51
|
Luttrell SM, Smith AST, Mack DL. Creating stem cell-derived neuromuscular junctions in vitro. Muscle Nerve 2021; 64:388-403. [PMID: 34328673 PMCID: PMC9292444 DOI: 10.1002/mus.27360] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Revised: 05/28/2021] [Accepted: 06/21/2021] [Indexed: 12/14/2022]
Abstract
Recent development of novel therapies has improved mobility and quality of life for people suffering from inheritable neuromuscular disorders. Despite this progress, the majority of neuromuscular disorders are still incurable, in part due to a lack of predictive models of neuromuscular junction (NMJ) breakdown. Improvement of predictive models of a human NMJ would be transformative in terms of expanding our understanding of the mechanisms that underpin development, maintenance, and disease, and as a testbed with which to evaluate novel therapeutics. Induced pluripotent stem cells (iPSCs) are emerging as a clinically relevant and non‐invasive cell source to create human NMJs to study synaptic development and maturation, as well as disease modeling and drug discovery. This review will highlight the recent advances and remaining challenges to generating an NMJ capable of eliciting contraction of stem cell‐derived skeletal muscle in vitro. We explore the advantages and shortcomings of traditional NMJ culturing platforms, as well as the pioneering technologies and novel, biomimetic culturing systems currently in use to guide development and maturation of the neuromuscular synapse and extracellular microenvironment. Then, we will explore how this NMJ‐in‐a‐dish can be used to study normal assembly and function of the efferent portion of the neuromuscular arc, and how neuromuscular disease‐causing mutations disrupt structure, signaling, and function.
Collapse
Affiliation(s)
- Shawn M Luttrell
- Department of Rehabilitation Medicine, University of Washington, Seattle, Washington, USA.,Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, Washington, USA
| | - Alec S T Smith
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, Washington, USA.,Department of Physiology and Biophysics, University of Washington, Seattle, Washington, USA
| | - David L Mack
- Department of Rehabilitation Medicine, University of Washington, Seattle, Washington, USA.,Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, Washington, USA.,Department of Physiology and Biophysics, University of Washington, Seattle, Washington, USA
| |
Collapse
|
52
|
Al Tanoury Z, Zimmerman JF, Rao J, Sieiro D, McNamara HM, Cherrier T, Rodríguez-delaRosa A, Hick-Colin A, Bousson F, Fugier-Schmucker C, Marchiano F, Habermann B, Chal J, Nesmith AP, Gapon S, Wagner E, Gupta VA, Bassel-Duby R, Olson EN, Cohen AE, Parker KK, Pourquié O. Prednisolone rescues Duchenne muscular dystrophy phenotypes in human pluripotent stem cell-derived skeletal muscle in vitro. Proc Natl Acad Sci U S A 2021; 118:e2022960118. [PMID: 34260377 PMCID: PMC8285911 DOI: 10.1073/pnas.2022960118] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is a devastating genetic disease leading to degeneration of skeletal muscles and premature death. How dystrophin absence leads to muscle wasting remains unclear. Here, we describe an optimized protocol to differentiate human induced pluripotent stem cells (iPSC) to a late myogenic stage. This allows us to recapitulate classical DMD phenotypes (mislocalization of proteins of the dystrophin-associated glycoprotein complex, increased fusion, myofiber branching, force contraction defects, and calcium hyperactivation) in isogenic DMD-mutant iPSC lines in vitro. Treatment of the myogenic cultures with prednisolone (the standard of care for DMD) can dramatically rescue force contraction, fusion, and branching defects in DMD iPSC lines. This argues that prednisolone acts directly on myofibers, challenging the largely prevalent view that its beneficial effects are caused by antiinflammatory properties. Our work introduces a human in vitro model to study the onset of DMD pathology and test novel therapeutic approaches.
Collapse
Affiliation(s)
- Ziad Al Tanoury
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, CNRS UMR 7104, INSERM U964, Université de Strasbourg, 67411 Illkirch Graffenstaden, France
- Department of Pathology, Brigham and Women's Hospital, Boston, MA 02115
- Department of Genetics, Harvard Medical School, Boston, MA 02115
- Harvard Stem Cell Institute, Harvard University, Boston, MA 02138
| | - John F Zimmerman
- Harvard Stem Cell Institute, Harvard University, Boston, MA 02138
- Disease Biophysics Group, Wyss Institute for Biologically Inspired Engineering, Harvard John A. Paulson School of Engineering and Applied Sciences, Boston, MA 02134
| | - Jyoti Rao
- Department of Pathology, Brigham and Women's Hospital, Boston, MA 02115
- Department of Genetics, Harvard Medical School, Boston, MA 02115
- Harvard Stem Cell Institute, Harvard University, Boston, MA 02138
| | - Daniel Sieiro
- Department of Pathology, Brigham and Women's Hospital, Boston, MA 02115
- Department of Genetics, Harvard Medical School, Boston, MA 02115
- Harvard Stem Cell Institute, Harvard University, Boston, MA 02138
| | - Harold M McNamara
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138
- Department of Physics, Harvard University, Cambridge, MA 02138
| | - Thomas Cherrier
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, CNRS UMR 7104, INSERM U964, Université de Strasbourg, 67411 Illkirch Graffenstaden, France
| | - Alejandra Rodríguez-delaRosa
- Department of Pathology, Brigham and Women's Hospital, Boston, MA 02115
- Department of Genetics, Harvard Medical School, Boston, MA 02115
- Harvard Stem Cell Institute, Harvard University, Boston, MA 02138
| | | | - Fanny Bousson
- Anagenesis Biotechnologies, 67400 Illkirch Graffenstaden, France
| | | | - Fabio Marchiano
- Aix-Marseille University, CNRS, Institut de Biologie du Développement de Marseille UMR 7288, The Turing Center for Living Systems, 13009 Marseille, France
| | - Bianca Habermann
- Aix-Marseille University, CNRS, Institut de Biologie du Développement de Marseille UMR 7288, The Turing Center for Living Systems, 13009 Marseille, France
| | - Jérome Chal
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, CNRS UMR 7104, INSERM U964, Université de Strasbourg, 67411 Illkirch Graffenstaden, France
- Department of Pathology, Brigham and Women's Hospital, Boston, MA 02115
- Department of Genetics, Harvard Medical School, Boston, MA 02115
- Harvard Stem Cell Institute, Harvard University, Boston, MA 02138
| | - Alexander P Nesmith
- Harvard Stem Cell Institute, Harvard University, Boston, MA 02138
- Disease Biophysics Group, Wyss Institute for Biologically Inspired Engineering, Harvard John A. Paulson School of Engineering and Applied Sciences, Boston, MA 02134
| | - Svetlana Gapon
- Department of Pathology, Brigham and Women's Hospital, Boston, MA 02115
| | - Erica Wagner
- Department of Pathology, Brigham and Women's Hospital, Boston, MA 02115
| | - Vandana A Gupta
- Division of Genetics, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115
| | - Rhonda Bassel-Duby
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390
- Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390
- Senator Paul D. Wellstone Muscular Dystrophy Cooperative Research Center, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Eric N Olson
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390
- Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390
- Senator Paul D. Wellstone Muscular Dystrophy Cooperative Research Center, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Adam E Cohen
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138
- Department of Physics, Harvard University, Cambridge, MA 02138
| | - Kevin Kit Parker
- Harvard Stem Cell Institute, Harvard University, Boston, MA 02138
- Disease Biophysics Group, Wyss Institute for Biologically Inspired Engineering, Harvard John A. Paulson School of Engineering and Applied Sciences, Boston, MA 02134
| | - Olivier Pourquié
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, CNRS UMR 7104, INSERM U964, Université de Strasbourg, 67411 Illkirch Graffenstaden, France;
- Department of Pathology, Brigham and Women's Hospital, Boston, MA 02115
- Department of Genetics, Harvard Medical School, Boston, MA 02115
- Harvard Stem Cell Institute, Harvard University, Boston, MA 02138
| |
Collapse
|
53
|
Inhibition of the Combinatorial Signaling of Transforming Growth Factor-Beta and NOTCH Promotes Myotube Formation of Human Pluripotent Stem Cell-Derived Skeletal Muscle Progenitor Cells. Cells 2021; 10:cells10071649. [PMID: 34209364 PMCID: PMC8303216 DOI: 10.3390/cells10071649] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Revised: 06/21/2021] [Accepted: 06/26/2021] [Indexed: 12/16/2022] Open
Abstract
Understanding the signaling pathways that regulate the final differentiation of human myoblasts is essential for successful cell transplantation and drug screening for the treatment of muscular dystrophy. In an effort to improve myotube formation from hiPSC-derived myoblasts, we validated a collection of 13 small molecules in a newly established in vitro screening platform for the assessment of myotube formation. The analysis of myotube formation as measured by the fusion index showed that the combinational inhibition of the TGFβ signaling with NOTCH signaling enhances the ability of multi-nucleated myotube production. Combinational treatment of inhibitors for TGFβ and NOTCH signaling pathways improved myotube formation in a dose-dependent manner. This effect was achieved by inhibiting the combinatorial mechanism of signaling. The combination treatment of small molecules effective in inducing multinucleated myotubes was validated in healthy human primary myoblasts. In addition, it was also applied to DMD patient iPSC-derived myoblasts to enhance the generation of multinucleated myotubes.
Collapse
|
54
|
Madrid M, Sumen C, Aivio S, Saklayen N. Autologous Induced Pluripotent Stem Cell-Based Cell Therapies: Promise, Progress, and Challenges. Curr Protoc 2021; 1:e88. [PMID: 33725407 DOI: 10.1002/cpz1.88] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The promise of human induced pluripotent stem cells (iPSCs) lies in their ability to serve as a starting material for autologous, or patient-specific, stem cell-based therapies. Since the first publications describing the generation of iPSCs from human tissue in 2007, a Phase I/IIa clinical trial testing an autologous iPSC-derived cell therapy has been initiated in the U.S., and several other autologous iPSC-based therapies have advanced through various stages of development. Three single-patient in-human transplants of autologous iPSC-derived cells have taken place worldwide. None of the patients suffered serious adverse events, despite not undergoing immunosuppression. These promising outcomes support the proposed advantage of an autologous approach: a cell therapy product that can engraft without the risk of immune rejection, eliminating the need for immunosuppression and the associated side effects. Despite this advantage, there are currently more allogeneic than autologous iPSC-based cell therapy products in development due to the cost and complexity of scaling out manufacturing for each patient. In this review, we highlight recent progress toward clinical translation of autologous iPSC-based cell therapies. We also highlight technological advancements that would reduce the cost and complexity of autologous iPSC-based cell therapy production, enabling autologous iPSC-based therapies to become a more commonplace treatment modality for patients. © 2021 The Authors.
Collapse
Affiliation(s)
| | - Cenk Sumen
- Stemson Therapeutics, San Diego, California
| | | | | |
Collapse
|
55
|
Uchimura T, Asano T, Nakata T, Hotta A, Sakurai H. A muscle fatigue-like contractile decline was recapitulated using skeletal myotubes from Duchenne muscular dystrophy patient-derived iPSCs. CELL REPORTS MEDICINE 2021; 2:100298. [PMID: 34195678 PMCID: PMC8233665 DOI: 10.1016/j.xcrm.2021.100298] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 01/28/2021] [Accepted: 05/10/2021] [Indexed: 02/07/2023]
Abstract
Duchenne muscular dystrophy (DMD) is a muscle degenerating disease caused by dystrophin deficiency, for which therapeutic options are limited. To facilitate drug development, it is desirable to develop in vitro disease models that enable the evaluation of DMD declines in contractile performance. Here, we show MYOD1-induced differentiation of hiPSCs into functional skeletal myotubes in vitro with collagen gel and electrical field stimulation (EFS). Long-term EFS training (0.5 Hz, 20 V, 2 ms, continuous for 2 weeks) mimicking muscle overuse recapitulates declines in contractile performance in dystrophic myotubes. A screening of clinically relevant drugs using this model detects three compounds that ameliorate this decline. Furthermore, we validate the feasibility of adapting the model to a 96-well culture system using optogenetic technology for large-scale screening. Our results support a disease model using patient-derived iPSCs that allows for the recapitulation of the contractile pathogenesis of DMD and a screening strategy for drug development.
Collapse
Affiliation(s)
- Tomoya Uchimura
- Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Shogoin Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan.,Takeda-CiRA Joint Program, Fujisawa, Kanagawa 251-8555, Japan
| | - Toshifumi Asano
- Department of Cell Biology, Graduate School of Medical and Dental Science, Tokyo Medical and Dental University, Tokyo 113-8510, Japan.,The Center for Brain Integration Research, Tokyo Medical and Dental University, Tokyo 113-8510, Japan
| | - Takao Nakata
- Department of Cell Biology, Graduate School of Medical and Dental Science, Tokyo Medical and Dental University, Tokyo 113-8510, Japan.,The Center for Brain Integration Research, Tokyo Medical and Dental University, Tokyo 113-8510, Japan
| | - Akitsu Hotta
- Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Shogoin Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan.,Takeda-CiRA Joint Program, Fujisawa, Kanagawa 251-8555, Japan
| | - Hidetoshi Sakurai
- Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Shogoin Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan.,Takeda-CiRA Joint Program, Fujisawa, Kanagawa 251-8555, Japan
| |
Collapse
|
56
|
Alarcin E, Bal-Öztürk A, Avci H, Ghorbanpoor H, Dogan Guzel F, Akpek A, Yesiltas G, Canak-Ipek T, Avci-Adali M. Current Strategies for the Regeneration of Skeletal Muscle Tissue. Int J Mol Sci 2021; 22:5929. [PMID: 34072959 PMCID: PMC8198586 DOI: 10.3390/ijms22115929] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 05/21/2021] [Accepted: 05/26/2021] [Indexed: 12/11/2022] Open
Abstract
Traumatic injuries, tumor resections, and degenerative diseases can damage skeletal muscle and lead to functional impairment and severe disability. Skeletal muscle regeneration is a complex process that depends on various cell types, signaling molecules, architectural cues, and physicochemical properties to be successful. To promote muscle repair and regeneration, various strategies for skeletal muscle tissue engineering have been developed in the last decades. However, there is still a high demand for the development of new methods and materials that promote skeletal muscle repair and functional regeneration to bring approaches closer to therapies in the clinic that structurally and functionally repair muscle. The combination of stem cells, biomaterials, and biomolecules is used to induce skeletal muscle regeneration. In this review, we provide an overview of different cell types used to treat skeletal muscle injury, highlight current strategies in biomaterial-based approaches, the importance of topography for the successful creation of functional striated muscle fibers, and discuss novel methods for muscle regeneration and challenges for their future clinical implementation.
Collapse
Affiliation(s)
- Emine Alarcin
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Marmara University, 34854 Istanbul, Turkey;
| | - Ayca Bal-Öztürk
- Department of Analytical Chemistry, Faculty of Pharmacy, Istinye University, 34010 Istanbul, Turkey;
- Department of Stem Cell and Tissue Engineering, Institute of Health Sciences, Istinye University, 34010 Istanbul, Turkey
| | - Hüseyin Avci
- Department of Metallurgical and Materials Engineering, Eskisehir Osmangazi University, 26040 Eskisehir, Turkey;
- Cellular Therapy and Stem Cell Research Center, Eskisehir Osmangazi University, 26040 Eskisehir, Turkey
- AvciBio Research Group, Eskisehir Osmangazi University, 26040 Eskisehir, Turkey;
- Translational Medicine Research and Clinical Center, Eskisehir Osmangazi University, 26040 Eskisehir, Turkey
| | - Hamed Ghorbanpoor
- AvciBio Research Group, Eskisehir Osmangazi University, 26040 Eskisehir, Turkey;
- Department of Biomedical Engineering, Ankara Yildirim Beyazit University, 06010 Ankara, Turkey;
- Department of Biomedical Engineering, Eskisehir Osmangazi University, 26040 Eskisehir, Turkey
| | - Fatma Dogan Guzel
- Department of Biomedical Engineering, Ankara Yildirim Beyazit University, 06010 Ankara, Turkey;
| | - Ali Akpek
- Department of Bioengineering, Gebze Technical University, 41400 Gebze, Turkey; (A.A.); (G.Y.)
| | - Gözde Yesiltas
- Department of Bioengineering, Gebze Technical University, 41400 Gebze, Turkey; (A.A.); (G.Y.)
| | - Tuba Canak-Ipek
- Department of Thoracic and Cardiovascular Surgery, University Hospital Tuebingen, Calwerstraße 7/1, 72076 Tuebingen, Germany;
| | - Meltem Avci-Adali
- Department of Thoracic and Cardiovascular Surgery, University Hospital Tuebingen, Calwerstraße 7/1, 72076 Tuebingen, Germany;
| |
Collapse
|
57
|
Decreased YAP activity reduces proliferative ability in human induced pluripotent stem cell of duchenne muscular dystrophy derived cardiomyocytes. Sci Rep 2021; 11:10351. [PMID: 33990626 PMCID: PMC8121946 DOI: 10.1038/s41598-021-89603-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Accepted: 04/20/2021] [Indexed: 11/29/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is characterized by progressive muscle degeneration accompanied by dilated cardiomyopathy. Recently, abnormality of yes-associated protein (YAP) has been reported as the pathogenesis of muscle degeneration of DMD; however YAP activity remains unclear in dystrophic heart of DMD. Herein, we investigated YAP activity using disease-specific induced pluripotent stem cell (iPSC) derived cardiomyocytes (CMs) in DMD. DMD-iPSCs were generated from DMD patient with exon 48–54 deletion in DMD, and genome-edited (Ed)-DMD-iPSCs with in-frame (Ed-DMD-iPSCs) were created using CRISPR/Cas9. Nuclear translocation of YAP [nuclear (N)/cytoplasmic (C) ratio] was significantly lower in DMD-iPSC-CMs than in Ed-DMD-iPSC-CMs. In addition, Ki67 expression, indicating proliferative ability, was significantly lower in DMD-iPSC-CMs than Ed-DMD-iPSC-CMs. Therefore, immunofluorescent staining showed that actin stress fibers associated with YAP activity by mechanotransduction were disorganized in DMD-iPSC-CMs. Lysophosphatidic acid (LPA), a known lipid mediator on induction of actin polymerization, significantly increased YAP activity and actin dynamics in DMD-iPSC-CMs using live cell imaging. These results suggested that altered YAP activity due to impaired actin dynamics reduced proliferative ability in DMD-iPSC-CMs. Hence, decreased YAP activity in dystrophic heart may contribute to DMD-cardiomyopathy pathogenesis.
Collapse
|
58
|
Lim H, Choi IY, Hyun SH, Kim H, Lee G. Approaches to characterize the transcriptional trajectory of human myogenesis. Cell Mol Life Sci 2021; 78:4221-4234. [PMID: 33590269 PMCID: PMC11072395 DOI: 10.1007/s00018-021-03782-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 12/31/2020] [Accepted: 01/28/2021] [Indexed: 12/16/2022]
Abstract
Human pluripotent stem cells (hPSCs) have attracted considerable interest in understanding the cellular fate determination processes and modeling a number of intractable diseases. In vitro generation of skeletal muscle tissues using hPSCs provides an essential model to identify the molecular functions and gene regulatory networks controlling the differentiation of skeletal muscle progenitor cells. Such a genetic roadmap is not only beneficial to understanding human myogenesis but also to decipher the molecular pathology of many skeletal muscle diseases. The combination of established human in vitro myogenesis protocols and newly developed molecular profiling techniques offers extensive insight into the molecular signatures for the development of normal and disease human skeletal muscle tissues. In this review, we provide a comprehensive overview of the current progress of in vitro skeletal muscle generation from hPSCs and relevant examples of the transcriptional landscape and disease-related transcriptional aberrations involving signaling pathways during the development of skeletal muscle cells.
