1
|
Scherrer C, Loret C, Védrenne N, Buckley C, Lia AS, Kermene V, Sturtz F, Favreau F, Rovini A, Faye PA. From in vivo models to in vitro bioengineered neuromuscular junctions for the study of Charcot-Marie-Tooth disease. J Tissue Eng 2025; 16:20417314241310508. [PMID: 40078221 PMCID: PMC11898049 DOI: 10.1177/20417314241310508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Accepted: 12/14/2024] [Indexed: 03/14/2025] Open
Abstract
Peripheral neuropathies are disorders affecting the peripheral nervous system. Among them, Charcot-Marie-Tooth disease is an inherited sensorimotor neuropathy for which no effective treatment exists yet. Research on Charcot-Marie-Tooth disease has been hampered by difficulties in accessing relevant cells, such as sensory and motor neurons, Schwann cells, and myocytes, which interact at the neuromuscular junction, the specialized synapses formed between nerves and skeletal muscles. This review first outlines the various in vivo models and methods used to study neuromuscular junction deficiencies in Charcot-Marie-Tooth disease. We then explore novel in vitro techniques and models, including complex hiPSC-derived cultures, which offer promising isogenic and reproducible neuromuscular junction models. The adaptability of in vitro culture methods, including cell origin, cell-type combinations, and choice of culture format, adds complexity and excitement to this rapidly evolving field. This review aims to recapitulate available tools for studying Charcot-Marie-Tooth disease to understand its pathophysiological mechanisms and test potential therapies.
Collapse
Affiliation(s)
- Camille Scherrer
- University of Limoges, NeurIT UR 20218, GEIST Institute, Limoges, France
| | - Camille Loret
- University of Limoges, NeurIT UR 20218, GEIST Institute, Limoges, France
| | - Nicolas Védrenne
- University of Limoges, Inserm U1248 Pharmacology & Transplantation, Limoges, France
| | - Colman Buckley
- University of Limoges, XLIM, CNRS UMR 7252, Limoges, France
| | - Anne-Sophie Lia
- University of Limoges, NeurIT UR 20218, GEIST Institute, Limoges, France
- Department of Biochemistry and Molecular Genetics, CHU Limoges, Limoges, France
- Department of Bioinformatics, CHU Limoges, Limoges, France
| | | | - Franck Sturtz
- University of Limoges, NeurIT UR 20218, GEIST Institute, Limoges, France
- Department of Biochemistry and Molecular Genetics, CHU Limoges, Limoges, France
| | - Frédéric Favreau
- University of Limoges, NeurIT UR 20218, GEIST Institute, Limoges, France
- Department of Biochemistry and Molecular Genetics, CHU Limoges, Limoges, France
| | - Amandine Rovini
- University of Limoges, NeurIT UR 20218, GEIST Institute, Limoges, France
| | - Pierre-Antoine Faye
- University of Limoges, NeurIT UR 20218, GEIST Institute, Limoges, France
- Department of Biochemistry and Molecular Genetics, CHU Limoges, Limoges, France
| |
Collapse
|
2
|
Gazzola M, Martinat C. Unlocking the Complexity of Neuromuscular Diseases: Insights from Human Pluripotent Stem Cell-Derived Neuromuscular Junctions. Int J Mol Sci 2023; 24:15291. [PMID: 37894969 PMCID: PMC10607237 DOI: 10.3390/ijms242015291] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 09/26/2023] [Accepted: 10/16/2023] [Indexed: 10/29/2023] Open
Abstract
Over the past 20 years, the use of pluripotent stem cells to mimic the complexities of the human neuromuscular junction has received much attention. Deciphering the key mechanisms underlying the establishment and maturation of this complex synapse has been driven by the dual goals of addressing developmental questions and gaining insight into neuromuscular disorders. This review aims to summarise the evolution and sophistication of in vitro neuromuscular junction models developed from the first differentiation of human embryonic stem cells into motor neurons to recent neuromuscular organoids. We also discuss the potential offered by these models to decipher different neuromuscular diseases characterised by defects in the presynaptic compartment, the neuromuscular junction, and the postsynaptic compartment. Finally, we discuss the emerging field that considers the use of these techniques in drug screening assay and the challenges they will face in the future.
Collapse
Affiliation(s)
- Morgan Gazzola
- INSERM U861, Institute for Stem Cell Therapy and Exploration of Monogenic Diseases, 91100 Corbeil-Essonnes, France;
| | | |
Collapse
|
3
|
Badu-Mensah A, Guo X, Mendez R, Parsaud H, Hickman JJ. The Effect of Skeletal Muscle-Specific Creatine Treatment on ALS NMJ Integrity and Function. Int J Mol Sci 2023; 24:13519. [PMID: 37686322 PMCID: PMC10487911 DOI: 10.3390/ijms241713519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 08/16/2023] [Accepted: 08/19/2023] [Indexed: 09/10/2023] Open
Abstract
Although skeletal muscle (hSKM) has been proven to be actively involved in Amyotrophic Lateral Sclerosis (ALS) neuromuscular junction (NMJ) dysfunction, it is rarely considered as a pharmacological target in preclinical drug discovery. This project investigated how improving ALS hSKM viability and function effects NMJ integrity. Phenotypic ALS NMJ human-on-a-chip models developed from patient-derived induced pluripotent stem cells (iPSCs) were used to study the effect of hSKM-specific creatine treatment on clinically relevant functional ALS NMJ parameters, such as NMJ numbers, fidelity, stability, and fatigue index. Results indicated comparatively enhanced NMJ numbers, fidelity, and stability, as well as reduced fatigue index, across all hSKM-specific creatine-treated systems. Immunocytochemical analysis of the NMJs also revealed improved post-synaptic nicotinic Acetylcholine receptor (AChR) clustering and cluster size in systems supplemented with creatine relative to the un-dosed control. This work strongly suggests hSKM as a therapeutic target in ALS drug discovery. It also demonstrates the need to consider all tissues involved in multi-systemic diseases, such as ALS, in drug discovery efforts. Finally, this work further establishes the BioMEMs NMJ platform as an effective means of performing mutation-specific drug screening, which is a step towards personalized medicine for rare diseases.
Collapse
Affiliation(s)
- Agnes Badu-Mensah
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway, Suite 400, Orlando, FL 32826, USA; (A.B.-M.); (X.G.); (R.M.); (H.P.)
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32816, USA
| | - Xiufang Guo
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway, Suite 400, Orlando, FL 32826, USA; (A.B.-M.); (X.G.); (R.M.); (H.P.)
| | - Roxana Mendez
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway, Suite 400, Orlando, FL 32826, USA; (A.B.-M.); (X.G.); (R.M.); (H.P.)
| | - Hemant Parsaud
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway, Suite 400, Orlando, FL 32826, USA; (A.B.-M.); (X.G.); (R.M.); (H.P.)
| | - James J. Hickman
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway, Suite 400, Orlando, FL 32826, USA; (A.B.-M.); (X.G.); (R.M.); (H.P.)
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32816, USA
| |
Collapse
|
4
|
Rupar MJ, Sasserath T, Smith E, Comiter B, Sriram N, Long CJ, McAleer CW, Hickman JJ. Development of a human malaria-on-a-chip disease model for drug efficacy and off-target toxicity evaluation. Sci Rep 2023; 13:10509. [PMID: 37380653 DOI: 10.1038/s41598-023-35694-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 05/22/2023] [Indexed: 06/30/2023] Open
Abstract
A functional, multi-organ, serum-free system was developed for the culture of P. falciparum in an attempt to establish innovative platforms for therapeutic drug development. It contains 4 human organ constructs including hepatocytes, splenocytes, endothelial cells, as well as recirculating red blood cells which allow for infection with the parasite. Two strains of P. falciparum were used: the 3D7 strain, which is sensitive to chloroquine; and the W2 strain, which is resistant to chloroquine. The maintenance of functional cells was successfully demonstrated both in healthy and diseased conditions for 7 days in the recirculating microfluidic model. To demonstrate an effective platform for therapeutic development, systems infected with the 3D7 strain were treated with chloroquine which significantly decreased parasitemia, with recrudescence observed after 5 days. Conversely, when the W2 systems were dosed with chloroquine, parasitemia levels were moderately decreased when compared to the 3D7 model. The system also allows for the concurrent evaluation of off-target toxicity for the anti-malarial treatment in a dose dependent manner which indicates this model could be utilized for therapeutic index determination. The work described here establishes a new approach to the evaluation of anti-malarial therapeutics in a realistic human model with recirculating blood cells for 7 days.
Collapse
Affiliation(s)
- Michael J Rupar
- Hesperos, Inc., 12501 Research Parkway, Suite 100, Orlando, FL, 32826, USA
| | - Trevor Sasserath
- Hesperos, Inc., 12501 Research Parkway, Suite 100, Orlando, FL, 32826, USA
| | - Ethan Smith
- Hesperos, Inc., 12501 Research Parkway, Suite 100, Orlando, FL, 32826, USA
| | - Brandon Comiter
- Hesperos, Inc., 12501 Research Parkway, Suite 100, Orlando, FL, 32826, USA
| | - Narasimhan Sriram
- Hesperos, Inc., 12501 Research Parkway, Suite 100, Orlando, FL, 32826, USA
| | - Christopher J Long
- Hesperos, Inc., 12501 Research Parkway, Suite 100, Orlando, FL, 32826, USA
| | | | - James J Hickman
- Hesperos, Inc., 12501 Research Parkway, Suite 100, Orlando, FL, 32826, USA.
| |
Collapse
|
5
|
Strickland JB, Davis-Anderson K, Micheva-Viteva S, Twary S, Iyer R, Harris JF, Solomon EA. Optimization of Application-Driven Development of In Vitro Neuromuscular Junction Models. TISSUE ENGINEERING. PART B, REVIEWS 2022; 28:1180-1191. [PMID: 35018825 PMCID: PMC9805869 DOI: 10.1089/ten.teb.2021.0204] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Neuromuscular junctions (NMJs) are specialized synapses responsible for signal transduction between motor neurons (MNs) and skeletal muscle tissue. Malfunction at this site can result from developmental disorders, toxic environmental exposures, and neurodegenerative diseases leading to severe neurological dysfunction. Exploring these conditions in human or animal subjects is restricted by ethical concerns and confounding environmental factors. Therefore, in vitro NMJ models provide exciting opportunities for advancements in tissue engineering. In the last two decades, multiple NMJ prototypes and platforms have been reported, and each model system design is strongly tied to a specific application: exploring developmental physiology, disease modeling, or high-throughput screening. Directing the differentiation of stem cells into mature MNs and/or skeletal muscle for NMJ modeling has provided critical cues to recapitulate early-stage development. Patient-derived inducible pluripotent stem cells provide a personalized approach to investigating NMJ disease, especially when disease etiology cannot be resolved down to a specific gene mutation. Having reproducible NMJ culture replicates is useful for high-throughput screening to evaluate drug toxicity and determine the impact of environmental threat exposures. Cutting-edge bioengineering techniques have propelled this field forward with innovative microfabrication and design approaches allowing both two-dimensional and three-dimensional NMJ culture models. Many of these NMJ systems require further validation for broader application by regulatory agencies, pharmaceutical companies, and the general research community. In this summary, we present a comprehensive review on the current state-of-art research in NMJ models and discuss their ability to provide valuable insight into cell and tissue interactions. Impact statement In vitro neuromuscular junction (NMJ) models reveal the specialized mechanisms of communication between neurons and muscle tissue. This site can be disrupted by developmental disorders, toxic environmental exposures, or neurodegenerative diseases, which often lead to fatal outcomes and is therefore of critical importance to the medical community. Many bioengineering approaches for in vitro NMJ modeling have been designed to mimic development and disease; other approaches include in vitro NMJ models for high-throughput toxicology screening, providing a platform to limit or replace animal testing. This review describes various NMJ applications and the bioengineering advancements allowing for human NMJ characteristics to be more accurately recapitulated. While the extensive range of NMJ device structures has hindered standardization attempts, there is still a need to harmonize these devices for broader application and to continue advancing the field of NMJ modeling.
