1
|
Modi PS, Singh A, Chaturvedi A, Agarwal S, Dutta R, Nayak R, Singh AK. Tissue chips as headway model and incitement technology. Synth Syst Biotechnol 2024; 10:86-101. [PMID: 39286054 PMCID: PMC11403008 DOI: 10.1016/j.synbio.2024.08.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 08/12/2024] [Accepted: 08/26/2024] [Indexed: 09/19/2024] Open
Abstract
Tissue on a chip or organ-on-chip (OOC) is a technology that's dignified to form a transformation in drug discovery through the use of advanced platforms. These are 3D in-vitro cell culture models that mimic micro-environment of human organs or tissues on artificial microstructures built on a portable microfluidic chip without involving sacrificial humans or animals. This review article aims to offer readers a thorough and insightful understanding of technology. It begins with an in-depth understanding of chip design and instrumentation, underlining its pivotal role and the imperative need for its development in the modern scientific landscape. The review article explores into the myriad applications of OOC technology, showcasing its transformative impact on fields such as radiobiology, drug discovery and screening, and its pioneering use in space research. In addition to highlighting these diverse applications, the article provides a critical analysis of the current challenges that OOC technology faces. It examines both the biological and technical limitations that hinder its progress and efficacy and discusses the potential advancements and innovations that could drive the OOC technology forward. Through this comprehensive review, readers will gain a deep appreciation of the significance, capabilities, and evolving landscape of OOC technology.
Collapse
Affiliation(s)
- Prerna Suchitan Modi
- Amity Institute of Biotechnology, Amity University Uttar Pradesh, Noida, Uttar Pradesh, India
| | - Abhishek Singh
- Amity Institute of Biotechnology, Amity University Uttar Pradesh, Noida, Uttar Pradesh, India
| | - Awyang Chaturvedi
- Amity Institute of Biotechnology, Amity University Uttar Pradesh, Noida, Uttar Pradesh, India
| | - Shailly Agarwal
- Amity Institute of Biotechnology, Amity University Uttar Pradesh, Noida, Uttar Pradesh, India
| | - Raghav Dutta
- Amity Institute of Biotechnology, Amity University Uttar Pradesh, Noida, Uttar Pradesh, India
| | - Ranu Nayak
- Amity Institute of Nanotechnology, Amity University Uttar Pradesh, Noida, Uttar Pradesh, India
| | - Alok Kumar Singh
- Amity Institute of Biotechnology, Amity University Uttar Pradesh, Noida, Uttar Pradesh, India
| |
Collapse
|
2
|
Zhou Z, Liu J, Xiong T, Liu Y, Tuan RS, Li ZA. Engineering Innervated Musculoskeletal Tissues for Regenerative Orthopedics and Disease Modeling. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2310614. [PMID: 38200684 DOI: 10.1002/smll.202310614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Revised: 12/28/2023] [Indexed: 01/12/2024]
Abstract
Musculoskeletal (MSK) disorders significantly burden patients and society, resulting in high healthcare costs and productivity loss. These disorders are the leading cause of physical disability, and their prevalence is expected to increase as sedentary lifestyles become common and the global population of the elderly increases. Proper innervation is critical to maintaining MSK function, and nerve damage or dysfunction underlies various MSK disorders, underscoring the potential of restoring nerve function in MSK disorder treatment. However, most MSK tissue engineering strategies have overlooked the significance of innervation. This review first expounds upon innervation in the MSK system and its importance in maintaining MSK homeostasis and functions. This will be followed by strategies for engineering MSK tissues that induce post-implantation in situ innervation or are pre-innervated. Subsequently, research progress in modeling MSK disorders using innervated MSK organoids and organs-on-chips (OoCs) is analyzed. Finally, the future development of engineering innervated MSK tissues to treat MSK disorders and recapitulate disease mechanisms is discussed. This review provides valuable insights into the underlying principles, engineering methods, and applications of innervated MSK tissues, paving the way for the development of targeted, efficacious therapies for various MSK conditions.
Collapse
Affiliation(s)
- Zhilong Zhou
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR, P. R. China
| | - Jun Liu
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR, P. R. China
- Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, Shatin, NT, Hong Kong SAR, P. R. China
| | - Tiandi Xiong
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR, P. R. China
- Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, Shatin, NT, Hong Kong SAR, P. R. China
| | - Yuwei Liu
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR, P. R. China
- Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong, 518000, P. R. China
| | - Rocky S Tuan
- Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, Shatin, NT, Hong Kong SAR, P. R. China
- School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR, P. R. China
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR, P. R. China
| | - Zhong Alan Li
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR, P. R. China
- Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, Shatin, NT, Hong Kong SAR, P. R. China
- School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR, P. R. China
- Key Laboratory of Regenerative Medicine, Ministry of Education, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR, P. R. China
- Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, Guangdong, 518057, P. R. China
| |
Collapse
|
3
|
Barakat N, Jangir H, Gallo L, Grillo M, Guo X, Hickman J. Inhibition of Metalloproteinases Extends Longevity and Function of In Vitro Human iPSC-Derived Skeletal Muscle. Biomedicines 2024; 12:856. [PMID: 38672210 PMCID: PMC11047953 DOI: 10.3390/biomedicines12040856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 04/02/2024] [Accepted: 04/03/2024] [Indexed: 04/28/2024] Open
Abstract
In vitro culture longevity has long been a concern for disease modeling and drug testing when using contractable cells. The dynamic nature of certain cells, such as skeletal muscle, contributes to cell surface release, which limits the system's ability to conduct long-term studies. This study hypothesized that regulating the extracellular matrix (ECM) dynamics should be able to prolong cell attachment on a culture surface. Human induced pluripotent stem cell (iPSC)-derived skeletal muscle (SKM) culture was utilized to test this hypothesis due to its forceful contractions in mature muscle culture, which can cause cell detachment. By specifically inhibiting matrix metalloproteinases (MMPs) that work to digest components of the ECM, it was shown that the SKM culture remained adhered for longer periods of time, up to 80 days. Functional testing of myofibers indicated that cells treated with the MMP inhibitors, tempol, and doxycycline, displayed a significantly reduced fatigue index, although the fidelity was not affected, while those treated with the MMP inducer, PMA, indicated a premature detachment and increased fatigue index. The MMP-modulating activity by the inhibitors and inducer was further validated by gel zymography analysis, where the MMP inhibitor showed minimally active MMPs, while the inducer-treated cells indicated high MMP activity. These data support the hypotheses that regulating the ECM dynamics can help maximize in vitro myotube longevity. This proof-of-principle strategy would benefit the modeling of diseases that require a long time to develop and the evaluation of chronic effects of potential therapeutics.
Collapse
Affiliation(s)
- Natali Barakat
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway, Suite 400, Orlando, FL 32826, USA; (N.B.); (H.J.); (L.G.); (M.G.); (X.G.)
- Department of Chemistry, University of Central Florida, Orlando, FL 32828, USA
| | - Himanshi Jangir
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway, Suite 400, Orlando, FL 32826, USA; (N.B.); (H.J.); (L.G.); (M.G.); (X.G.)
| | - Leandro Gallo
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway, Suite 400, Orlando, FL 32826, USA; (N.B.); (H.J.); (L.G.); (M.G.); (X.G.)
| | - Marcella Grillo
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway, Suite 400, Orlando, FL 32826, USA; (N.B.); (H.J.); (L.G.); (M.G.); (X.G.)
| | - Xiufang Guo
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway, Suite 400, Orlando, FL 32826, USA; (N.B.); (H.J.); (L.G.); (M.G.); (X.G.)
| | - James Hickman
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway, Suite 400, Orlando, FL 32826, USA; (N.B.); (H.J.); (L.G.); (M.G.); (X.G.)
- Department of Chemistry, University of Central Florida, Orlando, FL 32828, USA
| |
Collapse
|
4
|
De Vitis E, Stanzione A, Romano A, Quattrini A, Gigli G, Moroni L, Gervaso F, Polini A. The Evolution of Technology-Driven In Vitro Models for Neurodegenerative Diseases. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2304989. [PMID: 38366798 DOI: 10.1002/advs.202304989] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 01/15/2024] [Indexed: 02/18/2024]
Abstract
The alteration in the neural circuits of both central and peripheral nervous systems is closely related to the onset of neurodegenerative disorders (NDDs). Despite significant research efforts, the knowledge regarding NDD pathological processes, and the development of efficacious drugs are still limited due to the inability to access and reproduce the components of the nervous system and its intricate microenvironment. 2D culture systems are too simplistic to accurately represent the more complex and dynamic situation of cells in vivo and have therefore been surpassed by 3D systems. However, both models suffer from various limitations that can be overcome by employing two innovative technologies: organ-on-chip and 3D printing. In this review, an overview of the advantages and shortcomings of both microfluidic platforms and extracellular matrix-like biomaterials will be given. Then, the combination of microfluidics and hydrogels as a new synergistic approach to study neural disorders by analyzing the latest advances in 3D brain-on-chip for neurodegenerative research will be explored.
Collapse
Affiliation(s)
- Eleonora De Vitis
- CNR NANOTEC-Institute of Nanotechnology, Campus Ecotekn, via Monteroni, Lecce, 73100, Italy
| | - Antonella Stanzione
- CNR NANOTEC-Institute of Nanotechnology, Campus Ecotekn, via Monteroni, Lecce, 73100, Italy
| | - Alessandro Romano
- IRCCS San Raffaele Scientific Institute, Division of Neuroscience, Institute of Experimental Neurology, Milan, 20132, Italy
| | - Angelo Quattrini
- IRCCS San Raffaele Scientific Institute, Division of Neuroscience, Institute of Experimental Neurology, Milan, 20132, Italy
| | - Giuseppe Gigli
- CNR NANOTEC-Institute of Nanotechnology, Campus Ecotekn, via Monteroni, Lecce, 73100, Italy
- Dipartimento di Medicina Sperimentale, Università Del Salento, Campus Ecotekne, via Monteroni, Lecce, 73100, Italy
| | - Lorenzo Moroni
- CNR NANOTEC-Institute of Nanotechnology, Campus Ecotekn, via Monteroni, Lecce, 73100, Italy
- Complex Tissue Regeneration, Maastricht University, Universiteitssingel 40, Maastricht, 6229 ER, Netherlands
| | - Francesca Gervaso
- CNR NANOTEC-Institute of Nanotechnology, Campus Ecotekn, via Monteroni, Lecce, 73100, Italy
| | - Alessandro Polini
- CNR NANOTEC-Institute of Nanotechnology, Campus Ecotekn, via Monteroni, Lecce, 73100, Italy
| |
Collapse
|
5
|
Amartumur S, Nguyen H, Huynh T, Kim TS, Woo RS, Oh E, Kim KK, Lee LP, Heo C. Neuropathogenesis-on-chips for neurodegenerative diseases. Nat Commun 2024; 15:2219. [PMID: 38472255 PMCID: PMC10933492 DOI: 10.1038/s41467-024-46554-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Accepted: 02/28/2024] [Indexed: 03/14/2024] Open
Abstract
Developing diagnostics and treatments for neurodegenerative diseases (NDs) is challenging due to multifactorial pathogenesis that progresses gradually. Advanced in vitro systems that recapitulate patient-like pathophysiology are emerging as alternatives to conventional animal-based models. In this review, we explore the interconnected pathogenic features of different types of ND, discuss the general strategy to modelling NDs using a microfluidic chip, and introduce the organoid-on-a-chip as the next advanced relevant model. Lastly, we overview how these models are being applied in academic and industrial drug development. The integration of microfluidic chips, stem cells, and biotechnological devices promises to provide valuable insights for biomedical research and developing diagnostic and therapeutic solutions for NDs.
Collapse
Affiliation(s)
- Sarnai Amartumur
- Department of Biophysics, Institute of Quantum Biophysics, Sungkyunkwan University, Suwon, 16419, Korea
| | - Huong Nguyen
- Department of Biophysics, Institute of Quantum Biophysics, Sungkyunkwan University, Suwon, 16419, Korea
| | - Thuy Huynh
- Department of Biophysics, Institute of Quantum Biophysics, Sungkyunkwan University, Suwon, 16419, Korea
| | - Testaverde S Kim
- Center for Integrated Nanostructure Physics (CINAP), Institute for Basic Science (IBS), Suwon, 16419, Korea
| | - Ran-Sook Woo
- Department of Anatomy and Neuroscience, College of Medicine, Eulji University, Daejeon, 34824, Korea
| | - Eungseok Oh
- Department of Neurology, Chungnam National University Hospital, Daejeon, 35015, Korea
| | - Kyeong Kyu Kim
- Department of Precision Medicine, Graduate School of Basic Medical Science (GSBMS), Institute for Anti-microbial Resistance Research and Therapeutics, Sungkyunkwan University School of Medicine, Suwon, 16419, Korea
| | - Luke P Lee
- Department of Biophysics, Institute of Quantum Biophysics, Sungkyunkwan University, Suwon, 16419, Korea.