Collapse
Affiliation(s)
- HoTae Lim
- Laboratory of Veterinary Embryology and Biotechnology (VETEMBIO), College of Veterinary Medicine, Chungbuk National University, Cheongju, 28644, Republic of Korea
- School of Medicine, Institute for Cell Engineering, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - In Young Choi
- School of Medicine, Institute for Cell Engineering, Johns Hopkins University, Baltimore, MD, 21205, USA
- Department of Pathology, Graduate School, School of Medicine, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Sang-Hwan Hyun
- Laboratory of Veterinary Embryology and Biotechnology (VETEMBIO), College of Veterinary Medicine, Chungbuk National University, Cheongju, 28644, Republic of Korea
- School of Medicine, Institute for Cell Engineering, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Hyesoo Kim
- School of Medicine, Institute for Cell Engineering, Johns Hopkins University, Baltimore, MD, 21205, USA
- Department of Neurology, School of Medicine, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Gabsang Lee
- School of Medicine, Institute for Cell Engineering, Johns Hopkins University, Baltimore, MD, 21205, USA.
- Department of Neurology, School of Medicine, Johns Hopkins University, Baltimore, MD, 21205, USA.
- The Solomon H. Snyder Department of Neuroscience, School of Medicine, Johns Hopkins University, Baltimore, MD, 21205, USA.
| |
Collapse
|
59
|
da Silva HNM, Covatti C, da Rocha GL, Mizobuti DS, Mâncio RD, Hermes TDA, Kido LA, Cagnon VHA, Pereira ECL, Minatel E. Oxidative Stress, Inflammation, and Activators of Mitochondrial Biogenesis: Tempol Targets in the Diaphragm Muscle of Exercise Trained- mdx Mice. Front Physiol 2021; 12:649793. [PMID: 33981250 PMCID: PMC8107395 DOI: 10.3389/fphys.2021.649793] [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: 01/05/2021] [Accepted: 03/17/2021] [Indexed: 12/18/2022] Open
Abstract
The mdx mouse phenotype aggravated by chronic exercise on a treadmill makes this murine model more reliable for the study of muscular dystrophy. Thus, to better assess the Tempol effect on dystrophic pathways, the analyses in this study were performed in the blood samples and diaphragm muscle from treadmill trained adult (7–11-weeks old) mdx animals. The mdx mice were divided into three groups: mdxSed, sedentary controls (n = 28); mdxEx, exercise-trained animals (n = 28); and mdxEx+T, exercise-trained animals with the Tempol treatment (n = 28). The results demonstrated that the Tempol treatment promoted muscle strength gain, prevented muscle damage, reduced the inflammatory process, oxidative stress, and angiogenesis regulator, and up regulated the activators of mitochondrial biogenesis. The main new findings of this study are that Tempol reduced the NF-κB and increased the PGC1-α and PPARδ levels in the exercise-trained-mdx mice, which are probably related to the ability of this antioxidant to scavenge excessive ROS. These results reinforce the use of Tempol as a potential therapeutic strategy in DMD.
Collapse
Affiliation(s)
| | - Caroline Covatti
- Department of Structural and Functional Biology, Institute of Biology, State University of Campinas (UNICAMP), Campinas, Brazil
| | - Guilherme Luiz da Rocha
- Department of Structural and Functional Biology, Institute of Biology, State University of Campinas (UNICAMP), Campinas, Brazil
| | - Daniela Sayuri Mizobuti
- Department of Structural and Functional Biology, Institute of Biology, State University of Campinas (UNICAMP), Campinas, Brazil
| | - Rafael Dias Mâncio
- Department of Structural and Functional Biology, Institute of Biology, State University of Campinas (UNICAMP), Campinas, Brazil
| | - Túlio de Almeida Hermes
- Department of Structural and Functional Biology, Institute of Biology, State University of Campinas (UNICAMP), Campinas, Brazil
| | - Larissa Akemi Kido
- Department of Structural and Functional Biology, Institute of Biology, State University of Campinas (UNICAMP), Campinas, Brazil
| | - Valéria Helena Alves Cagnon
- Department of Structural and Functional Biology, Institute of Biology, State University of Campinas (UNICAMP), Campinas, Brazil
| | - Elaine Cristina Leite Pereira
- Department of Structural and Functional Biology, Institute of Biology, State University of Campinas (UNICAMP), Campinas, Brazil.,Faculty of Ceilândia, University of Brasília (UnB), Brasília, Brazil
| | - Elaine Minatel
- Department of Structural and Functional Biology, Institute of Biology, State University of Campinas (UNICAMP), Campinas, Brazil
| |
Collapse
|
60
|
Khodabukus A. Tissue-Engineered Skeletal Muscle Models to Study Muscle Function, Plasticity, and Disease. Front Physiol 2021; 12:619710. [PMID: 33716768 PMCID: PMC7952620 DOI: 10.3389/fphys.2021.619710] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Accepted: 01/25/2021] [Indexed: 12/20/2022] Open
Abstract
Skeletal muscle possesses remarkable plasticity that permits functional adaptations to a wide range of signals such as motor input, exercise, and disease. Small animal models have been pivotal in elucidating the molecular mechanisms regulating skeletal muscle adaptation and plasticity. However, these small animal models fail to accurately model human muscle disease resulting in poor clinical success of therapies. Here, we review the potential of in vitro three-dimensional tissue-engineered skeletal muscle models to study muscle function, plasticity, and disease. First, we discuss the generation and function of in vitro skeletal muscle models. We then discuss the genetic, neural, and hormonal factors regulating skeletal muscle fiber-type in vivo and the ability of current in vitro models to study muscle fiber-type regulation. We also evaluate the potential of these systems to be utilized in a patient-specific manner to accurately model and gain novel insights into diseases such as Duchenne muscular dystrophy (DMD) and volumetric muscle loss. We conclude with a discussion on future developments required for tissue-engineered skeletal muscle models to become more mature, biomimetic, and widely utilized for studying muscle physiology, disease, and clinical use.
Collapse
Affiliation(s)
- Alastair Khodabukus
- Department of Biomedical Engineering, Duke University, Durham, NC, United States
| |
Collapse
|
61
|
Kazuki Y, Uno N, Abe S, Kajitani N, Kazuki K, Yakura Y, Sawada C, Takata S, Sugawara M, Nagashima Y, Okada A, Hiratsuka M, Osaki M, Ferrari G, Tedesco FS, Nishikawa S, Fukumoto K, Takayanagi SI, Kunisato A, Kaneko S, Oshimura M, Tomizuka K. Engineering of human induced pluripotent stem cells via human artificial chromosome vectors for cell therapy and disease modeling. MOLECULAR THERAPY. NUCLEIC ACIDS 2021; 23:629-639. [PMID: 33552683 PMCID: PMC7819819 DOI: 10.1016/j.omtn.2020.12.012] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 12/11/2020] [Indexed: 02/04/2023]
Abstract
Genetic engineering of induced pluripotent stem cells (iPSCs) holds great promise for gene and cell therapy as well as drug discovery. However, there are potential concerns regarding the safety and control of gene expression using conventional vectors such as viruses and plasmids. Although human artificial chromosome (HAC) vectors have several advantages as a gene delivery vector, including stable episomal maintenance and the ability to carry large gene inserts, the full potential of HAC transfer into iPSCs still needs to be explored. Here, we provide evidence of a HAC transfer into human iPSCs by microcell-mediated chromosome transfer via measles virus envelope proteins for various applications, including gene and cell therapy, establishment of versatile human iPSCs capable of gene loading and differentiation into T cells, and disease modeling for aneuploidy syndrome. Thus, engineering of human iPSCs via desired HAC vectors is expected to be widely applied in biomedical research.
Collapse
Affiliation(s)
- Yasuhiro Kazuki
- Division of Genome and Cellular Functions, Department of Molecular and Cellular Biology, School of Life Science, Faculty of Medicine, Tottori University, 86 Nishi-cho, Yonago, Tottori 683-8503, Japan
- Chromosome Engineering Research Center (CERC), Tottori University, 86 Nishi-cho, Yonago, Tottori 683-8503, Japan
| | - Narumi Uno
- Division of Genome and Cellular Functions, Department of Molecular and Cellular Biology, School of Life Science, Faculty of Medicine, Tottori University, 86 Nishi-cho, Yonago, Tottori 683-8503, Japan
- Chromosome Engineering Research Center (CERC), Tottori University, 86 Nishi-cho, Yonago, Tottori 683-8503, Japan
- Laboratory of Bioengineering, Tokyo University of Pharmacy and Life Sciences, 1432-1, Horinouchi, Hachioji, Tokyo 192-0392, Japan
| | - Satoshi Abe
- Chromosome Engineering Research Center (CERC), Tottori University, 86 Nishi-cho, Yonago, Tottori 683-8503, Japan
| | - Naoyo Kajitani
- Chromosome Engineering Research Center (CERC), Tottori University, 86 Nishi-cho, Yonago, Tottori 683-8503, Japan
| | - Kanako Kazuki
- Chromosome Engineering Research Center (CERC), Tottori University, 86 Nishi-cho, Yonago, Tottori 683-8503, Japan
| | - Yuwna Yakura
- Chromosome Engineering Research Center (CERC), Tottori University, 86 Nishi-cho, Yonago, Tottori 683-8503, Japan
| | - Chiaki Sawada
- Division of Genome and Cellular Functions, Department of Molecular and Cellular Biology, School of Life Science, Faculty of Medicine, Tottori University, 86 Nishi-cho, Yonago, Tottori 683-8503, Japan
| | - Shuta Takata
- Division of Genome and Cellular Functions, Department of Molecular and Cellular Biology, School of Life Science, Faculty of Medicine, Tottori University, 86 Nishi-cho, Yonago, Tottori 683-8503, Japan
| | - Masaki Sugawara
- Division of Genome and Cellular Functions, Department of Molecular and Cellular Biology, School of Life Science, Faculty of Medicine, Tottori University, 86 Nishi-cho, Yonago, Tottori 683-8503, Japan
| | - Yuichi Nagashima
- Division of Genome and Cellular Functions, Department of Molecular and Cellular Biology, School of Life Science, Faculty of Medicine, Tottori University, 86 Nishi-cho, Yonago, Tottori 683-8503, Japan
| | - Akane Okada
- Division of Genome and Cellular Functions, Department of Molecular and Cellular Biology, School of Life Science, Faculty of Medicine, Tottori University, 86 Nishi-cho, Yonago, Tottori 683-8503, Japan
| | - Masaharu Hiratsuka
- Division of Genome and Cellular Functions, Department of Molecular and Cellular Biology, School of Life Science, Faculty of Medicine, Tottori University, 86 Nishi-cho, Yonago, Tottori 683-8503, Japan
| | - Mitsuhiko Osaki
- Division of Experimental Pathology, Faculty of Medicine, Tottori University, 86 Nishi-cho, Yonago, Tottori 683-8503, Japan
| | - Giulia Ferrari
- Department of Cell and Developmental Biology, University College London, London WC1E 6DE, UK
| | - Francesco Saverio Tedesco
- Department of Cell and Developmental Biology, University College London, London WC1E 6DE, UK
- Dubowitz Neuromuscular Centre, Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK
- The Francis Crick Institute, London NW1 1AT, UK
| | - Satoshi Nishikawa
- Regenerative Medicine Research Laboratories, Research Functions Unit, R&D Division, Kyowa Kirin, Co., Ltd. 3-6-6, Asahi-machi, Machida-shi, Tokyo 194-8533, Japan
| | - Ken Fukumoto
- Cell Therapy Project, R&D Division, Kirin Holdings, Co., Ltd. 1-13-5, Fukuura Kanazawa-ku, Yokohama, Kanagawa 236-0004 Japan
| | - Shin-ichiro Takayanagi
- Cell Therapy Project, R&D Division, Kirin Holdings, Co., Ltd. 1-13-5, Fukuura Kanazawa-ku, Yokohama, Kanagawa 236-0004 Japan
| | - Atsushi Kunisato
- Project Planning Section, Kirin Holdings, Co., Ltd., 4-10-2 Nakano, Nakano-ku, Tokyo 164-0001 Japan
| | - Shin Kaneko
- Shin Kaneko Laboratory, Department of Cell Growth and Differentiation, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Mitsuo Oshimura
- Chromosome Engineering Research Center (CERC), Tottori University, 86 Nishi-cho, Yonago, Tottori 683-8503, Japan
| | - Kazuma Tomizuka
- Laboratory of Bioengineering, Tokyo University of Pharmacy and Life Sciences, 1432-1, Horinouchi, Hachioji, Tokyo 192-0392, Japan
| |
Collapse
|
62
|
Fortunato F, Rossi R, Falzarano MS, Ferlini A. Innovative Therapeutic Approaches for Duchenne Muscular Dystrophy. J Clin Med 2021; 10:jcm10040820. [PMID: 33671409 PMCID: PMC7922390 DOI: 10.3390/jcm10040820] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 02/10/2021] [Accepted: 02/12/2021] [Indexed: 02/06/2023] Open
Abstract
Duchenne muscular dystrophy (DMD) is the most common childhood muscular dystrophy affecting ~1:5000 live male births. Following the identification of pathogenic variations in the dystrophin gene in 1986, the underlining genotype/phenotype correlations emerged and the role of the dystrophin protein was elucidated in skeletal, smooth, and cardiac muscles, as well as in the brain. When the dystrophin protein is absent or quantitatively or qualitatively modified, the muscle cannot sustain the stress of repeated contractions. Dystrophin acts as a bridging and anchoring protein between the sarcomere and the sarcolemma, and its absence or reduction leads to severe muscle damage that eventually cannot be repaired, with its ultimate substitution by connective tissue and fat. The advances of an understanding of the molecular pathways affected in DMD have led to the development of many therapeutic strategies that tackle different aspects of disease etiopathogenesis, which have recently led to the first successful approved orphan drugs for this condition. The therapeutic advances in this field have progressed exponentially, with second-generation drugs now entering in clinical trials as gene therapy, potentially providing a further effective approach to the condition.
Collapse
|
63
|
Sinenko SA, Ponomartsev SV, Tomilin AN. Pluripotent stem cell-based gene therapy approach: human de novo synthesized chromosomes. Cell Mol Life Sci 2021; 78:1207-1220. [PMID: 33011821 PMCID: PMC11072874 DOI: 10.1007/s00018-020-03653-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 09/14/2020] [Accepted: 09/22/2020] [Indexed: 02/06/2023]
Abstract
A novel approach in gene therapy was introduced 20 years ago since artificial non-integrative chromosome-based vectors containing gene loci size inserts were engineered. To date, different human artificial chromosomes (HAC) were generated with the use of de novo construction or "top-down" engineering approaches. The HAC-based therapeutic approach includes ex vivo gene transferring and correction of pluripotent stem cells (PSCs) or highly proliferative modified stem cells. The current progress in the technology of induced PSCs, integrating with the HAC technology, resulted in a novel platform of stem cell-based tissue replacement therapy for the treatment of genetic disease. Nowadays, the sophisticated and laborious HAC technology has significantly improved and is now closer to clinical studies. In here, we reviewed the achievements in the technology of de novo synthesized HACs for a chromosome transfer for developing gene therapy tissue replacement models of monogenic human diseases.
Collapse
Affiliation(s)
- Sergey A Sinenko
- Institute of Cytology, Russian Academy of Sciences, 4 Tikhoretsky Ave, St-Petersburg, 194064, Russia.
| | - Sergey V Ponomartsev
- Institute of Cytology, Russian Academy of Sciences, 4 Tikhoretsky Ave, St-Petersburg, 194064, Russia
| | - Alexey N Tomilin
- Institute of Cytology, Russian Academy of Sciences, 4 Tikhoretsky Ave, St-Petersburg, 194064, Russia.
- Institute of Translational Biomedicine, St-Petersburg State University, 7-9, Universitetskaya Emb, St-Petersburg, 199034, Russia.
| |
Collapse
|
64
|
Mournetas V, Massouridès E, Dupont JB, Kornobis E, Polvèche H, Jarrige M, Dorval ARL, Gosselin MRF, Manousopoulou A, Garbis SD, Górecki DC, Pinset C. Myogenesis modelled by human pluripotent stem cells: a multi-omic study of Duchenne myopathy early onset. J Cachexia Sarcopenia Muscle 2021; 12:209-232. [PMID: 33586340 PMCID: PMC7890274 DOI: 10.1002/jcsm.12665] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 11/25/2020] [Accepted: 12/16/2020] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Duchenne muscular dystrophy (DMD) causes severe disability of children and death of young men, with an incidence of approximately 1/5000 male births. Symptoms appear in early childhood, with a diagnosis made mostly around 4 years old, a time where the amount of muscle damage is already significant, preventing early therapeutic interventions that could be more efficient at halting disease progression. In the meantime, the precise moment at which disease phenotypes arise-even asymptomatically-is still unknown. Thus, there is a critical need to better define DMD onset as well as its first manifestations, which could help identify early disease biomarkers and novel therapeutic targets. METHODS We have used both human tissue-derived myoblasts and human induced pluripotent stem cells (hiPSCs) from DMD patients to model skeletal myogenesis and compared their differentiation dynamics with that of healthy control cells by a comprehensive multi-omic analysis at seven time points. Results were strengthened with the analysis of isogenic CRISPR-edited human embryonic stem cells and through comparisons against published transcriptomic and proteomic datasets from human DMD muscles. The study was completed with DMD knockdown/rescue experiments in hiPSC-derived skeletal muscle progenitor cells and adenosine triphosphate measurement in hiPSC-derived myotubes. RESULTS Transcriptome and miRnome comparisons combined with protein analyses demonstrated that hiPSC differentiation (i) leads to embryonic/foetal myotubes that mimic described DMD phenotypes at the differentiation endpoint and (ii) homogeneously and robustly recapitulates key developmental steps-mesoderm, somite, and skeletal muscle. Starting at the somite stage, DMD dysregulations concerned almost 10% of the transcriptome. These include mitochondrial genes whose dysregulations escalate during differentiation. We also describe fibrosis as an intrinsic feature of DMD skeletal muscle cells that begins early during myogenesis. All the omics data are available online for exploration through a graphical interface at https://muscle-dmd.omics.ovh/. CONCLUSIONS Our data argue for an early developmental manifestation of DMD whose onset is triggered before the entry into the skeletal muscle compartment, data leading to a necessary reconsideration of dystrophin roles during muscle development. This hiPSC model of skeletal muscle differentiation offers the possibility to explore these functions as well as find earlier DMD biomarkers and therapeutic targets.
