behrens YL, Bär C, Thum T, Hoepfner J. CRISPR/Cas9-based GLA knockout to generate the female Fabry disease human induced pluripotent stem cell line MHHi001-A-15. Stem Cell Res 2024; 79:103478. [PMID: 38905814 DOI: 10.1016/j.scr.2024.103478] [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: 05/27/2024] [Accepted: 06/18/2024] [Indexed: 06/23/2024] Open
Abstract
The X-linked lysosomal storage disorder Fabry disease originates from GLA gene mutations causing α-galactosidase A enzyme deficiency. Here we generated the GLA knockout hiPSC line MHHi001-A-15 (GLA-KOhiPSC) as an in vitro Fabry disease model by targeting exon 2 of the GLA gene by CRISPR/Cas9 in the established control hiPSC line MHHi001-A. GLA-KOhiPSCs retained the expression of pluripotency markers, trilineage differentiation potential, as well as normal karyotype and stem cell morphology but lacked α-galactosidase A enzyme activity. The GLA-KOhiPSCs represent a potent resource to not only study the Fabry disease manifestation but also screen for novel treatment options.
Collapse
Affiliation(s)
- Malte Juchem
- Institute of Molecular and Translational Therapeutic Strategies, Hannover Medical School, Hannover, Germany; Fraunhofer Institute for Toxicology and Experimental Medicine (ITEM), Hannover, Germany
| | - Nele Lehmann
- Institute of Molecular and Translational Therapeutic Strategies, Hannover Medical School, Hannover, Germany
| | | | - Christian Bär
- Institute of Molecular and Translational Therapeutic Strategies, Hannover Medical School, Hannover, Germany; Fraunhofer Institute for Toxicology and Experimental Medicine (ITEM), Hannover, Germany; Center of Translational Regenerative Medicine, Hannover Medical School, Hannover, Germany
| | - Thomas Thum
- Institute of Molecular and Translational Therapeutic Strategies, Hannover Medical School, Hannover, Germany; Center of Translational Regenerative Medicine, Hannover Medical School, Hannover, Germany
| | - Jeannine Hoepfner
- Institute of Molecular and Translational Therapeutic Strategies, Hannover Medical School, Hannover, Germany.
| |
Collapse
|
3
|
Han JW, Chang HS, Park SC, Yang JY, Kim YJ, Kim JH, Park HS, Jeong H, Lee J, Yoon CK, Yu HG, Woo SJ, Lyu J, Park TK. Early Developmental Characteristics and Features of a Three-Dimensional Retinal Organoid Model of X-Linked Juvenile Retinoschisis. Int J Mol Sci 2024; 25:8203. [PMID: 39125773 PMCID: PMC11311801 DOI: 10.3390/ijms25158203] [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: 06/28/2024] [Revised: 07/24/2024] [Accepted: 07/25/2024] [Indexed: 08/12/2024] Open
Abstract
X-linked juvenile retinoschisis (XLRS) is a hereditary retinal degeneration affecting young males caused by mutations in the retinoschisin (RS1) gene. We generated human induced pluripotent stem cells (hiPSCs) from XLRS patients and established three-dimensional retinal organoids (ROs) for disease investigation. This disease model recapitulates the characteristics of XLRS, exhibiting defects in RS1 protein production and photoreceptor cell development. XLRS ROs also revealed dysregulation of Na/K-ATPase due to RS1 deficiency and increased ERK signaling pathway activity. Transcriptomic analyses of XLRS ROs showed decreased expression of retinal cells, particularly photoreceptor cells. Furthermore, relevant recovery of the XLRS phenotype was observed when co-cultured with control ROs derived from healthy subject during the early stages of differentiation. In conclusion, our in vitro XLRS RO model presents a valuable tool for elucidating the pathophysiological mechanisms underlying XLRS, offering insights into disease progression. Additionally, this model serves as a robust platform for the development and optimization of targeted therapeutic strategies, potentially improving treatment outcomes for patients with XLRS.
Collapse
Affiliation(s)
- Jung Woo Han
- Department of Ophthalmology, Soonchunhyang University Bucheon Hospital, Soonchunhyang University College of Medicine, 170, Jomaru-ro, Bucheon 14584, Republic of Korea; (J.W.H.); (S.C.P.); (J.H.K.); (H.S.P.)
- Department of Ophthalmology, Soonchunhyang University College of Medicine, Cheonan 31151, Republic of Korea
| | - Hun Soo Chang
- Department of Microbiology, Soonchunhyang University College of Medicine, Cheonan 31151, Republic of Korea;
- Department of Interdisciplinary Program in Biomedical Science, Soonchunhyang Graduate School, Soonchunhyang University Bucheon Hospital, Bucheon 14584, Republic of Korea;
| | - Sung Chul Park
- Department of Ophthalmology, Soonchunhyang University Bucheon Hospital, Soonchunhyang University College of Medicine, 170, Jomaru-ro, Bucheon 14584, Republic of Korea; (J.W.H.); (S.C.P.); (J.H.K.); (H.S.P.)
| | - Jin Young Yang
- Laboratory of Molecular Therapy for Retinal Degeneration, Soonchunhyang University Bucheon Hospital, Soonchunhyang University College of Medicine, Bucheon 14584, Republic of Korea;
| | - Ye Ji Kim
- Department of Interdisciplinary Program in Biomedical Science, Soonchunhyang Graduate School, Soonchunhyang University Bucheon Hospital, Bucheon 14584, Republic of Korea;
| | - Jin Ha Kim
- Department of Ophthalmology, Soonchunhyang University Bucheon Hospital, Soonchunhyang University College of Medicine, 170, Jomaru-ro, Bucheon 14584, Republic of Korea; (J.W.H.); (S.C.P.); (J.H.K.); (H.S.P.)
- Department of Ophthalmology, Soonchunhyang University College of Medicine, Cheonan 31151, Republic of Korea
| | - Hyo Song Park
- Department of Ophthalmology, Soonchunhyang University Bucheon Hospital, Soonchunhyang University College of Medicine, 170, Jomaru-ro, Bucheon 14584, Republic of Korea; (J.W.H.); (S.C.P.); (J.H.K.); (H.S.P.)
- Department of Ophthalmology, Soonchunhyang University College of Medicine, Cheonan 31151, Republic of Korea
| | - Han Jeong
- Institute of Vision Research, Department of Ophthalmology, Severance Eye Hospital, Yonsei University College of Medicine, Seoul 03722, Republic of Korea;
- Brain Korea 21 Project for Medical Science, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Junwon Lee
- Institute of Vision Research, Department of Ophthalmology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul 06273, Republic of Korea;
| | - Chang Ki Yoon
- Department of Ophthalmology, Seoul National University Hospital, Seoul National University College of Medicine, Seoul 03080, Republic of Korea;
| | - Hyung Gon Yu
- Retina Center, The Sky Eye Institute, Seoul 06536, Republic of Korea;
| | - Se Joon Woo
- Department of Ophthalmology, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam 13620, Republic of Korea;
| | - Jungmook Lyu
- Department of Medical Science, Konyang University, Daejun 32992, Republic of Korea;
| | - Tae Kwann Park
- Department of Ophthalmology, Soonchunhyang University Bucheon Hospital, Soonchunhyang University College of Medicine, 170, Jomaru-ro, Bucheon 14584, Republic of Korea; (J.W.H.); (S.C.P.); (J.H.K.); (H.S.P.)
- Department of Ophthalmology, Soonchunhyang University College of Medicine, Cheonan 31151, Republic of Korea
- Department of Interdisciplinary Program in Biomedical Science, Soonchunhyang Graduate School, Soonchunhyang University Bucheon Hospital, Bucheon 14584, Republic of Korea;
- Laboratory of Molecular Therapy for Retinal Degeneration, Soonchunhyang University Bucheon Hospital, Soonchunhyang University College of Medicine, Bucheon 14584, Republic of Korea;
| |
Collapse
|
4
|
Kurdi H, Lavalle L, Moon JCC, Hughes D. Inflammation in Fabry disease: stages, molecular pathways, and therapeutic implications. Front Cardiovasc Med 2024; 11:1420067. [PMID: 38932991 PMCID: PMC11199868 DOI: 10.3389/fcvm.2024.1420067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 05/28/2024] [Indexed: 06/28/2024] Open
Abstract
Fabry disease, a multisystem X-linked disorder caused by mutations in the alpha-galactosidase gene. This leads to the accumulation of globotriaosylceramide (Gb3) and globotriaosylsphingosine (Lyso-Gb3), culminating in various clinical signs and symptoms that significantly impact quality of life. Although treatments such as enzyme replacement, oral chaperone, and emerging therapies like gene therapy exist; delayed diagnosis often curtails their effectiveness. Our review highlights the importance of delineating the stages of inflammation in Fabry disease to enhance the timing and efficacy of diagnosis and interventions, particularly before the progression to fibrosis, where treatment options are less effective. Inflammation is emerging as an important aspect of the pathogenesis of Fabry disease. This is thought to be predominantly mediated by the innate immune response, with growing evidence pointing towards the potential involvement of adaptive immune mechanisms that remain poorly understood. Highlighted by the fact that Fabry disease shares immune profiles with systemic autoinflammatory diseases, blurring the distinctions between these disorders and highlighting the need for a nuanced understanding of immune dynamics. This insight is crucial for developing targeted therapies and improving the administration of current treatments like enzyme replacement. Moreover, our review discusses the complex interplay between these inflammatory processes and current treatments, such as the challenges posed by anti-drug antibodies. These antibodies can attenuate the effectiveness of therapies, necessitating more refined approaches to mitigate their impact. By advancing our understanding of the molecular changes, inflammatory mediators and causative factors that drive inflammation in Fabry disease, we aim to clarify their role in the disease's progression. This improved understanding will help us see how these processes fit into the current landscape of Fabry disease. Additionally, it will guide the development of more effective diagnostic and therapeutic approaches, ultimately improving patient care.
Collapse
Affiliation(s)
- Hibba Kurdi
- Institute of Cardiovascular Science, University College London, London, United Kingdom
- Cardiovascular Imaging Department, Barts Heart Centre, London, United Kingdom
| | - Lucia Lavalle
- Institute of Cardiovascular Science, University College London, London, United Kingdom
- Lysosomal Storage Disorders Unit, The Royal Free Hospital, London, United Kingdom
| | - James C. C. Moon
- Institute of Cardiovascular Science, University College London, London, United Kingdom
- Cardiovascular Imaging Department, Barts Heart Centre, London, United Kingdom
| | - Derralynn Hughes
- Institute of Cardiovascular Science, University College London, London, United Kingdom
- Lysosomal Storage Disorders Unit, The Royal Free Hospital, London, United Kingdom
| |
Collapse
|
5
|
Dababneh S, Hamledari H, Maaref Y, Jayousi F, Hosseini DB, Khan A, Jannati S, Jabbari K, Arslanova A, Butt M, Roston TM, Sanatani S, Tibbits GF. Advances in Hypertrophic Cardiomyopathy Disease Modelling Using hiPSC-Derived Cardiomyocytes. Can J Cardiol 2024; 40:766-776. [PMID: 37952715 DOI: 10.1016/j.cjca.2023.11.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 10/21/2023] [Accepted: 11/07/2023] [Indexed: 11/14/2023] Open
Abstract
The advent of human induced pluripotent stem cells (hiPSCs) and their capacity to be differentiated into beating human cardiomyocytes (CMs) in vitro has revolutionized human disease modelling, genotype-phenotype predictions, and therapeutic testing. Hypertrophic cardiomyopathy (HCM) is a common inherited cardiomyopathy and the leading known cause of sudden cardiac arrest in young adults and athletes. On a molecular level, HCM is often driven by single pathogenic genetic variants, usually in sarcomeric proteins, that can alter the mechanical, electrical, signalling, and transcriptional properties of the cell. A deeper knowledge of these alterations is critical to better understanding HCM manifestation, progression, and treatment. Leveraging hiPSC-CMs to investigate the molecular mechanisms driving HCM presents a unique opportunity to dissect the consequences of genetic variants in a sophisticated and controlled manner. In this review, we summarize the molecular underpinnings of HCM and the role of hiPSC-CM studies in advancing our understanding, and we highlight the advances in hiPSC-CM-based modelling of HCM, including maturation, contractility, multiomics, and genome editing, with the notable exception of electrophysiology, which has been previously covered.