Collapse
Affiliation(s)
- Julie B. Strickland
- Bioscience Division, Los Alamos National Laboratory, Los Alamos, New Mexico, USA
| | - Katie Davis-Anderson
- Bioscience Division, Los Alamos National Laboratory, Los Alamos, New Mexico, USA
| | | | - Scott Twary
- Bioscience Division, Los Alamos National Laboratory, Los Alamos, New Mexico, USA
| | - Rashi Iyer
- Information System and Modeling, Los Alamos National Laboratory, Los Alamos, New Mexico, USA
| | | | - Emilia A. Solomon
- Bioscience Division, Los Alamos National Laboratory, Los Alamos, New Mexico, USA.,Address correspondence to: Emilia A. Solomon, PhD, Bioscience Division, Los Alamos National Laboratory, PO Box 1663 MS M888, Los Alamos, NM 87545, USA
| |
Collapse
|
6
|
ALS mutations in both human skeletal muscle and motoneurons differentially affects neuromuscular junction integrity and function. Biomaterials 2022; 289:121752. [DOI: 10.1016/j.biomaterials.2022.121752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 07/05/2022] [Accepted: 08/16/2022] [Indexed: 11/19/2022]
|
7
|
Takahashi H, Oikawa F, Takeda N, Shimizu T. Contraction Control of Aligned Myofiber Sheet Tissue by Parallel Oriented iPS Cell-derived Neurons. Tissue Eng Part A 2022; 28:661-671. [PMID: 35057641 DOI: 10.1089/ten.tea.2021.0202] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Fabrication and application of engineered complex tissues composed of different types of cells is a crucial milestone in the next phase of tissue engineering. The delicate organization structure of each tissue component and their physiological connections enable all the functions in the human body. In this study, cell sheet-based engineering allowed us to fabricate a complex myofiber sheet tissue using motor neurons derived from human iPS cells. In contrast with previous studies of other groups, a myofiber sheet with a biomimetic aligned structure was produced from human myoblasts using a striped-patterned thermoresponsive dish, which enabled manipulation of the sheet tissue by simply lowering the culture temperature. The myofiber sheet was transferred onto a gel that promotes functional maturation of human myofibers, resulting in production of contractile human muscle tissue. Just by seeding motor neurons onto the sheet tissue, all the neurons physically contacted to the aligned myofibers, and autonomously elongated in parallel to the myofiber orientation. In addition, the neurite outgrowth was enlarged by co-culturing on the myofiber sheet. The presence of the neurons enhanced clustering of myofiber acetylcholine receptors (AChR), typically found at the neuromuscular junctions (NMJs). Consequently, contraction behaviors of the myofiber sheet were regulated by neuronal signal transduction through NMJs. Muscle contraction was induced when the motor neurons were stimulated by glutamic acid, and effectively blocked by administration of d-tubocurarine as an antagonistic inhibitor for the AChR. The fibrin-based gel was useful as a culture environment for tissue maturation and as a favorable substrate for unobstructed contractions. Our neuron-muscle sheet tissue will be scalable by simply enlarging the micropatterned substrate and manipulable three-dimensionally; fabrication of a thick tissue and a bundle-like structured tissue will be possible just by layering multiple sheets or rolling up the sheet. Simplified control over self-orientation of neurite elongation will be advantageous for fabrication of such a large and complex tissue. Therefore, our methodology, established in this study, will be instrumental in future applications of regenerative medicine for locomotion apparatus.
Collapse
Affiliation(s)
| | - Fumiko Oikawa
- Waseda University, 13148, Shinjuku-ku, Tokyo, Japan;
| | - Naoya Takeda
- Waseda University, 13148, Shinjuku-ku, Tokyo, Japan;
| | | |
Collapse
|
8
|
Arjmand B, Kokabi Hamidpour S, Rabbani Z, Tayanloo-Beik A, Rahim F, Aghayan HR, Larijani B. Organ on a Chip: A Novel in vitro Biomimetic Strategy in Amyotrophic Lateral Sclerosis (ALS) Modeling. Front Neurol 2022; 12:788462. [PMID: 35111126 PMCID: PMC8802668 DOI: 10.3389/fneur.2021.788462] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Accepted: 12/20/2021] [Indexed: 12/20/2022] Open
Abstract
Amyotrophic lateral sclerosis is a pernicious neurodegenerative disorder that is associated with the progressive degeneration of motor neurons, the disruption of impulse transmission from motor neurons to muscle cells, and the development of mobility impairments. Clinically, muscle paralysis can spread to other parts of the body. Hence it may have adverse effects on swallowing, speaking, and even breathing, which serves as major problems facing these patients. According to the available evidence, no definite treatment has been found for amyotrophic lateral sclerosis (ALS) that results in a significant outcome, although some pharmacological and non-pharmacological treatments are currently applied that are accompanied by some positive effects. In other words, available therapies are only used to relieve symptoms without any significant treatment effects that highlight the importance of seeking more novel therapies. Unfortunately, the process of discovering new drugs with high therapeutic potential for ALS treatment is fraught with challenges. The lack of a broad view of the disease process from early to late-stage and insufficiency of preclinical studies for providing validated results prior to conducting clinical trials are other reasons for the ALS drug discovery failure. However, increasing the combined application of different fields of regenerative medicine, especially tissue engineering and stem cell therapy can be considered as a step forward to develop more novel technologies. For instance, organ on a chip is one of these technologies that can provide a platform to promote a comprehensive understanding of neuromuscular junction biology and screen candidate drugs for ALS in combination with pluripotent stem cells (PSCs). The structure of this technology is based on the use of essential components such as iPSC- derived motor neurons and iPSC-derived skeletal muscle cells on a single miniaturized chip for ALS modeling. Accordingly, an organ on a chip not only can mimic ALS complexities but also can be considered as a more cost-effective and time-saving disease modeling platform in comparison with others. Hence, it can be concluded that lab on a chip can make a major contribution as a biomimetic micro-physiological system in the treatment of neurodegenerative disorders such as ALS.
Collapse
Affiliation(s)
- Babak Arjmand
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
- *Correspondence: Babak Arjmand
| | - Shayesteh Kokabi Hamidpour
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Zahra Rabbani
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Akram Tayanloo-Beik
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Fakher Rahim
- Health Research Institute, Thalassemia, and Hemoglobinopathies Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Hamid Reza Aghayan
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Bagher Larijani
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
- Bagher Larijani
| |
Collapse
|
9
|
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: 1.8] [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
|
10
|
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: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [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
|
11
|
Solomon EA, Rooney AM, Rodriguez AM, Micheva-Viteva S, Bashir R, Iyer R, Harris JF. Neuromuscular Junction Model Optimized for Electrical Platforms. Tissue Eng Part C Methods 2021; 27:242-252. [PMID: 33599165 DOI: 10.1089/ten.tec.2020.0292] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Neuromuscular junctions (NMJs), specialized synapses between motor neurons and muscle fibers, are essential for muscle activity. A simple and reproducible cell-based in vitro NMJ platform is needed to test the impact of chemicals on the neuron-muscle communication. Our platform utilizes genetically modified neurons and muscle cells, optimized culture conditions, and commercially available multielectrode array system for recording action potentials. Neuronal cells (NSC34) were optogenetically modified with channelrhodopsin chimera to allow for simultaneous, light-mediated, millisecond-precise activation of neuronal population. This signal is propagated through functional synapses to the muscle fibers. Muscle cells (C2C12) were modified by incorporating gap junction protein (Connexin-43) to improve intracellular communication without affecting muscle differentiation. This communication between muscle fibers resulted in better signal propagation and signal strength. Optimized culture medium facilitated the growth and differentiation of both cell types together. Our system was validated using vecuronium, a muscle relaxant, which abolished the muscle response. This in vitro model provides a unique tool for establishing a NMJ platform that is easy to record and analyze. Potential applications include nondestructive long-term screening of drugs affecting the NMJ.
Collapse
Affiliation(s)
- Emilia A Solomon
- Bioscience Division, Los Alamos National Laboratory, Los Alamos, New Mexico, USA
| | - Allison M Rooney
- Bioscience Division, Los Alamos National Laboratory, Los Alamos, New Mexico, USA
| | - Arasely M Rodriguez
- Bioscience Division, Los Alamos National Laboratory, Los Alamos, New Mexico, USA
| | | | - Rashid Bashir
- Department of Bioengineering, Nick J. Holonyak Micro and Nanotechnology Laboratory, and Carle Illinois College of Medicine, University of Illinois Urbana-Champaign, Urbana, Illinois, USA
| | - Rashi Iyer
- Information System and Modeling, Los Alamos National Laboratory, Los Alamos, New Mexico, USA
| | | |
Collapse
|
12
|
Marshall KL, Farah MH. Axonal regeneration and sprouting as a potential therapeutic target for nervous system disorders. Neural Regen Res 2021; 16:1901-1910. [PMID: 33642358 PMCID: PMC8343323 DOI: 10.4103/1673-5374.308077] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
Nervous system disorders are prevalent health issues that will only continue to increase in frequency as the population ages. Dying-back axonopathy is a hallmark of many neurologic diseases and leads to axonal disconnection from their targets, which in turn leads to functional impairment. During the course of many of neurologic diseases, axons can regenerate or sprout in an attempt to reconnect with the target and restore synapse function. In amyotrophic lateral sclerosis (ALS), distal motor axons retract from neuromuscular junctions early in the disease-course before significant motor neuron death. There is evidence of compensatory motor axon sprouting and reinnervation of neuromuscular junctions in ALS that is usually quickly overtaken by the disease course. Potential drugs that enhance compensatory sprouting and encourage reinnervation may slow symptom progression and retain muscle function for a longer period of time in ALS and in other diseases that exhibit dying-back axonopathy. There remain many outstanding questions as to the impact of distinct disease-causing mutations on axonal outgrowth and regeneration, especially in regards to motor neurons derived from patient induced pluripotent stem cells. Compartmentalized microfluidic chambers are powerful tools for studying the distal axons of human induced pluripotent stem cells-derived motor neurons, and have recently been used to demonstrate striking regeneration defects in human motor neurons harboring ALS disease-causing mutations. Modeling the human neuromuscular circuit with human induced pluripotent stem cells-derived motor neurons will be critical for developing drugs that enhance axonal regeneration, sprouting, and reinnervation of neuromuscular junctions. In this review we will discuss compensatory axonal sprouting as a potential therapeutic target for ALS, and the use of compartmentalized microfluidic devices to find drugs that enhance regeneration and axonal sprouting of motor axons.
Collapse
Affiliation(s)
| | - Mohamed H Farah
- Department of Neurology at Johns Hopkins School of Medicine, Baltimore, MD, USA
| |
Collapse
|
13
|
Barrett P, Quick TJ, Mudera V, Player DJ. Generating intrafusal skeletal muscle fibres in vitro: Current state of the art and future challenges. J Tissue Eng 2020; 11:2041731420985205. [PMID: 34956586 PMCID: PMC8693220 DOI: 10.1177/2041731420985205] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Accepted: 12/12/2020] [Indexed: 01/18/2023] Open
Abstract
Intrafusal fibres are a specialised cell population in skeletal muscle, found within the muscle spindle. These fibres have a mechano-sensory capacity, forming part of the monosynaptic stretch-reflex arc, a key component responsible for proprioceptive function. Impairment of proprioception and associated dysfunction of the muscle spindle is linked with many neuromuscular diseases. Research to-date has largely been undertaken in vivo or using ex vivo preparations. These studies have provided a foundation for our understanding of muscle spindle physiology, however, the cellular and molecular mechanisms which underpin physiological changes are yet to be fully elucidated. Therefrom, the use of in vitro models has been proposed, whereby intrafusal fibres can be generated de novo. Although there has been progress, it is predominantly a developing and evolving area of research. This narrative review presents the current state of art in this area and proposes the direction of future work, with the aim of providing novel pre-clinical and clinical applications.
Collapse
Affiliation(s)
- Philip Barrett
- Centre for 3D Models of Health and Disease, Division of Surgery and Interventional Science, Faculty of Medical Sciences, University College London, London, UK
| | - Tom J Quick
- Peripheral Nerve Injury Research Unit, Royal National Orthopaedic Hospital, Stanmore, UK
- UCL Centre for Nerve Engineering, University College London, London, UK
| | - Vivek Mudera
- Centre for 3D Models of Health and Disease, Division of Surgery and Interventional Science, Faculty of Medical Sciences, University College London, London, UK
| | - Darren J Player
- Centre for 3D Models of Health and Disease, Division of Surgery and Interventional Science, Faculty of Medical Sciences, University College London, London, UK
| |
Collapse
|
14
|
Besser RR, Bowles AC, Alassaf A, Carbonero D, Maciel R, Saporta M, Agarwal A. A Chemically Defined Common Medium for Culture of C2C12 Skeletal Muscle and Human Induced Pluripotent Stem Cell Derived Spinal Spheroids. Cell Mol Bioeng 2020; 13:605-619. [PMID: 33281990 PMCID: PMC7704992 DOI: 10.1007/s12195-020-00624-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Accepted: 06/03/2020] [Indexed: 01/14/2023] Open
Abstract
INTRODUCTION Multicellular platforms and linked multi organ on chip devices are powerful tools for drug discovery, and basic mechanistic studies. Often, a critical constraint is defining a culture medium optimal for all cells present in the system. In this study, we focused on the key cells of the neuromuscular junction i.e., skeletal muscle and motor neurons. METHODS Formulation of a chemically defined medium for the co-culture of C2C12 skeletal muscle cells and human induced pluripotent stem cell (hiPSC) derived spinal spheroids (SpS) was optimized. C2C12 cells in 10 experimental media conditions and 2 topographies were evaluated over a 14-day maturation period to determine the ideal medium formulation for skeletal muscle tissue development. RESULTS During early maturation, overexpression of genes for myogenesis and myopathy was observed for several media conditions, corresponding to muscle delamination and death. Together, we identified 3 media formulations that allowed for more controlled differentiation, healthier muscle tissue, and long-term culture duration. This evidence was then used to select media formulations to culture SpS and subsequently assessed axonal growth. As axonal growth in SpS cultures was comparable in all selected media conditions, our data suggest that the neuronal basal medium with no added supplements is the ideal medium formulation for both cell types. CONCLUSIONS Optimization using both topographical cues and culture media formulations provides a comprehensive analyses of culture conditions that are vital to future applications for in vitro NMJ models.