Collapse
Affiliation(s)
| | | | | | - Etienne Kornobis
- Biomics, C2RT, Institut Pasteur, Paris, France.,Hub de Bioinformatique et Biostatistique - Département BiologieComputationnelle, Paris, France
| | | | | | | | - Maxime R F Gosselin
- Molecular Medicine, School of Pharmacy and Biomedical Sciences, University of Portsmouth, Portsmouth, UK
| | - Antigoni Manousopoulou
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA, USA
| | - Spiros D Garbis
- Unit for Cancer Sciences, Centre for Proteomics Research, Institute for Life Sciences, University of Southampton, Southampton, UK.,Proteas Bioanalytics Inc., BioLabs at The Lundquist Institute, Torrance, CA, USA
| | - Dariusz C Górecki
- Molecular Medicine, School of Pharmacy and Biomedical Sciences, University of Portsmouth, Portsmouth, UK.,Military Institute of Hygiene and Epidemiology, Warsaw, Poland
| | | |
Collapse
|
65
|
Xu B, Siehr A, Shen W. Functional skeletal muscle constructs from transdifferentiated human fibroblasts. Sci Rep 2020; 10:22047. [PMID: 33328524 PMCID: PMC7744552 DOI: 10.1038/s41598-020-78987-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Accepted: 11/24/2020] [Indexed: 12/14/2022] Open
Abstract
Transdifferentiation of human non-muscle cells directly into myogenic cells by forced expression of MyoD represents one route to obtain highly desirable human myogenic cells. However, functional properties of the tissue constructs derived from these transdifferentiated cells have been rarely studied. Here, we report that three-dimensional (3D) tissue constructs engineered with iMyoD-hTERT-NHDFs, normal human dermal fibroblasts transduced with genes encoding human telomerase reverse transcriptase and doxycycline-inducible MyoD, generate detectable contractile forces in response to electrical stimuli upon MyoD expression. Withdrawal of doxycycline in the middle of 3D culture results in 3.05 and 2.28 times increases in twitch and tetanic forces, respectively, suggesting that temporally-controlled MyoD expression benefits functional myogenic differentiation of transdifferentiated myoblast-like cells. Treatment with CHIR99021, a Wnt activator, and DAPT, a Notch inhibitor, leads to further enhanced contractile forces. The ability of these abundant and potentially patient-specific and disease-specific cells to develop into functional skeletal muscle constructs makes them highly valuable for many applications, such as disease modeling.
Collapse
Affiliation(s)
- Bin Xu
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Allison Siehr
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Wei Shen
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN, 55455, USA.
- Stem Cell Institute, University of Minnesota, Minneapolis, MN, 55455, USA.
- Institute for Engineering in Medicine, University of Minnesota, Minneapolis, MN, 55455, USA.
| |
Collapse
|
66
|
Hermes TDA, Mizobuti DS, da Rocha GL, da Silva HNM, Covatti C, Pereira ECL, Ferretti R, Minatel E. Tempol improves redox status in mdx dystrophic diaphragm muscle. Int J Exp Pathol 2020; 101:289-297. [PMID: 33098599 DOI: 10.1111/iep.12376] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 08/10/2020] [Accepted: 08/23/2020] [Indexed: 12/15/2022] Open
Abstract
Oxidative stress is a critical element in relationship to the pathophysiology of Duchenne muscular dystrophy (DMD). In the mice the diaphragm (DIA) is most resembles the dystrophic human pathology. In this study we have evaluated the consequences of a synthetic antioxidant (tempol) on oxidative stress parameters in the DIA muscle of mdx mice. The mdx mice were separated into two groups: mdx, the control group receiving intraperitoneal (i.p.) injections of saline solution (100 µL), and mdxT, the treated group receiving i.p. injections of tempol (100 mg/kg). The tempol-treated group showed reduced oxidative stress markers, decreasing the dihydroethidium reaction (DHE) area; autofluorescent lipofuscin granules; and 4-hydroxynonenal (4-HNE)-protein adduct levels. DIA muscle of mdx mice. At the same time, the manganese-superoxide dismutase 2 (SOD2) levels were increased in the tempol-treated group. In addition, the tempol-treated group showed reduced levels of glutathione-disulphide reductase (GSR), glutathione peroxidase 1 (GPx1) and catalase (CAT) in immunoblots. The tempol-treated group has also shown lower relative gene expression of SOD1, CAT and GPx than the non-treated group. Our data demonstrated that tempol treatment reduced oxidant parameters and increased anti-oxidant SOD2 levels in the DIA muscle of mdx mice, which may contribute to the normalization of the redox homeostasis of dystrophic muscles.
Collapse
Affiliation(s)
- Túlio de Almeida Hermes
- Department of Structural and Functional Biology, Institute of Biology, State University of Campinas (UNICAMP), Campinas, Brazil
| | - Daniela Sayuri Mizobuti
- Department of Structural and Functional Biology, Institute of Biology, State University of Campinas (UNICAMP), Campinas, Brazil
| | - Guilherme Luiz da Rocha
- Department of Structural and Functional Biology, Institute of Biology, State University of Campinas (UNICAMP), Campinas, Brazil
| | | | - Caroline Covatti
- Department of Structural and Functional Biology, Institute of Biology, State University of Campinas (UNICAMP), Campinas, Brazil
| | - Elaine Cristina Leite Pereira
- Department of Structural and Functional Biology, Institute of Biology, State University of Campinas (UNICAMP), Campinas, Brazil.,Faculty of Ceilandia, University of Brasilia (UnB), Brasília, Brazil
| | - Renato Ferretti
- Department of Anatomy, Institute of Bioscience of Botucatu, São Paulo State University (UNESP), Botucatu, Brazil
| | - Elaine Minatel
- Department of Structural and Functional Biology, Institute of Biology, State University of Campinas (UNICAMP), Campinas, Brazil
| |
Collapse
|
67
|
A Concise Review on Induced Pluripotent Stem Cell-Derived Cardiomyocytes for Personalized Regenerative Medicine. Stem Cell Rev Rep 2020; 17:748-776. [PMID: 33098306 DOI: 10.1007/s12015-020-10061-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/16/2020] [Indexed: 02/07/2023]
Abstract
The induced pluripotent stem cells (iPSCs) are derived from somatic cells by using reprogramming factors such as Oct4, Sox2, Klf4, and c-Myc (OSKM) or Oct4, Sox2, Nanog and Lin28 (OSNL). They resemble embryonic stem cells (ESCs) and have the ability to differentiate into cell lineage of all three germ-layer, including cardiomyocytes (CMs). The CMs can be generated from iPSCs by inducing embryoid bodies (EBs) formation and treatment with activin A, bone morphogenic protein 4 (BMP4), and inhibitors of Wnt signaling. However, these iPSC-derived CMs are a heterogeneous population of cells and require purification and maturation to mimic the in vivo CMs. The matured CMs can be used for various therapeutic purposes in regenerative medicine by cardiomyoplasty or through the development of tissue-engineered cardiac patches. In recent years, significant advancements have been made in the isolation of iPSC and their differentiation, purification, and maturation into clinically usable CMs. Newer small molecules have also been identified to substitute the reprogramming factors for iPSC generation as well as for direct differentiation of somatic cells into CMs without an intermediary pluripotent state. This review provides a concise update on the generation of iPSC-derived CMs and their application in personalized cardiac regenerative medicine. It also discusses the current limitations and challenges in the application of iPSC-derived CMs. Graphical abstract.
Collapse
|
68
|
Modelling Neuromuscular Diseases in the Age of Precision Medicine. J Pers Med 2020; 10:jpm10040178. [PMID: 33080928 PMCID: PMC7712305 DOI: 10.3390/jpm10040178] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 10/14/2020] [Accepted: 10/15/2020] [Indexed: 12/20/2022] Open
Abstract
Advances in knowledge resulting from the sequencing of the human genome, coupled with technological developments and a deeper understanding of disease mechanisms of pathogenesis are paving the way for a growing role of precision medicine in the treatment of a number of human conditions. The goal of precision medicine is to identify and deliver effective therapeutic approaches based on patients’ genetic, environmental, and lifestyle factors. With the exception of cancer, neurological diseases provide the most promising opportunity to achieve treatment personalisation, mainly because of accelerated progress in gene discovery, deep clinical phenotyping, and biomarker availability. Developing reproducible, predictable and reliable disease models will be key to the rapid delivery of the anticipated benefits of precision medicine. Here we summarize the current state of the art of preclinical models for neuromuscular diseases, with particular focus on their use and limitations to predict safety and efficacy treatment outcomes in clinical trials.
Collapse
|
69
|
Farr GH, Morris M, Gomez A, Pham T, Kilroy E, Parker EU, Said S, Henry C, Maves L. A novel chemical-combination screen in zebrafish identifies epigenetic small molecule candidates for the treatment of Duchenne muscular dystrophy. Skelet Muscle 2020; 10:29. [PMID: 33059738 PMCID: PMC7559456 DOI: 10.1186/s13395-020-00251-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Accepted: 10/05/2020] [Indexed: 02/06/2023] Open
Abstract
Background Duchenne muscular dystrophy (DMD) is a severe neuromuscular disorder and is one of the most common muscular dystrophies. There are currently few effective therapies to treat the disease, although many small-molecule approaches are being pursued. Certain histone deacetylase inhibitors (HDACi) have been shown to ameliorate DMD phenotypes in mouse and zebrafish animal models. The HDACi givinostat has shown promise for DMD in clinical trials. However, beyond a small group of HDACi, other classes of epigenetic small molecules have not been broadly and systematically studied for their benefits for DMD. Methods We used an established animal model for DMD, the zebrafish dmd mutant strain sapje. A commercially available library of epigenetic small molecules was used to treat embryonic-larval stages of dmd mutant zebrafish. We used a quantitative muscle birefringence assay in order to assess and compare the effects of small-molecule treatments on dmd mutant zebrafish skeletal muscle structure. Results We performed a novel chemical-combination screen of a library of epigenetic compounds using the zebrafish dmd model. We identified candidate pools of epigenetic compounds that improve skeletal muscle structure in dmd mutant zebrafish. We then identified a specific combination of two HDACi compounds, oxamflatin and salermide, that ameliorated dmd mutant zebrafish skeletal muscle degeneration. We validated the effects of oxamflatin and salermide on dmd mutant zebrafish in an independent laboratory. Furthermore, we showed that the combination of oxamflatin and salermide caused increased levels of histone H4 acetylation in zebrafish larvae. Conclusions Our results provide novel, effective methods for performing a combination of small-molecule screen in zebrafish. Our results also add to the growing evidence that epigenetic small molecules may be promising candidates for treating DMD.
Collapse
Affiliation(s)
- Gist H Farr
- Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, Seattle, WA, USA
| | - Melanie Morris
- Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, Seattle, WA, USA.,Medical Student Research Training Program, University of Washington School of Medicine, Seattle, WA, USA
| | - Arianna Gomez
- Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, Seattle, WA, USA.,Molecular Medicine and Mechanisms of Disease Program, Department of Pathology, University of Washington, Seattle, WA, USA.,Department of Pediatrics, University of Washington, Seattle, WA, USA
| | - Thao Pham
- Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, Seattle, WA, USA.,Molecular Medicine and Mechanisms of Disease Program, Department of Pathology, University of Washington, Seattle, WA, USA
| | - Elisabeth Kilroy
- Graduate School of Biomedical Science and Engineering, University of Maine, Orono, ME, USA
| | - Elizabeth U Parker
- Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, Seattle, WA, USA.,Medical Student Research Training Program, University of Washington School of Medicine, Seattle, WA, USA
| | - Shery Said
- Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, Seattle, WA, USA
| | - Clarissa Henry
- School of Biology and Ecology, University of Maine, Orono, ME, USA
| | - Lisa Maves
- Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, Seattle, WA, USA. .,Department of Pediatrics, University of Washington, Seattle, WA, USA.
| |
Collapse
|
70
|
May V, Arnold AA, Pagad S, Somagutta MR, Sridharan S, Nanthakumaran S, Malik BH. Duchenne's Muscular Dystrophy: The Role of Induced Pluripotent Stem Cells and Genomic Editing on Muscle Regeneration. Cureus 2020; 12:e10600. [PMID: 33123420 PMCID: PMC7584317 DOI: 10.7759/cureus.10600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
There are two types of well-known muscular dystrophies: Duchenne's muscular dystrophy (DMD) and Becker's muscular dystrophy. This article focuses on the X-linked recessive disorder of Duchenne's muscular dystrophy, which primarily affects children at age four, with a shortened life span of up to 40 years. A defective dystrophin protein lacking the gene dystrophin is the primary cause of the disease pathophysiology. This defect causes cardiac and skeletal muscle down-regulation of dystrophin, leading to weak and fibrotic muscles. The disease is currently untreatable, so most kids die due to cardiac failure in their late 30's. This review presents current treatment options, based on previous studies conducted over the last five years. We used the PubMed database to analyze and review the most important investigations. We also included an analysis of induced pluripotent stem cell therapy vs. genetic therapy using the mdx mouse model. We have discovered promising results on mdx mouse models to date and excited about the potential for where further clinical human trials can go.
Collapse
Affiliation(s)
- Vanessa May
- Department of Research, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Ashley A Arnold
- Department of Research, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Sukrut Pagad
- Department of Internal Medicine, Larkin Community Hospital, Hialeah, USA
| | - Manoj R Somagutta
- Department of Research, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Saijanakan Sridharan
- Department of Research, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Saruja Nanthakumaran
- Department of Research, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Bilal Haider Malik
- Department of Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| |
Collapse
|
71
|
Sanjurjo-Rodríguez C, Castro-Viñuelas R, Piñeiro-Ramil M, Rodríguez-Fernández S, Fuentes-Boquete I, Blanco FJ, Díaz-Prado S. Versatility of Induced Pluripotent Stem Cells (iPSCs) for Improving the Knowledge on Musculoskeletal Diseases. Int J Mol Sci 2020; 21:ijms21176124. [PMID: 32854405 PMCID: PMC7504376 DOI: 10.3390/ijms21176124] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 08/06/2020] [Accepted: 08/20/2020] [Indexed: 12/13/2022] Open
Abstract
Induced pluripotent stem cells (iPSCs) represent an unlimited source of pluripotent cells capable of differentiating into any cell type of the body. Several studies have demonstrated the valuable use of iPSCs as a tool for studying the molecular and cellular mechanisms underlying disorders affecting bone, cartilage and muscle, as well as their potential for tissue repair. Musculoskeletal diseases are one of the major causes of disability worldwide and impose an important socio-economic burden. To date there is neither cure nor proven approach for effectively treating most of these conditions and therefore new strategies involving the use of cells have been increasingly investigated in the recent years. Nevertheless, some limitations related to the safety and differentiation protocols among others remain, which humpers the translational application of these strategies. Nonetheless, the potential is indisputable and iPSCs are likely to be a source of different types of cells useful in the musculoskeletal field, for either disease modeling or regenerative medicine. In this review, we aim to illustrate the great potential of iPSCs by summarizing and discussing the in vitro tissue regeneration preclinical studies that have been carried out in the musculoskeletal field by using iPSCs.
Collapse
Affiliation(s)
- Clara Sanjurjo-Rodríguez
- Cell Therapy and Regenerative Medicine Group, Department of Physiotherapy, Medicine and Biomedical Sciences, Faculty of Health Sciences, University of A Coruña (UDC), 15006 A Coruña, Galicia, Spain; (R.C.-V.); (M.P.-R.); (S.R.-F.); (I.F.-B.)
- Institute of Biomedical Research of A Coruña (INIBIC), University Hospital Complex A Coruña (CHUAC), Galician Health Service (SERGAS), 15006 A Coruña, Galicia, Spain;
- Centro de Investigación Biomédica en Red (CIBER) de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), 28029 Madrid, Spain
- Centro de Investigaciones Científicas Avanzadas (CICA), Agrupación estratégica CICA-INIBIC, University of A Coruña, 15008 A Coruña, Galicia, Spain
- Correspondence: (C.S.-R.); (S.D.-P.)
| | - Rocío Castro-Viñuelas
- Cell Therapy and Regenerative Medicine Group, Department of Physiotherapy, Medicine and Biomedical Sciences, Faculty of Health Sciences, University of A Coruña (UDC), 15006 A Coruña, Galicia, Spain; (R.C.-V.); (M.P.-R.); (S.R.-F.); (I.F.-B.)
- Institute of Biomedical Research of A Coruña (INIBIC), University Hospital Complex A Coruña (CHUAC), Galician Health Service (SERGAS), 15006 A Coruña, Galicia, Spain;
- Centro de Investigaciones Científicas Avanzadas (CICA), Agrupación estratégica CICA-INIBIC, University of A Coruña, 15008 A Coruña, Galicia, Spain
| | - María Piñeiro-Ramil
- Cell Therapy and Regenerative Medicine Group, Department of Physiotherapy, Medicine and Biomedical Sciences, Faculty of Health Sciences, University of A Coruña (UDC), 15006 A Coruña, Galicia, Spain; (R.C.-V.); (M.P.-R.); (S.R.-F.); (I.F.-B.)
- Institute of Biomedical Research of A Coruña (INIBIC), University Hospital Complex A Coruña (CHUAC), Galician Health Service (SERGAS), 15006 A Coruña, Galicia, Spain;
- Centro de Investigaciones Científicas Avanzadas (CICA), Agrupación estratégica CICA-INIBIC, University of A Coruña, 15008 A Coruña, Galicia, Spain
| | - Silvia Rodríguez-Fernández
- Cell Therapy and Regenerative Medicine Group, Department of Physiotherapy, Medicine and Biomedical Sciences, Faculty of Health Sciences, University of A Coruña (UDC), 15006 A Coruña, Galicia, Spain; (R.C.-V.); (M.P.-R.); (S.R.-F.); (I.F.-B.)
- Institute of Biomedical Research of A Coruña (INIBIC), University Hospital Complex A Coruña (CHUAC), Galician Health Service (SERGAS), 15006 A Coruña, Galicia, Spain;
- Centro de Investigaciones Científicas Avanzadas (CICA), Agrupación estratégica CICA-INIBIC, University of A Coruña, 15008 A Coruña, Galicia, Spain
| | - Isaac Fuentes-Boquete
- Cell Therapy and Regenerative Medicine Group, Department of Physiotherapy, Medicine and Biomedical Sciences, Faculty of Health Sciences, University of A Coruña (UDC), 15006 A Coruña, Galicia, Spain; (R.C.-V.); (M.P.-R.); (S.R.-F.); (I.F.-B.)
- Institute of Biomedical Research of A Coruña (INIBIC), University Hospital Complex A Coruña (CHUAC), Galician Health Service (SERGAS), 15006 A Coruña, Galicia, Spain;
- Centro de Investigaciones Científicas Avanzadas (CICA), Agrupación estratégica CICA-INIBIC, University of A Coruña, 15008 A Coruña, Galicia, Spain
| | - Francisco J. Blanco
- Institute of Biomedical Research of A Coruña (INIBIC), University Hospital Complex A Coruña (CHUAC), Galician Health Service (SERGAS), 15006 A Coruña, Galicia, Spain;
- Centro de Investigación Biomédica en Red (CIBER) de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), 28029 Madrid, Spain
- Centro de Investigaciones Científicas Avanzadas (CICA), Agrupación estratégica CICA-INIBIC, University of A Coruña, 15008 A Coruña, Galicia, Spain
- Tissular Bioengineering and Cell Therapy Unit (GBTTC-CHUAC), Rheumatology Group, 15006 A Coruña, Galicia, Spain
| | - Silvia Díaz-Prado
- Cell Therapy and Regenerative Medicine Group, Department of Physiotherapy, Medicine and Biomedical Sciences, Faculty of Health Sciences, University of A Coruña (UDC), 15006 A Coruña, Galicia, Spain; (R.C.-V.); (M.P.-R.); (S.R.-F.); (I.F.-B.)