Collapse
Affiliation(s)
- Saif Dababneh
- Cellular and Regenerative Medicine Centre, BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada; Department of Cellular and Physiological Sciences, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Homa Hamledari
- Cellular and Regenerative Medicine Centre, BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada; Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Yasaman Maaref
- Cellular and Regenerative Medicine Centre, BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada; Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Farah Jayousi
- Cellular and Regenerative Medicine Centre, BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada; Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Dina B Hosseini
- Cellular and Regenerative Medicine Centre, BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada; Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Aasim Khan
- Cellular and Regenerative Medicine Centre, BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada; Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Shayan Jannati
- Faculty of Engineering, University of British Columbia, Vancouver, British Columbia, Canada
| | - Kosar Jabbari
- Cellular and Regenerative Medicine Centre, BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada; Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Alia Arslanova
- Cellular and Regenerative Medicine Centre, BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada; Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Mariam Butt
- Cellular and Regenerative Medicine Centre, BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada; Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Thomas M Roston
- Division of Cardiology and Centre for Cardiovascular Innovation, University of British Columbia, Vancouver, British Columbia, Canada
| | - Shubhayan Sanatani
- Cellular and Regenerative Medicine Centre, BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada
| | - Glen F Tibbits
- Cellular and Regenerative Medicine Centre, BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada; Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, British Columbia, Canada; School of Biomedical Engineering, University of British Columbia, Vancouver, British Columbia, Canada; Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, British Columbia, Canada.
| |
Collapse
|
6
|
Weissman D, Dudek J, Sequeira V, Maack C. Fabry Disease: Cardiac Implications and Molecular Mechanisms. Curr Heart Fail Rep 2024; 21:81-100. [PMID: 38289538 PMCID: PMC10923975 DOI: 10.1007/s11897-024-00645-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/05/2024] [Indexed: 03/09/2024]
Abstract
PURPOSE OF REVIEW This review explores the interplay among metabolic dysfunction, oxidative stress, inflammation, and fibrosis in Fabry disease, focusing on their potential implications for cardiac involvement. We aim to discuss the biochemical processes that operate in parallel to sphingolipid accumulation and contribute to disease pathogenesis, emphasizing the importance of a comprehensive understanding of these processes. RECENT FINDINGS Beyond sphingolipid accumulation, emerging studies have revealed that mitochondrial dysfunction, oxidative stress, and chronic inflammation could be significant contributors to Fabry disease and cardiac involvement. These factors promote cardiac remodeling and fibrosis and may predispose Fabry patients to conduction disturbances, ventricular arrhythmias, and heart failure. While current treatments, such as enzyme replacement therapy and pharmacological chaperones, address disease progression and symptoms, their effectiveness is limited. Our review uncovers the potential relationships among metabolic disturbances, oxidative stress, inflammation, and fibrosis in Fabry disease-related cardiac complications. Current findings suggest that beyond sphingolipid accumulation, other mechanisms may significantly contribute to disease pathogenesis. This prompts the exploration of innovative therapeutic strategies and underscores the importance of a holistic approach to understanding and managing Fabry disease.
Collapse
Affiliation(s)
- David Weissman
- Department of Translational Research, Comprehensive Heart Failure Center, University Hospital Würzburg, Am Schwarzenberg 15, Haus A15, 97078, Würzburg, Germany
| | - Jan Dudek
- Department of Translational Research, Comprehensive Heart Failure Center, University Hospital Würzburg, Am Schwarzenberg 15, Haus A15, 97078, Würzburg, Germany
| | - Vasco Sequeira
- Department of Translational Research, Comprehensive Heart Failure Center, University Hospital Würzburg, Am Schwarzenberg 15, Haus A15, 97078, Würzburg, Germany
| | - Christoph Maack
- Department of Translational Research, Comprehensive Heart Failure Center, University Hospital Würzburg, Am Schwarzenberg 15, Haus A15, 97078, Würzburg, Germany.
| |
Collapse
|
7
|
Zhang Z, Liu Q, Deng Z, Liu J, Li S, Hong M, Peng Y. Evaluating the Metabolic Basis of α-Gal A mRNA Therapy for Fabry Disease. BIOLOGY 2024; 13:106. [PMID: 38392324 PMCID: PMC10886685 DOI: 10.3390/biology13020106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 02/02/2024] [Accepted: 02/06/2024] [Indexed: 02/24/2024]
Abstract
mRNA injection-based protein supplementation has emerged as a feasible treatment for Fabry disease. However, whether the introduction of LNP-encapsulated mRNA results in the alteration of metabolomics in an in vivo system remains largely unknown. In the present study, α-galactosidase A (α-Gal A) mRNA was generated and injected into the Fabry disease mouse model. The α-Gal A protein was successfully expressed. The level of globotriaosylsphingosine (Lyso-Gb3), a biomarker for Fabry disease, as well as pro-inflammatory cytokines such as nuclear factor kappa-B (NF-κB), interleukin 6 (IL-6), and tumor necrosis factor-α (TNF-α), were greatly decreased compared to the untreated control, indicating the therapeutic outcome of the mRNA drug. Metabolomics analysis found that the level of 20 metabolites was significantly altered in the plasma of mRNA-injected mice. These compounds are primarily enriched in the arachidonic acid metabolism, alanine, aspartate and glutamate metabolism, and glycolysis/gluconeogenesis pathways. Arachidonic acid and 5-hydroxyeicosatetraenoic acid (5-HETE), both of which are important components in the eicosanoid pathway and related to inflammation response, were significantly increased in the injected mice, possibly due to the presence of lipid nanoparticles. Moreover, mRNA can effectively alter the level of metabolites in the amino acid and energy metabolic pathways that are commonly found to be suppressed in Fabry disease. Taken together, the present study demonstrated that in addition to supplementing the deficient α-Gal A protein, the mRNA-based therapeutic agent can also affect levels of metabolites that may help in the recovery of metabolic homeostasis in the full body system.
Collapse
Affiliation(s)
- Zhendong Zhang
- Liverna Therapeutics Inc., Zhuhai 519000, China
- College of Life Sciences, South China Agricultural University, Guangzhou 510642, China
| | - Qi Liu
- Liverna Therapeutics Inc., Zhuhai 519000, China
| | - Zhiwen Deng
- Liverna Therapeutics Inc., Zhuhai 519000, China
| | - Jun Liu
- Liverna Therapeutics Inc., Zhuhai 519000, China
| | - Shuang Li
- Liverna Therapeutics Inc., Zhuhai 519000, China
| | - Mei Hong
- College of Life Sciences, South China Agricultural University, Guangzhou 510642, China
| | - Yucai Peng
- Liverna Therapeutics Inc., Zhuhai 519000, China
| |
Collapse
|
8
|
Chirico N, Kessler EL, Maas RGC, Fang J, Qin J, Dokter I, Daniels M, Šarić T, Neef K, Buikema JW, Lei Z, Doevendans PA, Sluijter JPG, van Mil A. Small molecule-mediated rapid maturation of human induced pluripotent stem cell-derived cardiomyocytes. Stem Cell Res Ther 2022; 13:531. [PMID: 36575473 PMCID: PMC9795728 DOI: 10.1186/s13287-022-03209-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Accepted: 12/01/2022] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Human induced pluripotent stem cell (iPSC)-derived cardiomyocytes (iPSC-CMs) do not display all hallmarks of mature primary cardiomyocytes, especially the ability to use fatty acids (FA) as an energy source, containing high mitochondrial mass, presenting binucleation and increased DNA content per nuclei (polyploidism), and synchronized electrical conduction. This immaturity represents a bottleneck to their application in (1) disease modelling-as most cardiac (genetic) diseases have a middle-age onset-and (2) clinically relevant models, where integration and functional coupling are key. So far, several methods have been reported to enhance iPSC-CM maturation; however, these protocols are laborious, costly, and not easily scalable. Therefore, we developed a simple, low-cost, and rapid protocol to promote cardiomyocyte maturation using two small molecule activators of the peroxisome proliferator-activated receptor β/δ and gamma coactivator 1-alpha (PPAR/PGC-1α) pathway: asiatic acid (AA) and GW501516 (GW). METHODS AND RESULTS: Monolayers of iPSC-CMs were incubated with AA or GW every other day for ten days resulting in increased expression of FA metabolism-related genes and markers for mitochondrial activity. AA-treated iPSC-CMs responsiveness to the mitochondrial respiratory chain inhibitors increased and exhibited higher flexibility in substrate utilization. Additionally, structural maturity improved after treatment as demonstrated by an increase in mRNA expression of sarcomeric-related genes and higher nuclear polyploidy in AA-treated samples. Furthermore, treatment led to increased ion channel gene expression and protein levels. CONCLUSIONS Collectively, we developed a fast, easy, and economical method to induce iPSC-CMs maturation via PPAR/PGC-1α activation. Treatment with AA or GW led to increased metabolic, structural, functional, and electrophysiological maturation, evaluated using a multiparametric quality assessment.