- Harvard Medical School, Division of Engineering in Medicine and Renal Division, Department of Medicine, Brigham and Women's Hospital, Boston, MA, 02115, USA.
- Department of Bioengineering, Department of Electrical Engineering and Computer Science, University of California, Berkeley, CA, 94720, USA.
| | - Chaejeong Heo
- Department of Biophysics, Institute of Quantum Biophysics, Sungkyunkwan University, Suwon, 16419, Korea.
- Center for Integrated Nanostructure Physics (CINAP), Institute for Basic Science (IBS), Suwon, 16419, Korea.
| |
Collapse
|
6
|
Chen D, Philippidou P, Brenha BDF, Schaffer AE, Miranda HC. Scalable, optically-responsive human neuromuscular junction model reveals convergent mechanisms of synaptic dysfunction in familial ALS. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.11.575304. [PMID: 38260655 PMCID: PMC10802619 DOI: 10.1101/2024.01.11.575304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
Neuromuscular junctions (NMJs) are specialized synapses that mediate communication between motor neurons and skeletal muscles and are essential for movement. The degeneration of this system can lead to symptoms observed in neuromuscular and motor neuron diseases. Studying these synapses and their degeneration has proven challenging. Prior NMJ studies heavily relied upon the use of mouse, chick, or isolated primary human cells, which have demonstrated limited fidelity for disease modeling. To enable the study of NMJ dysfunction and model genetic diseases, we, and others, have developed methods to generate human NMJs from pluripotent stem cells (PSCs), embryonic stem cells, and induced pluripotent stem cells. However, published studies have highlighted technical limitations associated with these complex in vitro NMJ models. In this study, we developed a robust PSC-derived motor neuron and skeletal muscle co-culture method, and demonstrated its sensitivity in modeling motor neuron disease. Our method spontaneously and reproducibly forms human NMJs. We developed multiwell-multielectrode array (MEA) parameters to quantify the activity of PSC-derived skeletal muscles, as well as measured the electrophysiological activity of functional human PSC-derived NMJs. We further leveraged our method to morphologically and functionally assess NMJs from the familial amyotrophic lateral sclerosis (fALS) PSCs, C9orf72 hexanucleotide (G4C2)n repeat expansion (HRE), SOD1 A5V , and TDP43 G298S to define the reproducibility and sensitivity of our system. We observed a significant decrease in the numbers and activity of PSC-derived NMJs developed from the different ALS lines compared to their respective controls. Furthermore, we evaluated a therapeutic candidate undergoing clinical trials and observed a variant-dependent rescue of functionality of NMJs. Our newly developed method provides a platform for the systematic investigation of genetic causes of NMJ neurodegeneration and highlights the need for therapeutic avenues to consider patient genotype.
Collapse
|
7
|
Wang H, Ning X, Zhao F, Zhao H, Li D. Human organoids-on-chips for biomedical research and applications. Theranostics 2024; 14:788-818. [PMID: 38169573 PMCID: PMC10758054 DOI: 10.7150/thno.90492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Accepted: 12/09/2023] [Indexed: 01/05/2024] Open
Abstract
Human organoids-on-chips (OrgOCs) are the synergism of human organoids (HOs) technology and microfluidic organs-on-chips (OOCs). OOCs can mimic extrinsic characteristics of organs, such as environmental clues of living tissue, while HOs are more amenable to biological analysis and genetic manipulation. By spatial cooperation, OrgOCs served as 3D organotypic living models allowing them to recapitulate critical tissue-specific properties and forecast human responses and outcomes. It represents a giant leap forward from the regular 2D cell monolayers and animal models in the improved human ecological niche modeling. In recent years, OrgOCs have offered potential promises for clinical studies and advanced the preclinical-to-clinical translation in medical and industrial fields. In this review, we highlight the cutting-edge achievements in OrgOCs, introduce the key features of OrgOCs architectures, and share the revolutionary applications in basic biology, disease modeling, preclinical assay and precision medicine. Furthermore, we discuss how to combine a wide range of disciplines with OrgOCs and accelerate translational applications, as well as the challenges and opportunities of OrgOCs in biomedical research and applications.
Collapse
Affiliation(s)
- Hui Wang
- Department of Interventional & Vascular Surgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, China
- Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China
| | - Xiufan Ning
- Department of Interventional & Vascular Surgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, China
| | - Feng Zhao
- College of Basic Medical Sciences, Dalian Medical University, Dalian 116044, China
| | - Hui Zhao
- Department of Interventional & Vascular Surgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, China
| | - Dong Li
- Department of Interventional & Vascular Surgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, China
| |
Collapse
|
8
|
Urzi A, Lahmann I, Nguyen LVN, Rost BR, García-Pérez A, Lelievre N, Merritt-Garza ME, Phan HC, Bassell GJ, Rossoll W, Diecke S, Kunz S, Schmitz D, Gouti M. Efficient generation of a self-organizing neuromuscular junction model from human pluripotent stem cells. Nat Commun 2023; 14:8043. [PMID: 38114482 PMCID: PMC10730704 DOI: 10.1038/s41467-023-43781-3] [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: 01/19/2023] [Accepted: 11/20/2023] [Indexed: 12/21/2023] Open
Abstract
The complex neuromuscular network that controls body movements is the target of severe diseases that result in paralysis and death. Here, we report the development of a robust and efficient self-organizing neuromuscular junction (soNMJ) model from human pluripotent stem cells that can be maintained long-term in simple adherent conditions. The timely application of specific patterning signals instructs the simultaneous development and differentiation of position-specific brachial spinal neurons, skeletal muscles, and terminal Schwann cells. High-content imaging reveals self-organized bundles of aligned muscle fibers surrounded by innervating motor neurons that form functional neuromuscular junctions. Optogenetic activation and pharmacological interventions show that the spinal neurons actively instruct the synchronous skeletal muscle contraction. The generation of a soNMJ model from spinal muscular atrophy patient-specific iPSCs reveals that the number of NMJs and muscle contraction is severely affected, resembling the patient's pathology. In the future, the soNMJ model could be used for high-throughput studies in disease modeling and drug development. Thus, this model will allow us to address unmet needs in the neuromuscular disease field.
Collapse
Affiliation(s)
- Alessia Urzi
- Stem Cell Modeling of Development & Disease Group, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), 13125, Berlin, Germany
| | - Ines Lahmann
- Stem Cell Modeling of Development & Disease Group, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), 13125, Berlin, Germany
| | - Lan Vi N Nguyen
- Stem Cell Modeling of Development & Disease Group, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), 13125, Berlin, Germany
| | - Benjamin R Rost
- German Center for Neurodegenerative Diseases (DZNE), Berlin, Germany
| | - Angélica García-Pérez
- Stem Cell Modeling of Development & Disease Group, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), 13125, Berlin, Germany
| | - Noemie Lelievre
- Stem Cell Modeling of Development & Disease Group, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), 13125, Berlin, Germany
| | - Megan E Merritt-Garza
- Department of Cell Biology, Laboratory for Translational Cell Biology, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Han C Phan
- Department of Pediatrics, University of Alabama, Birmingham, AL, 35294, USA
| | - Gary J Bassell
- Department of Cell Biology, Laboratory for Translational Cell Biology, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Wilfried Rossoll
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, 32224, USA
- Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Sebastian Diecke
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Technology Platform Pluripotent Stem Cells, 13125, Berlin, Germany
| | - Severine Kunz
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Technology Platform Electron Microscopy, 13125, Berlin, Germany
| | - Dietmar Schmitz
- German Center for Neurodegenerative Diseases (DZNE), Berlin, Germany
- Berlin Institute of Health, NeuroCure Cluster of Excellence, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Mina Gouti
- Stem Cell Modeling of Development & Disease Group, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), 13125, Berlin, Germany.
| |
Collapse
|
9
|
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: 1.0] [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
|
10
|
Leng Y, Li X, Zheng F, Liu H, Wang C, Wang X, Liao Y, Liu J, Meng K, Yu J, Zhang J, Wang B, Tan Y, Liu M, Jia X, Li D, Li Y, Gu Z, Fan Y. Advances in In Vitro Models of Neuromuscular Junction: Focusing on Organ-on-a-Chip, Organoids, and Biohybrid Robotics. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2211059. [PMID: 36934404 DOI: 10.1002/adma.202211059] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Revised: 02/18/2023] [Indexed: 06/18/2023]
Abstract
The neuromuscular junction (NMJ) is a peripheral synaptic connection between presynaptic motor neurons and postsynaptic skeletal muscle fibers that enables muscle contraction and voluntary motor movement. Many traumatic, neurodegenerative, and neuroimmunological diseases are classically believed to mainly affect either the neuronal or the muscle side of the NMJ, and treatment options are lacking. Recent advances in novel techniques have helped develop in vitro physiological and pathophysiological models of the NMJ as well as enable precise control and evaluation of its functions. This paper reviews the recent developments in in vitro NMJ models with 2D or 3D cultures, from organ-on-a-chip and organoids to biohybrid robotics. Related derivative techniques are introduced for functional analysis of the NMJ, such as the patch-clamp technique, microelectrode arrays, calcium imaging, and stimulus methods, particularly optogenetic-mediated light stimulation, microelectrode-mediated electrical stimulation, and biochemical stimulation. Finally, the applications of the in vitro NMJ models as disease models or for drug screening related to suitable neuromuscular diseases are summarized and their future development trends and challenges are discussed.
Collapse
Affiliation(s)
- Yubing Leng
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, and with the School of Engineering Medicine, Beihang University, Beijing, 100083, China
| | - Xiaorui Li
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, and with the School of Engineering Medicine, Beihang University, Beijing, 100083, China
| | - Fuyin Zheng
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, and with the School of Engineering Medicine, Beihang University, Beijing, 100083, China
| | - Hui Liu
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, and with the School of Engineering Medicine, Beihang University, Beijing, 100083, China
| | - Chunyan Wang
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, 100094, China
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
| | - Xudong Wang
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, and with the School of Engineering Medicine, Beihang University, Beijing, 100083, China
| | - Yulong Liao
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, and with the School of Engineering Medicine, Beihang University, Beijing, 100083, China
| | - Jiangyue Liu
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, and with the School of Engineering Medicine, Beihang University, Beijing, 100083, China
| | - Kaiqi Meng
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, and with the School of Engineering Medicine, Beihang University, Beijing, 100083, China
| | - Jiaheng Yu
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, and with the School of Engineering Medicine, Beihang University, Beijing, 100083, China
| | - Jingyi Zhang
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, and with the School of Engineering Medicine, Beihang University, Beijing, 100083, China
| | - Binyu Wang
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, and with the School of Engineering Medicine, Beihang University, Beijing, 100083, China
| | - Yingjun Tan
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, 100094, China
| | - Meili Liu
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, and with the School of Engineering Medicine, Beihang University, Beijing, 100083, China
| | - Xiaoling Jia
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, and with the School of Engineering Medicine, Beihang University, Beijing, 100083, China
| | - Deyu Li
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, and with the School of Engineering Medicine, Beihang University, Beijing, 100083, China
| | - Yinghui Li
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, 100094, China
| | - Zhongze Gu
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
| | - Yubo Fan
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, and with the School of Engineering Medicine, Beihang University, Beijing, 100083, China
| |
Collapse
|
11
|
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
|
12
|
Ambrin G, Cai S, Singh BR. Critical analysis in the advancement of cell-based assays for botulinum neurotoxin. Crit Rev Microbiol 2023; 49:1-17. [PMID: 35212259 DOI: 10.1080/1040841x.2022.2035315] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
The study on botulinum neurotoxins (BoNTs) has rapidly evolved for their structure and functions as opposed to them being poisons or cures. Since their discoveries, the scientific community has come a long way in understanding BoNTs' structure and biological activity. Given its current application as a tool for understanding neurocellular activity and as a drug against over 800 neurological disorders, relevant and sensitive assays have become critical for biochemical, physiological, and pharmacological studies. The natural entry of the toxin being ingestion, it has also become important to examine its mechanism while crossing the epithelial cell barrier. Several techniques and methodologies have been developed, for its entry, pharmacokinetics, and biological activity for identification, and drug efficacy both in vivo and in vitro conditions. However, each of them presents its own challenges. The cell-based assay is a platform that exceeds the sensitivity of mouse bioassay while encompassing all the steps of intoxication including cell binding, transcytosis, endocytosis, translocation and proteolytic activity. In this article we review in detail both the neuronal and nonneuronal based cellular interaction of BoNT involving its transportation, and interaction with the targeted cells, and intracellular activities.