Collapse
Affiliation(s)
- Rachel R. Besser
- Department of Biomedical Engineering, DJTMF Biomedical Nanotechnology Institute, University of Miami, 1251 Memorial Dr, MEA 203, Coral Gables, FL 33146 USA
| | - Annie C. Bowles
- Department of Biomedical Engineering, DJTMF Biomedical Nanotechnology Institute, University of Miami, 1251 Memorial Dr, MEA 203, Coral Gables, FL 33146 USA
| | - Ahmad Alassaf
- Department of Biomedical Engineering, DJTMF Biomedical Nanotechnology Institute, University of Miami, 1251 Memorial Dr, MEA 203, Coral Gables, FL 33146 USA
- Department of Medical Equipment Technology, College of Applied Medical Sciences, Majmaah University, Al-Majmaah, 11952 Saudi Arabia
| | - Daniel Carbonero
- Department of Biomedical Engineering, DJTMF Biomedical Nanotechnology Institute, University of Miami, 1251 Memorial Dr, MEA 203, Coral Gables, FL 33146 USA
| | - Renata Maciel
- Department of Neurology, University of Miami Miller School of Medicine, 1120 NW 14th St, Suite 1310, Miami, FL 33136 USA
| | - Mario Saporta
- Department of Neurology, University of Miami Miller School of Medicine, 1120 NW 14th St, Suite 1310, Miami, FL 33136 USA
| | - Ashutosh Agarwal
- Department of Biomedical Engineering, DJTMF Biomedical Nanotechnology Institute, University of Miami, 1251 Memorial Dr, MEA 203, Coral Gables, FL 33146 USA
| |
Collapse
|
15
|
Barbeau S, Tahraoui-Bories J, Legay C, Martinat C. Building neuromuscular junctions in vitro. Development 2020; 147:147/22/dev193920. [PMID: 33199350 DOI: 10.1242/dev.193920] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The neuromuscular junction (NMJ) has been the model of choice to understand the principles of communication at chemical synapses. Following groundbreaking experiments carried out over 60 years ago, many studies have focused on the molecular mechanisms underlying the development and physiology of these synapses. This Review summarizes the progress made to date towards obtaining faithful models of NMJs in vitro We provide a historical approach discussing initial experiments investigating NMJ development and function from Xenopus to mice, the creation of chimeric co-cultures, in vivo approaches and co-culture methods from ex vivo and in vitro derived cells, as well as the most recent developments to generate human NMJs. We discuss the benefits of these techniques and the challenges to be addressed in the future for promoting our understanding of development and human disease.
Collapse
Affiliation(s)
- Susie Barbeau
- Université de Paris, CNRS, SPPIN - Saints-Pères Paris Institute for the Neurosciences, F-75006 Paris, France
| | - Julie Tahraoui-Bories
- INSERM/UEPS UMR 861, Paris Saclay Université, I-STEM, 91100 Corbeil-Essonnes, France
| | - Claire Legay
- Université de Paris, CNRS, SPPIN - Saints-Pères Paris Institute for the Neurosciences, F-75006 Paris, France
| | - Cécile Martinat
- INSERM/UEPS UMR 861, Paris Saclay Université, I-STEM, 91100 Corbeil-Essonnes, France
| |
Collapse
|
16
|
Guo X, Smith V, Jackson M, Tran M, Thomas M, Patel A, Lorusso E, Nimbalkar S, Cai Y, McAleer CW, Wang Y, Long CJ, Hickman JJ. A Human-Based Functional NMJ System for Personalized ALS Modeling and Drug Testing. ADVANCED THERAPEUTICS 2020; 3:2000133. [PMID: 33709015 PMCID: PMC7942691 DOI: 10.1002/adtp.202000133] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Indexed: 01/26/2023]
Abstract
Loss of the neuromuscular junction (NMJ) is an early and critical hallmark in all forms of ALS. The study design was to develop a functional NMJ disease model by integrating motoneurons (MNs) differentiated from multiple ALS-patients' induced pluripotent stem cells (iPSCs) and primary human muscle into a chambered system. NMJ functionality was tested by recording myotube contractions while stimulating MNs by field electrodes and a set of clinically relevant parameters were defined to characterize the NMJ function. Three ALS lines were analyzed, 2 with SOD1 mutations and 1 with a FUS mutation. The ALS-MNs reproduced pathological phenotypes, including increased axonal varicosities, reduced axonal branching and elongation and increased excitability. These MNs formed functional NMJs with wild type muscle, but with significant deficits in NMJ quantity, fidelity and fatigue index. Furthermore, treatment with the Deana protocol was found to correct the NMJ deficits in all the ALS mutant lines tested. Quantitative analysis also revealed the variations inherent in each mutant lines. This functional NMJ system provides a platform for the study of both fALS and sALS and has the capability of being adapted into subtype-specific or patient-specific models for ALS etiological investigation and patient stratification for drug testing.
Collapse
Affiliation(s)
- Xiufang Guo
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway, Suite 400, Orlando, FL 32826, USA
| | - Virginia Smith
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway, Suite 400, Orlando, FL 32826, USA
| | - Max Jackson
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway, Suite 400, Orlando, FL 32826, USA
| | - My Tran
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway, Suite 400, Orlando, FL 32826, USA
| | - Michael Thomas
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway, Suite 400, Orlando, FL 32826, USA
| | - Aakash Patel
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway, Suite 400, Orlando, FL 32826, USA
| | - Eric Lorusso
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway, Suite 400, Orlando, FL 32826, USA
| | - Siddharth Nimbalkar
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway, Suite 400, Orlando, FL 32826, USA
| | - Yunqing Cai
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway, Suite 400, Orlando, FL 32826, USA
| | - Christopher W. McAleer
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway, Suite 400, Orlando, FL 32826, USA
| | - Ying Wang
- Department of Biomedical Engineering, 305 Weill Hall, Cornell University, Ithaca, NY, 14853, USA
| | - Christopher J. Long
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway, Suite 400, Orlando, FL 32826, USA
| | - James J. Hickman
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway, Suite 400, Orlando, FL 32826, USA
| |
Collapse
|
17
|
Castellanos-Montiel MJ, Velasco I, Escobedo-Avila I. Modeling the neuromuscular junction in vitro: an approach to study neuromuscular junction disorders. Ann N Y Acad Sci 2020; 1488:3-15. [PMID: 33040338 DOI: 10.1111/nyas.14504] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 08/24/2020] [Accepted: 09/09/2020] [Indexed: 12/14/2022]
Abstract
The neuromuscular junction (NMJ) is a specialized structure that works as an interface to translate the action potential of the presynaptic motor neuron (MN) in the contraction of the postsynaptic myofiber. The design of appropriate experimental models is essential to have efficient and reliable approaches to study NMJ development and function, but also to generate conditions that recapitulate distinct features of diseases. Initial studies relied on the use of tissue slices maintained under the same environment and in which single motor axons were difficult to trace. Later, MNs and muscle cells were obtained from primary cultures or differentiation of progenitors and cocultured as monolayers; however, the tissue architecture was lost. Current approaches include self-assembling 3D structures or the incorporation of biomaterials with cells to generate engineered tissues, although the incorporation of Schwann cells remains a challenge. Thus, numerous investigations have established different NMJ models, some of which are quite complex and challenging. Our review summarizes the in vitro models that have emerged in recent years to coculture MNs and skeletal muscle, trying to mimic the healthy and diseased NMJ. We expect our review may serve as a reference for choosing the appropriate experimental model for the required purposes of investigation.
Collapse
Affiliation(s)
- María José Castellanos-Montiel
- Instituto de Fisiología Celular-Neurociencias, Universidad Nacional Autónoma de México (UNAM), Mexico City, Mexico.,Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montréal, Quebec, Canada
| | - Iván Velasco
- Instituto de Fisiología Celular-Neurociencias, Universidad Nacional Autónoma de México (UNAM), Mexico City, Mexico.,Laboratorio de Reprogramación Celular, Instituto Nacional de Neurología y Neurocirugía "Manuel Velasco Suárez", Mexico City, Mexico
| | - Itzel Escobedo-Avila
- Instituto de Fisiología Celular-Neurociencias, Universidad Nacional Autónoma de México (UNAM), Mexico City, Mexico
| |
Collapse
|
18
|
Saini J, Faroni A, Reid AJ, Mamchaoui K, Mouly V, Butler-Browne G, Lightfoot AP, McPhee JS, Degens H, Al-Shanti N. A Novel Bioengineered Functional Motor Unit Platform to Study Neuromuscular Interaction. J Clin Med 2020; 9:jcm9103238. [PMID: 33050427 PMCID: PMC7599749 DOI: 10.3390/jcm9103238] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Revised: 10/01/2020] [Accepted: 10/06/2020] [Indexed: 12/18/2022] Open
Abstract
Background: In many neurodegenerative and muscular disorders, and loss of innervation in sarcopenia, improper reinnervation of muscle and dysfunction of the motor unit (MU) are key pathogenic features. In vivo studies of MUs are constrained due to difficulties isolating and extracting functional MUs, so there is a need for a simplified and reproducible system of engineered in vitro MUs. Objective: to develop and characterise a functional MU model in vitro, permitting the analysis of MU development and function. Methods: an immortalised human myoblast cell line was co-cultured with rat embryo spinal cord explants in a serum-free/growth fact media. MUs developed and the morphology of their components (neuromuscular junction (NMJ), myotubes and motor neurons) were characterised using immunocytochemistry, phase contrast and confocal microscopy. The function of the MU was evaluated through live observations and videography of spontaneous myotube contractions after challenge with cholinergic antagonists and glutamatergic agonists. Results: blocking acetylcholine receptors with α-bungarotoxin resulted in complete, cessation of myotube contractions, which was reversible with tubocurarine. Furthermore, myotube activity was significantly higher with the application of L-glutamic acid. All these observations indicate the formed MU are functional. Conclusion: a functional nerve-muscle co-culture model was established that has potential for drug screening and pathophysiological studies of neuromuscular interactions.
Collapse
Affiliation(s)
- Jasdeep Saini
- Musculoskeletal Science & Sports Medicine Research Centre, Department of Life Sciences, Manchester Metropolitan University, Manchester M1 5GD, UK; (J.S.); (A.P.L.); (H.D.)
| | - Alessandro Faroni
- Manchester Academic Health Science Centre, Blond McIndoe Laboratories, Division of Cell Matrix Biology and Regenerative Medicine, Faculty of Biology Medicine and Health, School of Biological Sciences, University of Manchester, Manchester M1 7DN, UK; (A.F.); (A.J.R.)
- Manchester Academic Health Science Centre, Department of Plastic Surgery & Burns, Manchester University NHS Foundation Trust, Wythenshawe Hospital, Manchester M23 9LT, UK
| | - Adam J. Reid
- Manchester Academic Health Science Centre, Blond McIndoe Laboratories, Division of Cell Matrix Biology and Regenerative Medicine, Faculty of Biology Medicine and Health, School of Biological Sciences, University of Manchester, Manchester M1 7DN, UK; (A.F.); (A.J.R.)
- Manchester Academic Health Science Centre, Department of Plastic Surgery & Burns, Manchester University NHS Foundation Trust, Wythenshawe Hospital, Manchester M23 9LT, UK
| | - Kamel Mamchaoui
- Center for Research in Myology, Sorbonne Université-INSERM, 75013 Paris, France; (K.M.); (V.M.); (G.B.-B.)
| | - Vincent Mouly
- Center for Research in Myology, Sorbonne Université-INSERM, 75013 Paris, France; (K.M.); (V.M.); (G.B.-B.)
| | - Gillian Butler-Browne
- Center for Research in Myology, Sorbonne Université-INSERM, 75013 Paris, France; (K.M.); (V.M.); (G.B.-B.)
| | - Adam P. Lightfoot
- Musculoskeletal Science & Sports Medicine Research Centre, Department of Life Sciences, Manchester Metropolitan University, Manchester M1 5GD, UK; (J.S.); (A.P.L.); (H.D.)
| | - Jamie S. McPhee
- Department of Sport and Exercise Sciences, Manchester Metropolitan University, Manchester M1 5GD, UK;
| | - Hans Degens
- Musculoskeletal Science & Sports Medicine Research Centre, Department of Life Sciences, Manchester Metropolitan University, Manchester M1 5GD, UK; (J.S.); (A.P.L.); (H.D.)
- Institute of Sport Science and Innovations, Lithuanian Sports University, LT-44221 Kaunas, Lithuania
| | - Nasser Al-Shanti
- Musculoskeletal Science & Sports Medicine Research Centre, Department of Life Sciences, Manchester Metropolitan University, Manchester M1 5GD, UK; (J.S.); (A.P.L.); (H.D.)