Collapse
Affiliation(s)
- Nino Chirico
- grid.5477.10000000120346234Circulatory Health Laboratory, Regenerative Medicine Center Utrecht, University Utrecht, University Medical Center Utrecht, Utrecht, The Netherlands ,grid.7692.a0000000090126352Laboratory of Experimental Cardiology, Department of Cardiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Elise L. Kessler
- grid.5477.10000000120346234Circulatory Health Laboratory, Regenerative Medicine Center Utrecht, University Utrecht, University Medical Center Utrecht, Utrecht, The Netherlands ,grid.7692.a0000000090126352Laboratory of Experimental Cardiology, Department of Cardiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Renée G. C. Maas
- grid.5477.10000000120346234Circulatory Health Laboratory, Regenerative Medicine Center Utrecht, University Utrecht, University Medical Center Utrecht, Utrecht, The Netherlands ,grid.7692.a0000000090126352Laboratory of Experimental Cardiology, Department of Cardiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Juntao Fang
- grid.5477.10000000120346234Circulatory Health Laboratory, Regenerative Medicine Center Utrecht, University Utrecht, University Medical Center Utrecht, Utrecht, The Netherlands ,grid.7692.a0000000090126352Laboratory of Experimental Cardiology, Department of Cardiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Jiabin Qin
- grid.5477.10000000120346234Circulatory Health Laboratory, Regenerative Medicine Center Utrecht, University Utrecht, University Medical Center Utrecht, Utrecht, The Netherlands ,grid.7692.a0000000090126352Laboratory of Experimental Cardiology, Department of Cardiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Inge Dokter
- grid.5477.10000000120346234Circulatory Health Laboratory, Regenerative Medicine Center Utrecht, University Utrecht, University Medical Center Utrecht, Utrecht, The Netherlands ,grid.7692.a0000000090126352Laboratory of Experimental Cardiology, Department of Cardiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Mark Daniels
- grid.5477.10000000120346234Circulatory Health Laboratory, Regenerative Medicine Center Utrecht, University Utrecht, University Medical Center Utrecht, Utrecht, The Netherlands ,grid.7692.a0000000090126352Laboratory of Experimental Cardiology, Department of Cardiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Tomo Šarić
- grid.6190.e0000 0000 8580 3777Center for Physiology and Pathophysiology, Institute for Neurophysiology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Klaus Neef
- grid.7692.a0000000090126352Laboratory of Experimental Cardiology, Department of Cardiology, University Medical Center Utrecht, Utrecht, The Netherlands ,grid.491096.3Department of Cardiology, Amsterdam Medical Centre, 1105 AZ Amsterdam, The Netherlands
| | - Jan-Willem Buikema
- grid.5477.10000000120346234Circulatory Health Laboratory, Regenerative Medicine Center Utrecht, University Utrecht, University Medical Center Utrecht, Utrecht, The Netherlands ,grid.7692.a0000000090126352Laboratory of Experimental Cardiology, Department of Cardiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Zhiyong Lei
- grid.5477.10000000120346234Circulatory Health Laboratory, Regenerative Medicine Center Utrecht, University Utrecht, University Medical Center Utrecht, Utrecht, The Netherlands ,grid.7692.a0000000090126352Laboratory of Experimental Cardiology, Department of Cardiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Pieter A. Doevendans
- grid.5477.10000000120346234Circulatory Health Laboratory, Regenerative Medicine Center Utrecht, University Utrecht, University Medical Center Utrecht, Utrecht, The Netherlands ,grid.7692.a0000000090126352Laboratory of Experimental Cardiology, Department of Cardiology, University Medical Center Utrecht, Utrecht, The Netherlands ,grid.411737.7Netherlands Heart Institute, Utrecht, The Netherlands
| | - Joost P. G. Sluijter
- grid.5477.10000000120346234Circulatory Health Laboratory, Regenerative Medicine Center Utrecht, University Utrecht, University Medical Center Utrecht, Utrecht, The Netherlands ,grid.7692.a0000000090126352Laboratory of Experimental Cardiology, Department of Cardiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Alain van Mil
- grid.5477.10000000120346234Circulatory Health Laboratory, Regenerative Medicine Center Utrecht, University Utrecht, University Medical Center Utrecht, Utrecht, The Netherlands ,grid.7692.a0000000090126352Laboratory of Experimental Cardiology, Department of Cardiology, University Medical Center Utrecht, Utrecht, The Netherlands
| |
Collapse
|
9
|
Kaneski CR, Hanover JA, Schueler Hoffman UH. Generation of GLA-knockout human embryonic stem cell lines to model peripheral neuropathy in Fabry disease. Mol Genet Metab Rep 2022; 33:100914. [PMID: 36092250 PMCID: PMC9449667 DOI: 10.1016/j.ymgmr.2022.100914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 08/22/2022] [Accepted: 08/24/2022] [Indexed: 11/28/2022] Open
Abstract
Fabry disease is an X-linked glycolipid storage disorder caused by mutations in the GLA gene which result in a deficiency in the lysosomal enzyme alpha galactosidase A (AGA). As a result, the glycolipid substrate Gb3 accumulates in critical tissues and organs producing a progressive debilitating disease. In Fabry disease up to 80% of patients experience life-long neuropathic pain that is difficult to treat and greatly affects their quality of life. The molecular mechanisms by which deficiency of AGA leads to neuropathic pain are not well understood, due in part to a lack of in vitro models that can be used to study the underlying pathology at the cellular level. Using CRISPR-Cas9 gene editing, we generated two clones with mutations in the GLA gene from a human embryonic stem cell line. Our clonal cell lines maintained normal stem cell morphology and markers for pluripotency, and showed the phenotypic characteristics of Fabry disease including absent AGA activity and intracellular accumulation of Gb3. Mutations in the predicted locations in exon 1 of the GLA gene were confirmed. Using established techniques for dual-SMAD inhibition/WNT activation, we were able to show that our AGA-deficient clones, as well as wild-type controls, could be differentiated to peripheral-type sensory neurons that express pain receptors. This genetically and physiologically relevant human model system offers a new and promising tool for investigating the cellular mechanisms of peripheral neuropathy in Fabry disease and may assist in the development of new therapeutic strategies to help lessen the burden of this disease.
Collapse
Key Words
- 4-MU, 4-methylumbelliferone
- AGA, alpha-galactosidase A
- Alpha-galactosidase
- BDNF, brain-derived neurotrophic factor
- BRN3A, brain-specific homeobox/POU domain protein 3A
- CRISPR-Cas9
- DAPI, 4′,6-diamidino-2-phenylindole
- DRG, dorsal root ganglion
- EDTA, ethylene diamine tetracetic acid
- ERT, enzyme replacement therapy
- Fabry disease
- GAPDH, glyceraldehyde-3-phosphate dehydrogenase
- GDNF, glia-derived neurotrophic factor
- GLA, alpha-galactosidase A gene
- Gb3, globotriaosylceramide
- HEX, beta-hexosaminidase
- Human embryonic stem cells
- NGF, nerve growth factor
- Neuropathy
- PAM, protospacer adjacent motif
- PBS, phosphate buffered saline
- RNP, ribonucleoprotein
- Sensory neurons
- SgRNA, single guide RNA
- TNA-alpha, Tumor Necrosis Factor- alpha
- TRPV1, transient receptor potential vanilloid family-1
- eGFP, green fluorescent protein
- hESC, human embryonic stem cell
- iPSC, induced pluripotent stem cell
Collapse
Affiliation(s)
- Christine R. Kaneski
- National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - John A. Hanover
- National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Ulrike H. Schueler Hoffman
- National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
10
|
Vučković S, Dinani R, Nollet EE, Kuster DWD, Buikema JW, Houtkooper RH, Nabben M, van der Velden J, Goversen B. Characterization of cardiac metabolism in iPSC-derived cardiomyocytes: lessons from maturation and disease modeling. STEM CELL RESEARCH & THERAPY 2022; 13:332. [PMID: 35870954 PMCID: PMC9308297 DOI: 10.1186/s13287-022-03021-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 06/25/2022] [Indexed: 12/02/2022]
Abstract
Background Induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs) have emerged as a powerful tool for disease modeling, though their immature nature currently limits translation into clinical practice. Maturation strategies increasingly pay attention to cardiac metabolism because of its pivotal role in cardiomyocyte development and function. Moreover, aberrances in cardiac metabolism are central to the pathogenesis of cardiac disease. Thus, proper modeling of human cardiac disease warrants careful characterization of the metabolic properties of iPSC-CMs. Methods Here, we examined the effect of maturation protocols on healthy iPSC-CMs applied in 23 studies and compared fold changes in functional metabolic characteristics to assess the level of maturation. In addition, pathological metabolic remodeling was assessed in 13 iPSC-CM studies that focus on hypertrophic cardiomyopathy (HCM), which is characterized by abnormalities in metabolism. Results Matured iPSC-CMs were characterized by mitochondrial maturation, increased oxidative capacity and enhanced fatty acid use for energy production. HCM iPSC-CMs presented varying degrees of metabolic remodeling ranging from compensatory to energy depletion stages, likely due to the different types of mutations and clinical phenotypes modeled. HCM further displayed early onset hypertrophy, independent of the type of mutation or disease stage. Conclusions Maturation strategies improve the metabolic characteristics of iPSC-CMs, but not to the level of the adult heart. Therefore, a combination of maturation strategies might prove to be more effective. Due to early onset hypertrophy, HCM iPSC-CMs may be less suitable to detect early disease modifiers in HCM and might prove more useful to examine the effects of gene editing and new drugs in advanced disease stages. With this review, we provide an overview of the assays used for characterization of cardiac metabolism in iPSC-CMs and advise on which metabolic assays to include in future maturation and disease modeling studies.
Supplementary Information The online version contains supplementary material available at 10.1186/s13287-022-03021-9.
Collapse
|
11
|
Li X, Ren X, Zhang Y, Ding L, Huo M, Li Q. Fabry disease: Mechanism and therapeutics strategies. Front Pharmacol 2022; 13:1025740. [PMID: 36386210 PMCID: PMC9643830 DOI: 10.3389/fphar.2022.1025740] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 10/10/2022] [Indexed: 12/04/2022] Open
Abstract
Fabry disease is a monogenic disease characterized by a deficiency or loss of the α-galactosidase A (GLA). The resulting impairment in lysosomal GLA enzymatic activity leads to the pathogenic accumulation of enzymatic substrate and, consequently, the progressive appearance of clinical symptoms in target organs, including the heart, kidney, and brain. However, the mechanisms involved in Fabry disease-mediated organ damage are largely ambiguous and poorly understood, which hinders the development of therapeutic strategies for the treatment of this disorder. Although currently available clinical approaches have shown some efficiency in the treatment of Fabry disease, they all exhibit limitations that need to be overcome. In this review, we first introduce current mechanistic knowledge of Fabry disease and discuss potential therapeutic strategies for its treatment. We then systemically summarize and discuss advances in research on therapeutic approaches, including enzyme replacement therapy (ERT), gene therapy, and chaperone therapy, as well as strategies targeting subcellular compartments, such as lysosomes, the endoplasmic reticulum, and the nucleus. Finally, the future development of potential therapeutic strategies is discussed based on the results of mechanistic studies and the limitations associated with these therapeutic approaches.
Collapse
Affiliation(s)
- Xi Li
- Department of Anesthesiology, West China Hospital of Sichuan University, Chengdu, China
| | - Xiangyi Ren
- Core Facilities of West China Hospital, Sichuan University, Chengdu, China
| | - Yabing Zhang
- Department of Anesthesiology, West China Hospital of Sichuan University, Chengdu, China
| | - Lin Ding
- Department of Anesthesiology, West China Hospital of Sichuan University, Chengdu, China
| | - Minfeng Huo
- Shanghai Tenth People’s Hospital, Shanghai Frontiers Science Center of Nanocatalytic Medicine, School of Medicine, Tongji University, Shanghai, China
- *Correspondence: Qian Li, ; Minfeng Huo,
| | - Qian Li
- Department of Anesthesiology, West China Hospital of Sichuan University, Chengdu, China
- *Correspondence: Qian Li, ; Minfeng Huo,
| |
Collapse
|
12
|
Human Induced Pluripotent Stem Cell as a Disease Modeling and Drug Development Platform-A Cardiac Perspective. Cells 2021; 10:cells10123483. [PMID: 34943991 PMCID: PMC8699880 DOI: 10.3390/cells10123483] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 12/03/2021] [Accepted: 12/06/2021] [Indexed: 02/07/2023] Open
Abstract
A comprehensive understanding of the pathophysiology and cellular responses to drugs in human heart disease is limited by species differences between humans and experimental animals. In addition, isolation of human cardiomyocytes (CMs) is complicated because cells obtained by biopsy do not proliferate to provide sufficient numbers of cells for preclinical studies in vitro. Interestingly, the discovery of human-induced pluripotent stem cell (hiPSC) has opened up the possibility of generating and studying heart disease in a culture dish. The combination of reprogramming and genome editing technologies to generate a broad spectrum of human heart diseases in vitro offers a great opportunity to elucidate gene function and mechanisms. However, to exploit the potential applications of hiPSC-derived-CMs for drug testing and studying adult-onset cardiac disease, a full functional characterization of maturation and metabolic traits is required. In this review, we focus on methods to reprogram somatic cells into hiPSC and the solutions for overcome immaturity of the hiPSC-derived-CMs to mimic the structure and physiological properties of the adult human CMs to accurately model disease and test drug safety. Finally, we discuss how to improve the culture, differentiation, and purification of CMs to obtain sufficient numbers of desired types of hiPSC-derived-CMs for disease modeling and drug development platform.