Collapse
Affiliation(s)
- Ghuncha Ambrin
- Department of Biomedical Engineering and Biotechnology, University of Massachusetts, Dartmouth, MA, USA.,Department of Chemistry and Biochemistry, University of Massachusetts, Dartmouth, MA, USA
| | - Shuowei Cai
- Department of Chemistry and Biochemistry, University of Massachusetts, Dartmouth, MA, USA
| | - Bal Ram Singh
- Institute of Advanced Sciences, Botulinum Research Center, Dartmouth, MA, USA
| |
Collapse
|
13
|
Gao Y. Using Human iPSC-Derived Peripheral Nervous System Disease Models for Drug Discovery. Handb Exp Pharmacol 2023; 281:191-205. [PMID: 37815594 DOI: 10.1007/164_2023_690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/11/2023]
Abstract
Induced pluripotent stem cells (IPSCs), with their remarkable ability to differentiate into various cell types, including peripheral nervous system cells such as neurons and glial cells, offer an excellent platform for in vitro disease modeling. These iPSC-derived disease models have proven valuable in drug discovery, as they provide more precise simulations of a patient's disease state and allow for the assessment of potential therapeutic effectiveness and safety.
Collapse
Affiliation(s)
- Yuan Gao
- Translational Medicine Research Center, Shanxi Medical University, Taiyuan, China
| |
Collapse
|
14
|
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: 1.5] [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
|
15
|
Hyperglycemia Negatively Affects IPSC-Derived Myoblast Proliferation and Skeletal Muscle Regeneration and Function. Cells 2022; 11:cells11223674. [PMID: 36429100 PMCID: PMC9688533 DOI: 10.3390/cells11223674] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 10/17/2022] [Accepted: 10/28/2022] [Indexed: 11/22/2022] Open
Abstract
Diabetic myopathy is a co-morbidity diagnosed in most diabetes mellitus patients, yet its pathogenesis is still understudied, which hinders the development of effective therapies. This project aimed to investigate the effect of hyperglycemia on human myoblast physiology, devoid of other complicating factors, by utilizing human myoblasts derived from induced pluripotent stem cells (iPSCs), in a defined in vitro system. IPSC-derived myoblasts were expanded under three glucose conditions: low (5 mM), medium (17.5 mM) or high (25 mM). While hyperglycemic myoblasts demonstrated upregulation of Glut4 relative to the euglycemic control, myoblast proliferation demonstrated a glucose dose-dependent impedance. Further cellular analysis revealed a retarded cell cycle progression trapped at the S phase and G2/M phase and an impaired mitochondrial function in hyperglycemic myoblasts. Terminal differentiation of these hyperglycemic myoblasts resulted in significantly hypertrophic and highly branched myotubes with disturbed myosin heavy chain arrangement. Lastly, functional assessment of these myofibers derived from hyperglycemic myoblasts demonstrated comparatively increased fatigability. Collectively, the hyperglycemic myoblasts demonstrated deficient muscle regeneration capability and functionality, which falls in line with the sarcopenia symptoms observed in diabetic myopathy patients. This human-based iPSC-derived skeletal muscle hyperglycemic model provides a valuable platform for mechanistic investigation of diabetic myopathy and therapeutic development.
Collapse
|
16
|
Sasserath T, Robertson AL, Mendez R, Hays TT, Smith E, Cooper H, Akanda N, Rumsey JW, Guo X, Farkhondeh A, Pradhan M, Baumgaertel K, Might M, Rodems S, Zheng W, Hickman JJ. An induced pluripotent stem cell-derived NMJ platform for study of the NGLY1-Congenital Disorder of Deglycosylation. ADVANCED THERAPEUTICS 2022; 5:2200009. [PMID: 36589922 PMCID: PMC9798846 DOI: 10.1002/adtp.202200009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Indexed: 01/05/2023]
Abstract
There are many neurological rare diseases where animal models have proven inadequate or do not currently exist. NGLY1 Deficiency, a congenital disorder of deglycosylation, is a rare disease that predominantly affects motor control, especially control of neuromuscular action. In this study, NGLY1-deficient, patient-derived induced pluripotent stem cells (iPSCs) were differentiated into motoneurons (MNs) to identify disease phenotypes analogous to clinical disease pathology with significant deficits apparent in the NGLY1-deficient lines compared to the control. A neuromuscular junction (NMJ) model was developed using patient and wild type (WT) MNs to study functional differences between healthy and diseased NMJs. Reduced axon length, increased and shortened axon branches, MN action potential (AP) bursting and decreased AP firing rate and amplitude were observed in the NGLY1-deficient MNs in monoculture. When transitioned to the NMJ-coculture system, deficits in NMJ number, stability, failure rate, and synchronicity with indirect skeletal muscle (SkM) stimulation were observed. This project establishes a phenotypic NGLY1 model for investigation of possible therapeutics and investigations into mechanistic deficits in the system.
Collapse
Affiliation(s)
- Trevor Sasserath
- Hesperos, Inc., 12501 Research Parkway, Suite 100, Orlando, FL 32826 USA
| | - Ashley L Robertson
- Hesperos, Inc., 12501 Research Parkway, Suite 100, Orlando, FL 32826 USA
| | - Roxana Mendez
- University of Central Florida, NanoScience Technology Center, 12424 Research Parkway, Suite 400, Orlando, FL 32826 USA
| | - Tristan T Hays
- Hesperos, Inc., 12501 Research Parkway, Suite 100, Orlando, FL 32826 USA
| | - Ethan Smith
- Hesperos, Inc., 12501 Research Parkway, Suite 100, Orlando, FL 32826 USA
| | - Helena Cooper
- Hesperos, Inc., 12501 Research Parkway, Suite 100, Orlando, FL 32826 USA
| | - Nesar Akanda
- University of Central Florida, NanoScience Technology Center, 12424 Research Parkway, Suite 400, Orlando, FL 32826 USA
| | - John W Rumsey
- Hesperos, Inc., 12501 Research Parkway, Suite 100, Orlando, FL 32826 USA
| | - Xiufang Guo
- University of Central Florida, NanoScience Technology Center, 12424 Research Parkway, Suite 400, Orlando, FL 32826 USA
| | - Atena Farkhondeh
- National Center for Advancing Translational Sciences, National Institutes of Health, 9800 Medical Center Drive, Building C, Room 310W Rockville, MD 20850, USA
| | - Manisha Pradhan
- National Center for Advancing Translational Sciences, National Institutes of Health, 9800 Medical Center Drive, Building C, Room 310W Rockville, MD 20850, USA
| | - Karsten Baumgaertel
- Travere Therapeutics, 3611 Valley Centre Drive, Suite 300, San Diego, CA, USA
| | - Matthew Might
- University of Alabama at Birmingham, Hugh Kaul Precision Medicine Institute, 510 20th St S, Office 858B, Birmingham, AL 35210, USA
| | - Steven Rodems
- Travere Therapeutics, 3611 Valley Centre Drive, Suite 300, San Diego, CA, USA
| | - Wei Zheng
- National Center for Advancing Translational Sciences, National Institutes of Health, 9800 Medical Center Drive, Building C, Room 310W Rockville, MD 20850, USA
| | - James J Hickman
- Hesperos, Inc., 12501 Research Parkway, Suite 100, Orlando, FL 32826 USA
- University of Central Florida, NanoScience Technology Center, 12424 Research Parkway, Suite 400, Orlando, FL 32826 USA
| |
Collapse
|
17
|
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]
|
18
|
Miny L, Maisonneuve BGC, Quadrio I, Honegger T. Modeling Neurodegenerative Diseases Using In Vitro Compartmentalized Microfluidic Devices. Front Bioeng Biotechnol 2022; 10:919646. [PMID: 35813998 PMCID: PMC9263267 DOI: 10.3389/fbioe.2022.919646] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 05/31/2022] [Indexed: 01/27/2023] Open
Abstract
The human brain is a complex organ composed of many different types of cells interconnected to create an organized system able to efficiently process information. Dysregulation of this delicately balanced system can lead to the development of neurological disorders, such as neurodegenerative diseases (NDD). To investigate the functionality of human brain physiology and pathophysiology, the scientific community has been generated various research models, from genetically modified animals to two- and three-dimensional cell culture for several decades. These models have, however, certain limitations that impede the precise study of pathophysiological features of neurodegeneration, thus hindering therapeutical research and drug development. Compartmentalized microfluidic devices provide in vitro minimalistic environments to accurately reproduce neural circuits allowing the characterization of the human central nervous system. Brain-on-chip (BoC) is allowing our capability to improve neurodegeneration models on the molecular and cellular mechanism aspects behind the progression of these troubles. This review aims to summarize and discuss the latest advancements of microfluidic models for the investigations of common neurodegenerative disorders, such as Alzheimer's disease, Parkinson's disease, and amyotrophic lateral sclerosis.
Collapse
Affiliation(s)
- Louise Miny
- NETRI, Lyon, France
- BIORAN Team, Lyon Neuroscience Research Center, CNRS UMR 5292, INSERM U1028, Lyon 1 University, Bron, France
| | | | - Isabelle Quadrio
- BIORAN Team, Lyon Neuroscience Research Center, CNRS UMR 5292, INSERM U1028, Lyon 1 University, Bron, France
- Laboratory of Neurobiology and Neurogenetics, Department of Biochemistry and Molecular Biology, Lyon University Hospital, Bron, France
| | | |
Collapse
|
19
|
Filippi M, Buchner T, Yasa O, Weirich S, Katzschmann RK. Microfluidic Tissue Engineering and Bio-Actuation. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2108427. [PMID: 35194852 DOI: 10.1002/adma.202108427] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 02/07/2022] [Indexed: 06/14/2023]
Abstract
Bio-hybrid technologies aim to replicate the unique capabilities of biological systems that could surpass advanced artificial technologies. Soft bio-hybrid robots consist of synthetic and living materials and have the potential to self-assemble, regenerate, work autonomously, and interact safely with other species and the environment. Cells require a sufficient exchange of nutrients and gases, which is guaranteed by convection and diffusive transport through liquid media. The functional development and long-term survival of biological tissues in vitro can be improved by dynamic flow culture, but only microfluidic flow control can develop tissue with fine structuring and regulation at the microscale. Full control of tissue growth at the microscale will eventually lead to functional macroscale constructs, which are needed as the biological component of soft bio-hybrid technologies. This review summarizes recent progress in microfluidic techniques to engineer biological tissues, focusing on the use of muscle cells for robotic bio-actuation. Moreover, the instances in which bio-actuation technologies greatly benefit from fusion with microfluidics are highlighted, which include: the microfabrication of matrices, biomimicry of cell microenvironments, tissue maturation, perfusion, and vascularization.