- Correspondence:
| |
Collapse
|
19
|
Vila OF, Qu Y, Vunjak-Novakovic G. In vitro models of neuromuscular junctions and their potential for novel drug discovery and development. Expert Opin Drug Discov 2019; 15:307-317. [PMID: 31846349 DOI: 10.1080/17460441.2020.1700225] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Introduction: Neuromuscular Junctions (NMJs) are the synapses between motor neurons and skeletal muscle fibers, and they are responsible for voluntary motor function. NMJs are affected at early stages of numerous neurodegenerative and neuroimmunological diseases. Due to the difficulty of systematically studying and manipulating NMJs in live subjects, in vitro systems with human tissue models would provide a powerful complement to simple cell cultures and animal models for mechanistic and drug development studies.Areas covered: The authors review the latest advances in in vitro models of NMJs, from traditional cell co-culture systems to novel tissue culture approaches, with focus on disease modeling and drug testing.Expert opinion: In recent years, more sophisticated in vitro models of human NMJs have been established. The combination of human stem cell technology with advanced tissue culture systems has resulted in systems that better recapitulate the human NMJ structure and function, and thereby allow for high-throughput quantitative functional measurements under both healthy and diseased conditions. Although they still have limitations, these advanced systems are increasingly demonstrating their utility for evaluating new therapies for motoneuron and autoimmune neuromuscular diseases, and we expect them to become an integral part of the drug discovery process in the near future.
Collapse
Affiliation(s)
- Olaia F Vila
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Yihuai Qu
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | | |
Collapse
|
20
|
Najjar SA, Smith AST, Long CJ, McAleer CW, Cai Y, Srinivasan B, Martin C, Vandenburgh HH, Hickman JJ. A multiplexed in vitro assay system for evaluating human skeletal muscle functionality in response to drug treatment. Biotechnol Bioeng 2019; 117:736-747. [PMID: 31758543 DOI: 10.1002/bit.27231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Revised: 10/29/2019] [Accepted: 11/19/2019] [Indexed: 11/07/2022]
Abstract
In vitro systems that mimic organ functionality have become increasingly important tools in drug development studies. Systems that measure the functional properties of skeletal muscle are beneficial to compound screening studies and also for integration into multiorgan devices. To date, no studies have investigated human skeletal muscle responses to drug treatments at the single myotube level in vitro. This report details a microscale cantilever chip-based assay system for culturing individual human myotubes. The cantilevers, along with a laser and photo-detector system, enable measurement of myotube contractions in response to broad-field electrical stimulation. This system was used to obtain baseline functional parameters for untreated human myotubes, including peak contractile force and time-to-fatigue data. The cultured myotubes were then treated with known myotoxic compounds and the resulting functional changes were compared to baseline measurements as well as known physiological responses in vivo. The collected data demonstrate the system's capacity for screening direct effects of compound action on individual human skeletal myotubes in a reliable, reproducible, and noninvasive manner. Furthermore, it has the potential to be utilized for high-content screening, disease modeling, and exercise studies of human skeletal muscle performance utilizing iPSCs derived from specific patient populations such as the muscular dystrophies.
Collapse
Affiliation(s)
- Sarah A Najjar
- NanoScience Technology Center, University of Central Florida, Orlando, Florida
| | - Alexander S T Smith
- NanoScience Technology Center, University of Central Florida, Orlando, Florida
| | - Christopher J Long
- NanoScience Technology Center, University of Central Florida, Orlando, Florida
| | | | - Yunqing Cai
- NanoScience Technology Center, University of Central Florida, Orlando, Florida
| | - Balaji Srinivasan
- NanoScience Technology Center, University of Central Florida, Orlando, Florida
| | - Candace Martin
- NanoScience Technology Center, University of Central Florida, Orlando, Florida
| | - Herman H Vandenburgh
- Department of Pathology and Laboratory Medicine, Brown University, Providence, Rhode Island
| | - James J Hickman
- NanoScience Technology Center, University of Central Florida, Orlando, Florida
| |
Collapse
|
21
|
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: 70] [Impact Index Per Article: 11.7] [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
|
22
|
Natarajan A, Sethumadhavan A, Krishnan UM. Toward Building the Neuromuscular Junction: In Vitro Models To Study Synaptogenesis and Neurodegeneration. ACS OMEGA 2019; 4:12969-12977. [PMID: 31460423 PMCID: PMC6682064 DOI: 10.1021/acsomega.9b00973] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Accepted: 07/04/2019] [Indexed: 06/10/2023]
Abstract
The neuromuscular junction (NMJ) is a unique, specialized chemical synapse that plays a crucial role in transmitting and amplifying information from spinal motor neurons to skeletal muscles. NMJ complexity ensures closely intertwined interactions between numerous synaptic vesicles, signaling molecules, ion channels, motor neurons, glia, and muscle fibers, making it difficult to dissect the underlying mechanisms and factors affecting neurodegeneration and muscle loss. Muscle fiber or motor neuron cell death followed by rapid axonal degeneration due to injury or disease has a debilitating effect on movement and behavior, which adversely affects the quality of life. It thus becomes imperative to study the synapse and intercellular signaling processes that regulate plasticity at the NMJ and elucidate mechanisms and pathways at the cellular level. Studies using in vitro 2D cell cultures have allowed us to gain a fundamental understanding of how the NMJ functions. However, they do not provide information on the intricate signaling networks that exist between NMJs and the biological environment. The advent of 3D cell cultures and microfluidic lab-on-a-chip technologies has opened whole new avenues to explore the NMJ. In this perspective, we look at the challenges involved in building a functional NMJ and the progress made in generating models for studying the NMJ, highlighting the current and future applications of these models.
Collapse
Affiliation(s)
- Anupama Natarajan
- Centre
for Nanotechnology & Advanced Biomaterials (CeNTAB), School of Chemical
& Biotechnology, and School of Arts, Science & Humanities, SASTRA Deemed University, Thanjavur 613 401, India
| | - Anjali Sethumadhavan
- Centre
for Nanotechnology & Advanced Biomaterials (CeNTAB), School of Chemical
& Biotechnology, and School of Arts, Science & Humanities, SASTRA Deemed University, Thanjavur 613 401, India
| | - Uma Maheswari Krishnan
- Centre
for Nanotechnology & Advanced Biomaterials (CeNTAB), School of Chemical
& Biotechnology, and School of Arts, Science & Humanities, SASTRA Deemed University, Thanjavur 613 401, India
| |
Collapse
|
23
|
Arrigoni C, Petta D, Bersini S, Mironov V, Candrian C, Moretti M. Engineering complex muscle-tissue interfaces through microfabrication. Biofabrication 2019; 11:032004. [PMID: 31042682 DOI: 10.1088/1758-5090/ab1e7c] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Skeletal muscle is a tissue with a complex and hierarchical architecture that influences its functional properties. In order to exert its contractile function, muscle tissue is connected to neural, vascular and connective compartments, comprising finely structured interfaces which are orchestrated by multiple signalling pathways. Pathological conditions such as dystrophies and trauma, or physiological situations such as exercise and aging, modify the architectural organization of these structures, hence affecting muscle functionality. To overcome current limitations of in vivo and standard in vitro models, microfluidics and biofabrication techniques have been applied to better reproduce the microarchitecture and physicochemical environment of human skeletal muscle tissue. In the present review, we aim to critically discuss the role of those techniques, taken individually or in combination, in the generation of models that mimic the complex interfaces between muscle tissue and neural/vascular/tendon compartments. The exploitation of either microfluidics or biofabrication to model different muscle interfaces has led to the development of constructs with an improved spatial organization, thus presenting a better functionality as compared to standard models. However, the achievement of models replicating muscle-tissue interfaces with adequate architecture, presence of fundamental proteins and recapitulation of signalling pathways is still far from being achieved. Increased integration between microfluidics and biofabrication, providing the possibility to pattern cells in predetermined structures with higher resolution, will help to reproduce the hierarchical and heterogeneous structure of skeletal muscle interfaces. Such strategies will further improve the functionality of these techniques, providing a key contribution towards the study of skeletal muscle functions in physiology and pathology.
Collapse
Affiliation(s)
- Chiara Arrigoni
- Regenerative Medicine Technologies Lab, Ente Ospedaliero Cantonale (EOC), Via Tesserete 46, 6900 Lugano, Switzerland
| | | | | | | | | | | |
Collapse
|
24
|
Singh T, Vazquez M. Time-Dependent Addition of Neuronal and Schwann Cells Increase Myotube Viability and Length in an In Vitro Tri-culture Model of the Neuromuscular Junction. REGENERATIVE ENGINEERING AND TRANSLATIONAL MEDICINE 2019. [DOI: 10.1007/s40883-019-00095-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
25
|
Luo B, Tian L, Chen N, Ramakrishna S, Thakor N, Yang IH. Electrospun nanofibers facilitate better alignment, differentiation, and long-term culture in an in vitro model of the neuromuscular junction (NMJ). Biomater Sci 2019; 6:3262-3272. [PMID: 30402630 DOI: 10.1039/c8bm00720a] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The neuromuscular junction (NMJ) is a specialized synapse between motor neurons and the muscle fibers they innervate. Due to the complexity of various signalling molecules and pathways, in vivo NMJs are difficult to study. Therefore, in vitro motor neuron-muscle co-culture plays a pivotal role in studying the mechanisms of NMJ formation associated with neurodegenerative diseases. There is a growing need to develop novel methodologies that can be used to develop long-term cultures of NMJs. To date, there have been few studies on NMJ development and long-term maintenance of the system, which is also the main challenge for the current in vitro models of NMJs. In this study, we demonstrate a long-term co-culture system of primary embryonic motor neurons from Sprague-Dawley rats and C2C12 cells on both random and aligned electrospun polylactic acid (PLA) nanofibrous scaffolds. This is the first study to explore the role of electrospun nanofibers in the long-term maintenance of NMJs. PLA nanofibrous scaffolds provide better contact guidance for C2C12 cells aligning along the fibers, thus guiding myotube formation. We can only maintain the co-culture system on a conventional glass substrate for 2 weeks, whilst 55% and 70% of the cells still survived on random and aligned PLA substrates after 7 weeks. Our nanofiber-based long-term co-culture system is used as an important tool for the fundamental research of NMJs.
Collapse
Affiliation(s)
- Baiwen Luo
- Singapore Institute for Neurotechnology, National University of Singapore, 28 Medical Drive, #05-COR, Singapore 119077. inhong.yang.@ku.ac.ae
| | | | | | | | | | | |
Collapse
|
26
|
Abd Al Samid M, McPhee JS, Saini J, McKay TR, Fitzpatrick LM, Mamchaoui K, Bigot A, Mouly V, Butler-Browne G, Al-Shanti N. A functional human motor unit platform engineered from human embryonic stem cells and immortalized skeletal myoblasts. STEM CELLS AND CLONING-ADVANCES AND APPLICATIONS 2018; 11:85-93. [PMID: 30519053 PMCID: PMC6233953 DOI: 10.2147/sccaa.s178562] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Background Although considerable research on neuromuscular junctions (NMJs) has been conducted, the prospect of in vivo NMJ studies is limited and these studies are challenging to implement. Therefore, there is a clear unmet need to develop a feasible, robust, and physiologically relevant in vitro NMJ model. Objective We aimed to establish a novel functional human NMJs platform, which is serum and neural complex media/neural growth factor-free, using human immortalized myoblasts and human embryonic stem cells (hESCs)-derived neural progenitor cells (NPCs) that can be used to understand the mechanisms of NMJ development and degeneration. Methods Immortalized human myoblasts were co-cultured with hESCs derived committed NPCs. Over the course of the 7 days myoblasts differentiated into myotubes and NPCs differentiated into motor neurons. Results Neuronal axon sprouting branched to form multiple NMJ innervation sites along the myotubes and the myotubes showed extensive, spontaneous contractile activity. Choline acetyltransferase and βIII-tubulin immunostaining confirmed that the NPCs had matured into cholinergic motor neurons. Postsynaptic site of NMJs was further characterized by staining dihydropyridine receptors, ryanodine receptors, and acetylcholine receptors by α-bungarotoxin. Conclusion We established a functional human motor unit platform for in vitro investigations. Thus, this co-culture system can be used as a novel platform for 1) drug discovery in the treatment of neuromuscular disorders, 2) deciphering vital features of NMJ formation, regulation, maintenance, and repair, and 3) exploring neuromuscular diseases, age-associated degeneration of the NMJ, muscle aging, and diabetic neuropathy and myopathy.