Collapse
|
13
|
Tumor Necrosis Factor-Alpha Exacerbates Viral Entry in SARS-CoV2-Infected iPSC-Derived Cardiomyocytes. Int J Mol Sci 2021; 22:ijms22189869. [PMID: 34576032 PMCID: PMC8470197 DOI: 10.3390/ijms22189869] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 08/28/2021] [Accepted: 09/07/2021] [Indexed: 12/27/2022] Open
Abstract
The coronavirus disease 2019 (COVID-19) pandemic with high infectivity and mortality has caused severe social and economic impacts worldwide. Growing reports of COVID-19 patients with multi-organ damage indicated that severe acute respiratory syndrome coronavirus 2 (SARS-CoV2) may also disturb the cardiovascular system. Herein, we used human induced pluripotent stem cell (iPSC)-derived cardiomyocytes (iCMs) as the in vitro platform to examine the consequence of SARS-CoV2 infection on iCMs. Differentiated iCMs expressed the primary SARS-CoV2 receptor angiotensin-converting enzyme-II (ACE2) and the transmembrane protease serine type 2 (TMPRSS2) receptor suggesting the susceptibility of iCMs to SARS-CoV2. Following the infection of iCMs with SARS-CoV2, the viral nucleocapsid (N) protein was detected in the host cells, demonstrating the successful infection. Bioinformatics analysis revealed that the SARS-CoV2 infection upregulates several inflammation-related genes, including the proinflammatory cytokine tumor necrosis factor-α (TNF-α). The pretreatment of iCMs with TNF-α for 24 h, significantly increased the expression of ACE2 and TMPRSS2, SASR-CoV2 entry receptors. The TNF-α pretreatment enhanced the entry of GFP-expressing SARS-CoV2 pseudovirus into iCMs, and the neutralization of TNF-α ameliorated the TNF-α-enhanced viral entry. Collectively, SARS-CoV2 elevated TNF-α expression, which in turn enhanced the SARS-CoV2 viral entry. Our findings suggest that, TNF-α may participate in the cytokine storm and aggravate the myocardial damage in COVID-19 patients.
Collapse
|
14
|
Chien CS, Chien Y, Lin YY, Tsai PH, Chou SJ, Yarmishyn AA, Rastegari E, Wang TX, Leu HB, Yang YP, Wang ML, Jheng YC, Lai HIAM, Ching LJ, Huo TI, Cherng JY, Wang CY. Dual DNA Transfection Using 1,6-Hexanedithiol-Conjugated Maleimide-Functionalized PU-PEI 600 For Gene Correction in a Patient iPSC-Derived Fabry Cardiomyopathy Model. Front Cell Dev Biol 2021; 9:634190. [PMID: 34422789 PMCID: PMC8371449 DOI: 10.3389/fcell.2021.634190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Accepted: 07/14/2021] [Indexed: 11/25/2022] Open
Abstract
Non-viral gene delivery holds promises for treating inherited diseases. However, the limited cloning capacity of plasmids may hinder the co-delivery of distinct genes to the transfected cells. Previously, the conjugation of maleimide-functionalized polyurethane grafted with small molecular weight polyethylenimine (PU-PEI600-Mal) using 1,6-hexanedithiol (HDT) could promote the co-delivery and extensive co-expression of two different plasmids in target cells. Herein, we designed HDT-conjugated PU-PEI600-Mal for the simultaneous delivery of CRISPR/Cas9 components to achieve efficient gene correction in the induced pluripotent stem cell (iPSC)-derived model of Fabry cardiomyopathy (FC) harboring GLA IVS4 + 919 G > A mutation. This FC in vitro model recapitulated several clinical FC features, including cardiomyocyte hypertrophy and lysosomal globotriaosylceramide (Gb3) deposition. As evidenced by the expression of two reporter genes, GFP and mCherry, the addition of HDT conjugated two distinct PU-PEI600-Mal/DNA complexes and promoted the co-delivery of sgRNA/Cas9 and homology-directed repair DNA template into target cells to achieve an effective gene correction of IVS4 + 919 G > A mutation. PU-PEI600-Mal/DNA with or without HDT-mediated conjugation consistently showed neither the cytotoxicity nor an adverse effect on cardiac induction of transfected FC-iPSCs. After the gene correction and cardiac induction, disease features, including cardiomyocyte hypertrophy, the mis-regulated gene expressions, and Gb3 deposition, were remarkably rescued in the FC-iPSC-differentiated cardiomyocytes. Collectively, HDT-conjugated PU-PEI600-Mal-mediated dual DNA transfection system can be an ideal approach to improve the concurrent transfection of non-viral-based gene editing system in inherited diseases with specific mutations.
Collapse
Affiliation(s)
- Chian-Shiu Chien
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan.,School of Medicine, National Yang-Ming Medical University, Taipei, Taiwan
| | - Yueh Chien
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan.,School of Medicine, National Yang-Ming Medical University, Taipei, Taiwan
| | - Yi-Ying Lin
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan.,School of Medicine, National Yang-Ming Medical University, Taipei, Taiwan.,Institute of Pharmacology, National Yang-Ming University, Taipei, Taiwan
| | - Ping-Hsing Tsai
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan.,School of Medicine, National Yang-Ming Medical University, Taipei, Taiwan.,Institute of Pharmacology, National Yang-Ming University, Taipei, Taiwan
| | - Shih-Jie Chou
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan.,School of Medicine, National Yang-Ming Medical University, Taipei, Taiwan.,Institute of Pharmacology, National Yang-Ming University, Taipei, Taiwan
| | - Aliaksandr A Yarmishyn
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan.,School of Medicine, National Yang-Ming Medical University, Taipei, Taiwan
| | - Elham Rastegari
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Ting-Xian Wang
- Department of Chemistry and Biochemistry, National Chung-Cheng University, Chiayi County, Taiwan
| | - Hsin-Bang Leu
- School of Medicine, National Yang-Ming Medical University, Taipei, Taiwan.,Heath Care and Management Center, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Yi-Ping Yang
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan.,School of Medicine, National Yang-Ming Medical University, Taipei, Taiwan.,School of Pharmaceutical Sciences, Institute of Food Safety and Health Risk Assessment, National Yang-Ming University, Taipei, Taiwan
| | - Mong-Lien Wang
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan.,School of Medicine, National Yang-Ming Medical University, Taipei, Taiwan.,School of Pharmaceutical Sciences, Institute of Food Safety and Health Risk Assessment, National Yang-Ming University, Taipei, Taiwan
| | - Ying-Chun Jheng
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Henkie Isahwan Ahmad Mulyadi Lai
- Institute of Pharmacology, National Yang-Ming University, Taipei, Taiwan.,Department of Medical Laboratory, Faculty of Health Sciences, University of Selangor, Selangor, Malaysia
| | - Lo-Jei Ching
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Teh-Ia Huo
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan.,School of Medicine, National Yang-Ming Medical University, Taipei, Taiwan.,Institute of Pharmacology, National Yang-Ming University, Taipei, Taiwan.,Division of Gastroenterology, Department of Internal Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Jong-Yuh Cherng
- Department of Chemistry and Biochemistry, National Chung-Cheng University, Chiayi County, Taiwan
| | - Chien-Ying Wang
- School of Medicine, National Yang-Ming Medical University, Taipei, Taiwan.,Division of Trauma, Department of Emergency Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| |
Collapse
|
15
|
Castelli V, Stamerra CA, d'Angelo M, Cimini A, Ferri C. Current and experimental therapeutics for Fabry disease. Clin Genet 2021; 100:239-247. [PMID: 33997974 PMCID: PMC8453747 DOI: 10.1111/cge.13999] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 04/22/2021] [Accepted: 05/14/2021] [Indexed: 01/06/2023]
Abstract
Fabry (or Anderson‐Fabry) is a rare pan‐ethnic disease affecting males and females. Fabry is an X‐linked lysosomal storage disease, affecting glycosphingolipid metabolism, that is caused by mutations of the GLA gene that codes for α‐galactosidase A. Fabry disease (FD) can be classified into a severe, classical phenotype, most often seen in men with no residual enzyme activity, that usually appear before 18 years and a usually milder, nonclassical (later‐onset) phenotype that usually appear above 18 years. Affected patients show multifactorial complications, including renal failure, cardiovascular problems, and neuropathy. In this review, we briefly report the clinical trials so far performed with the available therapies, and then we focus on the in vitro and the in vivo experimental models of the disease, to highlight the relevance in improving the existing therapeutics and understand the mechanism of this rare disorder. Current available in vivo and in vitro models can assist in better comprehension of the pathogenesis and underlying mechanisms of FD, thus the existing therapeutic approaches can be optimized, and new options can be developed.
Collapse
Affiliation(s)
- Vanessa Castelli
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | - Cosimo Andrea Stamerra
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | - Michele d'Angelo
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | - Annamaria Cimini
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy.,Sbarro Institute for Cancer Research and Molecular Medicine, Department of Biology, Temple University, Philadelphia, Pennsylvania, USA
| | - Claudio Ferri
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| |
Collapse
|
16
|
Defining optimal enzyme and matrix combination for replating of human induced pluripotent stem cell-derived cardiomyocytes at different levels of maturity. Exp Cell Res 2021; 403:112599. [PMID: 33848551 DOI: 10.1016/j.yexcr.2021.112599] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 03/25/2021] [Accepted: 04/04/2021] [Indexed: 11/24/2022]
Abstract
Human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CM) create an unlimited cell source for basic and translational research. Depending on the maturity of cardiac cultures and the intended applications, obtaining hiPSC-CMs as a single-cell, monolayer or three-dimensional clusters can be challenging. Here, we defined strategies to replate hiPSC-CMs on early days (D15-30) or later more mature (D60-150) differentiation cultures. After generation of hiPSCs and derivation of cardiomyocytes, four dissociation reagents Collagenase A/B, Collagenase II, TrypLE, EDTA and five different extracellular matrix materials Laminin, iMatrix-511, Fibronectin, Matrigel, and Geltrex were comparatively evaluated by imaging, cell viability, and contraction analysis. For early cardiac differentiation cultures mimicking mostly the embryonic stage, the highest adhesion, cell viability, and beating frequencies were achieved by treatment with the TrypLE enzyme. Video-based contraction analysis demonstrated higher beating rates after replating compared to before treatment. For later differentiation days of more mature cardiac cultures, dissociation with EDTA and replating cells on Geltrex or Laminin-derivatives yielded better recovery. Cardiac clusters at various sizes were detected in several groups treated with collagenases. Collectively, our findings revealed the selection criteria of the dissociation approach and coating matrix for replating iPSC-CMs based on the maturity and the requirements of further downstream applications.