Collapse
Affiliation(s)
- Miriam Filippi
- Soft Robotics Laboratory, ETH Zurich, Tannenstrasse 3, Zurich, 8092, Switzerland
| | - Thomas Buchner
- Soft Robotics Laboratory, ETH Zurich, Tannenstrasse 3, Zurich, 8092, Switzerland
| | - Oncay Yasa
- Soft Robotics Laboratory, ETH Zurich, Tannenstrasse 3, Zurich, 8092, Switzerland
| | - Stefan Weirich
- Soft Robotics Laboratory, ETH Zurich, Tannenstrasse 3, Zurich, 8092, Switzerland
| | - Robert K Katzschmann
- Soft Robotics Laboratory, ETH Zurich, Tannenstrasse 3, Zurich, 8092, Switzerland
| |
Collapse
|
20
|
Cheesbrough A, Sciscione F, Riccio F, Harley P, R'Bibo L, Ziakas G, Darbyshire A, Lieberam I, Song W. Biobased Elastomer Nanofibers Guide Light-Controlled Human-iPSC-Derived Skeletal Myofibers. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2110441. [PMID: 35231133 PMCID: PMC9131876 DOI: 10.1002/adma.202110441] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 02/25/2022] [Indexed: 05/07/2023]
Abstract
Generating skeletal muscle tissue that mimics the cellular alignment, maturation, and function of native skeletal muscle is an ongoing challenge in disease modeling and regenerative therapies. Skeletal muscle cultures require extracellular guidance and mechanical support to stabilize contractile myofibers. Existing microfabrication-based solutions are limited by complex fabrication steps, low throughput, and challenges in measuring dynamic contractile function. Here, the synthesis and characterization of a new biobased nanohybrid elastomer, which is electrospun into aligned nanofiber sheets to mimic the skeletal muscle extracellular matrix, is presented. The polymer exhibits remarkable hyperelasticity well-matched to that of native skeletal muscle (≈11-50 kPa), with ultimate strain ≈1000%, and elastic modulus ≈25 kPa. Uniaxially aligned nanofibers guide myoblast alignment, enhance sarcomere formation, and promote a ≈32% increase in myotube fusion and ≈50% increase in myofiber maturation. The elastomer nanofibers stabilize optogenetically controlled human induced pluripotent stem cell derived skeletal myofibers. When activated by blue light, the myofiber-nanofiber hybrid constructs maintain a significantly higher (>200%) contraction velocity and specific force (>280%) compared to conventional culture methods. The engineered myofibers exhibit a power density of ≈35 W m-3 . This system is a promising new skeletal muscle tissue model for applications in muscular disease modeling, drug discovery, and muscle regeneration.
Collapse
Affiliation(s)
- Aimee Cheesbrough
- UCL Centre for Biomaterials in Surgical Reconstruction and RegenerationDepartment of Surgical BiotechnologyDivision of Surgery and Interventional ScienceUniversity College LondonLondonNW3 2PFUK
- Centre for Gene Therapy and Regenerative MedicineMRC Centre for Neurodevelopmental DisordersCentre for Developmental NeurobiologyKings College LondonLondonSE1 9RTUK
| | - Fabiola Sciscione
- UCL Centre for Biomaterials in Surgical Reconstruction and RegenerationDepartment of Surgical BiotechnologyDivision of Surgery and Interventional ScienceUniversity College LondonLondonNW3 2PFUK
| | - Federica Riccio
- Centre for Gene Therapy and Regenerative MedicineMRC Centre for Neurodevelopmental DisordersCentre for Developmental NeurobiologyKings College LondonLondonSE1 9RTUK
| | - Peter Harley
- Centre for Gene Therapy and Regenerative MedicineMRC Centre for Neurodevelopmental DisordersCentre for Developmental NeurobiologyKings College LondonLondonSE1 9RTUK
| | - Lea R'Bibo
- Centre for Gene Therapy and Regenerative MedicineMRC Centre for Neurodevelopmental DisordersCentre for Developmental NeurobiologyKings College LondonLondonSE1 9RTUK
| | - Georgios Ziakas
- UCL Centre for Biomaterials in Surgical Reconstruction and RegenerationDepartment of Surgical BiotechnologyDivision of Surgery and Interventional ScienceUniversity College LondonLondonNW3 2PFUK
| | - Arnold Darbyshire
- UCL Centre for Biomaterials in Surgical Reconstruction and RegenerationDepartment of Surgical BiotechnologyDivision of Surgery and Interventional ScienceUniversity College LondonLondonNW3 2PFUK
| | - Ivo Lieberam
- Centre for Gene Therapy and Regenerative MedicineMRC Centre for Neurodevelopmental DisordersCentre for Developmental NeurobiologyKings College LondonLondonSE1 9RTUK
| | - Wenhui Song
- UCL Centre for Biomaterials in Surgical Reconstruction and RegenerationDepartment of Surgical BiotechnologyDivision of Surgery and Interventional ScienceUniversity College LondonLondonNW3 2PFUK
| |
Collapse
|
21
|
Dubinskaya AD, Yurova OV, Fesyun AD, Kukshina AA. Correction of Psycho-Emotional Status Using Neuromuscular Relaxation of the Face. J Nerv Ment Dis 2022; 210:179-185. [PMID: 34456322 DOI: 10.1097/nmd.0000000000001413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
ABSTRACT This article is relevant, as most healthy women face the problem of increased facial muscle tone. The purpose of the study was to investigate current problems of the relationship between tonic activity of the facial muscles and psycho-emotional states. The study included a group of 151 women subjected to psychological testing. According to its results, they were randomized into five study groups depending on sex and age. The frequency and severity of psycho-emotional states and parameters of tonic activity of the facial muscles were studied. Currently accepted methods for the correction of psycho-emotional states based on the feedback mechanism were analyzed. The efficacy of myofascial facial massage as an instrument for psychological status correction was evaluated. The long-term effects of various massage techniques aimed at facial muscle relaxation on the psycho-emotional state were established. The most effective tools for muscle tone correction were proposed for psycho-emotional status correction.
Collapse
Affiliation(s)
- Anastasia D Dubinskaya
- Moscow Centre for Research and Practice in Medical Rehabilitation, Restorative and Sports Medicine of the Moscow Healthcare Department
| | - Olga V Yurova
- Moscow Centre for Research and Practice in Medical Rehabilitation, Restorative and Sports Medicine of the Moscow Healthcare Department
| | - Anatoliy D Fesyun
- Medical Research Center of Rehabilitation and Balneology of the Ministry of Health of Russian Federation, Moscow, Russian Federation
| | - Anastasia A Kukshina
- Moscow Centre for Research and Practice in Medical Rehabilitation, Restorative and Sports Medicine of the Moscow Healthcare Department
| |
Collapse
|
22
|
Lynch E, Peek E, Reilly M, FitzGibbons C, Robertson S, Suzuki M. Current Progress in the Creation, Characterization, and Application of Human Stem Cell-derived in Vitro Neuromuscular Junction Models. Stem Cell Rev Rep 2022; 18:768-780. [PMID: 34212303 PMCID: PMC8720113 DOI: 10.1007/s12015-021-10201-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/03/2021] [Indexed: 02/03/2023]
Abstract
Human pluripotent stem cells (PSCs) such as embryonic stem cells (ESCs) or induced pluripotent stem cells (iPSCs) are of great value for studying developmental processes, disease modeling, and drug testing. One area in which the use of human PSCs has become of great interest in recent years is for in vitro models of the neuromuscular junction (NMJ). The NMJ is a synapse at which a motor neuron releases acetylcholine to bind to skeletal muscle and stimulate contraction. Degeneration of the NMJ and subsequent loss of muscle function is a common feature of many neuromuscular diseases such as myasthenia gravis, spinal muscular atrophy, and amyotrophic lateral sclerosis. In order to develop new therapies for patients with neuromuscular diseases, it is essential to understand mechanisms taking place at the NMJ. However, we have limited ability to study the NMJ in living human patients, and animal models are limited by physiological relevance. Therefore, an in vitro model of the NMJ consisting of human cells is of great value. The use of stem cells for in vitro NMJ models is still in progress and requires further optimization in order to yield reliable, reproducible results. The objective of this review is (1) to outline the current progress towards fully PSC-derived in vitro co-culture models of the human NMJ and (2) to discuss future directions and challenges that must be overcome in order to create reproducible fully PSC-derived models that can be used for developmental studies, disease modeling, and drug testing.
Collapse
Affiliation(s)
- Eileen Lynch
- Department of Comparative Biosciences, University of Wisconsin-Madison, Wisconsin, USA
| | - Emma Peek
- Department of Comparative Biosciences, University of Wisconsin-Madison, Wisconsin, USA
| | - Megan Reilly
- Department of Comparative Biosciences, University of Wisconsin-Madison, Wisconsin, USA
| | - Claire FitzGibbons
- Department of Comparative Biosciences, University of Wisconsin-Madison, Wisconsin, USA
| | - Samantha Robertson
- Department of Comparative Biosciences, University of Wisconsin-Madison, Wisconsin, USA
| | - Masatoshi Suzuki
- Department of Comparative Biosciences, University of Wisconsin-Madison, Wisconsin, USA,Stem Cell and Regenerative Medicine Center, University of Wisconsin-Madison, Wisconsin, USA
| |
Collapse
|
23
|
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: 2.5] [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
|
24
|
Youhanna S, Kemas AM, Preiss L, Zhou Y, Shen JX, Cakal SD, Paqualini FS, Goparaju SK, Shafagh RZ, Lind JU, Sellgren CM, Lauschke VM. Organotypic and Microphysiological Human Tissue Models for Drug Discovery and Development-Current State-of-the-Art and Future Perspectives. Pharmacol Rev 2022; 74:141-206. [PMID: 35017176 DOI: 10.1124/pharmrev.120.000238] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 10/12/2021] [Indexed: 12/11/2022] Open
Abstract
The number of successful drug development projects has been stagnant for decades despite major breakthroughs in chemistry, molecular biology, and genetics. Unreliable target identification and poor translatability of preclinical models have been identified as major causes of failure. To improve predictions of clinical efficacy and safety, interest has shifted to three-dimensional culture methods in which human cells can retain many physiologically and functionally relevant phenotypes for extended periods of time. Here, we review the state of the art of available organotypic culture techniques and critically review emerging models of human tissues with key importance for pharmacokinetics, pharmacodynamics, and toxicity. In addition, developments in bioprinting and microfluidic multiorgan cultures to emulate systemic drug disposition are summarized. We close by highlighting important trends regarding the fabrication of organotypic culture platforms and the choice of platform material to limit drug absorption and polymer leaching while supporting the phenotypic maintenance of cultured cells and allowing for scalable device fabrication. We conclude that organotypic and microphysiological human tissue models constitute promising systems to promote drug discovery and development by facilitating drug target identification and improving the preclinical evaluation of drug toxicity and pharmacokinetics. There is, however, a critical need for further validation, benchmarking, and consolidation efforts ideally conducted in intersectoral multicenter settings to accelerate acceptance of these novel models as reliable tools for translational pharmacology and toxicology. SIGNIFICANCE STATEMENT: Organotypic and microphysiological culture of human cells has emerged as a promising tool for preclinical drug discovery and development that might be able to narrow the translation gap. This review discusses recent technological and methodological advancements and the use of these systems for hit discovery and the evaluation of toxicity, clearance, and absorption of lead compounds.