- Institute of Biomedical Research of A Coruña (INIBIC), University Hospital Complex A Coruña (CHUAC), Galician Health Service (SERGAS), 15006 A Coruña, Galicia, Spain;
- Centro de Investigación Biomédica en Red (CIBER) de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), 28029 Madrid, Spain
- Centro de Investigaciones Científicas Avanzadas (CICA), Agrupación estratégica CICA-INIBIC, University of A Coruña, 15008 A Coruña, Galicia, Spain
- Correspondence: (C.S.-R.); (S.D.-P.)
| |
Collapse
|
72
|
Zullo L, Bozzo M, Daya A, Di Clemente A, Mancini FP, Megighian A, Nesher N, Röttinger E, Shomrat T, Tiozzo S, Zullo A, Candiani S. The Diversity of Muscles and Their Regenerative Potential across Animals. Cells 2020; 9:cells9091925. [PMID: 32825163 PMCID: PMC7563492 DOI: 10.3390/cells9091925] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 08/14/2020] [Accepted: 08/17/2020] [Indexed: 02/06/2023] Open
Abstract
Cells with contractile functions are present in almost all metazoans, and so are the related processes of muscle homeostasis and regeneration. Regeneration itself is a complex process unevenly spread across metazoans that ranges from full-body regeneration to partial reconstruction of damaged organs or body tissues, including muscles. The cellular and molecular mechanisms involved in regenerative processes can be homologous, co-opted, and/or evolved independently. By comparing the mechanisms of muscle homeostasis and regeneration throughout the diversity of animal body-plans and life cycles, it is possible to identify conserved and divergent cellular and molecular mechanisms underlying muscle plasticity. In this review we aim at providing an overview of muscle regeneration studies in metazoans, highlighting the major regenerative strategies and molecular pathways involved. By gathering these findings, we wish to advocate a comparative and evolutionary approach to prompt a wider use of “non-canonical” animal models for molecular and even pharmacological studies in the field of muscle regeneration.
Collapse
Affiliation(s)
- Letizia Zullo
- Istituto Italiano di Tecnologia, Center for Micro-BioRobotics & Center for Synaptic Neuroscience and Technology (NSYN), 16132 Genova, Italy;
- IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy
- Correspondence: (L.Z.); (A.Z.)
| | - Matteo Bozzo
- Laboratory of Developmental Neurobiology, Department of Earth, Environment and Life Sciences, University of Genova, Viale Benedetto XV 5, 16132 Genova, Italy; (M.B.); (S.C.)
| | - Alon Daya
- Faculty of Marine Sciences, Ruppin Academic Center, Michmoret 40297, Israel; (A.D.); (N.N.); (T.S.)
| | - Alessio Di Clemente
- Istituto Italiano di Tecnologia, Center for Micro-BioRobotics & Center for Synaptic Neuroscience and Technology (NSYN), 16132 Genova, Italy;
- Department of Experimental Medicine, University of Genova, Viale Benedetto XV, 3, 16132 Genova, Italy
| | | | - Aram Megighian
- Department of Biomedical Sciences, University of Padova, 35131 Padova, Italy;
- Padova Neuroscience Center, University of Padova, 35131 Padova, Italy
| | - Nir Nesher
- Faculty of Marine Sciences, Ruppin Academic Center, Michmoret 40297, Israel; (A.D.); (N.N.); (T.S.)
| | - Eric Röttinger
- Institute for Research on Cancer and Aging (IRCAN), Université Côte d’Azur, CNRS, INSERM, 06107 Nice, France;
| | - Tal Shomrat
- Faculty of Marine Sciences, Ruppin Academic Center, Michmoret 40297, Israel; (A.D.); (N.N.); (T.S.)
| | - Stefano Tiozzo
- Laboratoire de Biologie du Développement de Villefranche-sur-Mer (LBDV), Sorbonne Université, CNRS, 06230 Paris, France;
| | - Alberto Zullo
- Department of Science and Technology, University of Sannio, 82100 Benevento, Italy;
- Correspondence: (L.Z.); (A.Z.)
| | - Simona Candiani
- Laboratory of Developmental Neurobiology, Department of Earth, Environment and Life Sciences, University of Genova, Viale Benedetto XV 5, 16132 Genova, Italy; (M.B.); (S.C.)
| |
Collapse
|
73
|
Choi IY, Lim H, Huynh A, Schofield J, Cho HJ, Lee H, Andersen P, Shin JH, Heo WD, Hyun SH, Kim YJ, Oh Y, Kim H, Lee G. Novel culture system via wirelessly controllable optical stimulation of the FGF signaling pathway for human and pig pluripotency. Biomaterials 2020; 269:120222. [PMID: 32736809 DOI: 10.1016/j.biomaterials.2020.120222] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2019] [Revised: 07/01/2020] [Accepted: 07/02/2020] [Indexed: 12/31/2022]
Abstract
Stem cell fate is largely determined by cellular signaling networks and is heavily dependent on the supplementation of exogenous recombinant proteins into culture media; however, uneven distribution and inconsistent stability of recombinant proteins are closely associated with the spontaneous differentiation of pluripotent stem cells (PSCs) and result in significant costs in large-scale manufacturing. Here, we report a novel PSC culture system via wirelessly controllable optical activation of the fibroblast growth factor (FGF) signaling pathway without the need for supplementation of recombinant FGF2 protein, a key molecule for maintaining pluripotency of PSCs. Using a fusion protein between the cytoplasmic region of the FGF receptor-1 and a light-oxygen-voltage domain, we achieved tunable, blue light-dependent activation of FGF signaling in human and porcine PSCs. Our data demonstrate that a highly controllable optical stimulation of the FGF signaling pathway is sufficient for long-term maintenance of PSCs, without the loss of differentiation potential into three germ layers. This culture system will be a cost-effective platform for a large-scale stem cell culture.
Collapse
Affiliation(s)
- In Young Choi
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Pathology, Graduate School, Kyung Hee University, School of Medicine, Seoul, Republic of Korea
| | - HoTae Lim
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA; College of Veterinary Medicine, Chungbuk National University, Chungbuk, Republic of Korea
| | - Alex Huynh
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - James Schofield
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Hyeon Jin Cho
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA
| | - Hosuk Lee
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Peter Andersen
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Joo Heon Shin
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA; Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Won Do Heo
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon, Republic of Korea; Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| | - Sang-Hwan Hyun
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA; College of Veterinary Medicine, Chungbuk National University, Chungbuk, Republic of Korea
| | - Yong Jun Kim
- Department of Pathology, Department of Biomedical Science, Graduate School, Kyung Hee University, School of Medicine, Seoul, Republic of Korea
| | - Yohan Oh
- Department of Biomedical Science, Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, Republic of Korea; Department of Biochemistry and Molecular Biology, College of Medicine, Hanyang University, Seoul, Republic of Korea.
| | - Hyesoo Kim
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - Gabsang Lee
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA; The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
74
|
Zhao M, Tazumi A, Takayama S, Takenaka-Ninagawa N, Nalbandian M, Nagai M, Nakamura Y, Nakasa M, Watanabe A, Ikeya M, Hotta A, Ito Y, Sato T, Sakurai H. Induced Fetal Human Muscle Stem Cells with High Therapeutic Potential in a Mouse Muscular Dystrophy Model. Stem Cell Reports 2020; 15:80-94. [PMID: 32619494 PMCID: PMC7363940 DOI: 10.1016/j.stemcr.2020.06.004] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Revised: 06/03/2020] [Accepted: 06/03/2020] [Indexed: 12/14/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is a progressive and fatal muscle-wasting disease caused by DYSTROPHIN deficiency. Cell therapy using muscle stem cells (MuSCs) is a potential cure. Here, we report a differentiation method to generate fetal MuSCs from human induced pluripotent stem cells (iPSCs) by monitoring MYF5 expression. Gene expression profiling indicated that MYF5-positive cells in the late stage of differentiation have fetal MuSC characteristics, while MYF5-positive cells in the early stage of differentiation have early myogenic progenitor characteristics. Moreover, late-stage MYF5-positive cells demonstrated good muscle regeneration potential and produced DYSTROPHIN in vivo after transplantation into DMD model mice, resulting in muscle function recovery. The engrafted cells also generated PAX7-positive MuSC-like cells under the basal lamina of DYSTROPHIN-positive fibers. These findings suggest that MYF5-positive fetal MuSCs induced in the late stage of iPSC differentiation have cell therapy potential for DMD. Wnt agonists at high dose and long term induces dermomyotome cells effectively MYF5+ cell characteristics vary between early- and late-stage differentiation Late-stage MYF5+ cells acquire characteristics resembling fetal muscle stem cells MYF5+ cells recover dystrophin and improves muscular function
Collapse
Affiliation(s)
- Mingming Zhao
- Department of Clinical Application, Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan.
| | - Atsutoshi Tazumi
- Department of Clinical Application, Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan; Asahi Kasei Co., Ltd., 1-105 Jinbo-cho, Kanda, Chiyoda-ku, Tokyo, Japan
| | - Satoru Takayama
- Department of Clinical Application, Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan; Asahi Kasei Co., Ltd., 1-105 Jinbo-cho, Kanda, Chiyoda-ku, Tokyo, Japan
| | - Nana Takenaka-Ninagawa
- Department of Clinical Application, Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Minas Nalbandian
- Department of Clinical Application, Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Miki Nagai
- Department of Clinical Application, Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Yumi Nakamura
- Department of Clinical Application, Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Masanori Nakasa
- Department of Clinical Application, Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Akira Watanabe
- Department of Life Science Frontiers, Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Makoto Ikeya
- Department of Clinical Application, Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Akitsu Hotta
- Department of Clinical Application, Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Yuta Ito
- Faculty of Rehabilitation Science, Nagoya Gakuin University, 1350 Kamishinano-cho, Seto City, Aichi 480-1298, Japan
| | - Takahiko Sato
- Department of Anatomy, Fujita Health University, 1-98 Dengakugakubo, Kutsukake-cho, Toyoake, Aichi 470-1192, Japan
| | - Hidetoshi Sakurai
- Department of Clinical Application, Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan.
| |
Collapse
|
75
|
Selvaraj S, Kyba M, Perlingeiro RCR. Pluripotent Stem Cell-Based Therapeutics for Muscular Dystrophies. Trends Mol Med 2020; 25:803-816. [PMID: 31473142 DOI: 10.1016/j.molmed.2019.07.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Revised: 06/30/2019] [Accepted: 07/08/2019] [Indexed: 02/06/2023]
Abstract
Pluripotent stem cells (PSCs) represent an attractive cell source for treating muscular dystrophies (MDs) since they easily allow for the generation of large numbers of highly regenerative myogenic progenitors. Using reprogramming technology, patient-specific PSCs have been derived for several types of MDs, and genome editing has allowed correction of mutations, opening the opportunity for their therapeutic application in an autologous transplantation setting. However, there has been limited progress on preclinical studies that validate the therapeutic potential of these gene corrected PSC-derived myogenic progenitors. In this review, we highlight the major research advances, challenges, and future prospects towards the development of PSC-based therapeutics for MDs.
Collapse
Affiliation(s)
- Sridhar Selvaraj
- Lillehei Heart Institute, Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Michael Kyba
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, USA; Stem Cell Institute, University of Minnesota, Minneapolis, MN, USA
| | - Rita C R Perlingeiro
- Lillehei Heart Institute, Department of Medicine, University of Minnesota, Minneapolis, MN, USA; Stem Cell Institute, University of Minnesota, Minneapolis, MN, USA.
| |
Collapse
|
76
|
Al Tanoury Z, Rao J, Tassy O, Gobert B, Gapon S, Garnier JM, Wagner E, Hick A, Hall A, Gussoni E, Pourquié O. Differentiation of the human PAX7-positive myogenic precursors/satellite cell lineage in vitro. Development 2020; 147:dev187344. [PMID: 32541004 PMCID: PMC7328153 DOI: 10.1242/dev.187344] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Accepted: 05/29/2020] [Indexed: 12/12/2022]
Abstract
Satellite cells (SC) are muscle stem cells that can regenerate adult muscles upon injury. Most SC originate from PAX7+ myogenic precursors set aside during development. Although myogenesis has been studied in mouse and chicken embryos, little is known about human muscle development. Here, we report the generation of human induced pluripotent stem cell (iPSC) reporter lines in which fluorescent proteins have been introduced into the PAX7 and MYOG loci. We use single cell RNA sequencing to analyze the developmental trajectory of the iPSC-derived PAX7+ myogenic precursors. We show that the PAX7+ cells generated in culture can produce myofibers and self-renew in vitro and in vivo Together, we demonstrate that cells exhibiting characteristics of human fetal satellite cells can be produced in vitro from iPSC, opening interesting avenues for muscular dystrophy cell therapy. This work provides significant insights into the development of the human myogenic lineage.
Collapse
Affiliation(s)
- Ziad Al Tanoury
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Department of Development and Stem Cells, CNRS (UMR 7104), Inserm U964, Université de Strasbourg, 67404, Illkirch Graffenstaden, France
- Department of Pathology, Brigham and Women's Hospital, 60 Fenwood Road, Boston, MA 02115, USA
- Department of Genetics, Harvard Medical School, 60 Fenwood Road, Boston, MA 02115, USA
| | - Jyoti Rao
- Department of Pathology, Brigham and Women's Hospital, 60 Fenwood Road, Boston, MA 02115, USA
- Department of Genetics, Harvard Medical School, 60 Fenwood Road, Boston, MA 02115, USA
| | - Olivier Tassy
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Department of Development and Stem Cells, CNRS (UMR 7104), Inserm U964, Université de Strasbourg, 67404, Illkirch Graffenstaden, France
| | - Bénédicte Gobert
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Department of Development and Stem Cells, CNRS (UMR 7104), Inserm U964, Université de Strasbourg, 67404, Illkirch Graffenstaden, France
- Anagenesis Biotechnologies, Parc d'innovation - BioParc 3, 850 Boulevard Sébastien Brandt, 67400 Illkirch Graffenstaden, France
| | - Svetlana Gapon
- Department of Pathology, Brigham and Women's Hospital, 60 Fenwood Road, Boston, MA 02115, USA
| | - Jean-Marie Garnier
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Department of Development and Stem Cells, CNRS (UMR 7104), Inserm U964, Université de Strasbourg, 67404, Illkirch Graffenstaden, France
| | - Erica Wagner
- Department of Pathology, Brigham and Women's Hospital, 60 Fenwood Road, Boston, MA 02115, USA
| | - Aurore Hick
- Anagenesis Biotechnologies, Parc d'innovation - BioParc 3, 850 Boulevard Sébastien Brandt, 67400 Illkirch Graffenstaden, France
| | - Arielle Hall
- Division of Genetics and Genomics, Boston Children's Hospital, 3 Blackfan Circle, CLS, Boston, MA 15021, USA
| | - Emanuela Gussoni
- Division of Genetics and Genomics, Boston Children's Hospital, 3 Blackfan Circle, CLS, Boston, MA 15021, USA
| | - Olivier Pourquié
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Department of Development and Stem Cells, CNRS (UMR 7104), Inserm U964, Université de Strasbourg, 67404, Illkirch Graffenstaden, France
- Department of Pathology, Brigham and Women's Hospital, 60 Fenwood Road, Boston, MA 02115, USA
- Department of Genetics, Harvard Medical School, 60 Fenwood Road, Boston, MA 02115, USA
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138, USA
| |
Collapse
|
77
|
Mazaleyrat K, Badja C, Broucqsault N, Chevalier R, Laberthonnière C, Dion C, Baldasseroni L, El-Yazidi C, Thomas M, Bachelier R, Altié A, Nguyen K, Lévy N, Robin JD, Magdinier F. Multilineage Differentiation for Formation of Innervated Skeletal Muscle Fibers from Healthy and Diseased Human Pluripotent Stem Cells. Cells 2020; 9:cells9061531. [PMID: 32585982 PMCID: PMC7349825 DOI: 10.3390/cells9061531] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2020] [Revised: 06/16/2020] [Accepted: 06/16/2020] [Indexed: 12/19/2022] Open
Abstract
Induced pluripotent stem cells (iPSCs) obtained by reprogramming primary somatic cells have revolutionized the fields of cell biology and disease modeling. However, the number protocols for generating mature muscle fibers with sarcolemmal organization using iPSCs remain limited, and partly mimic the complexity of mature skeletal muscle. Methods: We used a novel combination of small molecules added in a precise sequence for the simultaneous codifferentiation of human iPSCs into skeletal muscle cells and motor neurons. Results: We show that the presence of both cell types reduces the production time for millimeter-long multinucleated muscle fibers with sarcolemmal organization. Muscle fiber contractions are visible in 19–21 days, and can be maintained over long period thanks to the production of innervated multinucleated mature skeletal muscle fibers with autonomous cell regeneration of PAX7-positive cells and extracellular matrix synthesis. The sequential addition of specific molecules recapitulates key steps of human peripheral neurogenesis and myogenesis. Furthermore, this organoid-like culture can be used for functional evaluation and drug screening. Conclusion: Our protocol, which is applicable to hiPSCs from healthy individuals, was validated in Duchenne Muscular Dystrophy, Myotonic Dystrophy, Facio-Scapulo-Humeral Dystrophy and type 2A Limb-Girdle Muscular Dystrophy, opening new paths for the exploration of muscle differentiation, disease modeling and drug discovery.
Collapse
Affiliation(s)
- Kilian Mazaleyrat
- Aix-Marseille University, INSERM, MMG, Marseille Medical Genetics, 13385 Marseille, France; (K.M.); (C.B.); (N.B.); (R.C.); (C.L.); (C.D.); (L.B.); (C.E.-Y.); (M.T.); (K.N.); (N.L.); (J.D.R.)
| | - Cherif Badja
- Aix-Marseille University, INSERM, MMG, Marseille Medical Genetics, 13385 Marseille, France; (K.M.); (C.B.); (N.B.); (R.C.); (C.L.); (C.D.); (L.B.); (C.E.-Y.); (M.T.); (K.N.); (N.L.); (J.D.R.)
| | - Natacha Broucqsault
- Aix-Marseille University, INSERM, MMG, Marseille Medical Genetics, 13385 Marseille, France; (K.M.); (C.B.); (N.B.); (R.C.); (C.L.); (C.D.); (L.B.); (C.E.-Y.); (M.T.); (K.N.); (N.L.); (J.D.R.)
| | - Raphaël Chevalier
- Aix-Marseille University, INSERM, MMG, Marseille Medical Genetics, 13385 Marseille, France; (K.M.); (C.B.); (N.B.); (R.C.); (C.L.); (C.D.); (L.B.); (C.E.-Y.); (M.T.); (K.N.); (N.L.); (J.D.R.)
| | - Camille Laberthonnière
- Aix-Marseille University, INSERM, MMG, Marseille Medical Genetics, 13385 Marseille, France; (K.M.); (C.B.); (N.B.); (R.C.); (C.L.); (C.D.); (L.B.); (C.E.-Y.); (M.T.); (K.N.); (N.L.); (J.D.R.)
| | - Camille Dion
- Aix-Marseille University, INSERM, MMG, Marseille Medical Genetics, 13385 Marseille, France; (K.M.); (C.B.); (N.B.); (R.C.); (C.L.); (C.D.); (L.B.); (C.E.-Y.); (M.T.); (K.N.); (N.L.); (J.D.R.)
| | - Lyla Baldasseroni
- Aix-Marseille University, INSERM, MMG, Marseille Medical Genetics, 13385 Marseille, France; (K.M.); (C.B.); (N.B.); (R.C.); (C.L.); (C.D.); (L.B.); (C.E.-Y.); (M.T.); (K.N.); (N.L.); (J.D.R.)
| | - Claire El-Yazidi
- Aix-Marseille University, INSERM, MMG, Marseille Medical Genetics, 13385 Marseille, France; (K.M.); (C.B.); (N.B.); (R.C.); (C.L.); (C.D.); (L.B.); (C.E.-Y.); (M.T.); (K.N.); (N.L.); (J.D.R.)
| | - Morgane Thomas
- Aix-Marseille University, INSERM, MMG, Marseille Medical Genetics, 13385 Marseille, France; (K.M.); (C.B.); (N.B.); (R.C.); (C.L.); (C.D.); (L.B.); (C.E.-Y.); (M.T.); (K.N.); (N.L.); (J.D.R.)
| | - Richard Bachelier
- Aix-Marseille University, INSERM, INRA, C2VN, 13385 Marseille, France; (R.B.); (A.A.)
| | - Alexandre Altié
- Aix-Marseille University, INSERM, INRA, C2VN, 13385 Marseille, France; (R.B.); (A.A.)
| | - Karine Nguyen
- Aix-Marseille University, INSERM, MMG, Marseille Medical Genetics, 13385 Marseille, France; (K.M.); (C.B.); (N.B.); (R.C.); (C.L.); (C.D.); (L.B.); (C.E.-Y.); (M.T.); (K.N.); (N.L.); (J.D.R.)