Collapse
|
17
|
Reversal of the Inflammatory Responses in Fabry Patient iPSC-Derived Cardiovascular Endothelial Cells by CRISPR/Cas9-Corrected Mutation. Int J Mol Sci 2021; 22:ijms22052381. [PMID: 33673551 PMCID: PMC7956852 DOI: 10.3390/ijms22052381] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 02/20/2021] [Accepted: 02/22/2021] [Indexed: 11/23/2022] Open
Abstract
The late-onset type of Fabry disease (FD) with GLA IVS4 + 919G > A mutation has been shown to lead to cardiovascular dysfunctions. In order to eliminate variations in other aspects of the genetic background, we established the isogenic control of induced pluripotent stem cells (iPSCs) for the identification of the pathogenetic factors for FD phenotypes through CRISPR/Cas9 genomic editing. We adopted droplet digital PCR (ddPCR) to efficiently capture mutational events, thus enabling isolation of the corrected FD from FD-iPSCs. Both of these exhibited the characteristics of pluripotency and phenotypic plasticity, and they can be differentiated into endothelial cells (ECs). We demonstrated the phenotypic abnormalities in FD iPSC-derived ECs (FD-ECs), including intracellular Gb3 accumulation, autophagic flux impairment, and reactive oxygen species (ROS) production, and these abnormalities were rescued in isogenic control iPSC-derived ECs (corrected FD-ECs). Microarray profiling revealed that corrected FD-derived endothelial cells reversed the enrichment of genes in the pro-inflammatory pathway and validated the downregulation of NF-κB and the MAPK signaling pathway. Our findings highlighted the critical role of ECs in FD-associated vascular dysfunctions by establishing a reliable isogenic control and providing information on potential cellular targets to reduce the morbidity and mortality of FD patients with vascular complications.
Collapse
|
18
|
Weissmann C, Albanese AA, Contreras NE, Gobetto MN, Castellanos LCS, Uchitel OD. Ion channels and pain in Fabry disease. Mol Pain 2021; 17:17448069211033172. [PMID: 34284652 PMCID: PMC8299890 DOI: 10.1177/17448069211033172] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 06/24/2021] [Accepted: 06/28/2021] [Indexed: 12/29/2022] Open
Abstract
Fabry disease (FD) is a progressive, X-linked inherited disorder of glycosphingolipid metabolism due to deficient or absent lysosomal α-galactosidase A (α-Gal A) activity which results in progressive accumulation of globotriaosylceramide (Gb3) and related metabolites. One prominent feature of Fabry disease is neuropathic pain. Accumulation of Gb3 has been documented in dorsal root ganglia (DRG) as well as other neurons, and has lately been associated with the mechanism of pain though the pathophysiology is still unclear. Small fiber (SF) neuropathy in FD differs from other entities in several aspects related to the perception of pain, alteration of fibers as well as drug therapies used in the practice with patients, with therapies far from satisfying. In order to develop better treatments, more information on the underlying mechanisms of pain is needed. Research in neuropathy has gained momentum from the development of preclinical models where different aspects of pain can be modelled and further analyzed. This review aims at describing the different in vitro and FD animal models that have been used so far, as well as some of the insights gained from their use. We focus especially in recent findings associated with ion channel alterations -that apart from the vascular alterations-, could provide targets for improved therapies in pain.
Collapse
Affiliation(s)
- Carina Weissmann
- Instituto de Fisiología, Biología Molecular y Neurociencias (IFIBYNE-UBA-CONICET) and Departamento de Fisiología, Biología Molecular y Celular, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Ciudad Universitaria, Buenos Aires C1428EHA, Argentina
| | - Adriana A Albanese
- Instituto de Fisiología, Biología Molecular y Neurociencias (IFIBYNE-UBA-CONICET) and Departamento de Fisiología, Biología Molecular y Celular, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Ciudad Universitaria, Buenos Aires C1428EHA, Argentina
| | - Natalia E Contreras
- Instituto de Fisiología, Biología Molecular y Neurociencias (IFIBYNE-UBA-CONICET) and Departamento de Fisiología, Biología Molecular y Celular, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Ciudad Universitaria, Buenos Aires C1428EHA, Argentina
| | - María N Gobetto
- Instituto de Fisiología, Biología Molecular y Neurociencias (IFIBYNE-UBA-CONICET) and Departamento de Fisiología, Biología Molecular y Celular, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Ciudad Universitaria, Buenos Aires C1428EHA, Argentina
| | - Libia C Salinas Castellanos
- Instituto de Fisiología, Biología Molecular y Neurociencias (IFIBYNE-UBA-CONICET) and Departamento de Fisiología, Biología Molecular y Celular, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Ciudad Universitaria, Buenos Aires C1428EHA, Argentina
| | - Osvaldo D Uchitel
- Instituto de Fisiología, Biología Molecular y Neurociencias (IFIBYNE-UBA-CONICET) and Departamento de Fisiología, Biología Molecular y Celular, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Ciudad Universitaria, Buenos Aires C1428EHA, Argentina
| |
Collapse
|
19
|
Mittal K, Schrenk-Siemens K. Lessons from iPSC research: Insights on peripheral nerve disease. Neurosci Lett 2020; 738:135358. [PMID: 32898616 DOI: 10.1016/j.neulet.2020.135358] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 08/24/2020] [Accepted: 09/02/2020] [Indexed: 01/13/2023]
Abstract
With the publication of their breakthrough discovery describing the induction of pluripotent stem cells (iPSCs) from mouse and human fibroblasts, Takahashi and Yamanaka have changed the scientific landscape. The possibility of deriving human pluripotent stem cells from almost any somatic cell has provided the unprecedented opportunity to study specific hereditary diseases in human cells. In the context of diseases affecting peripheral nerves, iPSC platforms are now being increasingly utilized to investigate the underlying pathology as well as regenerative strategies. Peripheral neuropathies result in peripheral nerve damage, leading to - among other things - the degeneration of affected nerve fibers accompanied by severe sensory, motor and autonomic symptoms, often including intense pain. The generation of iPSCs from hereditary forms of peripheral neuropathies and their directed differentiation into cell types most affected by the disease can be instrumental to better understanding the pathological mechanisms underlying these disorders and to investigating cell replacement strategies for repair. In this minireview, we highlight studies that have used iPSCs to investigate the therapeutic potential of iPSC-derived Schwann cell-like cells for nerve regeneration, as well as studies using patient iPSC derivatives to investigate their contribution to disease pathology.
Collapse
Affiliation(s)
- Kritika Mittal
- Department of Anatomy, University of Heidelberg, Im Neuenheimer Feld 307, 69120, Heidelberg, Germany
| | - Katrin Schrenk-Siemens
- Department of Pharmacology, University of Heidelberg, Im Neuenheimer Feld 366, 69120, Heidelberg, Germany.
| |
Collapse
|
20
|
Do HS, Park SW, Im I, Seo D, Yoo HW, Go H, Kim YH, Koh GY, Lee BH, Han YM. Enhanced thrombospondin-1 causes dysfunction of vascular endothelial cells derived from Fabry disease-induced pluripotent stem cells. EBioMedicine 2020; 52:102633. [PMID: 31981984 PMCID: PMC6992938 DOI: 10.1016/j.ebiom.2020.102633] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Revised: 01/04/2020] [Accepted: 01/07/2020] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Fabry disease (FD) is a recessive X-linked lysosomal storage disorder caused by α-galactosidase A (GLA) deficiency. Although the mechanism is unclear, GLA deficiency causes an accumulation of globotriaosylceramide (Gb3), leading to vasculopathy. METHODS To explore the relationship between the accumulation of Gb3 and vasculopathy, induced pluripotent stem cells generated from four Fabry patients (FD-iPSCs) were differentiated into vascular endothelial cells (VECs). Genome editing using CRISPR-Cas9 system was carried out to correct the GLA mutation or to delete Thrombospondin-1 (TSP-1). Global transcriptomes were compared between wild-type (WT)- and FD-VECs by RNA-sequencing analysis. FINDINGS Here, we report that overexpression of TSP-1 contributes to the dysfunction of VECs in FD. VECs originating from FD-iPSCs (FD-VECs) showed aberrant angiogenic functionality even upon treatment with recombinant α-galactosidase. Intriguingly, FD-VECs produced more p-SMAD2 and TSP-1 than WT-VECs. We also found elevated TSP-1 in the peritubular capillaries of renal tissues biopsied from FD patients. Inhibition of SMAD2 signaling or knock out of TSP-1 (TSP-1-/-) rescues normal vascular functionality in FD-VECs, like in gene-corrected FD-VECs. In addition, the enhanced oxygen consumption rate is reduced in TSP-1-/- FD-VECs. INTERPRETATION The overexpression of TSP-1 secondary to Gb3 accumulation is primarily responsible for the observed FD-VEC dysfunction. Our findings implicate dysfunctional VEC angiogenesis in the peritubular capillaries in some of the complications of Fabry disease. FUNDING This study was supported by grant 2018M3A9H1078330 from the National Research Foundation of the Republic of Korea.
Collapse
Affiliation(s)
- Hyo-Sang Do
- Department of Biological Sciences, KAIST, Daejeon 34141, Republic of Korea
| | - Sang-Wook Park
- Department of Biological Sciences, KAIST, Daejeon 34141, Republic of Korea; New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu 41061, Republic of Korea
| | - Ilkyun Im
- Department of Predictive Toxicology, Korea Institute of Toxicology, Daejeon 34141, Republic of Korea
| | - Donghyuk Seo
- Department of Biological Sciences, KAIST, Daejeon 34141, Republic of Korea
| | - Han-Wook Yoo
- Department of Pediatrics, Asan Medical Center Children's Hospital, University of Ulsan College of Medicine, Seoul 05505, Republic of Korea
| | - Heounjeong Go
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Republic of Korea
| | - Yoo Hyung Kim
- College of Natural Sciences, KAIST, Daejeon 34141, Republic of Korea; Center for Vascular Research, Institute for Basic Sciences, Daejeon 34141, Republic of Korea
| | - Gou Young Koh
- Graduate School of Medical Science and Engineering, KAIST, Daejeon 34141, Republic of Korea; Center for Vascular Research, Institute for Basic Sciences, Daejeon 34141, Republic of Korea
| | - Beom-Hee Lee
- Department of Pediatrics, Asan Medical Center Children's Hospital, University of Ulsan College of Medicine, Seoul 05505, Republic of Korea.
| | - Yong-Mahn Han
- Department of Biological Sciences, KAIST, Daejeon 34141, Republic of Korea.