Collapse
Affiliation(s)
- Sonia Youhanna
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.Y., A.M.K., L.P., Y.Z., J.X.S., S.K.G., R.Z.S., C.M.S., V.M.L.); Department of Drug Metabolism and Pharmacokinetics (DMPK), Merck KGaA, Darmstadt, Germany (L.P.); Department of Health Technology, Technical University of Denmark, Lyngby, Denmark (S.D.C., J.U.L.); Synthetic Physiology Laboratory, Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy (F.S.P.); Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden (Z.S.); and Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.)
| | - Aurino M Kemas
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.Y., A.M.K., L.P., Y.Z., J.X.S., S.K.G., R.Z.S., C.M.S., V.M.L.); Department of Drug Metabolism and Pharmacokinetics (DMPK), Merck KGaA, Darmstadt, Germany (L.P.); Department of Health Technology, Technical University of Denmark, Lyngby, Denmark (S.D.C., J.U.L.); Synthetic Physiology Laboratory, Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy (F.S.P.); Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden (Z.S.); and Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.)
| | - Lena Preiss
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.Y., A.M.K., L.P., Y.Z., J.X.S., S.K.G., R.Z.S., C.M.S., V.M.L.); Department of Drug Metabolism and Pharmacokinetics (DMPK), Merck KGaA, Darmstadt, Germany (L.P.); Department of Health Technology, Technical University of Denmark, Lyngby, Denmark (S.D.C., J.U.L.); Synthetic Physiology Laboratory, Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy (F.S.P.); Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden (Z.S.); and Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.)
| | - Yitian Zhou
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.Y., A.M.K., L.P., Y.Z., J.X.S., S.K.G., R.Z.S., C.M.S., V.M.L.); Department of Drug Metabolism and Pharmacokinetics (DMPK), Merck KGaA, Darmstadt, Germany (L.P.); Department of Health Technology, Technical University of Denmark, Lyngby, Denmark (S.D.C., J.U.L.); Synthetic Physiology Laboratory, Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy (F.S.P.); Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden (Z.S.); and Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.)
| | - Joanne X Shen
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.Y., A.M.K., L.P., Y.Z., J.X.S., S.K.G., R.Z.S., C.M.S., V.M.L.); Department of Drug Metabolism and Pharmacokinetics (DMPK), Merck KGaA, Darmstadt, Germany (L.P.); Department of Health Technology, Technical University of Denmark, Lyngby, Denmark (S.D.C., J.U.L.); Synthetic Physiology Laboratory, Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy (F.S.P.); Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden (Z.S.); and Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.)
| | - Selgin D Cakal
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.Y., A.M.K., L.P., Y.Z., J.X.S., S.K.G., R.Z.S., C.M.S., V.M.L.); Department of Drug Metabolism and Pharmacokinetics (DMPK), Merck KGaA, Darmstadt, Germany (L.P.); Department of Health Technology, Technical University of Denmark, Lyngby, Denmark (S.D.C., J.U.L.); Synthetic Physiology Laboratory, Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy (F.S.P.); Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden (Z.S.); and Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.)
| | - Francesco S Paqualini
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.Y., A.M.K., L.P., Y.Z., J.X.S., S.K.G., R.Z.S., C.M.S., V.M.L.); Department of Drug Metabolism and Pharmacokinetics (DMPK), Merck KGaA, Darmstadt, Germany (L.P.); Department of Health Technology, Technical University of Denmark, Lyngby, Denmark (S.D.C., J.U.L.); Synthetic Physiology Laboratory, Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy (F.S.P.); Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden (Z.S.); and Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.)
| | - Sravan K Goparaju
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.Y., A.M.K., L.P., Y.Z., J.X.S., S.K.G., R.Z.S., C.M.S., V.M.L.); Department of Drug Metabolism and Pharmacokinetics (DMPK), Merck KGaA, Darmstadt, Germany (L.P.); Department of Health Technology, Technical University of Denmark, Lyngby, Denmark (S.D.C., J.U.L.); Synthetic Physiology Laboratory, Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy (F.S.P.); Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden (Z.S.); and Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.)
| | - Reza Zandi Shafagh
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.Y., A.M.K., L.P., Y.Z., J.X.S., S.K.G., R.Z.S., C.M.S., V.M.L.); Department of Drug Metabolism and Pharmacokinetics (DMPK), Merck KGaA, Darmstadt, Germany (L.P.); Department of Health Technology, Technical University of Denmark, Lyngby, Denmark (S.D.C., J.U.L.); Synthetic Physiology Laboratory, Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy (F.S.P.); Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden (Z.S.); and Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.)
| | - Johan Ulrik Lind
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.Y., A.M.K., L.P., Y.Z., J.X.S., S.K.G., R.Z.S., C.M.S., V.M.L.); Department of Drug Metabolism and Pharmacokinetics (DMPK), Merck KGaA, Darmstadt, Germany (L.P.); Department of Health Technology, Technical University of Denmark, Lyngby, Denmark (S.D.C., J.U.L.); Synthetic Physiology Laboratory, Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy (F.S.P.); Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden (Z.S.); and Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.)
| | - Carl M Sellgren
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.Y., A.M.K., L.P., Y.Z., J.X.S., S.K.G., R.Z.S., C.M.S., V.M.L.); Department of Drug Metabolism and Pharmacokinetics (DMPK), Merck KGaA, Darmstadt, Germany (L.P.); Department of Health Technology, Technical University of Denmark, Lyngby, Denmark (S.D.C., J.U.L.); Synthetic Physiology Laboratory, Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy (F.S.P.); Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden (Z.S.); and Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.)
| | - Volker M Lauschke
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.Y., A.M.K., L.P., Y.Z., J.X.S., S.K.G., R.Z.S., C.M.S., V.M.L.); Department of Drug Metabolism and Pharmacokinetics (DMPK), Merck KGaA, Darmstadt, Germany (L.P.); Department of Health Technology, Technical University of Denmark, Lyngby, Denmark (S.D.C., J.U.L.); Synthetic Physiology Laboratory, Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy (F.S.P.); Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden (Z.S.); and Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.)
| |
Collapse
|
25
|
Xiang Y, Miller K, Guan J, Kiratitanaporn W, Tang M, Chen S. 3D bioprinting of complex tissues in vitro: state-of-the-art and future perspectives. Arch Toxicol 2022; 96:691-710. [PMID: 35006284 PMCID: PMC8850226 DOI: 10.1007/s00204-021-03212-y] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 12/20/2021] [Indexed: 12/15/2022]
Abstract
The pharmacology and toxicology of a broad variety of therapies and chemicals have significantly improved with the aid of the increasing in vitro models of complex human tissues. Offering versatile and precise control over the cell population, extracellular matrix (ECM) deposition, dynamic microenvironment, and sophisticated microarchitecture, which is desired for the in vitro modeling of complex tissues, 3D bio-printing is a rapidly growing technology to be employed in the field. In this review, we will discuss the recent advancement of printing techniques and bio-ink sources, which have been spurred on by the increasing demand for modeling tactics and have facilitated the development of the refined tissue models as well as the modeling strategies, followed by a state-of-the-art update on the specialized work on cancer, heart, muscle and liver. In the end, the toxicological modeling strategies, substantial challenges, and future perspectives for 3D printed tissue models were explored.
Collapse
Affiliation(s)
- Yi Xiang
- Department of NanoEngineering, University of California San Diego, La Jolla, USA
| | - Kathleen Miller
- Department of NanoEngineering, University of California San Diego, La Jolla, USA
| | - Jiaao Guan
- Department of Electrical and Computer Engineering, University of California San Diego, La Jolla, USA
| | | | - Min Tang
- Department of NanoEngineering, University of California San Diego, La Jolla, USA
| | - Shaochen Chen
- Department of NanoEngineering, University of California San Diego, La Jolla, USA.
- Department of Electrical and Computer Engineering, University of California San Diego, La Jolla, USA.
| |
Collapse
|
26
|
Hines TJ, Lutz C, Murray SA, Burgess RW. An Integrated Approach to Studying Rare Neuromuscular Diseases Using Animal and Human Cell-Based Models. Front Cell Dev Biol 2022; 9:801819. [PMID: 35047510 PMCID: PMC8762301 DOI: 10.3389/fcell.2021.801819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 11/30/2021] [Indexed: 11/13/2022] Open
Abstract
As sequencing technology improves, the identification of new disease-associated genes and new alleles of known genes is rapidly increasing our understanding of the genetic underpinnings of rare diseases, including neuromuscular diseases. However, precisely because these disorders are rare and often heterogeneous, they are difficult to study in patient populations. In parallel, our ability to engineer the genomes of model organisms, such as mice or rats, has gotten increasingly efficient through techniques such as CRISPR/Cas9 genome editing, allowing the creation of precision human disease models. Such in vivo model systems provide an efficient means for exploring disease mechanisms and identifying therapeutic strategies. Furthermore, animal models provide a platform for preclinical studies to test the efficacy of those strategies. Determining whether the same mechanisms are involved in the human disease and confirming relevant parameters for treatment ideally involves a human experimental system. One system currently being used is induced pluripotent stem cells (iPSCs), which can then be differentiated into the relevant cell type(s) for in vitro confirmation of disease mechanisms and variables such as target engagement. Here we provide a demonstration of these approaches using the example of tRNA-synthetase-associated inherited peripheral neuropathies, rare forms of Charcot-Marie-Tooth disease (CMT). Mouse models have led to a better understanding of both the genetic and cellular mechanisms underlying the disease. To determine if the mechanisms are similar in human cells, we will use genetically engineered iPSC-based models. This will allow comparisons of different CMT-associated GARS alleles in the same genetic background, reducing the variability found between patient samples and simplifying the availability of cell-based models for a rare disease. The necessity of integrating mouse and human models, strategies for accomplishing this integration, and the challenges of doing it at scale are discussed using recently published work detailing the cellular mechanisms underlying GARS-associated CMT as a framework.
Collapse
|
27
|
Abstract
This paper represents the first assessment of agent-induced hormetic dose responses in induced pluripotent stem cells and their derived cells. The hormetic dose responses were induced by a broad range of chemicals, including pharmaceuticals (eg, metformin), dietary supplements/extracts from medicinal plants (eg, curcumin), and endogenous agents (eg, melatonin). The paper assesses the mechanistic foundations of these induced hormetic dose responses, their therapeutic implications and comparison with hormetic responses in multiple adult and embryonic stem cells.
Collapse
Affiliation(s)
- Edward J Calabrese
- Department of Environmental Health Sciences, School of Public Health and Health Sciences, University of Massachusetts, Amherst, MA, USA
| |
Collapse
|
28
|
Ajalik RE, Alenchery RG, Cognetti JS, Zhang VZ, McGrath JL, Miller BL, Awad HA. Human Organ-on-a-Chip Microphysiological Systems to Model Musculoskeletal Pathologies and Accelerate Therapeutic Discovery. Front Bioeng Biotechnol 2022; 10:846230. [PMID: 35360391 PMCID: PMC8964284 DOI: 10.3389/fbioe.2022.846230] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Accepted: 02/21/2022] [Indexed: 12/12/2022] Open
Abstract
Human Microphysiological Systems (hMPS), otherwise known as organ- and tissue-on-a-chip models, are an emerging technology with the potential to replace in vivo animal studies with in vitro models that emulate human physiology at basic levels. hMPS platforms are designed to overcome limitations of two-dimensional (2D) cell culture systems by mimicking 3D tissue organization and microenvironmental cues that are physiologically and clinically relevant. Unlike animal studies, hMPS models can be configured for high content or high throughput screening in preclinical drug development. Applications in modeling acute and chronic injuries in the musculoskeletal system are slowly developing. However, the complexity and load bearing nature of musculoskeletal tissues and joints present unique challenges related to our limited understanding of disease mechanisms and the lack of consensus biomarkers to guide biological therapy development. With emphasis on examples of modeling musculoskeletal tissues, joints on chips, and organoids, this review highlights current trends of microphysiological systems technology. The review surveys state-of-the-art design and fabrication considerations inspired by lessons from bioreactors and biological variables emphasizing the role of induced pluripotent stem cells and genetic engineering in creating isogenic, patient-specific multicellular hMPS. The major challenges in modeling musculoskeletal tissues using hMPS chips are identified, including incorporating biological barriers, simulating joint compartments and heterogenous tissue interfaces, simulating immune interactions and inflammatory factors, simulating effects of in vivo loading, recording nociceptors responses as surrogates for pain outcomes, modeling the dynamic injury and healing responses by monitoring secreted proteins in real time, and creating arrayed formats for robotic high throughput screens. Overcoming these barriers will revolutionize musculoskeletal research by enabling physiologically relevant, predictive models of human tissues and joint diseases to accelerate and de-risk therapeutic discovery and translation to the clinic.
Collapse
Affiliation(s)
- Raquel E. Ajalik
- Center for Musculoskeletal Research, University of Rochester, Rochester, NY, United States
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, United States
| | - Rahul G. Alenchery
- Center for Musculoskeletal Research, University of Rochester, Rochester, NY, United States
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, United States
| | - John S. Cognetti
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, United States
| | - Victor Z. Zhang
- Center for Musculoskeletal Research, University of Rochester, Rochester, NY, United States
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, United States
| | - James L. McGrath
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, United States
| | - Benjamin L. Miller
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, United States
- Department of Dermatology, University of Rochester, Rochester, NY, United States
| | - Hani A. Awad
- Center for Musculoskeletal Research, University of Rochester, Rochester, NY, United States
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, United States
- *Correspondence: Hani A. Awad,
| |
Collapse
|
29
|
de Lamotte JD, Polentes J, Roussange F, Lesueur L, Feurgard P, Perrier A, Nicoleau C, Martinat C. Optogenetically controlled human functional motor endplate for testing botulinum neurotoxins. Stem Cell Res Ther 2021; 12:599. [PMID: 34865655 PMCID: PMC8647380 DOI: 10.1186/s13287-021-02665-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Accepted: 11/18/2021] [Indexed: 11/19/2022] Open
Abstract
Background The lack of physiologically relevant and predictive cell-based assays is one of the major obstacles for testing and developing botulinum neurotoxins (BoNTs) therapeutics. Human-induced pluripotent stem cells (hiPSCs)-derivatives now offer the opportunity to improve the relevance of cellular models and thus the translational value of preclinical data. Methods We investigated the potential of hiPSC-derived motor neurons (hMNs) optical stimulation combined with calcium imaging in cocultured muscle cells activity to investigate BoNT-sensitivity of an in vitro model of human muscle-nerve system. Results Functional muscle-nerve coculture system was developed using hMNs and human immortalized skeletal muscle cells. Our results demonstrated that hMNs can innervate myotubes and induce contractions and calcium transient in muscle cells, generating an in vitro human motor endplate showing dose-dependent sensitivity to BoNTs intoxication. The implementation of optogenetics combined with live calcium imaging allows to monitor the impact of BoNTs intoxication on synaptic transmission in human motor endplate model. Conclusions Altogether, our findings demonstrate the promise of optogenetically hiPSC-derived controlled muscle-nerve system for pharmaceutical BoNTs testing and development. Supplementary Information The online version contains supplementary material available at 10.1186/s13287-021-02665-3.