Collapse
Affiliation(s)
- Marwah Abd Al Samid
- Healthcare Science Research Institute, School of Healthcare Science, Manchester Metropolitan University, Manchester, UK,
| | - Jamie S McPhee
- Department of Sport and Exercise Science, Manchester Metropolitan University, Manchester, UK
| | - Jasdeep Saini
- Healthcare Science Research Institute, School of Healthcare Science, Manchester Metropolitan University, Manchester, UK,
| | - Tristan R McKay
- Healthcare Science Research Institute, School of Healthcare Science, Manchester Metropolitan University, Manchester, UK,
| | - Lorna M Fitzpatrick
- Healthcare Science Research Institute, School of Healthcare Science, Manchester Metropolitan University, Manchester, UK,
| | - Kamel Mamchaoui
- Center for Research in Myology, Sorbonne Université-INSERM, Paris, France
| | - Anne Bigot
- Center for Research in Myology, Sorbonne Université-INSERM, Paris, France
| | - Vincent Mouly
- Center for Research in Myology, Sorbonne Université-INSERM, Paris, France
| | | | - Nasser Al-Shanti
- Healthcare Science Research Institute, School of Healthcare Science, Manchester Metropolitan University, Manchester, UK,
| |
Collapse
|
27
|
Al Samid MA, Al-Shanti N, Odeh M. Motor Neuron-Skeletal Muscle Co Culture Model: A Potential Novel in Vitro and Computaional Platform to Investigate Cancer Cachexia. 2018 1ST INTERNATIONAL CONFERENCE ON CANCER CARE INFORMATICS (CCI) 2018. [DOI: 10.1109/cancercare.2018.8618261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/02/2023]
|
28
|
Development of Microplatforms to Mimic the In Vivo Architecture of CNS and PNS Physiology and Their Diseases. Genes (Basel) 2018; 9:genes9060285. [PMID: 29882823 PMCID: PMC6027402 DOI: 10.3390/genes9060285] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Revised: 05/28/2018] [Accepted: 05/31/2018] [Indexed: 12/16/2022] Open
Abstract
Understanding the mechanisms that govern nervous tissues function remains a challenge. In vitro two-dimensional (2D) cell culture systems provide a simplistic platform to evaluate systematic investigations but often result in unreliable responses that cannot be translated to pathophysiological settings. Recently, microplatforms have emerged to provide a better approximation of the in vivo scenario with better control over the microenvironment, stimuli and structure. Advances in biomaterials enable the construction of three-dimensional (3D) scaffolds, which combined with microfabrication, allow enhanced biomimicry through precise control of the architecture, cell positioning, fluid flows and electrochemical stimuli. This manuscript reviews, compares and contrasts advances in nervous tissues-on-a-chip models and their applications in neural physiology and disease. Microplatforms used for neuro-glia interactions, neuromuscular junctions (NMJs), blood-brain barrier (BBB) and studies on brain cancer, metastasis and neurodegenerative diseases are addressed. Finally, we highlight challenges that can be addressed with interdisciplinary efforts to achieve a higher degree of biomimicry. Nervous tissue microplatforms provide a powerful tool that is destined to provide a better understanding of neural health and disease.
Collapse
|
29
|
Santhanam N, Kumanchik L, Guo X, Sommerhage F, Cai Y, Jackson M, Martin C, Saad G, McAleer CW, Wang Y, Lavado A, Long CJ, Hickman JJ. Stem cell derived phenotypic human neuromuscular junction model for dose response evaluation of therapeutics. Biomaterials 2018; 166:64-78. [PMID: 29547745 PMCID: PMC5866791 DOI: 10.1016/j.biomaterials.2018.02.047] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Revised: 02/20/2018] [Accepted: 02/24/2018] [Indexed: 01/01/2023]
Abstract
There are currently no functional neuromuscular junction (hNMJ) systems composed of human cells that could be used for drug evaluations or toxicity testing in vitro. These systems are needed to evaluate NMJs for diseases such as amyotrophic lateral sclerosis, spinal muscular atrophy or other neurodegenerative diseases or injury states. There are certainly no model systems, animal or human, that allows for isolated treatment of motoneurons or muscle capable of generating dose response curves to evaluate pharmacological activity of these highly specialized functional units. A system was developed in which human myotubes and motoneurons derived from stem cells were cultured in a serum-free medium in a BioMEMS construct. The system is composed of two chambers linked by microtunnels to enable axonal outgrowth to the muscle chamber that allows separate stimulation of each component and physiological NMJ function and MN stimulated tetanus. The muscle's contractions, induced by motoneuron activation or direct electrical stimulation, were monitored by image subtraction video recording for both frequency and amplitude. Bungarotoxin, BOTOX® and curare dose response curves were generated to demonstrate pharmacological relevance of the phenotypic screening device. This quantifiable functional hNMJ system establishes a platform for generating patient-specific NMJ models by including patient-derived iPSCs.
Collapse
Affiliation(s)
- Navaneetha Santhanam
- Hybrid Systems Lab, NanoScience Technology Center, University of Central Florida, 12424 Research Parkway, Suite 400, Orlando, FL 32826, USA
| | - Lee Kumanchik
- Hybrid Systems Lab, NanoScience Technology Center, University of Central Florida, 12424 Research Parkway, Suite 400, Orlando, FL 32826, USA
| | - Xiufang Guo
- Hybrid Systems Lab, NanoScience Technology Center, University of Central Florida, 12424 Research Parkway, Suite 400, Orlando, FL 32826, USA
| | - Frank Sommerhage
- Hybrid Systems Lab, NanoScience Technology Center, University of Central Florida, 12424 Research Parkway, Suite 400, Orlando, FL 32826, USA
| | - Yunqing Cai
- Hybrid Systems Lab, NanoScience Technology Center, University of Central Florida, 12424 Research Parkway, Suite 400, Orlando, FL 32826, USA
| | - Max Jackson
- Hybrid Systems Lab, NanoScience Technology Center, University of Central Florida, 12424 Research Parkway, Suite 400, Orlando, FL 32826, USA
| | - Candace Martin
- Hybrid Systems Lab, NanoScience Technology Center, University of Central Florida, 12424 Research Parkway, Suite 400, Orlando, FL 32826, USA
| | - George Saad
- Hybrid Systems Lab, NanoScience Technology Center, University of Central Florida, 12424 Research Parkway, Suite 400, Orlando, FL 32826, USA
| | - Christopher W. McAleer
- Hybrid Systems Lab, NanoScience Technology Center, University of Central Florida, 12424 Research Parkway, Suite 400, Orlando, FL 32826, USA
| | - Ying Wang
- Hybrid Systems Lab, NanoScience Technology Center, University of Central Florida, 12424 Research Parkway, Suite 400, Orlando, FL 32826, USA,Department of Biomedical Engineering, 305 Weill Hall, Cornell University, Ithaca, NY, 14853, USA
| | - Andrea Lavado
- Hybrid Systems Lab, NanoScience Technology Center, University of Central Florida, 12424 Research Parkway, Suite 400, Orlando, FL 32826, USA
| | - Christopher J. Long
- Hybrid Systems Lab, NanoScience Technology Center, University of Central Florida, 12424 Research Parkway, Suite 400, Orlando, FL 32826, USA
| | - James J. Hickman
- Hybrid Systems Lab, NanoScience Technology Center, University of Central Florida, 12424 Research Parkway, Suite 400, Orlando, FL 32826, USA,correspondence:
| |
Collapse
|
30
|
Bucchia M, Merwin SJ, Re DB, Kariya S. Limitations and Challenges in Modeling Diseases Involving Spinal Motor Neuron Degeneration in Vitro. Front Cell Neurosci 2018; 12:61. [PMID: 29559895 PMCID: PMC5845677 DOI: 10.3389/fncel.2018.00061] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Accepted: 02/20/2018] [Indexed: 12/12/2022] Open
Abstract
Pathogenic conditions involving degeneration of spinal motor neurons (MNs), such as amyotrophic lateral sclerosis, sarcopenia, and spinal cord injury, mostly occur in individuals whose spinal MNs are fully mature. There is currently no effective treatment to prevent death or promote axonal regeneration of the spinal MNs affected in these patients. To increase our understanding and find a cure for such conditions, easily controllable and monitorable cell culture models allow for a better dissection of certain molecular and cellular events that cannot be teased apart in whole organism models. To date, various types of spinal MN cultures have been described. Yet these models are all based on the use of immature neurons or neurons uncharacterized for their degree of maturity after being isolated and cultured. Additionally, studying only MNs cannot give a comprehensive and complete view of the neurodegenerative processes usually involving other cell types. To date, there is no confirmed in vitro model faithfully emulating disease or injury of the mature spinal MNs. In this review, we summarize the different limitations of currently available culture models, and discuss the challenges that have to be overcome for developing more reliable and translational platforms for the in vitro study of spinal MN degeneration.
Collapse
Affiliation(s)
- Monica Bucchia
- Department of Neurology, Columbia University Medical Center, New York, NY, United States
| | - Samantha J Merwin
- Department of Environmental Health Sciences, Columbia University Medical Center, New York, NY, United States
| | - Diane B Re
- Department of Environmental Health Sciences, Columbia University Medical Center, New York, NY, United States
| | - Shingo Kariya
- Department of Neurology, Columbia University Medical Center, New York, NY, United States
| |
Collapse
|
31
|
Jodat YA, Kang MG, Kiaee K, Kim GJ, Martinez AFH, Rosenkranz A, Bae H, Shin SR. Human-Derived Organ-on-a-Chip for Personalized Drug Development. Curr Pharm Des 2018; 24:5471-5486. [PMID: 30854951 PMCID: PMC6587585 DOI: 10.2174/1381612825666190308150055] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Accepted: 02/26/2019] [Indexed: 12/22/2022]
Abstract
To reduce the required capital and time investment in the development of new pharmaceutical agents, there is an urgent need for preclinical drug testing models that are predictive of drug response in human tissues or organs. Despite tremendous advancements and rigorous multistage screening of drug candidates involving computational models, traditional cell culture platforms, animal models and most recently humanized animals, there is still a large deficit in our ability to predict drug response in patient groups and overall attrition rates from phase 1 through phase 4 of clinical studies remain well above 90%. Organ-on-a-chip (OOC) platforms have proven potential in providing tremendous flexibility and robustness in drug screening and development by employing engineering techniques and materials. More importantly, in recent years, there is a clear upward trend in studies that utilize human-induced pluripotent stem cell (hiPSC) to develop personalized tissue or organ models. Additionally, integrated multiple organs on the single chip with increasingly more sophisticated representation of absorption, distribution, metabolism, excretion and toxicity (ADMET) process are being utilized to better understand drug interaction mechanisms in the human body and thus showing great potential to better predict drug efficacy and safety. In this review, we summarize these advances, highlighting studies that took the next step to clinical trials and research areas with the utmost potential and discuss the role of the OOCs in the overall drug discovery process at a preclinical and clinical stage, as well as outline remaining challenges.
Collapse
Affiliation(s)
- Yasamin A Jodat
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA 02139, United States
- Department of Mechanical Engineering, Stevens Institute of Technology, New Jersey, 07030, United States
| | - Min G Kang
- Department of Bioindustrial Technologies, College of Animal Bioscience and Technology, Konkuk University, Seoul 05029, Korea
| | - Kiavash Kiaee
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA 02139, United States
- Department of Mechanical Engineering, Stevens Institute of Technology, New Jersey, 07030, United States
| | - Gyeong J Kim
- Department of Bioindustrial Technologies, College of Animal Bioscience and Technology, Konkuk University, Seoul 05029, Korea
| | - Angel F H Martinez
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA 02139, United States
- ALPHA Medical Leadership Program, Anahuac University, School of Medicine, Mexico
| | - Aliza Rosenkranz
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA 02139, United States
| | - Hojae Bae
- Department of Stem Cell and Regenerative Biotechnology, KU Convergence Science and Technololgy Institute, Konkuk University, Seoul, 05029, Korea
| | - Su R Shin
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA 02139, United States
| |
Collapse
|
32
|
Charoensook SN, Williams DJ, Chakraborty S, Leong KW, Vunjak-Novakovic G. Bioreactor model of neuromuscular junction with electrical stimulation for pharmacological potency testing. Integr Biol (Camb) 2017; 9:956-967. [PMID: 29168874 PMCID: PMC5725265 DOI: 10.1039/c7ib00144d] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
In vitro models of the neuromuscular junction (NMJ) are emerging as a valuable tool to study synaptogenesis, synaptic maintenance, and pathogenesis of neurodegenerative diseases. Many models have previously been developed using a variety of cell sources for skeletal muscle and motoneurons. These models can advanced by integrating beneficial features of the native developmental milieu of the NMJ. We created a functional in vitro model of NMJ by bioreactor cultivation of transdifferentiated myocytes and stem cell-derived motoneurons, in the presence of electrical stimulation. In conjunction with a coculture medium, electrical stimulation resulted in improved maturation and function of motoneurons and myocytes, as evidenced by mature cellular structures, increased expression of neuronal and muscular genes, clusterization of acetylcholine receptors (AChRs) in the vicinity of motoneurons, and the response to glutamate stimulation. To validate the model and demonstrate its utility for pharmacological testing, we documented the potency of drugs that affect key pathways during NMJ signal transduction: (i) acetylcholine (ACh) synthesis, (ii) ACh vesicular storage, (iii) ACh synaptic release, (iv) AChR activation, and (v) ACh inactivation in the synaptic cleft. The model properly responded to the drugs in a concentration-dependent manner. We thus propose that this in vitro model of NMJ could be used as a platform in pharmacological screening and controlled studies of neuromuscular diseases.