| |
Collapse
|
21
|
Huang KC, Wang ML, Chen SJ, Kuo JC, Wang WJ, Nhi Nguyen PN, Wahlin KJ, Lu JF, Tran AA, Shi M, Chien Y, Yarmishyn AA, Tsai PH, Yang TC, Jane WN, Chang CC, Peng CH, Schlaeger TM, Chiou SH. Morphological and Molecular Defects in Human Three-Dimensional Retinal Organoid Model of X-Linked Juvenile Retinoschisis. Stem Cell Reports 2019; 13:906-923. [PMID: 31668851 PMCID: PMC6895767 DOI: 10.1016/j.stemcr.2019.09.010] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Revised: 09/25/2019] [Accepted: 09/26/2019] [Indexed: 12/24/2022] Open
Abstract
X-linked juvenile retinoschisis (XLRS), linked to mutations in the RS1 gene, is a degenerative retinopathy with a retinal splitting phenotype. We generated human induced pluripotent stem cells (hiPSCs) from patients to study XLRS in a 3D retinal organoid in vitro differentiation system. This model recapitulates key features of XLRS including retinal splitting, defective retinoschisin production, outer-segment defects, abnormal paxillin turnover, and impaired ER-Golgi transportation. RS1 mutation also affects the development of photoreceptor sensory cilia and results in altered expression of other retinopathy-associated genes. CRISPR/Cas9 correction of the disease-associated C625T mutation normalizes the splitting phenotype, outer-segment defects, paxillin dynamics, ciliary marker expression, and transcriptome profiles. Likewise, mutating RS1 in control hiPSCs produces the disease-associated phenotypes. Finally, we show that the C625T mutation can be repaired precisely and efficiently using a base-editing approach. Taken together, our data establish 3D organoids as a valid disease model. hiPSC-derived retinal organoid model recapitulates key features of XLRS CRISPR/Cas9 correction normalizes RS1 secretion and retinal development Transcriptome analysis links XLRS to other hereditary retinopathies
Collapse
Affiliation(s)
- Kang-Chieh Huang
- Department of Medical Research, Taipei Veterans General Hospital, Taipei 11217, Taiwan; Institute of Pharmacology, National Yang-Ming University, Taipei 11221, Taiwan; Department of Biology, Indiana University-Purdue University Indianapolis, Indianapolis, IN 46202, USA
| | - Mong-Lien Wang
- Department of Medical Research, Taipei Veterans General Hospital, Taipei 11217, Taiwan; Institute of Pharmacology, National Yang-Ming University, Taipei 11221, Taiwan; School of Medicine, National Yang-Ming University, Taipei 11221, Taiwan; Institute of Food Safety and Health Risk Assessment, National Yang-Ming University, Taipei 11221, Taiwan
| | - Shih-Jen Chen
- School of Medicine, National Yang-Ming University, Taipei 11221, Taiwan; Department of Ophthalmology, Taipei Veterans General Hospital, Taipei 11217, Taiwan
| | - Jean-Cheng Kuo
- Institute of Biochemistry and Molecular Biology, National Yang-Ming University, Taipei 11221, Taiwan; Cancer Progression Research Center, National Yang-Ming University, Taipei 11221, Taiwan
| | - Won-Jing Wang
- Institute of Biochemistry and Molecular Biology, National Yang-Ming University, Taipei 11221, Taiwan
| | - Phan Nguyen Nhi Nguyen
- Department of Medical Research, Taipei Veterans General Hospital, Taipei 11217, Taiwan; Institute of Pharmacology, National Yang-Ming University, Taipei 11221, Taiwan
| | - Karl J Wahlin
- Shiley Eye Institute, University of California San Diego, La Jolla, CA 92093, USA
| | - Jyh-Feng Lu
- School of Medicine, Fu-Jen Catholic University, New Taipei City 24205, Taiwan
| | - Audrey A Tran
- Stem Cell Program and Division of Hematology/Oncology, Boston Children's Hospital and Dana-Farber Cancer Institute, Harvard Stem Cell Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Michael Shi
- Stem Cell Program and Division of Hematology/Oncology, Boston Children's Hospital and Dana-Farber Cancer Institute, Harvard Stem Cell Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Yueh Chien
- Department of Medical Research, Taipei Veterans General Hospital, Taipei 11217, Taiwan
| | | | - Ping-Hsing Tsai
- Department of Medical Research, Taipei Veterans General Hospital, Taipei 11217, Taiwan; Institute of Pharmacology, National Yang-Ming University, Taipei 11221, Taiwan
| | - Tien-Chun Yang
- Department of Medical Research, Taipei Veterans General Hospital, Taipei 11217, Taiwan; Institute of Pharmacology, National Yang-Ming University, Taipei 11221, Taiwan
| | - Wann-Neng Jane
- Institute of Plant and Microbial Biology, Academia Sinica, Taipei 11529, Taiwan
| | - Chia-Ching Chang
- Department of Biological Science and Technology, National Chiao Tung University, Hsinchu 300, Taiwan
| | - Chi-Hsien Peng
- Department of Ophthalmology, Shin Kong Wu Ho-Su Memorial Hospital & Fu-Jen Catholic University, Taipei 11101, Taiwan
| | - Thorsten M Schlaeger
- Stem Cell Program and Division of Hematology/Oncology, Boston Children's Hospital and Dana-Farber Cancer Institute, Harvard Stem Cell Institute, Harvard Medical School, Boston, MA 02115, USA.
| | - Shih-Hwa Chiou
- Department of Medical Research, Taipei Veterans General Hospital, Taipei 11217, Taiwan; Institute of Pharmacology, National Yang-Ming University, Taipei 11221, Taiwan; School of Medicine, National Yang-Ming University, Taipei 11221, Taiwan; Department of Ophthalmology, Taipei Veterans General Hospital, Taipei 11217, Taiwan; Genomic Research Center, Academia Sinica, Taipei 11529, Taiwan.
| |
Collapse
|
22
|
Kooman JP, Stenvinkel P, Shiels PG. Fabry Disease: A New Model of Premature Ageing? Nephron Clin Pract 2019; 144:1-4. [PMID: 31563917 DOI: 10.1159/000503290] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Accepted: 09/09/2019] [Indexed: 12/13/2022] Open
Affiliation(s)
- Jeroen P Kooman
- Maastricht University Medical Center, Department of Internal Medicine, Division of Nephrology, Maastricht, The Netherlands,
| | - Peter Stenvinkel
- Division of Renal Medicine, Department of Clinical Science Technology and Intervention, Karolinska Institutet, Stockholm, Sweden
| | - Paul G Shiels
- Institute of Cancer Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| |
Collapse
|
23
|
Brodehl A, Ebbinghaus H, Deutsch MA, Gummert J, Gärtner A, Ratnavadivel S, Milting H. Human Induced Pluripotent Stem-Cell-Derived Cardiomyocytes as Models for Genetic Cardiomyopathies. Int J Mol Sci 2019; 20:ijms20184381. [PMID: 31489928 PMCID: PMC6770343 DOI: 10.3390/ijms20184381] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Revised: 08/29/2019] [Accepted: 09/03/2019] [Indexed: 12/17/2022] Open
Abstract
In the last few decades, many pathogenic or likely pathogenic genetic mutations in over hundred different genes have been described for non-ischemic, genetic cardiomyopathies. However, the functional knowledge about most of these mutations is still limited because the generation of adequate animal models is time-consuming and challenging. Therefore, human induced pluripotent stem cells (iPSCs) carrying specific cardiomyopathy-associated mutations are a promising alternative. Since the original discovery that pluripotency can be artificially induced by the expression of different transcription factors, various patient-specific-induced pluripotent stem cell lines have been generated to model non-ischemic, genetic cardiomyopathies in vitro. In this review, we describe the genetic landscape of non-ischemic, genetic cardiomyopathies and give an overview about different human iPSC lines, which have been developed for the disease modeling of inherited cardiomyopathies. We summarize different methods and protocols for the general differentiation of human iPSCs into cardiomyocytes. In addition, we describe methods and technologies to investigate functionally human iPSC-derived cardiomyocytes. Furthermore, we summarize novel genome editing approaches for the genetic manipulation of human iPSCs. This review provides an overview about the genetic landscape of inherited cardiomyopathies with a focus on iPSC technology, which might be of interest for clinicians and basic scientists interested in genetic cardiomyopathies.
Collapse
Affiliation(s)
- Andreas Brodehl
- Erich and Hanna Klessmann Institute, Heart and Diabetes Center NRW, University Hospital of the Ruhr-University Bochum, Georgstrasse 11, D-32545 Bad Oeynhausen, Germany.
| | - Hans Ebbinghaus
- Erich and Hanna Klessmann Institute, Heart and Diabetes Center NRW, University Hospital of the Ruhr-University Bochum, Georgstrasse 11, D-32545 Bad Oeynhausen, Germany.
| | - Marcus-André Deutsch
- Department of Thoracic and Cardiovascular Surgery, Heart and Diabetes Center NRW, University Hospital Ruhr-University Bochum, Georgstrasse 11, D-32545 Bad Oeynhausen, Germany.
| | - Jan Gummert
- Erich and Hanna Klessmann Institute, Heart and Diabetes Center NRW, University Hospital of the Ruhr-University Bochum, Georgstrasse 11, D-32545 Bad Oeynhausen, Germany.
- Department of Thoracic and Cardiovascular Surgery, Heart and Diabetes Center NRW, University Hospital Ruhr-University Bochum, Georgstrasse 11, D-32545 Bad Oeynhausen, Germany.
| | - Anna Gärtner
- Erich and Hanna Klessmann Institute, Heart and Diabetes Center NRW, University Hospital of the Ruhr-University Bochum, Georgstrasse 11, D-32545 Bad Oeynhausen, Germany.
| | - Sandra Ratnavadivel
- Erich and Hanna Klessmann Institute, Heart and Diabetes Center NRW, University Hospital of the Ruhr-University Bochum, Georgstrasse 11, D-32545 Bad Oeynhausen, Germany.
| | - Hendrik Milting
- Erich and Hanna Klessmann Institute, Heart and Diabetes Center NRW, University Hospital of the Ruhr-University Bochum, Georgstrasse 11, D-32545 Bad Oeynhausen, Germany.
| |
Collapse
|
24
|
Birket MJ, Raibaud S, Lettieri M, Adamson AD, Letang V, Cervello P, Redon N, Ret G, Viale S, Wang B, Biton B, Guillemot JC, Mikol V, Leonard JP, Hanley NA, Orsini C, Itier JM. A Human Stem Cell Model of Fabry Disease Implicates LIMP-2 Accumulation in Cardiomyocyte Pathology. Stem Cell Reports 2019; 13:380-393. [PMID: 31378672 PMCID: PMC6700557 DOI: 10.1016/j.stemcr.2019.07.004] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Revised: 07/04/2019] [Accepted: 07/05/2019] [Indexed: 01/19/2023] Open
Abstract
Here, we have used patient-derived induced pluripotent stem cell (iPSC) and gene-editing technology to study the cardiac-related molecular and functional consequences of mutations in GLA causing the lysosomal storage disorder Fabry disease (FD), for which heart dysfunction is a major cause of mortality. Our in vitro model recapitulated clinical data with FD cardiomyocytes accumulating GL-3 and displaying an increased excitability, with altered electrophysiology and calcium handling. Quantitative proteomics enabled the identification of >5,500 proteins in the cardiomyocyte proteome and secretome, and revealed accumulation of the lysosomal protein LIMP-2 and secretion of cathepsin F and HSPA2/HSP70-2 in FD. Genetic correction reversed these changes. Overexpression of LIMP-2 directly induced the secretion of cathepsin F and HSPA2/HSP70-2, implying causative relationship, and led to massive vacuole accumulation. In summary, our study has revealed potential new cardiac biomarkers for FD, and provides valuable mechanistic insight into the earliest pathological events in FD cardiomyocytes.