Collapse
Affiliation(s)
- Juliette Duchesne de Lamotte
- IPSEN Innovation, 5 avenue du Canada, 91940, Les Ulis, France.,Université Evry-Paris Saclay/INSERM UMR861, Institut Des Cellules Souches Pour Le Traitement Et L'étude Des Maladies Monogéniques (I-Stem), 2 rue Henri Auguste Desbruères, 91100, Corbeil-Essonne, France
| | - Jérôme Polentes
- Université Evry-Paris Saclay/INSERM UMR861, Institut Des Cellules Souches Pour Le Traitement Et L'étude Des Maladies Monogéniques (I-Stem), 2 rue Henri Auguste Desbruères, 91100, Corbeil-Essonne, France
| | - Florine Roussange
- Université Evry-Paris Saclay/INSERM UMR861, Institut Des Cellules Souches Pour Le Traitement Et L'étude Des Maladies Monogéniques (I-Stem), 2 rue Henri Auguste Desbruères, 91100, Corbeil-Essonne, France
| | - Léa Lesueur
- Université Evry-Paris Saclay/INSERM UMR861, Institut Des Cellules Souches Pour Le Traitement Et L'étude Des Maladies Monogéniques (I-Stem), 2 rue Henri Auguste Desbruères, 91100, Corbeil-Essonne, France
| | - Pauline Feurgard
- Université Evry-Paris Saclay/INSERM UMR861, Institut Des Cellules Souches Pour Le Traitement Et L'étude Des Maladies Monogéniques (I-Stem), 2 rue Henri Auguste Desbruères, 91100, Corbeil-Essonne, France
| | - Anselme Perrier
- Université Evry-Paris Saclay/INSERM UMR861, Institut Des Cellules Souches Pour Le Traitement Et L'étude Des Maladies Monogéniques (I-Stem), 2 rue Henri Auguste Desbruères, 91100, Corbeil-Essonne, France.,Laboratoire Des Maladies Neurodégénératives: Mécanismes, thérapies, imagerie, Université Paris Saclay/CEA/CNRS UMR9199, MIRCen, Bâtiment 61, CEA-Fontenay-Aux-Roses, 18 route du Panorama, 92265, Fontenay-aux-Roses, France
| | | | - Cécile Martinat
- Université Evry-Paris Saclay/INSERM UMR861, Institut Des Cellules Souches Pour Le Traitement Et L'étude Des Maladies Monogéniques (I-Stem), 2 rue Henri Auguste Desbruères, 91100, Corbeil-Essonne, France.
| |
Collapse
|
30
|
Jalal S, Dastidar S, Tedesco FS. Advanced models of human skeletal muscle differentiation, development and disease: Three-dimensional cultures, organoids and beyond. Curr Opin Cell Biol 2021; 73:92-104. [PMID: 34384976 PMCID: PMC8692266 DOI: 10.1016/j.ceb.2021.06.004] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Accepted: 06/23/2021] [Indexed: 02/08/2023]
Abstract
Advanced in vitro models of human skeletal muscle tissue are increasingly needed to model complex developmental dynamics and disease mechanisms not recapitulated in animal models or in conventional monolayer cell cultures. There has been impressive progress towards creating such models by using tissue engineering approaches to recapitulate a range of physical and biochemical components of native human skeletal muscle tissue. In this review, we discuss recent studies focussed on developing complex in vitro models of human skeletal muscle beyond monolayer cell cultures, involving skeletal myogenic differentiation from human primary myoblasts or pluripotent stem cells, often in the presence of structural scaffolding support. We conclude with our outlook on the future of advanced skeletal muscle three-dimensional cultures (e.g. organoids and biofabrication) to produce physiologically and clinically relevant platforms for disease modelling and therapy development in musculoskeletal and neuromuscular disorders.
Collapse
Affiliation(s)
- Salma Jalal
- Department of Cell and Developmental Biology, University College London, WC1E 6DE London, United Kingdom
| | - Sumitava Dastidar
- Department of Cell and Developmental Biology, University College London, WC1E 6DE London, United Kingdom
| | - Francesco Saverio Tedesco
- Department of Cell and Developmental Biology, University College London, WC1E 6DE London, United Kingdom; The Francis Crick Institute, 1 Midland Road, London NW1 1AT, United Kingdom; Dubowitz Neuromuscular Centre, Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, United Kingdom; Department of Paediatric Neurology, Great Ormond Street Hospital for Children, WC1N 3JH London, United Kingdom.
| |
Collapse
|
31
|
Hörner SJ, Couturier N, Bruch R, Koch P, Hafner M, Rudolf R. hiPSC-Derived Schwann Cells Influence Myogenic Differentiation in Neuromuscular Cocultures. Cells 2021; 10:cells10123292. [PMID: 34943800 PMCID: PMC8699767 DOI: 10.3390/cells10123292] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 11/20/2021] [Accepted: 11/21/2021] [Indexed: 12/13/2022] Open
Abstract
Motoneurons, skeletal muscle fibers, and Schwann cells form synapses, termed neuromuscular junctions (NMJs). These control voluntary body movement and are affected in numerous neuromuscular diseases. Therefore, a variety of NMJ in vitro models have been explored to enable mechanistic and pharmacological studies. So far, selective integration of Schwann cells in these models has been hampered, due to technical limitations. Here we present robust protocols for derivation of Schwann cells from human induced pluripotent stem cells (hiPSC) and their coculture with hiPSC-derived motoneurons and C2C12 muscle cells. Upon differentiation with tuned BMP signaling, Schwann cells expressed marker proteins, S100b, Gap43, vimentin, and myelin protein zero. Furthermore, they displayed typical spindle-shaped morphologies with long processes, which often aligned with motoneuron axons. Inclusion of Schwann cells in coculture experiments with hiPSC-derived motoneurons and C2C12 myoblasts enhanced myotube growth and affected size and number of acetylcholine receptor plaques on myotubes. Altogether, these data argue for the availability of a consistent differentiation protocol for Schwann cells and their amenability for functional integration into neuromuscular in vitro models, fostering future studies of neuromuscular mechanisms and disease.
Collapse
Affiliation(s)
- Sarah Janice Hörner
- Institute of Molecular and Cell Biology, Mannheim University of Applied Sciences, 68163 Mannheim, Germany; (S.J.H.); (N.C.); (R.B.); (M.H.)
- Interdisciplinary Center for Neurosciences, Heidelberg University, 69120 Heidelberg, Germany
| | - Nathalie Couturier
- Institute of Molecular and Cell Biology, Mannheim University of Applied Sciences, 68163 Mannheim, Germany; (S.J.H.); (N.C.); (R.B.); (M.H.)
| | - Roman Bruch
- Institute of Molecular and Cell Biology, Mannheim University of Applied Sciences, 68163 Mannheim, Germany; (S.J.H.); (N.C.); (R.B.); (M.H.)
| | - Philipp Koch
- Central Institute of Mental Health, Medical Faculty Mannheim of Heidelberg University, 68159 Mannheim, Germany;
- Hector Institute for Translational Brain Research (HITBR gGmbH), 68159 Mannheim, Germany
- German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Mathias Hafner
- Institute of Molecular and Cell Biology, Mannheim University of Applied Sciences, 68163 Mannheim, Germany; (S.J.H.); (N.C.); (R.B.); (M.H.)
- Institute of Medical Technology, Mannheim University of Applied Sciences and Heidelberg University, 68163 Mannheim, Germany
| | - Rüdiger Rudolf
- Institute of Molecular and Cell Biology, Mannheim University of Applied Sciences, 68163 Mannheim, Germany; (S.J.H.); (N.C.); (R.B.); (M.H.)
- Interdisciplinary Center for Neurosciences, Heidelberg University, 69120 Heidelberg, Germany
- Institute of Medical Technology, Mannheim University of Applied Sciences and Heidelberg University, 68163 Mannheim, Germany
- Correspondence:
| |
Collapse
|
32
|
Smith VM, Nguyen H, Rumsey JW, Long CJ, Shuler ML, Hickman JJ. A Functional Human-on-a-Chip Autoimmune Disease Model of Myasthenia Gravis for Development of Therapeutics. Front Cell Dev Biol 2021; 9:745897. [PMID: 34881241 PMCID: PMC8645836 DOI: 10.3389/fcell.2021.745897] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Accepted: 10/14/2021] [Indexed: 11/13/2022] Open
Abstract
Myasthenia gravis (MG) is a chronic and progressive neuromuscular disease where autoantibodies target essential proteins such as the nicotinic acetylcholine receptor (nAChR) at the neuromuscular junction (NMJ) causing muscle fatigue and weakness. Autoantibodies directed against nAChRs are proposed to work by three main pathological mechanisms of receptor disruption: blocking, receptor internalization, and downregulation. Current in vivo models using experimental autoimmune animal models fail to recapitulate the disease pathology and are limited in clinical translatability due to disproportionate disease severity and high animal death rates. The development of a highly sensitive antibody assay that mimics human disease pathology is desirable for clinical advancement and therapeutic development. To address this lack of relevant models, an NMJ platform derived from human iPSC differentiated motoneurons and primary skeletal muscle was used to investigate the ability of an anti-nAChR antibody to induce clinically relevant MG pathology in the serum-free, spatially organized, functionally mature NMJ platform. Treatment of the NMJ model with the anti-nAChR antibody revealed decreasing NMJ stability as measured by the number of NMJs before and after the synchrony stimulation protocol. This decrease in NMJ stability was dose-dependent over a concentration range of 0.01-20 μg/mL. Immunocytochemical (ICC) analysis was used to distinguish between pathological mechanisms of antibody-mediated receptor disruption including blocking, receptor internalization and downregulation. Antibody treatment also activated the complement cascade as indicated by complement protein 3 deposition near the nAChRs. Additionally, complement cascade activation significantly altered other readouts of NMJ function including the NMJ fidelity parameter as measured by the number of muscle contractions missed in response to increasing motoneuron stimulation frequencies. This synchrony readout mimics the clinical phenotype of neurological blocking that results in failure of muscle contractions despite motoneuron stimulations. Taken together, these data indicate the establishment of a relevant disease model of MG that mimics reduction of functional nAChRs at the NMJ, decreased NMJ stability, complement activation and blocking of neuromuscular transmission. This system is the first functional human in vitro model of MG to be used to simulate three potential disease mechanisms as well as to establish a preclinical platform for evaluation of disease modifying treatments (etiology).