- APHM, Département de Génétique Médicale, Hôpital de la Timone Enfants, 13385 Marseille, France
| | - Nicolas Lévy
- Aix-Marseille University, INSERM, MMG, Marseille Medical Genetics, 13385 Marseille, France; (K.M.); (C.B.); (N.B.); (R.C.); (C.L.); (C.D.); (L.B.); (C.E.-Y.); (M.T.); (K.N.); (N.L.); (J.D.R.)
- APHM, Département de Génétique Médicale, Hôpital de la Timone Enfants, 13385 Marseille, France
| | - Jérôme D. Robin
- Aix-Marseille University, INSERM, MMG, Marseille Medical Genetics, 13385 Marseille, France; (K.M.); (C.B.); (N.B.); (R.C.); (C.L.); (C.D.); (L.B.); (C.E.-Y.); (M.T.); (K.N.); (N.L.); (J.D.R.)
| | - Frédérique Magdinier
- Aix-Marseille University, INSERM, MMG, Marseille Medical Genetics, 13385 Marseille, France; (K.M.); (C.B.); (N.B.); (R.C.); (C.L.); (C.D.); (L.B.); (C.E.-Y.); (M.T.); (K.N.); (N.L.); (J.D.R.)
- Correspondence:
| |
Collapse
|
78
|
Baci D, Chirivì M, Pace V, Maiullari F, Milan M, Rampin A, Somma P, Presutti D, Garavelli S, Bruno A, Cannata S, Lanzuolo C, Gargioli C, Rizzi R, Bearzi C. Extracellular Vesicles from Skeletal Muscle Cells Efficiently Promote Myogenesis in Induced Pluripotent Stem Cells. Cells 2020; 9:cells9061527. [PMID: 32585911 PMCID: PMC7349204 DOI: 10.3390/cells9061527] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2020] [Revised: 06/13/2020] [Accepted: 06/15/2020] [Indexed: 12/11/2022] Open
Abstract
The recent advances, offered by cell therapy in the regenerative medicine field, offer a revolutionary potential for the development of innovative cures to restore compromised physiological functions or organs. Adult myogenic precursors, such as myoblasts or satellite cells, possess a marked regenerative capacity, but the exploitation of this potential still encounters significant challenges in clinical application, due to low rate of proliferation in vitro, as well as a reduced self-renewal capacity. In this scenario, induced pluripotent stem cells (iPSCs) can offer not only an inexhaustible source of cells for regenerative therapeutic approaches, but also a valuable alternative for in vitro modeling of patient-specific diseases. In this study we established a reliable protocol to induce the myogenic differentiation of iPSCs, generated from pericytes and fibroblasts, exploiting skeletal muscle-derived extracellular vesicles (EVs), in combination with chemically defined factors. This genetic integration-free approach generates functional skeletal myotubes maintaining the engraftment ability in vivo. Our results demonstrate evidence that EVs can act as biological "shuttles" to deliver specific bioactive molecules for a successful transgene-free differentiation offering new opportunities for disease modeling and regenerative approaches.
Collapse
Affiliation(s)
- Denisa Baci
- Institute of Biochemistry and Cell Biology, National Research Council, 00015 Rome, Italy; (D.B.); (M.C.); (V.P.); (M.M.); (A.R.); (D.P.)
- Department of Biotechnology and Life Sciences, University of Insubria, 21100 Varese, Italy
| | - Maila Chirivì
- Institute of Biochemistry and Cell Biology, National Research Council, 00015 Rome, Italy; (D.B.); (M.C.); (V.P.); (M.M.); (A.R.); (D.P.)
| | - Valentina Pace
- Institute of Biochemistry and Cell Biology, National Research Council, 00015 Rome, Italy; (D.B.); (M.C.); (V.P.); (M.M.); (A.R.); (D.P.)
| | | | - Marika Milan
- Institute of Biochemistry and Cell Biology, National Research Council, 00015 Rome, Italy; (D.B.); (M.C.); (V.P.); (M.M.); (A.R.); (D.P.)
| | - Andrea Rampin
- Institute of Biochemistry and Cell Biology, National Research Council, 00015 Rome, Italy; (D.B.); (M.C.); (V.P.); (M.M.); (A.R.); (D.P.)
| | - Paolo Somma
- Flow Cytometry Core, Humanitas Clinical and Research Center, 20089 Milan, Italy;
| | - Dario Presutti
- Institute of Biochemistry and Cell Biology, National Research Council, 00015 Rome, Italy; (D.B.); (M.C.); (V.P.); (M.M.); (A.R.); (D.P.)
| | - Silvia Garavelli
- Institute for Endocrinology and Oncology “Gaetano Salvatore”, National Research Council, 80131 Naples, Italy;
| | | | - Stefano Cannata
- Department of Biology, University of Rome Tor Vergata, 00133 Rome, Italy; (S.C.); (C.G.)
| | - Chiara Lanzuolo
- Institute of Biomedical Technologies, National Research Council, 20090 Milan, Italy;
- Fondazione Istituto Nazionale di Genetica Molecolare, 20122 Milan, Italy
| | - Cesare Gargioli
- Department of Biology, University of Rome Tor Vergata, 00133 Rome, Italy; (S.C.); (C.G.)
| | - Roberto Rizzi
- Institute of Biomedical Technologies, National Research Council, 20090 Milan, Italy;
- Fondazione Istituto Nazionale di Genetica Molecolare, 20122 Milan, Italy
- Correspondence: (R.R.); (C.B.); Tel.: +39-02-0066-0230 (R.R.); +39-02-0066-0230 (C.B.)
| | - Claudia Bearzi
- Institute of Biochemistry and Cell Biology, National Research Council, 00015 Rome, Italy; (D.B.); (M.C.); (V.P.); (M.M.); (A.R.); (D.P.)
- Fondazione Istituto Nazionale di Genetica Molecolare, 20122 Milan, Italy
- Correspondence: (R.R.); (C.B.); Tel.: +39-02-0066-0230 (R.R.); +39-02-0066-0230 (C.B.)
| |
Collapse
|
79
|
Sun C, Choi IY, Gonzalez YIR, Andersen P, Talbot CC, Iyer SR, Lovering RM, Wagner KR, Lee G. Duchenne muscular dystrophy hiPSC-derived myoblast drug screen identifies compounds that ameliorate disease in mdx mice. JCI Insight 2020; 5:134287. [PMID: 32343677 PMCID: PMC7308059 DOI: 10.1172/jci.insight.134287] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Accepted: 04/23/2020] [Indexed: 12/18/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is the most common muscular dystrophy. In the present study, when human induced pluripotent stem cells (hiPSCs) were differentiated into myoblasts, the myoblasts derived from DMD patient hiPSCs (DMD hiPSC-derived myoblasts) exhibited an identifiable DMD-relevant phenotype: myogenic fusion deficiency. Based on this model, we developed a DMD hiPSC-derived myoblast screening platform employing a high-content imaging (BD Pathway 855) approach to generate parameters describing morphological as well as myogenic marker protein expression. Following treatment of the cells with 1524 compounds from the Johns Hopkins Clinical Compound Library, compounds that enhanced myogenic fusion of DMD hiPSC-derived myoblasts were identified. The final hits were ginsenoside Rd and fenofibrate. Transcriptional profiling revealed that ginsenoside Rd is functionally related to FLT3 signaling, while fenofibrate is linked to TGF-β signaling. Preclinical tests in mdx mice showed that treatment with these 2 hit compounds can significantly ameliorate some of the skeletal muscle phenotypes caused by dystrophin deficiency, supporting their therapeutic potential. Further study revealed that fenofibrate could inhibit mitochondrion-induced apoptosis in DMD hiPSC-derived cardiomyocytes. We have developed a platform based on DMD hiPSC-derived myoblasts for drug screening and identified 2 promising small molecules with in vivo efficacy.
Collapse
Affiliation(s)
- Congshan Sun
- Departments of Neurology and Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Center for Genetic Muscle Disorders, Hugo W. Moser Research Institute at Kennedy Krieger Institute, Baltimore, Maryland, USA
| | | | - Yazmin I. Rovira Gonzalez
- Center for Genetic Muscle Disorders, Hugo W. Moser Research Institute at Kennedy Krieger Institute, Baltimore, Maryland, USA
- Cellular and Molecular Medicine Graduate Program, and
| | - Peter Andersen
- Institute for Cell Engineering
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - C. Conover Talbot
- The Johns Hopkins School of Medicine Institute for Basic Biomedical Sciences, Baltimore, Maryland, USA
| | | | - Richard M. Lovering
- Department of Orthopaedics and
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Kathryn R. Wagner
- Departments of Neurology and Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Center for Genetic Muscle Disorders, Hugo W. Moser Research Institute at Kennedy Krieger Institute, Baltimore, Maryland, USA
| | - Gabsang Lee
- Departments of Neurology and Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Institute for Cell Engineering
| |
Collapse
|
80
|
Chemello F, Bassel-Duby R, Olson EN. Correction of muscular dystrophies by CRISPR gene editing. J Clin Invest 2020; 130:2766-2776. [PMID: 32478678 PMCID: PMC7259998 DOI: 10.1172/jci136873] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Muscular dystrophies are debilitating disorders that result in progressive weakness and degeneration of skeletal muscle. Although the genetic mutations and clinical abnormalities of a variety of neuromuscular diseases are well known, no curative therapies have been developed to date. The advent of genome editing technology provides new opportunities to correct the underlying mutations responsible for many monogenic neuromuscular diseases. For example, Duchenne muscular dystrophy, which is caused by mutations in the dystrophin gene, has been successfully corrected in mice, dogs, and human cells through CRISPR/Cas9 editing. In this Review, we focus on the potential for, and challenges of, correcting muscular dystrophies by editing disease-causing mutations at the genomic level. Ideally, because muscle tissues are extremely long-lived, CRISPR technology could offer a one-time treatment for muscular dystrophies by correcting the culprit genomic mutations and enabling normal expression of the repaired gene.
Collapse
|
81
|
Judson RN, Rossi FMV. Towards stem cell therapies for skeletal muscle repair. NPJ Regen Med 2020; 5:10. [PMID: 32411395 PMCID: PMC7214464 DOI: 10.1038/s41536-020-0094-3] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Accepted: 03/31/2020] [Indexed: 01/23/2023] Open
Abstract
Skeletal muscle is an ideal target for cell therapy. The use of its potent stem cell population in the form of autologous intramuscular transplantation represents a tantalizing strategy to slow the progression of congenital muscle diseases (such as Duchenne Muscular Dystrophy) or regenerate injured tissue following trauma. The syncytial nature of skeletal muscle uniquely permits the engraftment of stem/progenitor cells to contribute to new myonuclei and restore the expression of genes mutated in myopathies. Historically however, the implementation of this approach has been significantly limited by the inability to expand undifferentiated muscle stem cells (MuSCs) in culture whilst maintaining transplantation potential. This is crucial, as MuSC expansion and/or genetic manipulation is likely necessary for therapeutic applications. In this article, we review recent studies that have provided a number of important breakthroughs to tackle this problem. Progress towards this goal has been achieved by exploiting biochemical, biophysical and developmental paradigms to construct innovative in vitro strategies that are guiding stem cell therapies for muscle repair towards the clinic.
Collapse
Affiliation(s)
- Robert N Judson
- 1STEMCELL Technologies Inc, Vancouver, BC Canada.,2Biomedical Research Centre, Department of Medical Genetics, University of British Columbia, 2222 Health Sciences Mall, Vancouver, BC Canada
| | - Fabio M V Rossi
- 2Biomedical Research Centre, Department of Medical Genetics, University of British Columbia, 2222 Health Sciences Mall, Vancouver, BC Canada
| |
Collapse
|
82
|
Caputo L, Granados A, Lenzi J, Rosa A, Ait-Si-Ali S, Puri PL, Albini S. Acute conversion of patient-derived Duchenne muscular dystrophy iPSC into myotubes reveals constitutive and inducible over-activation of TGFβ-dependent pro-fibrotic signaling. Skelet Muscle 2020; 10:13. [PMID: 32359374 PMCID: PMC7195779 DOI: 10.1186/s13395-020-00224-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2019] [Accepted: 02/24/2020] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND In Duchenne muscular dystrophy (DMD), DYSTROPHIN deficiency exposes myofibers to repeated cycles of contraction/degeneration, ultimately leading to muscle loss and replacement by fibrotic tissue. DMD pathology is typically exacerbated by excessive secretion of TGFβ and consequent accumulation of pro-fibrotic components of the extra-cellular matrix (ECM), which in turn impairs compensatory regeneration and complicates the efficacy of therapeutic strategies. It is currently unclear whether DMD skeletal muscle fibers directly contribute to excessive activation of TGFβ. Development of skeletal myofibers from DMD patient-derived induced pluripotent stem cells (iPSC), as an "in dish" model of disease, can be exploited to determine the myofiber contribution to pathogenic TGFβ signaling in DMD and might provide a screening platform for the identification of anti-fibrotic interventions in DMD. METHODS We describe a rapid and efficient method for the generation of contractile human skeletal muscle cells from DMD patient-derived hiPSC, based on the inducible expression of MyoD and BAF60C (encoded by SMARCD3 gene), using an enhanced version of piggyBac (epB) transposone vectors. DMD iPSC-derived myotubes were tested as an "in dish" disease model and exposed to environmental and mechanical cues that recapitulate salient pathological features of DMD. RESULTS We show that DMD iPSC-derived myotubes exhibit a constitutive activation of TGFβ-SMAD2/3 signaling. High-content screening (HCS)-based quantification of nuclear phosphorylated SMAD2/3 signal revealed that DMD iPSC-derived myotubes also exhibit increased activation of the TGFβ-SMAD2/3 signaling following exposure to either recombinant TGFβ or electrical pacing-induced contraction. CONCLUSIONS Acute conversion of DMD patient-derived iPSC into skeletal muscles, by the ectopic expression of MyoD and BAF60C, provides a rapid and reliable protocol for an "in dish" DMD model that recapitulates key pathogenic features of disease pathology, such as the constitutive activation of the TGFβ/SMAD signaling as well as the deregulated response to pathogenic stimuli, e.g., ECM-derived signals or mechanical cues. Thus, this model is suitable for the identification of new therapeutic targets in DMD patient-specific muscles.
Collapse
Affiliation(s)
- Luca Caputo
- Development, Aging and Regeneration Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, 92037, USA
| | - Alice Granados
- Epigenetics and Cell Fate (EDC), Centre National de la Recherche Scientifique (CNRS), Université Paris Diderot, Université de Paris, F-75013, Paris, France
| | - Jessica Lenzi
- Department of Biology and Biotechnology Charles Darwin, Sapienza University of Rome, P.le Aldo Moro 5, Rome, Italy
| | - Alessandro Rosa
- Department of Biology and Biotechnology Charles Darwin, Sapienza University of Rome, P.le Aldo Moro 5, Rome, Italy.,Center for Life Nano Science, Istituto Italiano di Tecnologia, Viale Regina Elena 291, 00161, Rome, Italy
| | - Slimane Ait-Si-Ali
- Epigenetics and Cell Fate (EDC), Centre National de la Recherche Scientifique (CNRS), Université Paris Diderot, Université de Paris, F-75013, Paris, France
| | - Pier Lorenzo Puri
- Development, Aging and Regeneration Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, 92037, USA.
| | - Sonia Albini
- Epigenetics and Cell Fate (EDC), Centre National de la Recherche Scientifique (CNRS), Université Paris Diderot, Université de Paris, F-75013, Paris, France. .,Genethon, 1bis, Rue de l'Internationale, 91000, EVRY, France.
| |
Collapse
|
83
|
Vermersch E, Jouve C, Hulot JS. CRISPR/Cas9 gene-editing strategies in cardiovascular cells. Cardiovasc Res 2020; 116:894-907. [PMID: 31584620 DOI: 10.1093/cvr/cvz250] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Revised: 05/05/2019] [Accepted: 09/26/2019] [Indexed: 01/05/2024] Open
Abstract
Cardiovascular diseases are among the main causes of morbidity and mortality in Western countries and considered as a leading public health issue. Therefore, there is a strong need for new disease models to support the development of novel therapeutics approaches. The successive improvement of genome editing tools with zinc finger nucleases (ZFNs), transcription activator-like effector nucleases (TALENs), and more recently with clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated 9 (Cas9) has enabled the generation of genetically modified cells and organisms with much greater efficiency and precision than before. The simplicity of CRISPR/Cas9 technology made it especially suited for different studies, both in vitro and in vivo, and has been used in multiple studies evaluating gene functions, disease modelling, transcriptional regulation, and testing of novel therapeutic approaches. Notably, with the parallel development of human induced pluripotent stem cells (hiPSCs), the generation of knock-out and knock-in human cell lines significantly increased our understanding of mutation impacts and physiopathological mechanisms within the cardiovascular domain. Here, we review the recent development of CRISPR-Cas9 genome editing, the alternative tools, the available strategies to conduct genome editing in cardiovascular cells with a focus on its use for correcting mutations in vitro and in vivo both in germ and somatic cells. We will also highlight that, despite its potential, CRISPR/Cas9 technology comes with important technical and ethical limitations. The development of CRISPR/Cas9 genome editing for cardiovascular diseases indeed requires to develop a specific strategy in order to optimize the design of the genome editing tools, the manipulation of DNA repair mechanisms, the packaging and delivery of the tools to the studied organism, and the assessment of their efficiency and safety.