Collapse
Affiliation(s)
- Matthew J Birket
- Sanofi, Translational Sciences Unit, Sanofi, 13 quai Jules Guesdes, 94400 Vitry-sur-Seine, France; Faculty of Biology, Medicine and Health, Manchester Academic Health Sciences Centre, The University of Manchester, Oxford Road, Manchester M13 9PT, UK.
| | - Sophie Raibaud
- Sanofi, Translational Sciences Unit, Avenue Pierre Brossolette, 91380 Chilly-Mazarin, France
| | - Miriam Lettieri
- Faculty of Biology, Medicine and Health, Manchester Academic Health Sciences Centre, The University of Manchester, Oxford Road, Manchester M13 9PT, UK
| | - Antony D Adamson
- Faculty of Biology, Medicine and Health, Manchester Academic Health Sciences Centre, The University of Manchester, Oxford Road, Manchester M13 9PT, UK
| | - Valerie Letang
- Sanofi, Translational Sciences Unit, Avenue Pierre Brossolette, 91380 Chilly-Mazarin, France
| | - Pauline Cervello
- Sanofi, Translational Sciences Unit, Avenue Pierre Brossolette, 91380 Chilly-Mazarin, France
| | - Nicolas Redon
- Sanofi, Translational Sciences Unit, Avenue Pierre Brossolette, 91380 Chilly-Mazarin, France
| | - Gwenaelle Ret
- Sanofi, Translational Sciences Unit, Sanofi, 13 quai Jules Guesdes, 94400 Vitry-sur-Seine, France
| | - Sandra Viale
- Sanofi, Translational Sciences Unit, Sanofi, 13 quai Jules Guesdes, 94400 Vitry-sur-Seine, France
| | - Bing Wang
- Sanofi, GBD-Analytical R&D, 211 Second Avenue, Waltham, MA 02451, USA
| | - Bruno Biton
- Sanofi, Translational Sciences Unit, Avenue Pierre Brossolette, 91380 Chilly-Mazarin, France
| | - Jean-Claude Guillemot
- Sanofi, Translational Sciences Unit, Avenue Pierre Brossolette, 91380 Chilly-Mazarin, France
| | - Vincent Mikol
- Sanofi, Translational Sciences Unit, Avenue Pierre Brossolette, 91380 Chilly-Mazarin, France
| | - John P Leonard
- Sanofi, Rare Disease Science Unit, 153 Second Avenue, Waltham, MA 02451, USA
| | - Neil A Hanley
- Faculty of Biology, Medicine and Health, Manchester Academic Health Sciences Centre, The University of Manchester, Oxford Road, Manchester M13 9PT, UK; Endocrinology Department, Manchester University NHS Foundation Trust, Grafton Street, Manchester M13 9WU, UK
| | - Cecile Orsini
- Sanofi, Translational Sciences Unit, Sanofi, 13 quai Jules Guesdes, 94400 Vitry-sur-Seine, France
| | - Jean-Michel Itier
- Sanofi, Translational Sciences Unit, Sanofi, 13 quai Jules Guesdes, 94400 Vitry-sur-Seine, France.
| |
Collapse
|
25
|
van Mil A, Balk GM, Neef K, Buikema JW, Asselbergs FW, Wu SM, Doevendans PA, Sluijter JPG. Modelling inherited cardiac disease using human induced pluripotent stem cell-derived cardiomyocytes: progress, pitfalls, and potential. Cardiovasc Res 2018; 114:1828-1842. [PMID: 30169602 PMCID: PMC6887927 DOI: 10.1093/cvr/cvy208] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Revised: 06/06/2018] [Accepted: 08/28/2018] [Indexed: 12/17/2022] Open
Abstract
In the past few years, the use of specific cell types derived from induced pluripotent stem cells (iPSCs) has developed into a powerful approach to investigate the cellular pathophysiology of numerous diseases. Despite advances in therapy, heart disease continues to be one of the leading causes of death in the developed world. A major difficulty in unravelling the underlying cellular processes of heart disease is the extremely limited availability of viable human cardiac cells reflecting the pathological phenotype of the disease at various stages. Thus, the development of methods for directed differentiation of iPSCs to cardiomyocytes (iPSC-CMs) has provided an intriguing option for the generation of patient-specific cardiac cells. In this review, a comprehensive overview of the currently published iPSC-CM models for hereditary heart disease is compiled and analysed. Besides the major findings of individual studies, detailed methodological information on iPSC generation, iPSC-CM differentiation, characterization, and maturation is included. Both, current advances in the field and challenges yet to overcome emphasize the potential of using patient-derived cell models to mimic genetic cardiac diseases.
Collapse
Affiliation(s)
- Alain van Mil
- Division Heart and Lungs, Department of Cardiology, Experimental Cardiology Laboratory, Regenerative Medicine Center, University Medical Center Utrecht, Internal Mail No G03.550, GA Utrecht, the Netherlands
- Division Heart and Lungs, Department of Cardiology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Geerthe Margriet Balk
- Division Heart and Lungs, Department of Cardiology, Experimental Cardiology Laboratory, Regenerative Medicine Center, University Medical Center Utrecht, Internal Mail No G03.550, GA Utrecht, the Netherlands
- Division Heart and Lungs, Department of Cardiology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Klaus Neef
- Division Heart and Lungs, Department of Cardiology, Experimental Cardiology Laboratory, Regenerative Medicine Center, University Medical Center Utrecht, Internal Mail No G03.550, GA Utrecht, the Netherlands
- Division Heart and Lungs, Department of Cardiology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Jan Willem Buikema
- Division Heart and Lungs, Department of Cardiology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Folkert W Asselbergs
- Division Heart and Lungs, Department of Cardiology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
- Faculty of Population Health Sciences, Institute of Cardiovascular Science, University College London, London, UK
- Durrer Center for Cardiovascular Research, Netherlands Heart Institute, Utrecht, the Netherlands
- Farr Institute of Health Informatics Research and Institute of Health Informatics, University College London, London, UK
| | - Sean M Wu
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
- Division of Cardiovascular Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Institute of Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Pieter A Doevendans
- Division Heart and Lungs, Department of Cardiology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Joost P G Sluijter
- Division Heart and Lungs, Department of Cardiology, Experimental Cardiology Laboratory, Regenerative Medicine Center, University Medical Center Utrecht, Internal Mail No G03.550, GA Utrecht, the Netherlands
- Division Heart and Lungs, Department of Cardiology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| |
Collapse
|
26
|
Eschenhagen T, Carrier L. Cardiomyopathy phenotypes in human-induced pluripotent stem cell-derived cardiomyocytes-a systematic review. Pflugers Arch 2018; 471:755-768. [PMID: 30324321 PMCID: PMC6475632 DOI: 10.1007/s00424-018-2214-0] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Revised: 09/19/2018] [Accepted: 10/02/2018] [Indexed: 12/12/2022]
Abstract
Human-induced pluripotent stem cells (hiPSC) can be differentiated to cardiomyocytes at high efficiency and are increasingly used to study cardiac disease in a human context. This review evaluated 38 studies on hypertrophic (HCM) and dilated cardiomyopathy (DCM) of different genetic causes asking to which extent published data allow the definition of an in vitro HCM/DCM hiPSC-CM phenotype. The data are put in context with the prevailing hypotheses on HCM/DCM dysfunction and pathophysiology. Relatively consistent findings in HCM not reported in DCM were larger cell size (156 ± 85%, n = 15), more nuclear localization of nuclear factor of activated T cells (NFAT; 175 ± 65%, n = 3), and higher β-myosin heavy chain gene expression levels (500 ± 547%, n = 8) than respective controls. Conversely, DCM lines showed consistently less force development than controls (47 ± 23%, n = 9), while HCM forces scattered without clear trend. Both HCM and DCM lines often showed sarcomere disorganization, higher NPPA/NPPB expression levels, and arrhythmic beating behaviour. The data have to be taken with the caveat that reporting frequencies of the various parameters (e.g. cell size, NFAT expression) differ widely between HCM and DCM lines, in which data scatter is large and that only 9/38 studies used isogenic controls. Taken together, the current data provide interesting suggestions for disease-specific phenotypes in HCM/DCM hiPSC-CM but indicate that the field is still in its early days. Systematic, quantitative comparisons and robust, high content assays are warranted to advance the field.
Collapse
Affiliation(s)
- Thomas Eschenhagen
- Institute of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany. .,Partner Site Hamburg/Kiel/Lübeck, DZHK (German Centre for Cardiovascular Research), Hamburg, Germany.
| | - Lucie Carrier
- Institute of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany. .,Partner Site Hamburg/Kiel/Lübeck, DZHK (German Centre for Cardiovascular Research), Hamburg, Germany.
| |
Collapse
|
27
|
Durbin MD, Cadar AG, Chun YW, Hong CC. Investigating pediatric disorders with induced pluripotent stem cells. Pediatr Res 2018; 84:499-508. [PMID: 30065271 PMCID: PMC6265074 DOI: 10.1038/s41390-018-0064-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Revised: 05/02/2018] [Accepted: 05/07/2018] [Indexed: 12/14/2022]
Abstract
The study of disease pathophysiology has long relied on model systems, including animal models and cultured cells. In 2006, Shinya Yamanaka achieved a breakthrough by reprogramming somatic cells into induced pluripotent stem cells (iPSCs). This revolutionary discovery provided new opportunities for disease modeling and therapeutic intervention. With established protocols, investigators can generate iPSC lines from patient blood, urine, and tissue samples. These iPSCs retain ability to differentiate into every human cell type. Advances in differentiation and organogenesis move cellular in vitro modeling to a multicellular model capable of recapitulating physiology and disease. Here, we discuss limitations of traditional animal and tissue culture models, as well as the application of iPSC models. We highlight various techniques, including reprogramming strategies, directed differentiation, tissue engineering, organoid developments, and genome editing. We extensively summarize current established iPSC disease models that utilize these techniques. Confluence of these technologies will advance our understanding of pediatric diseases and help usher in new personalized therapies for patients.
Collapse
Affiliation(s)
- Matthew D. Durbin
- Department of Pediatrics – Division of Neonatal-Perinatal Medicine, Indiana University School of Medicine, Indianapolis, IN 46202
| | - Adrian G. Cadar
- Departments of Molecular Physiology & Biophysics, Vanderbilt University School of Medicine, Nashville, TN 37232
| | - Young W. Chun
- Department of Medicine - Cardiovascular Medicine Division University of Maryland School of Medicine, Baltimore, MD 21201
| | - Charles C. Hong
- Department of Medicine - Cardiovascular Medicine Division University of Maryland School of Medicine, Baltimore, MD 21201
| |
Collapse
|
28
|
Kuramoto Y, Naito AT, Tojo H, Sakai T, Ito M, Shibamoto M, Nakagawa A, Higo T, Okada K, Yamaguchi T, Lee JK, Miyagawa S, Sawa Y, Sakata Y, Komuro I. Generation of Fabry cardiomyopathy model for drug screening using induced pluripotent stem cell-derived cardiomyocytes from a female Fabry patient. J Mol Cell Cardiol 2018; 121:256-265. [PMID: 30048710 DOI: 10.1016/j.yjmcc.2018.07.246] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Revised: 07/04/2018] [Accepted: 07/21/2018] [Indexed: 11/30/2022]
Abstract
BACKGROUND Fabry disease is an X-linked disease caused by mutations in α-galactosidase A (GLA); these mutations result in the accumulation of its substrates, mainly globotriaosylceramide (Gb3). The accumulation of glycosphingolipids induces pathogenic changes in various organs, including the heart, and Fabry cardiomyopathy is the most frequent cause of death in patients with Fabry disease. Existing therapies to treat Fabry disease have limited efficacy, and new approaches to improve the prognosis of patients with Fabry cardiomyopathy are required. METHODS AND RESULTS We generated induced pluripotent stem cell (iPSC) lines from a female patient and her son. Each iPSC clone from the female patient showed either deficient or normal GLA activity, which could be used as a Fabry disease model or its isogenic control, respectively. Erosion of the inactivated X chromosome developed heterogeneously among clones, and mono-allelic expression of the GLA gene was maintained for a substantial period in a subset of iPSC clones. Gb3 accumulation was observed in iPSC-derived cardiomyocytes (iPS-CMs) from GLA activity-deficient iPSCs by mass-spectrometry and immunofluorescent staining. The expression of ANP was increased, but the cell surface area was decreased in iPS-CMs from the Fabry model, suggesting that cardiomyopathic change is ongoing at the molecular level in Fabry iPS-CMs. We also established an algorithm for selecting proper Gb3 staining that could be used for high-content analysis-based drug screening. CONCLUSIONS We generated a Fabry cardiomyopathy model and a drug screening system by using iPS-CMs from a female Fabry patient. Drug screening using our system may help discover new drugs that would improve the prognosis of patients with Fabry cardiomyopathy.