Collapse
Affiliation(s)
- Virginia M. Smith
- Hybrid Systems Lab, NanoScience Technology Center, University of Central Florida, Orlando, FL, United States
- Hesperos, Inc., Orlando, FL, United States
| | - Huan Nguyen
- Hybrid Systems Lab, NanoScience Technology Center, University of Central Florida, Orlando, FL, United States
| | | | | | | | - James J. Hickman
- Hybrid Systems Lab, NanoScience Technology Center, University of Central Florida, Orlando, FL, United States
- Hesperos, Inc., Orlando, FL, United States
| |
Collapse
|
33
|
Fralish Z, Lotz EM, Chavez T, Khodabukus A, Bursac N. Neuromuscular Development and Disease: Learning From in vitro and in vivo Models. Front Cell Dev Biol 2021; 9:764732. [PMID: 34778273 PMCID: PMC8579029 DOI: 10.3389/fcell.2021.764732] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Accepted: 10/06/2021] [Indexed: 01/02/2023] Open
Abstract
The neuromuscular junction (NMJ) is a specialized cholinergic synaptic interface between a motor neuron and a skeletal muscle fiber that translates presynaptic electrical impulses into motor function. NMJ formation and maintenance require tightly regulated signaling and cellular communication among motor neurons, myogenic cells, and Schwann cells. Neuromuscular diseases (NMDs) can result in loss of NMJ function and motor input leading to paralysis or even death. Although small animal models have been instrumental in advancing our understanding of the NMJ structure and function, the complexities of studying this multi-tissue system in vivo and poor clinical outcomes of candidate therapies developed in small animal models has driven the need for in vitro models of functional human NMJ to complement animal studies. In this review, we discuss prevailing models of NMDs and highlight the current progress and ongoing challenges in developing human iPSC-derived (hiPSC) 3D cell culture models of functional NMJs. We first review in vivo development of motor neurons, skeletal muscle, Schwann cells, and the NMJ alongside current methods for directing the differentiation of relevant cell types from hiPSCs. We further compare the efficacy of modeling NMDs in animals and human cell culture systems in the context of five NMDs: amyotrophic lateral sclerosis, myasthenia gravis, Duchenne muscular dystrophy, myotonic dystrophy, and Pompe disease. Finally, we discuss further work necessary for hiPSC-derived NMJ models to function as effective personalized NMD platforms.
Collapse
Affiliation(s)
- Zachary Fralish
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC, United States
| | - Ethan M Lotz
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC, United States
| | - Taylor Chavez
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC, United States
| | - Alastair Khodabukus
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC, United States
| | - Nenad Bursac
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC, United States
| |
Collapse
|
34
|
Duc P, Vignes M, Hugon G, Sebban A, Carnac G, Malyshev E, Charlot B, Rage F. Human neuromuscular junction on micro-structured microfluidic devices implemented with a custom micro electrode array (MEA). LAB ON A CHIP 2021; 21:4223-4236. [PMID: 34559171 DOI: 10.1039/d1lc00497b] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
In the neuromuscular system, signal transmission from motor neurons (MNs) to innervated muscle fibers is crucial for their synaptic function, viability, and maintenance. In order to better understand human neuromuscular junction (hNMJ) functionality, it is important to develop on-a-chip devices with human cells. To investigate this cell network, microfluidic platforms are useful to grow different cell types in isolated compartments. Such devices have already been developed to study in vitro neuronal circuitry. Here, we combined microfluidics with two techniques: soft lithography and custom microelectrodes array (MEA). Our goal was to create hNMJs on a specific pattern of electrodes to stimulate pre-synaptic axons and record post-synaptic muscle activity. Micromachining was used to create structurations to guide muscle growth above electrodes, without impairing axon propagation, therefore optimizing the effectiveness of activity recording. Electrodes were also arranged to be aligned with the microfluidic chambers in order to specifically stimulate axons that were growing between the two compartments. Isolation of the two cell types allows for the selective treatment of neurons or muscle fibers to assess NMJ functionality hallmarks. Altogether, this microfluidic/microstructured/MEA platform allowed mature and functional in vitro hNMJ modelling. We demonstrate that electrical activation of MNs can trigger recordable extracellular muscle action potentials. This study provides evidence for a physiologically relevant model to mimic a hNMJ that will in the future be a powerful tool, more sensitive than calcium imaging, to better understand and characterize NMJs and their disruption in neurodegenerative diseases.
Collapse
Affiliation(s)
- Pauline Duc
- IGMM, University of Montpellier, CNRS, Montpellier, France.
| | - Michel Vignes
- IBMM UMR5247, University of Montpellier, CNRS, Montpellier, France
| | - Gérald Hugon
- PhyMedExp, INSERM U1046, CNRS UMR9214, Université de Montpellier, Montpellier, France
| | - Audrey Sebban
- IES, CNRS University of Montpellier, Montpellier 34095, France
| | - Gilles Carnac
- PhyMedExp, INSERM U1046, CNRS UMR9214, Université de Montpellier, Montpellier, France
| | - Eugene Malyshev
- IES, CNRS University of Montpellier, Montpellier 34095, France
| | - Benoît Charlot
- IES, CNRS University of Montpellier, Montpellier 34095, France
| | - Florence Rage
- IGMM, University of Montpellier, CNRS, Montpellier, France.
| |
Collapse
|
35
|
Malik M, Yang Y, Fathi P, Mahler GJ, Esch MB. Critical Considerations for the Design of Multi-Organ Microphysiological Systems (MPS). Front Cell Dev Biol 2021; 9:721338. [PMID: 34568333 PMCID: PMC8459628 DOI: 10.3389/fcell.2021.721338] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2021] [Accepted: 08/05/2021] [Indexed: 12/19/2022] Open
Abstract
Identification and approval of new drugs for use in patients requires extensive preclinical studies and clinical trials. Preclinical studies rely on in vitro experiments and animal models of human diseases. The transferability of drug toxicity and efficacy estimates to humans from animal models is being called into question. Subsequent clinical studies often reveal lower than expected efficacy and higher drug toxicity in humans than that seen in animal models. Microphysiological systems (MPS), sometimes called organ or human-on-chip models, present a potential alternative to animal-based models used for drug toxicity screening. This review discusses multi-organ MPS that can be used to model diseases and test the efficacy and safety of drug candidates. The translation of an in vivo environment to an in vitro system requires physiologically relevant organ scaling, vascular dimensions, and appropriate flow rates. Even small changes in those parameters can alter the outcome of experiments conducted with MPS. With many MPS devices being developed, we have outlined some established standards for designing MPS devices and described techniques to validate the devices. A physiologically realistic mimic of the human body can help determine the dose response and toxicity effects of a new drug candidate with higher predictive power.
Collapse
Affiliation(s)
- Mridu Malik
- Department of Bioengineering, University of Maryland, College Park, College Park, MD, United States
- Biophysical and Biomedical Measurement Group, Physical Measurement Laboratory, Microsystems and Nanotechnology Division, National Institute of Standards and Technology, Gaithersburg, MD, United States
| | - Yang Yang
- Biophysical and Biomedical Measurement Group, Physical Measurement Laboratory, Microsystems and Nanotechnology Division, National Institute of Standards and Technology, Gaithersburg, MD, United States
- Department of Chemical Engineering, University of Maryland, College Park, College Park, MD, United States
| | - Parinaz Fathi
- Department of Bioengineering, Materials Science and Engineering, and Beckman Institute, University of Illinois at Urbana-Champaign, Champaign, IL, United States
| | - Gretchen J. Mahler
- Department of Biomedical Engineering, Binghamton University, Binghamton, NY, United States
| | - Mandy B. Esch
- Biophysical and Biomedical Measurement Group, Physical Measurement Laboratory, Microsystems and Nanotechnology Division, National Institute of Standards and Technology, Gaithersburg, MD, United States
| |
Collapse
|
36
|
Santoso JW, Li X, Gupta D, Suh GC, Hendricks E, Lin S, Perry S, Ichida JK, Dickman D, McCain ML. Engineering skeletal muscle tissues with advanced maturity improves synapse formation with human induced pluripotent stem cell-derived motor neurons. APL Bioeng 2021; 5:036101. [PMID: 34286174 PMCID: PMC8282350 DOI: 10.1063/5.0054984] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Accepted: 06/21/2021] [Indexed: 12/12/2022] Open
Abstract
To develop effective cures for neuromuscular diseases, human-relevant in vitro models of neuromuscular tissues are critically needed to probe disease mechanisms on a cellular and molecular level. However, previous attempts to co-culture motor neurons and skeletal muscle have resulted in relatively immature neuromuscular junctions (NMJs). In this study, NMJs formed by human induced pluripotent stem cell (hiPSC)-derived motor neurons were improved by optimizing the maturity of the co-cultured muscle tissue. First, muscle tissues engineered from the C2C12 mouse myoblast cell line, cryopreserved primary human myoblasts, and freshly isolated primary chick myoblasts on micromolded gelatin hydrogels were compared. After three weeks, only chick muscle tissues remained stably adhered to hydrogels and exhibited progressive increases in myogenic index and stress generation, approaching values generated by native muscle tissue. After three weeks of co-culture with hiPSC-derived motor neurons, engineered chick muscle tissues formed NMJs with increasing co-localization of pre- and postsynaptic markers as well as increased frequency and magnitude of synaptic activity, surpassing structural and functional maturity of previous in vitro models. Engineered chick muscle tissues also demonstrated increased expression of genes related to sarcomere maturation and innervation over time, revealing new insights into the molecular pathways that likely contribute to enhanced NMJ formation. These approaches for engineering advanced neuromuscular tissues with relatively mature NMJs and interrogating their structure and function have many applications in neuromuscular disease modeling and drug development.
Collapse
Affiliation(s)
- Jeffrey W. Santoso
- Laboratory for Living Systems Engineering, Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles, California 90089, USA
| | - Xiling Li
- Department of Biological Sciences, Dornsife College of Arts and Letters, University of Southern California, Los Angeles, California 90089, USA
| | - Divya Gupta
- Laboratory for Living Systems Engineering, Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles, California 90089, USA
| | - Gio C. Suh
- Laboratory for Living Systems Engineering, Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles, California 90089, USA
| | - Eric Hendricks
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine of USC, University of Southern California, Los Angeles, California 90033, USA
| | - Shaoyu Lin
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine of USC, University of Southern California, Los Angeles, California 90033, USA
| | - Sarah Perry
- Department of Biological Sciences, Dornsife College of Arts and Letters, University of Southern California, Los Angeles, California 90089, USA
| | - Justin K. Ichida
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine of USC, University of Southern California, Los Angeles, California 90033, USA
| | - Dion Dickman
- Department of Biological Sciences, Dornsife College of Arts and Letters, University of Southern California, Los Angeles, California 90089, USA
| | - Megan L. McCain
- Author to whom correspondence should be addressed:. Tel: +1 2138210791. URL:https://livingsystemsengineering.usc.edu
| |
Collapse
|
37
|
Pereira JD, DuBreuil DM, Devlin AC, Held A, Sapir Y, Berezovski E, Hawrot J, Dorfman K, Chander V, Wainger BJ. Human sensorimotor organoids derived from healthy and amyotrophic lateral sclerosis stem cells form neuromuscular junctions. Nat Commun 2021; 12:4744. [PMID: 34362895 PMCID: PMC8346474 DOI: 10.1038/s41467-021-24776-4] [Citation(s) in RCA: 77] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Accepted: 07/06/2021] [Indexed: 02/07/2023] Open
Abstract
Human induced pluripotent stem cells (iPSC) hold promise for modeling diseases in individual human genetic backgrounds and thus for developing precision medicine. Here, we generate sensorimotor organoids containing physiologically functional neuromuscular junctions (NMJs) and apply the model to different subgroups of amyotrophic lateral sclerosis (ALS). Using a range of molecular, genomic, and physiological techniques, we identify and characterize motor neurons and skeletal muscle, along with sensory neurons, astrocytes, microglia, and vasculature. Organoid cultures derived from multiple human iPSC lines generated from individuals with ALS and isogenic lines edited to harbor familial ALS mutations show impairment at the level of the NMJ, as detected by both contraction and immunocytochemical measurements. The physiological resolution of the human NMJ synapse, combined with the generation of major cellular cohorts exerting autonomous and non-cell autonomous effects in motor and sensory diseases, may prove valuable to understand the pathophysiological mechanisms of ALS.
Collapse
Affiliation(s)
- João D Pereira
- Department of Neurology, Sean M. Healey & AMG Center for ALS, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Daniel M DuBreuil
- Department of Neurology, Sean M. Healey & AMG Center for ALS, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Anna-Claire Devlin
- Department of Neurology, Sean M. Healey & AMG Center for ALS, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Aaron Held
- Department of Neurology, Sean M. Healey & AMG Center for ALS, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Yechiam Sapir
- Department of Neurology, Sean M. Healey & AMG Center for ALS, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Eugene Berezovski
- Department of Neurology, Sean M. Healey & AMG Center for ALS, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - James Hawrot
- Department of Neurology, Sean M. Healey & AMG Center for ALS, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Katherine Dorfman
- Department of Neurology, Sean M. Healey & AMG Center for ALS, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Vignesh Chander
- Department of Neurology, Sean M. Healey & AMG Center for ALS, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Brian J Wainger
- Department of Neurology, Sean M. Healey & AMG Center for ALS, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
- Department of Anesthesiology, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, MA, USA.