Collapse
Affiliation(s)
- Eva Vermersch
- Paris Cardiovascular Research Center PARCC, Université de Paris, INSERM, 56 Rue Leblanc, 75015 Paris, France
| | - Charlène Jouve
- Paris Cardiovascular Research Center PARCC, Université de Paris, INSERM, 56 Rue Leblanc, 75015 Paris, France
| | - Jean-Sébastien Hulot
- Paris Cardiovascular Research Center PARCC, Université de Paris, INSERM, 56 Rue Leblanc, 75015 Paris, France
- Centre d'Investigations Cliniques CIC1418, AP-HP, Hôpital Européen Georges Pompidou, 75015 Paris, France
| |
Collapse
|
84
|
Saha M, Rizzo SA, Ramanathan M, Hightower RM, Santostefano KE, Terada N, Finkel RS, Berg JS, Chahin N, Pacak CA, Wagner RE, Alexander MS, Draper I, Kang PB. Selective serotonin reuptake inhibitors ameliorate MEGF10 myopathy. Hum Mol Genet 2020; 28:2365-2377. [PMID: 31267131 DOI: 10.1093/hmg/ddz064] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Revised: 03/18/2019] [Accepted: 03/21/2019] [Indexed: 02/02/2023] Open
Abstract
MEGF10 myopathy is a rare inherited muscle disease that is named after the causative gene, MEGF10. The classic phenotype, early onset myopathy, areflexia, respiratory distress and dysphagia, is severe and immediately life-threatening. There are no disease-modifying therapies. We performed a small molecule screen and follow-up studies to seek a novel therapy. A primary in vitro drug screen assessed cellular proliferation patterns in Megf10-deficient myoblasts. Secondary evaluations were performed on primary screen hits using myoblasts derived from Megf10-/- mice, induced pluripotent stem cell-derived myoblasts from MEGF10 myopathy patients, mutant Drosophila that are deficient in the homologue of MEGF10 (Drpr) and megf10 mutant zebrafish. The screen yielded two promising candidates that are both selective serotonin reuptake inhibitors (SSRIs), sertraline and escitalopram. In depth follow-up analyses demonstrated that sertraline was highly effective in alleviating abnormalities across multiple models of the disease including mouse myoblast, human myoblast, Drosophila and zebrafish models. Sertraline also restored deficiencies of Notch1 in disease models. We conclude that SSRIs show promise as potential therapeutic compounds for MEGF10 myopathy, especially sertraline. The mechanism of action may involve the Notch pathway.
Collapse
Affiliation(s)
- Madhurima Saha
- Division of Pediatric Neurology, Department of Pediatrics, University of Florida College of Medicine, Gainesville, FL, USA
| | - Skylar A Rizzo
- Division of Pediatric Neurology, Department of Pediatrics, University of Florida College of Medicine, Gainesville, FL, USA.,Medosome Biotec, Alachua, FL, USA
| | - Manashwi Ramanathan
- Division of Pediatric Neurology, Department of Pediatrics, University of Florida College of Medicine, Gainesville, FL, USA
| | - Rylie M Hightower
- Department of Pediatrics, Division of Pediatric Neurology, Children's of Alabama and the University of Alabama at Birmingham, Birmingham, AL, USA.,University of Alabama Birmingham, Center for Exercise Medicine Birmingham, AL, USA
| | - Katherine E Santostefano
- Center for Cellular Reprogramming, Department of Pathology, Immunology and Laboratory Medicine, University of Florida College of Medicine, Gainesville, FL, USA
| | - Naohiro Terada
- Center for Cellular Reprogramming, Department of Pathology, Immunology and Laboratory Medicine, University of Florida College of Medicine, Gainesville, FL, USA
| | - Richard S Finkel
- Division of Pediatric Neurology, Nemours Children's Hospital, Orlando, FL, USA
| | - Jonathan S Berg
- Department of Genetics, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - Nizar Chahin
- Department of Neurology, Neuromuscular Division, Oregon Health and Science University, Portland, Oregon, USA
| | - Christina A Pacak
- Department of Pediatrics, University of Florida College of Medicine, Gainesville, FL, USA
| | | | - Matthew S Alexander
- Department of Pediatrics, Division of Pediatric Neurology, Children's of Alabama and the University of Alabama at Birmingham, Birmingham, AL, USA.,University of Alabama Birmingham, Center for Exercise Medicine Birmingham, AL, USA.,Department of Genetics, University of Alabama Birmingham, Birmingham, AL, USA.,Civitan International Research Center at University of Alabama Birmingham, Birmingham, AL, USA
| | - Isabelle Draper
- Department of Medicine, Tufts Medical Center, Molecular Cardiology Research Institute, Boston, MA, USA
| | - Peter B Kang
- Division of Pediatric Neurology, Department of Pediatrics, University of Florida College of Medicine, Gainesville, FL, USA.,Department of Molecular Genetics and Microbiology and Department of Neurology, University of Florida College of Medicine, Gainesville, FL, USA.,Genetics Institute and Myology Institute, University of Florida, Gainesville, FL, USA
| |
Collapse
|
85
|
Borok MJ, Mademtzoglou D, Relaix F. Bu-M-P-ing Iron: How BMP Signaling Regulates Muscle Growth and Regeneration. J Dev Biol 2020; 8:jdb8010004. [PMID: 32053985 PMCID: PMC7151139 DOI: 10.3390/jdb8010004] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Revised: 02/05/2020] [Accepted: 02/07/2020] [Indexed: 12/16/2022] Open
Abstract
The bone morphogenetic protein (BMP) pathway is best known for its role in promoting bone formation, however it has been shown to play important roles in both development and regeneration of many different tissues. Recent work has shown that the BMP proteins have a number of functions in skeletal muscle, from embryonic to postnatal development. Furthermore, complementary studies have recently demonstrated that specific components of the pathway are required for efficient muscle regeneration.
Collapse
Affiliation(s)
- Matthew J Borok
- Inserm, IMRB U955-E10, 94010 Créteil, France; (M.J.B.); (D.M.)
- Faculté de santé, Université Paris Est, 94000 Creteil, France
| | - Despoina Mademtzoglou
- Inserm, IMRB U955-E10, 94010 Créteil, France; (M.J.B.); (D.M.)
- Faculté de santé, Université Paris Est, 94000 Creteil, France
| | - Frederic Relaix
- Inserm, IMRB U955-E10, 94010 Créteil, France; (M.J.B.); (D.M.)
- Faculté de santé, Université Paris Est, 94000 Creteil, France
- Ecole Nationale Veterinaire d’Alfort, 94700 Maison Alfort, France
- Etablissement Français du Sang, 94017 Créteil, France
- APHP, Hopitaux Universitaires Henri Mondor, DHU Pepsy & Centre de Référence des Maladies Neuromusculaires GNMH, 94000 Créteil, France
- Correspondence: ; Tel.: +33-149-813-940
| |
Collapse
|
86
|
Wilkinson IVL, Perkins KJ, Dugdale H, Moir L, Vuorinen A, Chatzopoulou M, Squire SE, Monecke S, Lomow A, Geese M, Charles PD, Burch P, Tinsley JM, Wynne GM, Davies SG, Wilson FX, Rastinejad F, Mohammed S, Davies KE, Russell AJ. Chemical Proteomics and Phenotypic Profiling Identifies the Aryl Hydrocarbon Receptor as a Molecular Target of the Utrophin Modulator Ezutromid. Angew Chem Int Ed Engl 2020. [DOI: 10.1002/ange.201912392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Affiliation(s)
- Isabel V. L. Wilkinson
- Department of ChemistryUniversity of OxfordChemistry Research Laboratory Mansfield Road Oxford OX1 3TA UK
| | - Kelly J. Perkins
- Department of Physiology, Anatomy and GeneticsUniversity of OxfordSir Henry Wellcome Building of Gene Function South Parks Road Oxford OX1 3PT UK
| | - Hannah Dugdale
- Department of Physiology, Anatomy and GeneticsUniversity of OxfordSir Henry Wellcome Building of Gene Function South Parks Road Oxford OX1 3PT UK
| | - Lee Moir
- Department of Physiology, Anatomy and GeneticsUniversity of OxfordSir Henry Wellcome Building of Gene Function South Parks Road Oxford OX1 3PT UK
| | - Aini Vuorinen
- Department of ChemistryUniversity of OxfordChemistry Research Laboratory Mansfield Road Oxford OX1 3TA UK
| | - Maria Chatzopoulou
- Department of ChemistryUniversity of OxfordChemistry Research Laboratory Mansfield Road Oxford OX1 3TA UK
| | - Sarah E. Squire
- Department of Physiology, Anatomy and GeneticsUniversity of OxfordSir Henry Wellcome Building of Gene Function South Parks Road Oxford OX1 3PT UK
| | - Sebastian Monecke
- Evotec International GmbHManfred Eigen Campus Essener Bogen 7 22419 Hamburg Germany
| | - Alexander Lomow
- Evotec International GmbHManfred Eigen Campus Essener Bogen 7 22419 Hamburg Germany
| | - Marcus Geese
- Evotec International GmbHManfred Eigen Campus Essener Bogen 7 22419 Hamburg Germany
| | - Philip D. Charles
- Department of BiochemistryUniversity of Oxford South Parks Rd Oxford OX1 3QU UK
- Target Discovery InstituteUniversity of OxfordOld Road Campus Roosevelt Drive Oxford OX3 7FZ UK
| | - Peter Burch
- Summit Therapeutics plc. 136a Eastern Avenue, Milton Park Abingdon Oxfordshire OX14 4SB UK
| | - Jonathan M. Tinsley
- Summit Therapeutics plc. 136a Eastern Avenue, Milton Park Abingdon Oxfordshire OX14 4SB UK
| | - Graham M. Wynne
- Department of ChemistryUniversity of OxfordChemistry Research Laboratory Mansfield Road Oxford OX1 3TA UK
| | - Stephen G. Davies
- Department of ChemistryUniversity of OxfordChemistry Research Laboratory Mansfield Road Oxford OX1 3TA UK
| | - Francis X. Wilson
- Summit Therapeutics plc. 136a Eastern Avenue, Milton Park Abingdon Oxfordshire OX14 4SB UK
| | - Fraydoon Rastinejad
- Target Discovery InstituteUniversity of OxfordOld Road Campus Roosevelt Drive Oxford OX3 7FZ UK
| | - Shabaz Mohammed
- Department of ChemistryUniversity of OxfordChemistry Research Laboratory Mansfield Road Oxford OX1 3TA UK
- Department of BiochemistryUniversity of Oxford South Parks Rd Oxford OX1 3QU UK
| | - Kay E. Davies
- Department of Physiology, Anatomy and GeneticsUniversity of OxfordSir Henry Wellcome Building of Gene Function South Parks Road Oxford OX1 3PT UK
| | - Angela J. Russell
- Department of ChemistryUniversity of OxfordChemistry Research Laboratory Mansfield Road Oxford OX1 3TA UK
- Department of PharmacologyUniversity of Oxford Mansfield Road Oxford OX1 3PQ UK
| |
Collapse
|
87
|
Choi IY, Lim H, Cho HJ, Oh Y, Chou BK, Bai H, Cheng L, Kim YJ, Hyun S, Kim H, Shin JH, Lee G. Transcriptional landscape of myogenesis from human pluripotent stem cells reveals a key role of TWIST1 in maintenance of skeletal muscle progenitors. eLife 2020; 9:e46981. [PMID: 32011235 PMCID: PMC6996923 DOI: 10.7554/elife.46981] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Accepted: 01/14/2020] [Indexed: 12/15/2022] Open
Abstract
Generation of skeletal muscle cells with human pluripotent stem cells (hPSCs) opens new avenues for deciphering essential, but poorly understood aspects of transcriptional regulation in human myogenic specification. In this study, we characterized the transcriptional landscape of distinct human myogenic stages, including OCT4::EGFP+ pluripotent stem cells, MSGN1::EGFP+ presomite cells, PAX7::EGFP+ skeletal muscle progenitor cells, MYOG::EGFP+ myoblasts, and multinucleated myotubes. We defined signature gene expression profiles from each isolated cell population with unbiased clustering analysis, which provided unique insights into the transcriptional dynamics of human myogenesis from undifferentiated hPSCs to fully differentiated myotubes. Using a knock-out strategy, we identified TWIST1 as a critical factor in maintenance of human PAX7::EGFP+ putative skeletal muscle progenitor cells. Our data revealed a new role of TWIST1 in human skeletal muscle progenitors, and we have established a foundation to identify transcriptional regulations of human myogenic ontogeny (online database can be accessed in http://www.myogenesis.net/).
Collapse
Affiliation(s)
- In Young Choi
- The Institute for Cell EngineeringJohns Hopkins University, School of MedicineBaltimoreUnited States
- Department of Medicine, Graduate SchoolKyung Hee UniversitySeoulRepublic of Korea
| | - Hotae Lim
- The Institute for Cell EngineeringJohns Hopkins University, School of MedicineBaltimoreUnited States
- College of Veterinary MedicineChungbuk National UniversityChungbukRepublic of Korea
| | - Hyeon Jin Cho
- Lieber Institute for Brain Development, Johns Hopkins Medical CampusBaltimoreUnited States
| | - Yohan Oh
- The Institute for Cell EngineeringJohns Hopkins University, School of MedicineBaltimoreUnited States
| | - Bin-Kuan Chou
- The Institute for Cell EngineeringJohns Hopkins University, School of MedicineBaltimoreUnited States
- Division of Hematology, Department of MedicineJohns Hopkins University, School of MedicineBaltimoreUnited States
| | - Hao Bai
- The Institute for Cell EngineeringJohns Hopkins University, School of MedicineBaltimoreUnited States
- Division of Hematology, Department of MedicineJohns Hopkins University, School of MedicineBaltimoreUnited States
| | - Linzhao Cheng
- Division of Hematology, Department of MedicineJohns Hopkins University, School of MedicineBaltimoreUnited States
| | - Yong Jun Kim
- Department of Pathololgy, College of MedicineKyung Hee UniversitySeoulRepublic of Korea
| | - SangHwan Hyun
- The Institute for Cell EngineeringJohns Hopkins University, School of MedicineBaltimoreUnited States
- College of Veterinary MedicineChungbuk National UniversityChungbukRepublic of Korea
| | - Hyesoo Kim
- The Institute for Cell EngineeringJohns Hopkins University, School of MedicineBaltimoreUnited States
- Department of NeurologyJohns Hopkins University, School of MedicineBaltimoreUnited States
| | - Joo Heon Shin
- Lieber Institute for Brain Development, Johns Hopkins Medical CampusBaltimoreUnited States
| | - Gabsang Lee
- The Institute for Cell EngineeringJohns Hopkins University, School of MedicineBaltimoreUnited States
- Department of NeurologyJohns Hopkins University, School of MedicineBaltimoreUnited States
- The Solomon H. Synder Department of NeuroscienceJohns Hopkins University, School of MedicineBaltimoreUnited States
| |
Collapse
|
88
|
Wilkinson IVL, Perkins KJ, Dugdale H, Moir L, Vuorinen A, Chatzopoulou M, Squire SE, Monecke S, Lomow A, Geese M, Charles PD, Burch P, Tinsley JM, Wynne GM, Davies SG, Wilson FX, Rastinejad F, Mohammed S, Davies KE, Russell AJ. Chemical Proteomics and Phenotypic Profiling Identifies the Aryl Hydrocarbon Receptor as a Molecular Target of the Utrophin Modulator Ezutromid. Angew Chem Int Ed Engl 2020; 59:2420-2428. [PMID: 31755636 PMCID: PMC7003794 DOI: 10.1002/anie.201912392] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 11/13/2019] [Indexed: 12/20/2022]
Abstract
Duchenne muscular dystrophy (DMD) is a fatal muscle-wasting disease arising from mutations in the dystrophin gene. Upregulation of utrophin to compensate for the missing dystrophin offers a potential therapy independent of patient genotype. The first-in-class utrophin modulator ezutromid/SMT C1100 was developed from a phenotypic screen through to a Phase 2 clinical trial. Promising efficacy and evidence of target engagement was observed in DMD patients after 24 weeks of treatment, however trial endpoints were not met after 48 weeks. The objective of this study was to understand the mechanism of action of ezutromid which could explain the lack of sustained efficacy and help development of new generations of utrophin modulators. Using chemical proteomics and phenotypic profiling we show that the aryl hydrocarbon receptor (AhR) is a target of ezutromid. Several lines of evidence demonstrate that ezutromid binds AhR with an apparent KD of 50 nm and behaves as an AhR antagonist. Furthermore, other reported AhR antagonists also upregulate utrophin, showing that this pathway, which is currently being explored in other clinical applications including oncology and rheumatoid arthritis, could also be exploited in future DMD therapies.