Collapse
Affiliation(s)
- Surapon N Charoensook
- Department of Biomedical Engineering, Columbia University, 622 West 168th Street, VC12-234, New York, NY 10032, USA.
| | | | | | | | | |
Collapse
|
33
|
Colón A, Guo X, Akanda N, Cai Y, Hickman JJ. Functional analysis of human intrafusal fiber innervation by human γ-motoneurons. Sci Rep 2017; 7:17202. [PMID: 29222416 PMCID: PMC5722897 DOI: 10.1038/s41598-017-17382-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Accepted: 11/21/2017] [Indexed: 11/09/2022] Open
Abstract
Investigation of neuromuscular deficits and diseases such as SMA, as well as for next generation prosthetics, utilizing in vitro phenotypic models would benefit from the development of a functional neuromuscular reflex arc. The neuromuscular reflex arc is the system that integrates the proprioceptive information for muscle length and activity (sensory afferent), to modify motoneuron output to achieve graded muscle contraction (actuation efferent). The sensory portion of the arc is composed of proprioceptive sensory neurons and the muscle spindle, which is embedded in the muscle tissue and composed of intrafusal fibers. The gamma motoneurons (γ-MNs) that innervate these fibers regulate the intrafusal fiber's stretch so that they retain proper tension and sensitivity during muscle contraction or relaxation. This mechanism is in place to maintain the sensitivity of proprioception during dynamic muscle activity and to prevent muscular damage. In this study, a co-culture system was developed for innervation of intrafusal fibers by human γ-MNs and demonstrated by morphological and immunocytochemical analysis, then validated by functional electrophysiological evaluation. This human-based fusimotor model and its incorporation into the reflex arc allows for a more accurate recapitulation of neuromuscular function for applications in disease investigations, drug discovery, prosthetic design and neuropathic pain investigations.
Collapse
Affiliation(s)
- A Colón
- Hybrid Systems Lab, NanoScience Technology Center, University of Central Florida, 12424 Research Parkway, Suite 400, Orlando, FL, 32826, USA
| | - X Guo
- Hybrid Systems Lab, NanoScience Technology Center, University of Central Florida, 12424 Research Parkway, Suite 400, Orlando, FL, 32826, USA
| | - N Akanda
- Hybrid Systems Lab, NanoScience Technology Center, University of Central Florida, 12424 Research Parkway, Suite 400, Orlando, FL, 32826, USA
| | - Y Cai
- Hybrid Systems Lab, NanoScience Technology Center, University of Central Florida, 12424 Research Parkway, Suite 400, Orlando, FL, 32826, USA
| | - J J Hickman
- Hybrid Systems Lab, NanoScience Technology Center, University of Central Florida, 12424 Research Parkway, Suite 400, Orlando, FL, 32826, USA.
| |
Collapse
|
34
|
Cvetkovic C, Rich MH, Raman R, Kong H, Bashir R. A 3D-printed platform for modular neuromuscular motor units. MICROSYSTEMS & NANOENGINEERING 2017; 3:17015. [PMID: 31057862 PMCID: PMC6444989 DOI: 10.1038/micronano.2017.15] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Revised: 12/23/2016] [Accepted: 01/24/2017] [Indexed: 05/23/2023]
Abstract
A complex and functional living cellular system requires the interaction of one or more cell types to perform specific tasks, such as sensing, processing, or force production. Modular and flexible platforms for fabrication of such multi-cellular modules and their characterization have been lacking. Here, we present a modular cellular system, made up of multi-layered tissue rings containing integrated skeletal muscle and motor neurons (MNs) embedded in an extracellular matrix. The MNs were differentiated from mouse embryonic stem cells through the formation of embryoid bodies (EBs), which are spherical aggregations of cells grown in a suspension culture. The EBs were integrated into a tissue ring with skeletal muscle, which was differentiated in parallel, to create a co-culture amenable to both cell types. The multi-layered rings were then sequentially placed on a stationary three-dimensional-printed hydrogel structure resembling an anatomical muscle-tendon-bone organization. We demonstrate that the site-specific innervation of a group of muscle fibers in the multi-layered tissue rings allows for muscle contraction via chemical stimulation of MNs with glutamate, a major excitatory neurotransmitter in the mammalian nervous system, with the frequency of contraction increasing with glutamate concentration. The addition of tubocurarine chloride (a nicotinic receptor antagonist) halted the contractions, indicating that muscle contraction was MN induced. With a bio-fabricated system permitting controllable mechanical and geometric attributes in a range of length scales, our novel engineered cellular system can be utilized for easier integration of other modular "building blocks" in living cellular and biological machines.
Collapse
Affiliation(s)
- Caroline Cvetkovic
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Micro and Nanotechnology Laboratory, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Max H. Rich
- Department of Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Ritu Raman
- Micro and Nanotechnology Laboratory, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Department of Mechanical Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Hyunjoon Kong
- Department of Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Rashid Bashir
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Micro and Nanotechnology Laboratory, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| |
Collapse
|
35
|
Wang YI, Oleaga C, Long CJ, Esch MB, McAleer CW, Miller PG, Hickman JJ, Shuler ML. Self-contained, low-cost Body-on-a-Chip systems for drug development. Exp Biol Med (Maywood) 2017; 242:1701-1713. [PMID: 29065797 DOI: 10.1177/1535370217694101] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Integrated multi-organ microphysiological systems are an evolving tool for preclinical evaluation of the potential toxicity and efficacy of drug candidates. Such systems, also known as Body-on-a-Chip devices, have a great potential to increase the successful conversion of drug candidates entering clinical trials into approved drugs. Systems, to be attractive for commercial adoption, need to be inexpensive, easy to operate, and give reproducible results. Further, the ability to measure functional responses, such as electrical activity, force generation, and barrier integrity of organ surrogates, enhances the ability to monitor response to drugs. The ability to operate a system for significant periods of time (up to 28 d) will provide potential to estimate chronic as well as acute responses of the human body. Here we review progress towards a self-contained low-cost microphysiological system with functional measurements of physiological responses. Impact statement Multi-organ microphysiological systems are promising devices to improve the drug development process. The development of a pumpless system represents the ability to build multi-organ systems that are of low cost, high reliability, and self-contained. These features, coupled with the ability to measure electrical and mechanical response in addition to chemical or metabolic changes, provides an attractive system for incorporation into the drug development process. This will be the most complete review of the pumpless platform with recirculation yet written.
Collapse
Affiliation(s)
- Ying I Wang
- 1 Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Carlota Oleaga
- 2 NanoScience Technology Center, University of Central Florida, Orlando, FL 32826, USA
| | - Christopher J Long
- 2 NanoScience Technology Center, University of Central Florida, Orlando, FL 32826, USA.,3 Hesperos, Inc., Orlando, FL 32826, USA
| | - Mandy B Esch
- 4 Center for Nanoscale Science and Technology, National Institute of Standards and Technology, Gaithersburg, MD 20899, USA
| | - Christopher W McAleer
- 2 NanoScience Technology Center, University of Central Florida, Orlando, FL 32826, USA.,3 Hesperos, Inc., Orlando, FL 32826, USA
| | - Paula G Miller
- 1 Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA
| | - James J Hickman
- 2 NanoScience Technology Center, University of Central Florida, Orlando, FL 32826, USA.,3 Hesperos, Inc., Orlando, FL 32826, USA
| | - Michael L Shuler
- 1 Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA.,3 Hesperos, Inc., Orlando, FL 32826, USA
| |
Collapse
|
36
|
Chal J, Al Tanoury Z, Hestin M, Gobert B, Aivio S, Hick A, Cherrier T, Nesmith AP, Parker KK, Pourquié O. Generation of human muscle fibers and satellite-like cells from human pluripotent stem cells in vitro. Nat Protoc 2016; 11:1833-50. [PMID: 27583644 DOI: 10.1038/nprot.2016.110] [Citation(s) in RCA: 187] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Progress toward finding a cure for muscle diseases has been slow because of the absence of relevant cellular models and the lack of a reliable source of muscle progenitors for biomedical investigation. Here we report an optimized serum-free differentiation protocol to efficiently produce striated, millimeter-long muscle fibers together with satellite-like cells from human pluripotent stem cells (hPSCs) in vitro. By mimicking key signaling events leading to muscle formation in the embryo, in particular the dual modulation of Wnt and bone morphogenetic protein (BMP) pathway signaling, this directed differentiation protocol avoids the requirement for genetic modifications or cell sorting. Robust myogenesis can be achieved in vitro within 1 month by personnel experienced in hPSC culture. The differentiating culture can be subcultured to produce large amounts of myogenic progenitors amenable to numerous downstream applications. Beyond the study of myogenesis, this differentiation method offers an attractive platform for the development of relevant in vitro models of muscle dystrophies and drug screening strategies, as well as providing a source of cells for tissue engineering and cell therapy approaches.
Collapse
Affiliation(s)
- Jérome Chal
- Institut de Génétique et de Biologie Moléculaireet Cellulaire (IGBMC), CNRS (UMR 7104), Inserm U964, Université de Strasbourg, Illkirch-Graffenstaden, France
- Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts, USA
- Department of Genetics, Harvard Medical School, Boston, Massachusetts, USA
- Harvard Stem Cell Institute, Boston, Massachusetts, USA
| | - Ziad Al Tanoury
- Institut de Génétique et de Biologie Moléculaireet Cellulaire (IGBMC), CNRS (UMR 7104), Inserm U964, Université de Strasbourg, Illkirch-Graffenstaden, France
| | - Marie Hestin
- Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts, USA
- Department of Genetics, Harvard Medical School, Boston, Massachusetts, USA
- Harvard Stem Cell Institute, Boston, Massachusetts, USA
| | - Bénédicte Gobert
- Institut de Génétique et de Biologie Moléculaireet Cellulaire (IGBMC), CNRS (UMR 7104), Inserm U964, Université de Strasbourg, Illkirch-Graffenstaden, France
| | - Suvi Aivio
- Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts, USA
- Department of Genetics, Harvard Medical School, Boston, Massachusetts, USA
- Harvard Stem Cell Institute, Boston, Massachusetts, USA
| | - Aurore Hick
- Anagenesis Biotechnologies, Parc d'innovation, Illkirch-Graffenstaden, France
| | - Thomas Cherrier
- Institut de Génétique et de Biologie Moléculaireet Cellulaire (IGBMC), CNRS (UMR 7104), Inserm U964, Université de Strasbourg, Illkirch-Graffenstaden, France
| | - Alexander P Nesmith
- Disease Biophysics Group, Wyss Institute for Biologically Inspired Engineering, School of Engineering and Applied Sciences, Harvard University, Cambridge, Massachusetts, USA
| | - Kevin K Parker
- Disease Biophysics Group, Wyss Institute for Biologically Inspired Engineering, School of Engineering and Applied Sciences, Harvard University, Cambridge, Massachusetts, USA
| | - Olivier Pourquié
- Institut de Génétique et de Biologie Moléculaireet Cellulaire (IGBMC), CNRS (UMR 7104), Inserm U964, Université de Strasbourg, Illkirch-Graffenstaden, France
- Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts, USA
- Department of Genetics, Harvard Medical School, Boston, Massachusetts, USA
- Harvard Stem Cell Institute, Boston, Massachusetts, USA
| |
Collapse
|
37
|
Bursac N, Juhas M, Rando TA. Synergizing Engineering and Biology to Treat and Model Skeletal Muscle Injury and Disease. Annu Rev Biomed Eng 2016; 17:217-42. [PMID: 26643021 DOI: 10.1146/annurev-bioeng-071114-040640] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Although skeletal muscle is one of the most regenerative organs in our body, various genetic defects, alterations in extrinsic signaling, or substantial tissue damage can impair muscle function and the capacity for self-repair. The diversity and complexity of muscle disorders have attracted much interest from both cell biologists and, more recently, bioengineers, leading to concentrated efforts to better understand muscle pathology and develop more efficient therapies. This review describes the biological underpinnings of muscle development, repair, and disease, and discusses recent bioengineering efforts to design and control myomimetic environments, both to study muscle biology and function and to aid in the development of new drug, cell, and gene therapies for muscle disorders. The synergy between engineering-aided biological discovery and biology-inspired engineering solutions will be the path forward for translating laboratory results into clinical practice.