Collapse
Affiliation(s)
- Yuki Kuramoto
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita 565-0871, Japan
| | - Atsuhiko T Naito
- Department of Pharmacology, Faculty of Medicine, Toho University, 5-21-16 Omori-nishi, Ohta-ku, Tokyo, Japan.
| | - Hiromasa Tojo
- Department of Biophysics and Biochemistry, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita 565-0871, Japan
| | - Taku Sakai
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita 565-0871, Japan
| | - Masamichi Ito
- Department of Cardiovascular Medicine, the University of Tokyo Graduate School of Medicine, 7-3-1 Hongo, Tokyo 113-8655, Japan
| | - Masato Shibamoto
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita 565-0871, Japan
| | - Akito Nakagawa
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita 565-0871, Japan
| | - Tomoaki Higo
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita 565-0871, Japan
| | - Katsuki Okada
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita 565-0871, Japan
| | - Toshihiro Yamaguchi
- Department of Cardiovascular Medicine, the University of Tokyo Graduate School of Medicine, 7-3-1 Hongo, Tokyo 113-8655, Japan
| | - Jong-Kook Lee
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita 565-0871, Japan; Department of Advanced Cardiovascular Regenerative Medicine, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita 565-0871, Japan
| | - Shigeru Miyagawa
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita 565-0871, Japan
| | - Yoshiki Sawa
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita 565-0871, Japan
| | - Yasushi Sakata
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita 565-0871, Japan
| | - Issei Komuro
- Department of Cardiovascular Medicine, the University of Tokyo Graduate School of Medicine, 7-3-1 Hongo, Tokyo 113-8655, Japan
| |
Collapse
|
29
|
Chien Y, Chou SJ, Chang YL, Leu HB, Yang YP, Tsai PH, Lai YH, Chen KH, Chang WC, Sung SH, Yu WC. Inhibition of Arachidonate 12/15-Lipoxygenase Improves α-Galactosidase Efficacy in iPSC-Derived Cardiomyocytes from Fabry Patients. Int J Mol Sci 2018; 19:ijms19051480. [PMID: 29772700 PMCID: PMC5983630 DOI: 10.3390/ijms19051480] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Revised: 05/03/2018] [Accepted: 05/08/2018] [Indexed: 02/07/2023] Open
Abstract
(1) Background: A high incidence of intervening sequence (IVS)4+919 G>A mutation with later-onset cardiac phenotype have been reported in a majority of Taiwan Fabry cohorts. Some evidence indicated that conventional biomarkers failed to predict the long-term progression and therapeutic outcome; (2) Methods: In this study, we constructed an induced pluripotent stem cell (iPSC)-based platform from Fabry cardiomyopathy (FC) patients carrying IVS4+919 G>A mutation to screen for potential targets that may help the conventional treatment; (3) Results: The FC-patient-derived iPSC-differentiated cardiomyocytes (FC-iPSC-CMs) carried an expected IVS4+919 G>A genetic mutation and recapitulated several FC characteristics, including low α-galactosidase A enzyme activity and cellular hypertrophy. The proteomic analysis revealed that arachidonate 12/15-lipoxygenase (Alox12/15) was the most highly upregulated marker in FC-iPSC-CMs, and the metabolites of Alox12/15, 12(S)- and 15(S)-hydroxyeicosatetraenoic acid (HETE), were also elevated in the culture media. Late administration of Alox12/15 pharmacological inhibitor LOXBlock-1 combined with α-galactosidase, but not α-galactosidase alone, effectively reduced cardiomyocyte hypertrophy, the secretion of 12(S)- and 15(S)-HETE and the upregulation of fibrotic markers at the late phase of FC; (4) Conclusions: Our study demonstrates that cardiac Alox12/15 and circulating 12(S)-HETE/15(S)-HETE are involved in the pathogenesis of FC with IVS4+919 G>A mutation.
Collapse
Affiliation(s)
- Yueh Chien
- Institute of Pharmacology, School of Medicine, National Yang-Ming University, Taipei 11217, Taiwan.
- Department of Medical Research, Taipei Veterans General Hospital, Taipei 11217, Taiwan.
| | - Shih-Jie Chou
- Institute of Pharmacology, School of Medicine, National Yang-Ming University, Taipei 11217, Taiwan.
| | - Yuh-Lih Chang
- Institute of Pharmacology, School of Medicine, National Yang-Ming University, Taipei 11217, Taiwan.
- Department of Pharmacology, Taipei Veterans General Hospital, Taipei 11217, Taiwan.
| | - Hsin-Bang Leu
- Institute of Clinical Medicine, School of Medicine, National Yang-Ming University, Taipei 11217, Taiwan.
- Heath Care and Management Center, Taipei Veterans General Hospital, Taipei 11217, Taiwan.
- Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei 11217, Taiwan.
| | - Yi-Ping Yang
- Department of Medical Research, Taipei Veterans General Hospital, Taipei 11217, Taiwan.
| | - Ping-Hsing Tsai
- Department of Medical Research, Taipei Veterans General Hospital, Taipei 11217, Taiwan.
- Institute of Clinical Medicine, School of Medicine, National Yang-Ming University, Taipei 11217, Taiwan.
| | - Ying-Hsiu Lai
- Department of Medical Research, Taipei Veterans General Hospital, Taipei 11217, Taiwan.
| | - Kuan-Hsuan Chen
- Department of Pharmacology, Taipei Veterans General Hospital, Taipei 11217, Taiwan.
- Institute of Clinical Medicine, School of Medicine, National Yang-Ming University, Taipei 11217, Taiwan.
| | - Wei-Chao Chang
- Center for Molecular Medicine, China Medical University Hospital, Taichung 40447, Taiwan.
| | - Shih-Hsien Sung
- Institute of Clinical Medicine, School of Medicine, National Yang-Ming University, Taipei 11217, Taiwan.
- Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei 11217, Taiwan.
| | - Wen-Chung Yu
- Institute of Clinical Medicine, School of Medicine, National Yang-Ming University, Taipei 11217, Taiwan.
- Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei 11217, Taiwan.
| |
Collapse
|
30
|
Borger DK, McMahon B, Roshan Lal T, Serra-Vinardell J, Aflaki E, Sidransky E. Induced pluripotent stem cell models of lysosomal storage disorders. Dis Model Mech 2018; 10:691-704. [PMID: 28592657 PMCID: PMC5483008 DOI: 10.1242/dmm.029009] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2016] [Accepted: 04/28/2017] [Indexed: 01/30/2023] Open
Abstract
Induced pluripotent stem cells (iPSCs) have provided new opportunities to explore the cell biology and pathophysiology of human diseases, and the lysosomal storage disorder research community has been quick to adopt this technology. Patient-derived iPSC models have been generated for a number of lysosomal storage disorders, including Gaucher disease, Pompe disease, Fabry disease, metachromatic leukodystrophy, the neuronal ceroid lipofuscinoses, Niemann-Pick types A and C1, and several of the mucopolysaccharidoses. Here, we review the strategies employed for reprogramming and differentiation, as well as insights into disease etiology gleaned from the currently available models. Examples are provided to illustrate how iPSC-derived models can be employed to develop new therapeutic strategies for these disorders. We also discuss how models of these rare diseases could contribute to an enhanced understanding of more common neurodegenerative disorders such as Parkinson’s disease, and discuss key challenges and opportunities in this area of research. Summary: This Review discusses how induced pluripotent stem cells (iPSCs) provide new opportunities to explore the biology and pathophysiology of lysosomal storage diseases, and how iPSCs have illuminated the role of lysosomes in more common disorders.
Collapse
Affiliation(s)
- Daniel K Borger
- Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Benjamin McMahon
- Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Tamanna Roshan Lal
- Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jenny Serra-Vinardell
- Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Elma Aflaki
- Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Ellen Sidransky
- Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
31
|
Yoshida S, Nakanishi C, Okada H, Mori M, Yokawa J, Yoshimuta T, Ohta K, Konno T, Fujino N, Kawashiri MA, Yachie A, Yamagishi M, Hayashi K. Characteristics of induced pluripotent stem cells from clinically divergent female monozygotic twins with Danon disease. J Mol Cell Cardiol 2018; 114:234-242. [DOI: 10.1016/j.yjmcc.2017.11.019] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Revised: 11/04/2017] [Accepted: 11/22/2017] [Indexed: 12/20/2022]
|
32
|
Giacomelli E, Mummery CL, Bellin M. Human heart disease: lessons from human pluripotent stem cell-derived cardiomyocytes. Cell Mol Life Sci 2017; 74:3711-3739. [PMID: 28573431 PMCID: PMC5597692 DOI: 10.1007/s00018-017-2546-5] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Revised: 05/09/2017] [Accepted: 05/23/2017] [Indexed: 02/07/2023]
Abstract
Technical advances in generating and phenotyping cardiomyocytes from human pluripotent stem cells (hPSC-CMs) are now driving their wider acceptance as in vitro models to understand human heart disease and discover therapeutic targets that may lead to new compounds for clinical use. Current literature clearly shows that hPSC-CMs recapitulate many molecular, cellular, and functional aspects of human heart pathophysiology and their responses to cardioactive drugs. Here, we provide a comprehensive overview of hPSC-CMs models that have been described to date and highlight their most recent and remarkable contributions to research on cardiovascular diseases and disorders with cardiac traits. We conclude discussing immediate challenges, limitations, and emerging solutions.
Collapse
Affiliation(s)
- E Giacomelli
- Department of Anatomy and Embryology, Leiden University Medical Center, Einthovenweg 20, 2333 ZC, Leiden, The Netherlands
| | - C L Mummery
- Department of Anatomy and Embryology, Leiden University Medical Center, Einthovenweg 20, 2333 ZC, Leiden, The Netherlands
- Department of Applied Stem Cell Technologies, University of Twente, Building Zuidhorst, 7500 AE, Enschede, The Netherlands
| | - M Bellin
- Department of Anatomy and Embryology, Leiden University Medical Center, Einthovenweg 20, 2333 ZC, Leiden, The Netherlands.
| |
Collapse
|