Collapse
Affiliation(s)
- Isabel V. L. Wilkinson
- Department of ChemistryUniversity of OxfordChemistry Research LaboratoryMansfield RoadOxfordOX1 3TAUK
| | - Kelly J. Perkins
- Department of Physiology, Anatomy and GeneticsUniversity of OxfordSir Henry Wellcome Building of Gene FunctionSouth Parks RoadOxfordOX1 3PTUK
| | - Hannah Dugdale
- Department of Physiology, Anatomy and GeneticsUniversity of OxfordSir Henry Wellcome Building of Gene FunctionSouth Parks RoadOxfordOX1 3PTUK
| | - Lee Moir
- Department of Physiology, Anatomy and GeneticsUniversity of OxfordSir Henry Wellcome Building of Gene FunctionSouth Parks RoadOxfordOX1 3PTUK
| | - Aini Vuorinen
- Department of ChemistryUniversity of OxfordChemistry Research LaboratoryMansfield RoadOxfordOX1 3TAUK
| | - Maria Chatzopoulou
- Department of ChemistryUniversity of OxfordChemistry Research LaboratoryMansfield RoadOxfordOX1 3TAUK
| | - Sarah E. Squire
- Department of Physiology, Anatomy and GeneticsUniversity of OxfordSir Henry Wellcome Building of Gene FunctionSouth Parks RoadOxfordOX1 3PTUK
| | - Sebastian Monecke
- Evotec International GmbHManfred Eigen CampusEssener Bogen 722419HamburgGermany
| | - Alexander Lomow
- Evotec International GmbHManfred Eigen CampusEssener Bogen 722419HamburgGermany
| | - Marcus Geese
- Evotec International GmbHManfred Eigen CampusEssener Bogen 722419HamburgGermany
| | - Philip D. Charles
- Department of BiochemistryUniversity of OxfordSouth Parks RdOxfordOX1 3QUUK
- Target Discovery InstituteUniversity of OxfordOld Road CampusRoosevelt DriveOxfordOX3 7FZUK
| | - Peter Burch
- Summit Therapeutics plc.136a Eastern Avenue, Milton ParkAbingdonOxfordshireOX14 4SBUK
| | - Jonathan M. Tinsley
- Summit Therapeutics plc.136a Eastern Avenue, Milton ParkAbingdonOxfordshireOX14 4SBUK
| | - Graham M. Wynne
- Department of ChemistryUniversity of OxfordChemistry Research LaboratoryMansfield RoadOxfordOX1 3TAUK
| | - Stephen G. Davies
- Department of ChemistryUniversity of OxfordChemistry Research LaboratoryMansfield RoadOxfordOX1 3TAUK
| | - Francis X. Wilson
- Summit Therapeutics plc.136a Eastern Avenue, Milton ParkAbingdonOxfordshireOX14 4SBUK
| | - Fraydoon Rastinejad
- Target Discovery InstituteUniversity of OxfordOld Road CampusRoosevelt DriveOxfordOX3 7FZUK
| | - Shabaz Mohammed
- Department of ChemistryUniversity of OxfordChemistry Research LaboratoryMansfield RoadOxfordOX1 3TAUK
- Department of BiochemistryUniversity of OxfordSouth Parks RdOxfordOX1 3QUUK
| | - Kay E. Davies
- Department of Physiology, Anatomy and GeneticsUniversity of OxfordSir Henry Wellcome Building of Gene FunctionSouth Parks RoadOxfordOX1 3PTUK
| | - Angela J. Russell
- Department of ChemistryUniversity of OxfordChemistry Research LaboratoryMansfield RoadOxfordOX1 3TAUK
- Department of PharmacologyUniversity of OxfordMansfield RoadOxfordOX1 3PQUK
| |
Collapse
|
89
|
Moretti A, Fonteyne L, Giesert F, Hoppmann P, Meier AB, Bozoglu T, Baehr A, Schneider CM, Sinnecker D, Klett K, Fröhlich T, Rahman FA, Haufe T, Sun S, Jurisch V, Kessler B, Hinkel R, Dirschinger R, Martens E, Jilek C, Graf A, Krebs S, Santamaria G, Kurome M, Zakhartchenko V, Campbell B, Voelse K, Wolf A, Ziegler T, Reichert S, Lee S, Flenkenthaler F, Dorn T, Jeremias I, Blum H, Dendorfer A, Schnieke A, Krause S, Walter MC, Klymiuk N, Laugwitz KL, Wolf E, Wurst W, Kupatt C. Somatic gene editing ameliorates skeletal and cardiac muscle failure in pig and human models of Duchenne muscular dystrophy. Nat Med 2020; 26:207-214. [PMID: 31988462 PMCID: PMC7212064 DOI: 10.1038/s41591-019-0738-2] [Citation(s) in RCA: 155] [Impact Index Per Article: 38.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Accepted: 12/11/2019] [Indexed: 11/09/2022]
Abstract
Frameshift mutations in the DMD gene, encoding dystrophin, cause Duchenne muscular dystrophy (DMD), leading to terminal muscle and heart failure in patients. Somatic gene editing by sequence-specific nucleases offers new options for restoring the DMD reading frame, resulting in expression of a shortened but largely functional dystrophin protein. Here, we validated this approach in a pig model of DMD lacking exon 52 of DMD (DMDΔ52), as well as in a corresponding patient-derived induced pluripotent stem cell model. In DMDΔ52 pigs1, intramuscular injection of adeno-associated viral vectors of serotype 9 carrying an intein-split Cas9 (ref. 2) and a pair of guide RNAs targeting sequences flanking exon 51 (AAV9-Cas9-gE51) induced expression of a shortened dystrophin (DMDΔ51-52) and improved skeletal muscle function. Moreover, systemic application of AAV9-Cas9-gE51 led to widespread dystrophin expression in muscle, including diaphragm and heart, prolonging survival and reducing arrhythmogenic vulnerability. Similarly, in induced pluripotent stem cell-derived myoblasts and cardiomyocytes of a patient lacking DMDΔ52, AAV6-Cas9-g51-mediated excision of exon 51 restored dystrophin expression and amelioreate skeletal myotube formation as well as abnormal cardiomyocyte Ca2+ handling and arrhythmogenic susceptibility. The ability of Cas9-mediated exon excision to improve DMD pathology in these translational models paves the way for new treatment approaches in patients with this devastating disease.
Collapse
Affiliation(s)
- A Moretti
- Klinik und Poliklinik für Innere Medizin I, Klinikum rechts der Isar, Technical University Munich and German Center for Cardiovascular Research (DZHK), Munich Heart Alliance, Munich, Germany.
| | - L Fonteyne
- Chair for Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, LMU Munich, Munich, Germany
| | - F Giesert
- Institute of Developmental Genetics, Helmholtz Centre and Munich School of Life Sciences Weihenstephan, Technical University of Munich, Munich, Germany
| | - P Hoppmann
- Klinik und Poliklinik für Innere Medizin I, Klinikum rechts der Isar, Technical University Munich and German Center for Cardiovascular Research (DZHK), Munich Heart Alliance, Munich, Germany
| | - A B Meier
- Klinik und Poliklinik für Innere Medizin I, Klinikum rechts der Isar, Technical University Munich and German Center for Cardiovascular Research (DZHK), Munich Heart Alliance, Munich, Germany
| | - T Bozoglu
- Klinik und Poliklinik für Innere Medizin I, Klinikum rechts der Isar, Technical University Munich and German Center for Cardiovascular Research (DZHK), Munich Heart Alliance, Munich, Germany
| | - A Baehr
- Klinik und Poliklinik für Innere Medizin I, Klinikum rechts der Isar, Technical University Munich and German Center for Cardiovascular Research (DZHK), Munich Heart Alliance, Munich, Germany
| | - C M Schneider
- Klinik und Poliklinik für Innere Medizin I, Klinikum rechts der Isar, Technical University Munich and German Center for Cardiovascular Research (DZHK), Munich Heart Alliance, Munich, Germany
| | - D Sinnecker
- Klinik und Poliklinik für Innere Medizin I, Klinikum rechts der Isar, Technical University Munich and German Center for Cardiovascular Research (DZHK), Munich Heart Alliance, Munich, Germany
| | - K Klett
- Klinik und Poliklinik für Innere Medizin I, Klinikum rechts der Isar, Technical University Munich and German Center for Cardiovascular Research (DZHK), Munich Heart Alliance, Munich, Germany
| | - T Fröhlich
- Chair for Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, LMU Munich, Munich, Germany
- Laboratory for Functional Genome Analysis (LAFUGA), Gene Center, LMU Munich, Munich, Germany
| | - F Abdel Rahman
- Klinik und Poliklinik für Innere Medizin I, Klinikum rechts der Isar, Technical University Munich and German Center for Cardiovascular Research (DZHK), Munich Heart Alliance, Munich, Germany
| | - T Haufe
- Klinik und Poliklinik für Innere Medizin I, Klinikum rechts der Isar, Technical University Munich and German Center for Cardiovascular Research (DZHK), Munich Heart Alliance, Munich, Germany
| | - S Sun
- Klinik und Poliklinik für Innere Medizin I, Klinikum rechts der Isar, Technical University Munich and German Center for Cardiovascular Research (DZHK), Munich Heart Alliance, Munich, Germany
| | - V Jurisch
- Klinik und Poliklinik für Innere Medizin I, Klinikum rechts der Isar, Technical University Munich and German Center for Cardiovascular Research (DZHK), Munich Heart Alliance, Munich, Germany
| | - B Kessler
- Chair for Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, LMU Munich, Munich, Germany
- Laboratory for Functional Genome Analysis (LAFUGA), Gene Center, LMU Munich, Munich, Germany
| | - R Hinkel
- Klinik und Poliklinik für Innere Medizin I, Klinikum rechts der Isar, Technical University Munich and German Center for Cardiovascular Research (DZHK), Munich Heart Alliance, Munich, Germany
| | - R Dirschinger
- Klinik und Poliklinik für Innere Medizin I, Klinikum rechts der Isar, Technical University Munich and German Center for Cardiovascular Research (DZHK), Munich Heart Alliance, Munich, Germany
| | - E Martens
- Klinik und Poliklinik für Innere Medizin I, Klinikum rechts der Isar, Technical University Munich and German Center for Cardiovascular Research (DZHK), Munich Heart Alliance, Munich, Germany
| | - C Jilek
- Klinik und Poliklinik für Innere Medizin I, Klinikum rechts der Isar, Technical University Munich and German Center for Cardiovascular Research (DZHK), Munich Heart Alliance, Munich, Germany
| | - A Graf
- Chair for Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, LMU Munich, Munich, Germany
- Laboratory for Functional Genome Analysis (LAFUGA), Gene Center, LMU Munich, Munich, Germany
| | - S Krebs
- Chair for Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, LMU Munich, Munich, Germany
- Laboratory for Functional Genome Analysis (LAFUGA), Gene Center, LMU Munich, Munich, Germany
| | - G Santamaria
- Klinik und Poliklinik für Innere Medizin I, Klinikum rechts der Isar, Technical University Munich and German Center for Cardiovascular Research (DZHK), Munich Heart Alliance, Munich, Germany
| | - M Kurome
- Chair for Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, LMU Munich, Munich, Germany
- Laboratory for Functional Genome Analysis (LAFUGA), Gene Center, LMU Munich, Munich, Germany
| | - V Zakhartchenko
- Chair for Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, LMU Munich, Munich, Germany
- Laboratory for Functional Genome Analysis (LAFUGA), Gene Center, LMU Munich, Munich, Germany
| | - B Campbell
- Klinik und Poliklinik für Innere Medizin I, Klinikum rechts der Isar, Technical University Munich and German Center for Cardiovascular Research (DZHK), Munich Heart Alliance, Munich, Germany
| | - K Voelse
- Reseach Unit Apoptosis in Hemopoietic Stem Cells, Helmholtz Zentrum München, German Center for Environmental Health (HMGU), Munich, Germany
| | - A Wolf
- Klinik und Poliklinik für Innere Medizin I, Klinikum rechts der Isar, Technical University Munich and German Center for Cardiovascular Research (DZHK), Munich Heart Alliance, Munich, Germany
| | - T Ziegler
- Klinik und Poliklinik für Innere Medizin I, Klinikum rechts der Isar, Technical University Munich and German Center for Cardiovascular Research (DZHK), Munich Heart Alliance, Munich, Germany
| | - S Reichert
- Department of Neurology, Friedrich Baur Institute, LMU Munich, Munich, Germany
| | - S Lee
- Klinik und Poliklinik für Innere Medizin I, Klinikum rechts der Isar, Technical University Munich and German Center for Cardiovascular Research (DZHK), Munich Heart Alliance, Munich, Germany
| | - F Flenkenthaler
- Chair for Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, LMU Munich, Munich, Germany
- Laboratory for Functional Genome Analysis (LAFUGA), Gene Center, LMU Munich, Munich, Germany
| | - T Dorn
- Klinik und Poliklinik für Innere Medizin I, Klinikum rechts der Isar, Technical University Munich and German Center for Cardiovascular Research (DZHK), Munich Heart Alliance, Munich, Germany
| | - I Jeremias
- Reseach Unit Apoptosis in Hemopoietic Stem Cells, Helmholtz Zentrum München, German Center for Environmental Health (HMGU), Munich, Germany
| | - H Blum
- Chair for Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, LMU Munich, Munich, Germany
- Laboratory for Functional Genome Analysis (LAFUGA), Gene Center, LMU Munich, Munich, Germany
| | - A Dendorfer
- Walter Brendel Centre of Experimental Medicine, University Hospital, LMU Munich, Munich, Germany
| | - A Schnieke
- Chair of Livestock Biotechnology, School of Life Sciences Weihenstephan, Technical University of Munich, Munich, Germany
| | - S Krause
- Department of Neurology, Friedrich Baur Institute, LMU Munich, Munich, Germany
| | - M C Walter
- Department of Neurology, Friedrich Baur Institute, LMU Munich, Munich, Germany
| | - N Klymiuk
- Chair for Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, LMU Munich, Munich, Germany
- Laboratory for Functional Genome Analysis (LAFUGA), Gene Center, LMU Munich, Munich, Germany
| | - K L Laugwitz
- Klinik und Poliklinik für Innere Medizin I, Klinikum rechts der Isar, Technical University Munich and German Center for Cardiovascular Research (DZHK), Munich Heart Alliance, Munich, Germany
| | - E Wolf
- Chair for Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, LMU Munich, Munich, Germany
- Laboratory for Functional Genome Analysis (LAFUGA), Gene Center, LMU Munich, Munich, Germany
| | - W Wurst
- Institute of Developmental Genetics, Helmholtz Centre and Munich School of Life Sciences Weihenstephan, Technical University of Munich, Munich, Germany
- German Center for Neurodegenerative Diseases, Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - C Kupatt
- Klinik und Poliklinik für Innere Medizin I, Klinikum rechts der Isar, Technical University Munich and German Center for Cardiovascular Research (DZHK), Munich Heart Alliance, Munich, Germany.
| |
Collapse
|
90
|
Gilbert-Honick J, Grayson W. Vascularized and Innervated Skeletal Muscle Tissue Engineering. Adv Healthc Mater 2020; 9:e1900626. [PMID: 31622051 PMCID: PMC6986325 DOI: 10.1002/adhm.201900626] [Citation(s) in RCA: 80] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 09/27/2019] [Indexed: 12/12/2022]
Abstract
Volumetric muscle loss (VML) is a devastating loss of muscle tissue that overwhelms the native regenerative properties of skeletal muscle and results in lifelong functional deficits. There are currently no treatments for VML that fully recover the lost muscle tissue and function. Tissue engineering presents a promising solution for VML treatment and significant research has been performed using tissue engineered muscle constructs in preclinical models of VML with a broad range of defect locations and sizes, tissue engineered construct characteristics, and outcome measures. Due to the complex vascular and neural anatomy within skeletal muscle, regeneration of functional vasculature and nerves is vital for muscle recovery following VML injuries. This review aims to summarize the current state of the field of skeletal muscle tissue engineering using 3D constructs for VML treatment with a focus on studies that have promoted vascular and neural regeneration within the muscle tissue post-VML.
Collapse
Affiliation(s)
- Jordana Gilbert-Honick
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Warren Grayson
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Material Sciences & Engineering, Johns Hopkins University, School of Engineering, Baltimore, MD 21218, USA
- Institute for NanoBioTechnology (INBT), Johns Hopkins University School of Engineering, Baltimore, MD 21218, USA
| |
Collapse
|
91
|
Fusto A, Moyle LA, Gilbert PM, Pegoraro E. Cored in the act: the use of models to understand core myopathies. Dis Model Mech 2019; 12:dmm041368. [PMID: 31874912 PMCID: PMC6955215 DOI: 10.1242/dmm.041368] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The core myopathies are a group of congenital myopathies with variable clinical expression - ranging from early-onset skeletal-muscle weakness to later-onset disease of variable severity - that are identified by characteristic 'core-like' lesions in myofibers and the presence of hypothonia and slowly or rather non-progressive muscle weakness. The genetic causes are diverse; central core disease is most often caused by mutations in ryanodine receptor 1 (RYR1), whereas multi-minicore disease is linked to pathogenic variants of several genes, including selenoprotein N (SELENON), RYR1 and titin (TTN). Understanding the mechanisms that drive core development and muscle weakness remains challenging due to the diversity of the excitation-contraction coupling (ECC) proteins involved and the differential effects of mutations across proteins. Because of this, the use of representative models expressing a mature ECC apparatus is crucial. Animal models have facilitated the identification of disease progression mechanisms for some mutations and have provided evidence to help explain genotype-phenotype correlations. However, many unanswered questions remain about the common and divergent pathological mechanisms that drive disease progression, and these mechanisms need to be understood in order to identify therapeutic targets. Several new transgenic animals have been described recently, expanding the spectrum of core myopathy models, including mice with patient-specific mutations. Furthermore, recent developments in 3D tissue engineering are expected to enable the study of core myopathy disease progression and the effects of potential therapeutic interventions in the context of human cells. In this Review, we summarize the current landscape of core myopathy models, and assess the hurdles and opportunities of future modeling strategies.
Collapse
Affiliation(s)
- Aurora Fusto
- Department of Neuroscience, University of Padua, Padua 35128, Italy
| | - Louise A Moyle
- Donnelly Centre, University of Toronto, Toronto, ON M5S3E1, Canada
- Institute of Biomaterials and Biochemical Engineering, University of Toronto, Toronto, ON M5S3G9, Canada
| | - Penney M Gilbert
- Donnelly Centre, University of Toronto, Toronto, ON M5S3E1, Canada
- Institute of Biomaterials and Biochemical Engineering, University of Toronto, Toronto, ON M5S3G9, Canada
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON M5S3G5, Canada
- Department of Biochemistry, University of Toronto, Toronto, ON M5S1A8, Canada
| | - Elena Pegoraro
- Department of Neuroscience, University of Padua, Padua 35128, Italy
| |
Collapse
|
92
|
Wu J, Matthias N, Lo J, Ortiz-Vitali JL, Shieh AW, Wang SH, Darabi R. A Myogenic Double-Reporter Human Pluripotent Stem Cell Line Allows Prospective Isolation of Skeletal Muscle Progenitors. Cell Rep 2019; 25:1966-1981.e4. [PMID: 30428361 DOI: 10.1016/j.celrep.2018.10.067] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Revised: 10/08/2018] [Accepted: 10/18/2018] [Indexed: 02/06/2023] Open
Abstract
Myogenic differentiation of human pluripotent stem cells (hPSCs) has been done by gene overexpression or directed differentiation. However, viral integration, long-term culture, and the presence of unwanted cells are the main obstacles. By using CRISPR/Cas9n, a double-reporter human embryonic stem cell (hESC) line was generated for PAX7/MYF5, allowing prospective readout. This strategy allowed pathway screen to define efficient myogenic induction in hPSCs. Next, surface marker screen allowed identification of CD10 and CD24 for purification of myogenic progenitors and exclusion of non-myogenic cells. CD10 expression was also identified on human satellite cells and skeletal muscle progenitors. In vitro and in vivo studies using transgene and/or reporter-free hPSCs further validated myogenic potential of the cells by formation of new fibers expressing human dystrophin as well as donor-derived satellite cells in NSG-mdx4Cv mice. This study provides biological insights for myogenic differentiation of hPSCs using a double-reporter cell resource and defines an improved myogenic differentiation and purification strategy.
Collapse
Affiliation(s)
- Jianbo Wu
- Center for Stem Cell and Regenerative Medicine (CSCRM), The Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases (IMM), The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Nadine Matthias
- Center for Stem Cell and Regenerative Medicine (CSCRM), The Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases (IMM), The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Jonathan Lo
- Center for Stem Cell and Regenerative Medicine (CSCRM), The Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases (IMM), The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Jose L Ortiz-Vitali
- Center for Stem Cell and Regenerative Medicine (CSCRM), The Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases (IMM), The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Annie W Shieh
- Department of Psychiatry, SUNY Upstate Medical University, Syracuse, NY 13210, USA
| | - Sidney H Wang
- Center for Human Genetics, The Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases (IMM), The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Radbod Darabi
- Center for Stem Cell and Regenerative Medicine (CSCRM), The Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases (IMM), The University of Texas Health Science Center at Houston, Houston, TX 77030, USA.
| |
Collapse
|
93
|
Selvaraj S, Dhoke NR, Kiley J, Mateos-Aierdi AJ, Tungtur S, Mondragon-Gonzalez R, Killeen G, Oliveira VKP, López de Munain A, Perlingeiro RCR. Gene Correction of LGMD2A Patient-Specific iPSCs for the Development of Targeted Autologous Cell Therapy. Mol Ther 2019; 27:2147-2157. [PMID: 31501033 PMCID: PMC6904833 DOI: 10.1016/j.ymthe.2019.08.011] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Revised: 08/18/2019] [Accepted: 08/21/2019] [Indexed: 01/25/2023] Open
Abstract
Limb girdle muscular dystrophy type 2A (LGMD2A), caused by mutations in the Calpain 3 (CAPN3) gene, is an incurable autosomal recessive disorder that results in muscle wasting and loss of ambulation. To test the feasibility of an autologous induced pluripotent stem cell (iPSC)-based therapy for LGMD2A, here we applied CRISPR-Cas9-mediated genome editing to iPSCs from three LGMD2A patients to enable correction of mutations in the CAPN3 gene. Using a gene knockin approach, we genome edited iPSCs carrying three different CAPN3 mutations, and we demonstrated the rescue of CAPN3 protein in myotube derivatives in vitro. Transplantation of gene-corrected LGMD2A myogenic progenitors in a novel mouse model combining immunodeficiency and a lack of CAPN3 resulted in muscle engraftment and rescue of the CAPN3 mRNA. Thus, we provide here proof of concept for the integration of genome editing and iPSC technologies to develop a novel autologous cell therapy for LGMD2A.