Collapse
Affiliation(s)
- Nenad Bursac
- Department of Biomedical Engineering, Duke University, Durham, North Carolina 27708;
| | - Mark Juhas
- Department of Biomedical Engineering, Duke University, Durham, North Carolina 27708;
| | - Thomas A Rando
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California 94305.,Rehabilitation Research & Development Service, VA Palo Alto Health Care System, Palo Alto, California 94304
| |
Collapse
|
38
|
Uzel SGM, Platt RJ, Subramanian V, Pearl TM, Rowlands CJ, Chan V, Boyer LA, So PTC, Kamm RD. Microfluidic device for the formation of optically excitable, three-dimensional, compartmentalized motor units. SCIENCE ADVANCES 2016; 2:e1501429. [PMID: 27493991 PMCID: PMC4972469 DOI: 10.1126/sciadv.1501429] [Citation(s) in RCA: 148] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/12/2015] [Accepted: 07/06/2016] [Indexed: 05/21/2023]
Abstract
Motor units are the fundamental elements responsible for muscle movement. They are formed by lower motor neurons and their muscle targets, synapsed via neuromuscular junctions (NMJs). The loss of NMJs in neurodegenerative disorders (such as amyotrophic lateral sclerosis or spinal muscle atrophy) or as a result of traumatic injuries affects millions of lives each year. Developing in vitro assays that closely recapitulate the physiology of neuromuscular tissues is crucial to understand the formation and maturation of NMJs, as well as to help unravel the mechanisms leading to their degeneration and repair. We present a microfluidic platform designed to coculture myoblast-derived muscle strips and motor neurons differentiated from mouse embryonic stem cells (ESCs) within a three-dimensional (3D) hydrogel. The device geometry mimics the spinal cord-limb physical separation by compartmentalizing the two cell types, which also facilitates the observation of 3D neurite outgrowth and remote muscle innervation. Moreover, the use of compliant pillars as anchors for muscle strips provides a quantitative functional readout of force generation. Finally, photosensitizing the ESC provides a pool of source cells that can be differentiated into optically excitable motor neurons, allowing for spatiodynamic, versatile, and noninvasive in vitro control of the motor units.
Collapse
Affiliation(s)
- Sebastien G. M. Uzel
- Department of Mechanical Engineering, Massachusetts Institute of Technology (MIT), Cambridge, MA 02139, USA
| | - Randall J. Platt
- Department of Biological Engineering, MIT, Cambridge, MA 02139, USA
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | | | - Taylor M. Pearl
- Department of Biological Engineering, MIT, Cambridge, MA 02139, USA
| | | | - Vincent Chan
- Department of Mechanical Engineering, Massachusetts Institute of Technology (MIT), Cambridge, MA 02139, USA
| | | | - Peter T. C. So
- Department of Mechanical Engineering, Massachusetts Institute of Technology (MIT), Cambridge, MA 02139, USA
- Department of Biological Engineering, MIT, Cambridge, MA 02139, USA
- BioSystems and Micromechanics (BioSyM) IRG, Singapore-MIT Alliance for Research and Technology, Singapore, Singapore
| | - Roger D. Kamm
- Department of Mechanical Engineering, Massachusetts Institute of Technology (MIT), Cambridge, MA 02139, USA
- Department of Biological Engineering, MIT, Cambridge, MA 02139, USA
- BioSystems and Micromechanics (BioSyM) IRG, Singapore-MIT Alliance for Research and Technology, Singapore, Singapore
- Corresponding author.
| |
Collapse
|
39
|
Martin NRW, Passey SL, Player DJ, Mudera V, Baar K, Greensmith L, Lewis MP. Neuromuscular Junction Formation in Tissue-Engineered Skeletal Muscle Augments Contractile Function and Improves Cytoskeletal Organization. Tissue Eng Part A 2015; 21:2595-604. [PMID: 26166548 PMCID: PMC4605379 DOI: 10.1089/ten.tea.2015.0146] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Neuromuscular and neurodegenerative diseases are conditions that affect both motor neurons and the underlying skeletal muscle tissue. At present, the majority of neuromuscular research utilizes animal models and there is a growing need to develop novel methodologies that can be used to help understand and develop treatments for these diseases. Skeletal muscle tissue-engineered constructs exhibit many of the characteristics of the native tissue such as accurate fascicular structure and generation of active contractions. However, to date, there has been little consideration toward the integration of engineered skeletal muscle with motor neurons with the aim of neuromuscular junction (NMJ) formation, which would provide a model to investigate neuromuscular diseases and basic biology. In the present work we isolated primary embryonic motor neurons and neonatal myoblasts from Sprague-Dawley rats, and cocultured the two cell types in three-dimensional tissue-engineered fibrin hydrogels with the aim of NMJ formation. Immunohistochemistry revealed myotube formation in a fascicular arrangement and neurite outgrowth from motor neuron cell bodies toward the aligned myotubes. Furthermore, colocalization of pre- and postsynaptic proteins and chemical inhibition of spontaneous myotube twitch indicated the presence of NMJs in the innervated constructs. When electrical field stimulation was employed to evoke isometric contractions, maximal twitch and tetanic force were higher in the constructs cocultured with motor neurons, which may, in part, be explained by improved myotube cytoskeletal organization in these constructs. The fabrication of such constructs may be useful tools for investigating neuromuscular pharmaceuticals and improving the understanding of neuromuscular pathologies.
Collapse
Affiliation(s)
- Neil R W Martin
- 1 Arthritis Research UK Centre for Sport, Exercise and Osteoarthritis, National Centre for Sport and Exercise Medicine (NCSEM), School of Sport, Exercise and Health Sciences, Loughborough University , Loughborough, United Kingdom
| | - Samantha L Passey
- 1 Arthritis Research UK Centre for Sport, Exercise and Osteoarthritis, National Centre for Sport and Exercise Medicine (NCSEM), School of Sport, Exercise and Health Sciences, Loughborough University , Loughborough, United Kingdom .,2 Department of Pharmacology and Therapeutics, University of Melbourne , Parkville, Victoria, Australia
| | - Darren J Player
- 1 Arthritis Research UK Centre for Sport, Exercise and Osteoarthritis, National Centre for Sport and Exercise Medicine (NCSEM), School of Sport, Exercise and Health Sciences, Loughborough University , Loughborough, United Kingdom
| | - Vivek Mudera
- 3 Institute of Orthopedics and Musculoskeletal Sciences, University College London , London, United Kingdom
| | - Keith Baar
- 4 Division of Neurobiology, Physiology and Behavior, University of California Davis , Davis, California
| | - Linda Greensmith
- 5 The Sobell Department of Motor Neuroscience and Movement Disorders, University College London , London, United Kingdom
| | - Mark P Lewis
- 1 Arthritis Research UK Centre for Sport, Exercise and Osteoarthritis, National Centre for Sport and Exercise Medicine (NCSEM), School of Sport, Exercise and Health Sciences, Loughborough University , Loughborough, United Kingdom
| |
Collapse
|
40
|
McAleer CW, Rumsey JW, Stancescu M, Hickman JJ. Functional myotube formation from adult rat satellite cells in a defined serum-free system. Biotechnol Prog 2015; 31:997-1003. [PMID: 25683642 PMCID: PMC5015122 DOI: 10.1002/btpr.2063] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Revised: 01/28/2015] [Indexed: 12/28/2022]
Abstract
This manuscript describes the development of a culture system whereby mature contracting myotubes were formed from adult rat derived satellite cells. Satellite cells, extracted from the Tibialis Anterior of adult rats, were grown in defined serum-free growth and differentiation media, on a nonbiological substrate, N-1[3-trimethoxysilyl propyl] diethylenetriamine. Myotubes were evaluated morphologically and immunocytochemically, using MyHC specific antibodies, as well as functionally using patch clamp electrophysiology to measure ion channel activity. Results indicated the establishment of the rapid expression of adult myosin isoforms that contrasts to their slow development in embryonic cultures. This culture system has applications in the understanding and treatment of age-related muscle myopathy, muscular dystrophy, and for skeletal muscle engineering by providing a more relevant phenotype for both in vitro and in vivo applications.
Collapse
Affiliation(s)
- Christopher W McAleer
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway, Suite 400, Orlando, FL, 32826
| | - John W Rumsey
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway, Suite 400, Orlando, FL, 32826
| | - Maria Stancescu
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway, Suite 400, Orlando, FL, 32826
| | - James J Hickman
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway, Suite 400, Orlando, FL, 32826
| |
Collapse
|
41
|
Stoppel WL, Ghezzi CE, McNamara SL, Black LD, Kaplan DL. Clinical applications of naturally derived biopolymer-based scaffolds for regenerative medicine. Ann Biomed Eng 2015; 43:657-80. [PMID: 25537688 PMCID: PMC8196399 DOI: 10.1007/s10439-014-1206-2] [Citation(s) in RCA: 93] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2014] [Accepted: 11/26/2014] [Indexed: 01/05/2023]
Abstract
Naturally derived polymeric biomaterials, such as collagens, silks, elastins, alginates, and fibrins are utilized in tissue engineering due to their biocompatibility, bioactivity, and tunable mechanical and degradation kinetics. The use of these natural biopolymers in biomedical applications is advantageous because they do not release cytotoxic degradation products, are often processed using environmentally-friendly aqueous-based methods, and their degradation rates within biological systems can be manipulated by modifying the starting formulation or processing conditions. For these reasons, many recent in vivo investigations and FDA-approval of new biomaterials for clinical use have utilized natural biopolymers as matrices for cell delivery and as scaffolds for cell-free support of native tissues. This review highlights biopolymer-based scaffolds used in clinical applications for the regeneration and repair of native tissues, with a focus on bone, skeletal muscle, peripheral nerve, cardiac muscle, and cornea substitutes.
Collapse
Affiliation(s)
- Whitney L. Stoppel
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA
| | - Chiara E. Ghezzi
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA
| | - Stephanie L. McNamara
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA
- Cellular, Molecular and Developmental Biology Program, Sackler School of Graduate Biomedical Sciences, Tufts University School of Medicine, Boston, MA 02111, USA
- The Harvard/MIT MD-PhD Program, Harvard Medical School, Boston, MA 02115, USA
| | - Lauren D. Black
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA
- Cellular, Molecular and Developmental Biology Program, Sackler School of Graduate Biomedical Sciences, Tufts University School of Medicine, Boston, MA 02111, USA
| | - David L. Kaplan
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA
| |
Collapse
|
42
|
Smith AST, Long CJ, McAleer C, Guo X, Esch M, Prot JM, Shuler ML, Hickman JJ. ‘Body-on-a-Chip’ Technology and Supporting Microfluidics. HUMAN-BASED SYSTEMS FOR TRANSLATIONAL RESEARCH 2014. [DOI: 10.1039/9781782620136-00132] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
In order to effectively streamline current drug development protocols, there is a need to generate high information content preclinical screens capable of generating data with a predictive power in relation to the activity of novel therapeutics in humans. Given the poor predictive power of animal models, and the lack of complexity and interconnectivity of standard in vitro culture methodologies, many investigators are now moving toward the development of physiologically and functionally accurate culture platforms composed of human cells to investigate cellular responses to drug compounds in high-throughput preclinical studies. The generation of complex, multi-organ in vitro platforms, built to recapitulate physiological dimensions, flow rates and shear stresses, is being investigated as the logical extension of this drive. Production and application of a biologically accurate multi-organ platform, or ‘body-on-a-chip’, would facilitate the correct modelling of the dynamic and interconnected state of living systems for high-throughput drug studies as well as basic and applied biomolecular research. This chapter will discuss current technologies aimed at producing ‘body-on-a-chip’ models, as well as highlighting recent advances and important challenges still to be met in the development of biomimetic single-organ systems for drug development purposes.
Collapse
Affiliation(s)
- A. S. T. Smith
- NanoScience Technology Center, University of Central Florida Orlando FL 32826 USA
| | - C. J. Long
- NanoScience Technology Center, University of Central Florida Orlando FL 32826 USA
| | - C. McAleer
- NanoScience Technology Center, University of Central Florida Orlando FL 32826 USA
| | - X. Guo
- NanoScience Technology Center, University of Central Florida Orlando FL 32826 USA
| | - M. Esch
- Biomedical Engineering, Cornell University Ithaca NY USA
| | - J. M. Prot
- Biomedical Engineering, Cornell University Ithaca NY USA
| | - M. L. Shuler
- Biomedical Engineering, Cornell University Ithaca NY USA
| | - J. J. Hickman
- NanoScience Technology Center, University of Central Florida Orlando FL 32826 USA
| |
Collapse
|
43
|
McAleer CW, Smith AST, Najjar S, Pirozzi K, Long CJ, Hickman JJ. Mechanistic investigation of adult myotube response to exercise and drug treatment in vitro using a multiplexed functional assay system. J Appl Physiol (1985) 2014; 117:1398-405. [PMID: 25301895 DOI: 10.1152/japplphysiol.00612.2014] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
The ability to accurately measure skeletal muscle functional performance at the single-cell level would be advantageous for exercise physiology studies and disease modeling applications. To that end, this study characterizes the functional response of individual skeletal muscle myotubes derived from adult rodent tissue to creatine treatment and chronic exercise. The observed improvements to functional performance in response to these treatments appear to correlate with alterations in hypertrophic and mitochondrial biogenesis pathways, supporting previously published in vivo and in vitro data, which highlights the role of these pathways in augmenting skeletal muscle output. The developed system represents a multiplexed functional in vitro assay capable of long-term assessment of contractile cellular outputs in real-time that is compatible with concomitant molecular biology analysis. Adoption of this system in drug toxicity and efficacy studies would improve understanding of compound activity on physical cellular outputs and provide more streamlined and predictive data for future preclinical analyses.
Collapse
Affiliation(s)
- C W McAleer
- NanoScience Technology Center, University of Central Florida, Orlando, Florida
| | - A S T Smith
- NanoScience Technology Center, University of Central Florida, Orlando, Florida
| | - S Najjar
- NanoScience Technology Center, University of Central Florida, Orlando, Florida
| | - K Pirozzi
- NanoScience Technology Center, University of Central Florida, Orlando, Florida
| | - C J Long
- NanoScience Technology Center, University of Central Florida, Orlando, Florida
| | - J J Hickman
- NanoScience Technology Center, University of Central Florida, Orlando, Florida
| |
Collapse
|
44
|
Ostrovidov S, Hosseini V, Ahadian S, Fujie T, Parthiban SP, Ramalingam M, Bae H, Kaji H, Khademhosseini A. Skeletal muscle tissue engineering: methods to form skeletal myotubes and their applications. TISSUE ENGINEERING. PART B, REVIEWS 2014; 20:403-36. [PMID: 24320971 PMCID: PMC4193686 DOI: 10.1089/ten.teb.2013.0534] [Citation(s) in RCA: 181] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/02/2013] [Accepted: 12/05/2013] [Indexed: 12/25/2022]
Abstract
Skeletal muscle tissue engineering (SMTE) aims to repair or regenerate defective skeletal muscle tissue lost by traumatic injury, tumor ablation, or muscular disease. However, two decades after the introduction of SMTE, the engineering of functional skeletal muscle in the laboratory still remains a great challenge, and numerous techniques for growing functional muscle tissues are constantly being developed. This article reviews the recent findings regarding the methodology and various technical aspects of SMTE, including cell alignment and differentiation. We describe the structure and organization of muscle and discuss the methods for myoblast alignment cultured in vitro. To better understand muscle formation and to enhance the engineering of skeletal muscle, we also address the molecular basics of myogenesis and discuss different methods to induce myoblast differentiation into myotubes. We then provide an overview of different coculture systems involving skeletal muscle cells, and highlight major applications of engineered skeletal muscle tissues. Finally, potential challenges and future research directions for SMTE are outlined.
Collapse
Affiliation(s)
- Serge Ostrovidov
- WPI-Advanced Institute for Materials Research, Tohoku University, Sendai, Japan
| | - Vahid Hosseini
- Laboratory of Applied Mechanobiology, Department of Health Sciences and Technology, ETH, Zurich, Switzerland
| | - Samad Ahadian
- WPI-Advanced Institute for Materials Research, Tohoku University, Sendai, Japan
| | - Toshinori Fujie
- WPI-Advanced Institute for Materials Research, Tohoku University, Sendai, Japan
- Department of Life Science and Medical Bioscience, Graduate School of Advanced Science and Engineering, Waseda University, Tokyo, Japan
| | | | - Murugan Ramalingam
- WPI-Advanced Institute for Materials Research, Tohoku University, Sendai, Japan
- Faculté de Chirurgie Dentaire, Université de Strasbourg, Strasbourg Cedex, France
- Centre for Stem Cell Research, Christian Medical College Campus, Vellore, India
| | - Hojae Bae
- College of Animal Bioscience and Technology, Department of Bioindustrial Technologies, Konkuk University, Hwayang-dong, Kwangjin-gu, Seoul, Republic of Korea
| | - Hirokazu Kaji
- Department of Bioengineering and Robotics, Graduate School of Engineering, Tohoku University, Sendai, Japan
| | - Ali Khademhosseini
- WPI-Advanced Institute for Materials Research, Tohoku University, Sendai, Japan
- Department of Maxillofacial Biomedical Engineering, Institute of Oral Biology, School of Dentistry, Kyung Hee University, Seoul, Republic of Korea
- Center for Biomedical Engineering, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, Massachusetts, United States
- Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, Massachusetts, United States
- Department of Physics, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
45
|
Sung JH, Srinivasan B, Esch MB, McLamb WT, Bernabini C, Shuler ML, Hickman JJ. Using physiologically-based pharmacokinetic-guided "body-on-a-chip" systems to predict mammalian response to drug and chemical exposure. Exp Biol Med (Maywood) 2014; 239:1225-39. [PMID: 24951471 DOI: 10.1177/1535370214529397] [Citation(s) in RCA: 93] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The continued development of in vitro systems that accurately emulate human response to drugs or chemical agents will impact drug development, our understanding of chemical toxicity, and enhance our ability to respond to threats from chemical or biological agents. A promising technology is to build microscale replicas of humans that capture essential elements of physiology, pharmacology, and/or toxicology (microphysiological systems). Here, we review progress on systems for microscale models of mammalian systems that include two or more integrated cellular components. These systems are described as a "body-on-a-chip", and utilize the concept of physiologically-based pharmacokinetic (PBPK) modeling in the design. These microscale systems can also be used as model systems to predict whole-body responses to drugs as well as study the mechanism of action of drugs using PBPK analysis. In this review, we provide examples of various approaches to construct such systems with a focus on their physiological usefulness and various approaches to measure responses (e.g. chemical, electrical, or mechanical force and cellular viability and morphology). While the goal is to predict human response, other mammalian cell types can be utilized with the same principle to predict animal response. These systems will be evaluated on their potential to be physiologically accurate, to provide effective and efficient platform for analytics with accessibility to a wide range of users, for ease of incorporation of analytics, functional for weeks to months, and the ability to replicate previously observed human responses.
Collapse
Affiliation(s)
- Jong Hwan Sung
- Chemical Engineering, Hongik University, Seoul 121-791, Republic of Korea
| | - Balaji Srinivasan
- NanoScience Technology Center, University of Central Florida, Orlando, FL 32826, USA
| | - Mandy Brigitte Esch
- Department of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA
| | - William T McLamb
- NanoScience Technology Center, University of Central Florida, Orlando, FL 32826, USA
| | - Catia Bernabini
- NanoScience Technology Center, University of Central Florida, Orlando, FL 32826, USA
| | - Michael L Shuler
- Department of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA
| | - James J Hickman
- NanoScience Technology Center, University of Central Florida, Orlando, FL 32826, USA Biomolecular Science Center, Burnett School of Biomedical Sciences, University of Central Florida, Orlando, FL 32816, USA
| |
Collapse
|
46
|
Abstract
A multiorgan, functional, human in vitro assay system or 'Body-on-a-Chip' would be of tremendous benefit to the drug discovery and toxicology industries, as well as providing a more biologically accurate model for the study of disease as well as applied and basic biological research. Here, we describe the advances our team has made towards this goal, as well as the most pertinent issues facing further development of these systems. Description is given of individual organ models with appropriate cellular functionality, and our efforts to produce human iterations of each using primary and stem cell sources for eventual incorporation into this system. Advancement of the 'Body-on-a-Chip' field is predicated on the availability of abundant sources of human cells, capable of full differentiation and maturation to adult phenotypes, for which researchers are largely dependent on stem cells. Although this level of maturation is not yet achievable in all cell types, the work of our group highlights the high level of functionality that can be achieved using current technology, for a wide variety of cell types. As availability of functional human cell types for in vitro culture increases, the potential to produce a multiorgan in vitro system capable of accurately reproducing acute and chronic human responses to chemical and pathological challenge in real time will also increase.
Collapse
|
47
|
Morimoto Y, Kato-Negishi M, Onoe H, Takeuchi S. Three-dimensional neuron-muscle constructs with neuromuscular junctions. Biomaterials 2013; 34:9413-9. [PMID: 24041425 DOI: 10.1016/j.biomaterials.2013.08.062] [Citation(s) in RCA: 124] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2013] [Accepted: 08/20/2013] [Indexed: 12/13/2022]
Abstract
This paper describes a fabrication method of muscle tissue constructs driven by neurotransmitters released from activated motor neurons. The constructs consist of three-dimensional (3D) free-standing skeletal muscle fibers co-cultured with motor neurons. We differentiated mouse neural stem cells (mNSCs) cultured on the skeletal muscle fibers into neurons that extend their processes into the muscle fibers. We found that acetylcholine receptors (AChRs) were formed at the connection between the muscle fibers and the neurons. The neuron-muscle constructs consist of highly aligned, long and matured muscle fibers that facilitate wide contractions of muscle fibers in a single direction. The contractions of the neuron-muscle construct were observed after glutamic acid activation of the neurons. The contraction was stopped by treatment with curare, an neuromuscular junction (NMJ) antagonist. These results indicate that our method succeeded in the formation of NMJs in the neuron-muscle constructs. The neuron-muscle construct system can potentially be used in pharmacokinetic assays related to NMJ disease therapies and in soft-robotic actuators.
Collapse
Affiliation(s)
- Yuya Morimoto
- Center for International Research on Micronano Mechatronics (CIRMM), Institute of Industrial Science (IIS), The University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo 153-8505, Japan
| | | | | | | |
Collapse
|
48
|
Abstract
Rabies virus (RABV), which is transmitted via a bite wound caused by a rabid animal, infects peripheral nerves and then spreads to the central nervous system (CNS) before causing severe neurological symptoms and death in the infected individual. Despite the importance of this ability of the virus to spread from a peripheral site to the CNS (neuroinvasiveness) in the pathogenesis of rabies, little is known about the mechanism underlying the neuroinvasiveness of RABV. In this study, to obtain insights into the mechanism, we conducted comparative analysis of two fixed RABV strains, Nishigahara and the derivative strain Ni-CE, which cause lethal and asymptomatic infections, respectively, in mice after intramuscular inoculation. Examination of a series of chimeric viruses harboring the respective genes from Nishigahara in the genetic background of Ni-CE revealed that the Nishigahara phosphoprotein (P) gene plays a major role in the neuroinvasiveness by mediating infection of peripheral nerves. The results obtained from both in vivo and in vitro experiments strongly suggested that the Nishigahara P gene, but not the Ni-CE P gene, is important for stable viral replication in muscle cells. Further investigation based on the previous finding that RABV phosphoprotein counteracts the host interferon (IFN) system demonstrated that the Nishigahara P gene, but not the Ni-CE P gene, functions to suppress expression of the beta interferon (IFN-β) gene (Ifn-β) and IFN-stimulated genes in muscle cells. In conclusion, we provide the first data strongly suggesting that RABV phosphoprotein assists viral replication in muscle cells by counteracting the host IFN system and, consequently, enhances infection of peripheral nerves.
Collapse
|
49
|
Myelination and node of Ranvier formation on sensory neurons in a defined in vitro system. In Vitro Cell Dev Biol Anim 2013; 49:608-618. [PMID: 23949775 DOI: 10.1007/s11626-013-9647-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2012] [Accepted: 05/29/2013] [Indexed: 10/26/2022]
Abstract
One of the most important developmental modifications of the nervous system is Schwann cell myelination of axons. Schwann cells ensheath axons to create myelin segments to provide protection to the axon as well as increase the conduction of action potentials. In vitro neuronal systems provide a unique modality to study a variety of factors influencing myelination as well as diseases associated with myelin sheath degradation. This work details the development of a patterned in vitro myelinating dorsal root ganglion culture. This defined system utilized a serum-free medium in combination with a patterned substrate, utilizing the cytophobic and cytophilic molecules (poly)ethylene glycol (PEG) and N-1[3 (trimethoxysilyl) propyl] diethylenetriamine (DETA), respectively. Directional outgrowth of the neurites and subsequent myelination was controlled by surface modifications, and conformity to the pattern was measured over the duration of the experiments. The myelinated segments and nodal proteins were visualized and quantified using confocal microscopy. This tissue-engineered system provides a highly controlled, reproducible model for studying Schwann cell interactions with sensory neurons, as well as the myelination process, and its effect on neuronal plasticity and peripheral nerve regeneration. It is also compatible for use in bio-hybrid constructs to reproduce the stretch reflex arc on a chip because the media combination used is the same that we have used previously for motoneurons, muscle, and for neuromuscular junction (NMJ) formation. This work could have application for the study of demyelinating diseases such as diabetes induced peripheral neuropathy and could rapidly translate to a role in the discovery of drugs promoting enhanced peripheral nervous system (PNS) remyelination.
Collapse
|
50
|
Sung JH, Esch MB, Prot JM, Long CJ, Smith A, Hickman JJ, Shuler ML. Microfabricated mammalian organ systems and their integration into models of whole animals and humans. LAB ON A CHIP 2013; 13:1201-12. [PMID: 23388858 PMCID: PMC3593746 DOI: 10.1039/c3lc41017j] [Citation(s) in RCA: 169] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/02/2023]
Abstract
While in vitro cell based systems have been an invaluable tool in biology, they often suffer from a lack of physiological relevance. The discrepancy between the in vitro and in vivo systems has been a bottleneck in drug development process and biological sciences. The recent progress in microtechnology has enabled manipulation of cellular environment at a physiologically relevant length scale, which has led to the development of novel in vitro organ systems, often termed 'organ-on-a-chip' systems. By mimicking the cellular environment of in vivo tissues, various organ-on-a-chip systems have been reported to reproduce target organ functions better than conventional in vitro model systems. Ultimately, these organ-on-a-chip systems will converge into multi-organ 'body-on-a-chip' systems composed of functional tissues that reproduce the dynamics of the whole-body response. Such microscale in vitro systems will open up new possibilities in medical science and in the pharmaceutical industry.
Collapse
Affiliation(s)
- Jong H Sung
- Chemical Engineering, Hongik University, Seoul, Korea
| | | | | | | | | | | | | |
Collapse
|