Collapse
MESH Headings
- Animals
- Calpain/physiology
- Cell- and Tissue-Based Therapy/methods
- Cells, Cultured
- Humans
- Induced Pluripotent Stem Cells/cytology
- Induced Pluripotent Stem Cells/metabolism
- Male
- Mice
- Mice, Inbred NOD
- Mice, Knockout
- Mice, SCID
- Muscle Fibers, Skeletal/metabolism
- Muscle Fibers, Skeletal/pathology
- Muscle Proteins/physiology
- Muscle, Skeletal/metabolism
- Muscle, Skeletal/pathology
- Muscular Dystrophies, Limb-Girdle/genetics
- Muscular Dystrophies, Limb-Girdle/pathology
- Muscular Dystrophies, Limb-Girdle/therapy
- Mutation
- Transplantation, Autologous
Collapse
Affiliation(s)
- Sridhar Selvaraj
- Lillehei Heart Institute, Department of Medicine, University of Minnesota, Minneapolis, MN 55455, USA
| | - Neha R Dhoke
- Lillehei Heart Institute, Department of Medicine, University of Minnesota, Minneapolis, MN 55455, USA
| | - James Kiley
- Lillehei Heart Institute, Department of Medicine, University of Minnesota, Minneapolis, MN 55455, USA
| | - Alba Judith Mateos-Aierdi
- Neurosciences Department, Biodonostia Research Institute-University of the Basque Country UPV-EHU, San Sebastián 20014, Spain; CIBERNED, Institute Carlos III, Madrid 28029, Spain
| | - Sudheer Tungtur
- Lillehei Heart Institute, Department of Medicine, University of Minnesota, Minneapolis, MN 55455, USA
| | - Ricardo Mondragon-Gonzalez
- Lillehei Heart Institute, Department of Medicine, University of Minnesota, Minneapolis, MN 55455, USA; Departamento de Genética y Biología Molecular, Centro de Investigación y de Estudios Avanzados del IPN (CINVESTAV-IPN), 07360 Ciudad de México, Mexico
| | - Grace Killeen
- Lillehei Heart Institute, Department of Medicine, University of Minnesota, Minneapolis, MN 55455, USA
| | - Vanessa K P Oliveira
- Lillehei Heart Institute, Department of Medicine, University of Minnesota, Minneapolis, MN 55455, USA
| | - Adolfo López de Munain
- Neurosciences Department, Biodonostia Research Institute-University of the Basque Country UPV-EHU, San Sebastián 20014, Spain; CIBERNED, Institute Carlos III, Madrid 28029, Spain
| | - Rita C R Perlingeiro
- Lillehei Heart Institute, Department of Medicine, University of Minnesota, Minneapolis, MN 55455, USA; Stem Cell Institute, University of Minnesota, Minneapolis, MN 55455, USA.
| |
Collapse
|
94
|
Sun J, Ma X, Chu HT, Feng B, Tuan RS, Jiang Y. Biomaterials and Advanced Biofabrication Techniques in hiPSCs Based Neuromyopathic Disease Modeling. Front Bioeng Biotechnol 2019; 7:373. [PMID: 31850331 PMCID: PMC6895005 DOI: 10.3389/fbioe.2019.00373] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Accepted: 11/13/2019] [Indexed: 12/22/2022] Open
Abstract
Induced pluripotent stem cells (iPSCs) are reprogrammed somatic cells by defined factors, and have great application potentials in tissue regeneration and disease modeling. Biomaterials have been widely used in stem cell-based studies, and are involved in human iPSCs based studies, but they were not enough emphasized and recognized. Biomaterials can mimic the extracellular matrix and microenvironment, and act as powerful tools to promote iPSCs proliferation, differentiation, maturation, and migration. Many classic and advanced biofabrication technologies, such as cell-sheet approach, electrospinning, and 3D-bioprinting, are used to provide physical cues in macro-/micro-patterning, and in combination with other biological factors to support iPSCs applications. In this review, we highlight the biomaterials and fabrication technologies used in human iPSC-based tissue engineering to model neuromyopathic diseases, particularly those with genetic mutations, such as Duchenne Muscular Dystrophy (DMD), Congenital Heart Diseases (CHD) and Alzheimer's disease (AD).
Collapse
Affiliation(s)
- Jing Sun
- Faculty of Medicine, School of Biomedical Sciences, Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong, China.,Faculty of Medicine, School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Xun Ma
- Faculty of Medicine, School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Ho Ting Chu
- Faculty of Medicine, School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Bo Feng
- Faculty of Medicine, School of Biomedical Sciences, Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong, China.,Faculty of Medicine, School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China.,Key Laboratory for Regenerative Medicine, Ministry of Education, Faculty of Medicine, School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Rocky S Tuan
- Faculty of Medicine, School of Biomedical Sciences, Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong, China.,Faculty of Medicine, School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Yangzi Jiang
- Faculty of Medicine, School of Biomedical Sciences, Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong, China.,Faculty of Medicine, School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China
| |
Collapse
|
95
|
Tey SR, Robertson S, Lynch E, Suzuki M. Coding Cell Identity of Human Skeletal Muscle Progenitor Cells Using Cell Surface Markers: Current Status and Remaining Challenges for Characterization and Isolation. Front Cell Dev Biol 2019; 7:284. [PMID: 31828070 PMCID: PMC6890603 DOI: 10.3389/fcell.2019.00284] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Accepted: 11/01/2019] [Indexed: 12/12/2022] Open
Abstract
Skeletal muscle progenitor cells (SMPCs), also called myogenic progenitors, have been studied extensively in recent years because of their promising therapeutic potential to preserve and recover skeletal muscle mass and function in patients with cachexia, sarcopenia, and neuromuscular diseases. SMPCs can be utilized to investigate the mechanisms of natural and pathological myogenesis via in vitro modeling and in vivo experimentation. While various types of SMPCs are currently available from several sources, human pluripotent stem cells (PSCs) offer an efficient and cost-effective method to derive SMPCs. As human PSC-derived cells often display varying heterogeneity in cell types, cell enrichment using cell surface markers remains a critical step in current procedures to establish a pure population of SMPCs. Here we summarize the cell surface markers currently being used to detect human SMPCs, describing their potential application for characterizing, identifying and isolating human PSC-derived SMPCs. To date, several positive and negative markers have been used to enrich human SMPCs from differentiated PSCs by cell sorting. A careful analysis of current findings can broaden our understanding and reveal potential uses for these surface markers with SMPCs.
Collapse
Affiliation(s)
- Sin-Ruow Tey
- Department of Comparative Biosciences, University of Wisconsin, Madison, WI, United States
| | - Samantha Robertson
- Department of Comparative Biosciences, University of Wisconsin, Madison, WI, United States
| | - Eileen Lynch
- Department of Comparative Biosciences, University of Wisconsin, Madison, WI, United States
| | - Masatoshi Suzuki
- Department of Comparative Biosciences, University of Wisconsin, Madison, WI, United States.,The Stem Cell and Regenerative Medicine Center, University of Wisconsin, Madison, WI, United States
| |
Collapse
|
96
|
Guan J, Liu X, Zhang H, Yang X, Ma Y, Li Y, Gai Z, Liu Y. Reprogramming of human Peripheral Blood Mononuclear Cell (PBMC) from a Chinese patient suffering Duchenne muscular dystrophy to iPSC line (SDQLCHi007-A) carrying deletion of 49-50 exons in the DMD gene. Stem Cell Res 2019; 42:101666. [PMID: 31812072 DOI: 10.1016/j.scr.2019.101666] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Revised: 11/10/2019] [Accepted: 11/20/2019] [Indexed: 12/22/2022] Open
Abstract
Duchenne muscular dystrophy (DMD), an X-linked genetic disorder characterized by progressive muscle weakness and atrophies affecting skeletal and cardiac muscles, is caused by mutations in dystrophin (DMD) gene that spans 79 exons. Here, we generated iPSCs from a Chinese patient with 49-50 exons deletion in DMD gene by reprogramming peripheral blood mononuclear cells with non-integrating vectors. The generated iPSCs line (SDQLCHi007-A) carrying the identical deletion of 49-50 exons, expresses pluripotency markers, presents a normal karyotype and is able to differentiate into three germ layers.
Collapse
Affiliation(s)
- Jingyun Guan
- Pediatric Research Institute, Qilu Children's Hospital of Shandong University, Jinan, Shandong 250022, China
| | - Xinnong Liu
- Department of Vascular Surgery, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037,China
| | - Haiyan Zhang
- Pediatric Research Institute, Qilu Children's Hospital of Shandong University, Jinan, Shandong 250022, China
| | - Xiaomeng Yang
- Pediatric Research Institute, Qilu Children's Hospital of Shandong University, Jinan, Shandong 250022, China
| | - Yanyan Ma
- Pediatric Research Institute, Qilu Children's Hospital of Shandong University, Jinan, Shandong 250022, China
| | - Yue Li
- Pediatric Research Institute, Qilu Children's Hospital of Shandong University, Jinan, Shandong 250022, China
| | - Zhongtao Gai
- Pediatric Research Institute, Qilu Children's Hospital of Shandong University, Jinan, Shandong 250022, China.
| | - Yi Liu
- Pediatric Research Institute, Qilu Children's Hospital of Shandong University, Jinan, Shandong 250022, China.
| |
Collapse
|
97
|
Sun C, Serra C, Lee G, Wagner KR. Stem cell-based therapies for Duchenne muscular dystrophy. Exp Neurol 2019; 323:113086. [PMID: 31639376 DOI: 10.1016/j.expneurol.2019.113086] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 10/16/2019] [Accepted: 10/18/2019] [Indexed: 02/08/2023]
Abstract
Muscular dystrophies are a group of genetic muscle disorders that cause progressive muscle weakness and degeneration. Within this group, Duchenne muscular dystrophy (DMD) is the most common and one of the most severe. DMD is an X chromosome linked disease that occurs to 1 in 3500 to 1 in 5000 boys. The cause of DMD is a mutation in the dystrophin gene, whose encoded protein provides both structural support and cell signaling capabilities. So far, there are very limited therapeutic options available and there is no cure for this disease. In this review, we discuss the existing cell therapy research, especially stem cell-based, which utilize myoblasts, satellite cells, bone marrow cells, mesoangioblasts and CD133+ cells. Finally, we focus on human pluripotent stem cells (hPSCs) which hold great potential in treating DMD. hPSCs can be used for autologous transplantation after being specified to a myogenic lineage. Over the last few years, there has been a rapid development of isolation, as well as differentiation, techniques in order to achieve effective transplantation results of myogenic cells specified from hPSCs. In this review, we summarize the current methods of hPSCs myogenic commitment/differentiation, and describe the current status of hPSC-derived myogenic cell transplantation.
Collapse
Affiliation(s)
- Congshan Sun
- Departments of Neurology and Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA; Center for Genetic Muscle Disorders, Hugo W. Moser Research Institute at Kennedy Krieger Institute, Baltimore, MD 21205, USA.
| | - Carlo Serra
- Departments of Neurology and Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA; Center for Genetic Muscle Disorders, Hugo W. Moser Research Institute at Kennedy Krieger Institute, Baltimore, MD 21205, USA
| | - Gabsang Lee
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Kathryn R Wagner
- Departments of Neurology and Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA; Center for Genetic Muscle Disorders, Hugo W. Moser Research Institute at Kennedy Krieger Institute, Baltimore, MD 21205, USA
| |
Collapse
|
98
|
Gene expression profiling of skeletal myogenesis in human embryonic stem cells reveals a potential cascade of transcription factors regulating stages of myogenesis, including quiescent/activated satellite cell-like gene expression. PLoS One 2019; 14:e0222946. [PMID: 31560727 PMCID: PMC6764674 DOI: 10.1371/journal.pone.0222946] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Accepted: 09/10/2019] [Indexed: 01/05/2023] Open
Abstract
Human embryonic stem cell (hESC)-derived skeletal muscle progenitors (SMP)—defined as PAX7-expressing cells with myogenic potential—can provide an abundant source of donor material for muscle stem cell therapy. As in vitro myogenesis is decoupled from in vivo timing and 3D-embryo structure, it is important to characterize what stage or type of muscle is modeled in culture. Here, gene expression profiling is analyzed in hESCs over a 50 day skeletal myogenesis protocol and compared to datasets of other hESC-derived skeletal muscle and adult murine satellite cells. Furthermore, day 2 cultures differentiated with high or lower concentrations of CHIR99021, a GSK3A/GSK3B inhibitor, were contrasted. Expression profiling of the 50 day time course identified successively expressed gene subsets involved in mesoderm/paraxial mesoderm induction, somitogenesis, and skeletal muscle commitment/formation which could be regulated by a putative cascade of transcription factors. Initiating differentiation with higher CHIR99021 concentrations significantly increased expression of MSGN1 and TGFB-superfamily genes, notably NODAL, resulting in enhanced paraxial mesoderm and reduced ectoderm/neuronal gene expression. Comparison to adult satellite cells revealed that genes expressed in 50-day cultures correlated better with those expressed by quiescent or early activated satellite cells, which have the greatest therapeutic potential. Day 50 cultures were similar to other hESC-derived skeletal muscle and both expressed known and novel SMP surface proteins. Overall, a putative cascade of transcription factors has been identified which regulates four stages of myogenesis. Subsets of these factors were upregulated by high CHIR99021 or their binding sites were significantly over-represented during SMP activation, ranging from quiescent to late-activated stages. This analysis serves as a resource to further study the progression of in vitro skeletal myogenesis and could be mined to identify novel markers of pluripotent-derived SMPs or regulatory transcription/growth factors. Finally, 50-day hESC-derived SMPs appear similar to quiescent/early activated satellite cells, suggesting they possess therapeutic potential.
Collapse
|
99
|
Wang J, Khodabukus A, Rao L, Vandusen K, Abutaleb N, Bursac N. Engineered skeletal muscles for disease modeling and drug discovery. Biomaterials 2019; 221:119416. [PMID: 31419653 DOI: 10.1016/j.biomaterials.2019.119416] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2019] [Revised: 08/01/2019] [Accepted: 08/05/2019] [Indexed: 01/04/2023]
Abstract
Skeletal muscle is the largest organ of human body with several important roles in everyday movement and metabolic homeostasis. The limited ability of small animal models of muscle disease to accurately predict drug efficacy and toxicity in humans has prompted the development in vitro models of human skeletal muscle that fatefully recapitulate cell and tissue level functions and drug responses. We first review methods for development of three-dimensional engineered muscle tissues and organ-on-a-chip microphysiological systems and discuss their potential utility in drug discovery research and development of new regenerative therapies. Furthermore, we describe strategies to increase the functional maturation of engineered muscle, and motivate the importance of incorporating multiple tissue types on the same chip to model organ cross-talk and generate more predictive drug development platforms. Finally, we review the ability of available in vitro systems to model diseases such as type II diabetes, Duchenne muscular dystrophy, Pompe disease, and dysferlinopathy.
Collapse
Affiliation(s)
- Jason Wang
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | | | - Lingjun Rao
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Keith Vandusen
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Nadia Abutaleb
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Nenad Bursac
- Department of Biomedical Engineering, Duke University, Durham, NC, USA.
| |
Collapse
|
100
|
Jiwlawat N, Lynch EM, Napiwocki BN, Stempien A, Ashton RS, Kamp TJ, Crone WC, Suzuki M. Micropatterned substrates with physiological stiffness promote cell maturation and Pompe disease phenotype in human induced pluripotent stem cell-derived skeletal myocytes. Biotechnol Bioeng 2019; 116:2377-2392. [PMID: 31131875 DOI: 10.1002/bit.27075] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Revised: 04/19/2019] [Accepted: 05/21/2019] [Indexed: 12/23/2022]
Abstract
Recent advances in bioengineering have enabled cell culture systems that more closely mimic the native cellular environment. Here, we demonstrated that human induced pluripotent stem cell (iPSC)-derived myogenic progenitors formed highly-aligned myotubes and contracted when seeded on two-dimensional micropatterned platforms. The differentiated cells showed clear nuclear alignment and formed elongated myotubes dependent on the width of the micropatterned lanes. Topographical cues from micropatterning and physiological substrate stiffness improved the formation of well-aligned and multinucleated myotubes similar to myofibers. These aligned myotubes exhibited spontaneous contractions specifically along the long axis of the pattern. Notably, the micropatterned platforms developed bundle-like myotubes using patient-derived iPSCs with a background of Pompe disease (glycogen storage disease type II) and even enhanced the disease phenotype as shown through the specific pathology of abnormal lysosome accumulations. A highly-aligned formation of matured myotubes holds great potential in further understanding the process of human muscle development, as well as advancing in vitro pharmacological studies for skeletal muscle diseases.
Collapse
Affiliation(s)
- Nunnapas Jiwlawat
- Department of Comparative Biosciences, University of Wisconsin, Madison, Wisconsin
| | - Eileen M Lynch
- Department of Comparative Biosciences, University of Wisconsin, Madison, Wisconsin
| | - Brett N Napiwocki
- Wisconsin Institute for Discovery, University of Wisconsin, Madison, Wisconsin.,Department of Biomedical Engineering, University of Wisconsin, Madison, Wisconsin
| | - Alana Stempien
- Wisconsin Institute for Discovery, University of Wisconsin, Madison, Wisconsin.,Department of Biomedical Engineering, University of Wisconsin, Madison, Wisconsin
| | - Randolph S Ashton
- Wisconsin Institute for Discovery, University of Wisconsin, Madison, Wisconsin.,Department of Biomedical Engineering, University of Wisconsin, Madison, Wisconsin.,The Stem Cell and Regenerative Medicine Center, University of Wisconsin, Madison, Wisconsin
| | - Timothy J Kamp
- Department of Medicine, University of Wisconsin, Madison, Wisconsin.,The Stem Cell and Regenerative Medicine Center, University of Wisconsin, Madison, Wisconsin.,Department of Cell and Regenerative Biology, University of Wisconsin, Madison, Wisconsin
| | - Wendy C Crone
- Wisconsin Institute for Discovery, University of Wisconsin, Madison, Wisconsin.,Department of Biomedical Engineering, University of Wisconsin, Madison, Wisconsin.,The Stem Cell and Regenerative Medicine Center, University of Wisconsin, Madison, Wisconsin.,Department of Engineering Physics, University of Wisconsin, Madison, Wisconsin
| | - Masatoshi Suzuki
- Department of Comparative Biosciences, University of Wisconsin, Madison, Wisconsin.,Department of Biomedical Engineering, University of Wisconsin, Madison, Wisconsin.,The Stem Cell and Regenerative Medicine Center, University of Wisconsin, Madison, Wisconsin
| |
Collapse
|