- Harvard Stem Cell Institute, Cambridge, MA, USA.
- Broad Institute of Harvard University and MIT, Cambridge, MA, USA.
| |
Collapse
|
38
|
Vila OF, Chavez M, Ma SP, Yeager K, Zholudeva LV, Colón-Mercado JM, Qu Y, Nash TR, Lai C, Feliciano CM, Carter M, Kamm RD, Judge LM, Conklin BR, Ward ME, McDevitt TC, Vunjak-Novakovic G. Bioengineered optogenetic model of human neuromuscular junction. Biomaterials 2021; 276:121033. [PMID: 34403849 DOI: 10.1016/j.biomaterials.2021.121033] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 07/09/2021] [Accepted: 07/15/2021] [Indexed: 12/28/2022]
Abstract
Functional human tissues engineered from patient-specific induced pluripotent stem cells (hiPSCs) hold great promise for investigating the progression, mechanisms, and treatment of musculoskeletal diseases in a controlled and systematic manner. For example, bioengineered models of innervated human skeletal muscle could be used to identify novel therapeutic targets and treatments for patients with complex central and peripheral nervous system disorders. There is a need to develop standardized and objective quantitative methods for engineering and using these complex tissues, in order increase their robustness, reproducibility, and predictiveness across users. Here we describe a standardized method for engineering an isogenic, patient specific human neuromuscular junction (NMJ) that allows for automated quantification of NMJ function to diagnose disease using a small sample of blood serum and evaluate new therapeutic modalities. By combining tissue engineering, optogenetics, microfabrication, optoelectronics and video processing, we created a novel platform for the precise investigation of the development and degeneration of human NMJ. We demonstrate the utility of this platform for the detection and diagnosis of myasthenia gravis, an antibody-mediated autoimmune disease that disrupts the NMJ function.
Collapse
Affiliation(s)
- Olaia F Vila
- Columbia University, 622 W 168th St, New York, NY, 10032, USA; Gladstone Institutes, 1650 Owens St, San Francisco, CA, 94158, USA.
| | - Miguel Chavez
- Columbia University, 622 W 168th St, New York, NY, 10032, USA
| | - Stephen P Ma
- Columbia University, 622 W 168th St, New York, NY, 10032, USA
| | - Keith Yeager
- Columbia University, 622 W 168th St, New York, NY, 10032, USA
| | | | | | - Yihuai Qu
- Columbia University, 622 W 168th St, New York, NY, 10032, USA
| | - Trevor R Nash
- Columbia University, 622 W 168th St, New York, NY, 10032, USA
| | - Carmen Lai
- Gladstone Institutes, 1650 Owens St, San Francisco, CA, 94158, USA
| | - Carissa M Feliciano
- Gladstone Institutes, 1650 Owens St, San Francisco, CA, 94158, USA; Department of Pediatrics, UCSF, 550 16th St, Floor 5, San Francisco, CA, 94143, USA
| | - Matthew Carter
- Gladstone Institutes, 1650 Owens St, San Francisco, CA, 94158, USA
| | - Roger D Kamm
- Department of Mechanical Engineering and Biological Engineering, Massachusetts Institute of Technology, Cambridge MA, 02139, USA
| | - Luke M Judge
- Gladstone Institutes, 1650 Owens St, San Francisco, CA, 94158, USA; Department of Pediatrics, UCSF, 550 16th St, Floor 5, San Francisco, CA, 94143, USA
| | - Bruce R Conklin
- Gladstone Institutes, 1650 Owens St, San Francisco, CA, 94158, USA
| | - Michael E Ward
- National Institute of Neurological Disorders and Stroke, NIH, Bethesda, MD, 20892, USA
| | - Todd C McDevitt
- Gladstone Institutes, 1650 Owens St, San Francisco, CA, 94158, USA
| | | |
Collapse
|
39
|
Luttrell SM, Smith AST, Mack DL. Creating stem cell-derived neuromuscular junctions in vitro. Muscle Nerve 2021; 64:388-403. [PMID: 34328673 PMCID: PMC9292444 DOI: 10.1002/mus.27360] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Revised: 05/28/2021] [Accepted: 06/21/2021] [Indexed: 12/14/2022]
Abstract
Recent development of novel therapies has improved mobility and quality of life for people suffering from inheritable neuromuscular disorders. Despite this progress, the majority of neuromuscular disorders are still incurable, in part due to a lack of predictive models of neuromuscular junction (NMJ) breakdown. Improvement of predictive models of a human NMJ would be transformative in terms of expanding our understanding of the mechanisms that underpin development, maintenance, and disease, and as a testbed with which to evaluate novel therapeutics. Induced pluripotent stem cells (iPSCs) are emerging as a clinically relevant and non‐invasive cell source to create human NMJs to study synaptic development and maturation, as well as disease modeling and drug discovery. This review will highlight the recent advances and remaining challenges to generating an NMJ capable of eliciting contraction of stem cell‐derived skeletal muscle in vitro. We explore the advantages and shortcomings of traditional NMJ culturing platforms, as well as the pioneering technologies and novel, biomimetic culturing systems currently in use to guide development and maturation of the neuromuscular synapse and extracellular microenvironment. Then, we will explore how this NMJ‐in‐a‐dish can be used to study normal assembly and function of the efferent portion of the neuromuscular arc, and how neuromuscular disease‐causing mutations disrupt structure, signaling, and function.
Collapse
Affiliation(s)
- Shawn M Luttrell
- Department of Rehabilitation Medicine, University of Washington, Seattle, Washington, USA.,Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, Washington, USA
| | - Alec S T Smith
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, Washington, USA.,Department of Physiology and Biophysics, University of Washington, Seattle, Washington, USA
| | - David L Mack
- Department of Rehabilitation Medicine, University of Washington, Seattle, Washington, USA.,Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, Washington, USA.,Department of Physiology and Biophysics, University of Washington, Seattle, Washington, USA
| |
Collapse
|
40
|
Duchesne de Lamotte J, Perrier A, Martinat C, Nicoleau C. Emerging Opportunities in Human Pluripotent Stem-Cells Based Assays to Explore the Diversity of Botulinum Neurotoxins as Future Therapeutics. Int J Mol Sci 2021; 22:7524. [PMID: 34299143 PMCID: PMC8308099 DOI: 10.3390/ijms22147524] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 07/02/2021] [Accepted: 07/07/2021] [Indexed: 02/07/2023] Open
Abstract
Botulinum neurotoxins (BoNTs) are produced by Clostridium botulinum and are responsible for botulism, a fatal disorder of the nervous system mostly induced by food poisoning. Despite being one of the most potent families of poisonous substances, BoNTs are used for both aesthetic and therapeutic indications from cosmetic reduction of wrinkles to treatment of movement disorders. The increasing understanding of the biology of BoNTs and the availability of distinct toxin serotypes and subtypes offer the prospect of expanding the range of indications for these toxins. Engineering of BoNTs is considered to provide a new avenue for improving safety and clinical benefit from these neurotoxins. Robust, high-throughput, and cost-effective assays for BoNTs activity, yet highly relevant to the human physiology, have become indispensable for a successful translation of engineered BoNTs to the clinic. This review presents an emerging family of cell-based assays that take advantage of newly developed human pluripotent stem cells and neuronal function analyses technologies.
Collapse
Affiliation(s)
- Juliette Duchesne de Lamotte
- IPSEN Innovation, 91940 Les Ulis, France;
- I-STEM, INSERM UMR861, Université Evry-Paris Saclay, 91100 Corbeil-Essonne, France
| | - Anselme Perrier
- I-STEM, INSERM UMR861, Université Evry-Paris Saclay, 91100 Corbeil-Essonne, France
- Laboratoire des Maladies Neurodégénératives: Mécanismes, Thérapies, Imagerie, CEA/CNRS UMR9199, Université Paris Saclay, 92265 Fontenay-aux-Roses, France
| | - Cécile Martinat
- I-STEM, INSERM UMR861, Université Evry-Paris Saclay, 91100 Corbeil-Essonne, France
| | | |
Collapse
|
41
|
Whelan IT, Moeendarbary E, Hoey DA, Kelly DJ. Biofabrication of vasculature in microphysiological models of bone. Biofabrication 2021; 13. [PMID: 34034238 DOI: 10.1088/1758-5090/ac04f7] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Accepted: 05/25/2021] [Indexed: 11/12/2022]
Abstract
Bone contains a dense network of blood vessels that are essential to its homoeostasis, endocrine function, mineral metabolism and regenerative functions. In addition, bone vasculature is implicated in a number of prominent skeletal diseases, and bone has high affinity for metastatic cancers. Despite vasculature being an integral part of bone physiology and pathophysiology, it is often ignored or oversimplified inin vitrobone models. However, 3D physiologically relevant vasculature can now be engineeredin vitro, with microphysiological systems (MPS) increasingly being used as platforms for engineering this physiologically relevant vasculature. In recent years, vascularised models of bone in MPSs systems have been reported in the literature, representing the beginning of a possible technological step change in how bone is modelledin vitro. Vascularised bone MPSs is a subfield of bone research in its nascency, however given the impact of MPSs has had inin vitroorgan modelling, and the crucial role of vasculature to bone physiology, these systems stand to have a substantial impact on bone research. However, engineering vasculature within the specific design restraints of the bone niche is significantly challenging given the different requirements for engineering bone and vasculature. With this in mind, this paper aims to serve as technical guidance for the biofabrication of vascularised bone tissue within MPS devices. We first discuss the key engineering and biological considerations for engineering more physiologically relevant vasculaturein vitrowithin the specific design constraints of the bone niche. We next explore emerging applications of vascularised bone MPSs, and conclude with a discussion on the current status of vascularised bone MPS biofabrication and suggest directions for development of next generation vascularised bone MPSs.
Collapse
|
42
|
Guttenplan APM, Tahmasebi Birgani Z, Giselbrecht S, Truckenmüller RK, Habibović P. Chips for Biomaterials and Biomaterials for Chips: Recent Advances at the Interface between Microfabrication and Biomaterials Research. Adv Healthc Mater 2021; 10:e2100371. [PMID: 34033239 PMCID: PMC11468311 DOI: 10.1002/adhm.202100371] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 05/08/2021] [Indexed: 12/24/2022]
Abstract
In recent years, the use of microfabrication techniques has allowed biomaterials studies which were originally carried out at larger length scales to be miniaturized as so-called "on-chip" experiments. These miniaturized experiments have a range of advantages which have led to an increase in their popularity. A range of biomaterial shapes and compositions are synthesized or manufactured on chip. Moreover, chips are developed to investigate specific aspects of interactions between biomaterials and biological systems. Finally, biomaterials are used in microfabricated devices to replicate the physiological microenvironment in studies using so-called "organ-on-chip," "tissue-on-chip" or "disease-on-chip" models, which can reduce the use of animal models with their inherent high cost and ethical issues, and due to the possible use of human cells can increase the translation of research from lab to clinic. This review gives an overview of recent developments at the interface between microfabrication and biomaterials science, and indicates potential future directions that the field may take. In particular, a trend toward increased scale and automation is apparent, allowing both industrial production of micron-scale biomaterials and high-throughput screening of the interaction of diverse materials libraries with cells and bioengineered tissues and organs.
Collapse
Affiliation(s)
- Alexander P. M. Guttenplan
- Department of Instructive Biomaterials EngineeringMERLN Institute for Technology‐Inspired Regenerative MedicineMaastricht UniversityUniversiteitssingel 40Maastricht6229ERThe Netherlands
| | - Zeinab Tahmasebi Birgani
- Department of Instructive Biomaterials EngineeringMERLN Institute for Technology‐Inspired Regenerative MedicineMaastricht UniversityUniversiteitssingel 40Maastricht6229ERThe Netherlands
| | - Stefan Giselbrecht
- Department of Instructive Biomaterials EngineeringMERLN Institute for Technology‐Inspired Regenerative MedicineMaastricht UniversityUniversiteitssingel 40Maastricht6229ERThe Netherlands
| | - Roman K. Truckenmüller
- Department of Instructive Biomaterials EngineeringMERLN Institute for Technology‐Inspired Regenerative MedicineMaastricht UniversityUniversiteitssingel 40Maastricht6229ERThe Netherlands
| | - Pamela Habibović
- Department of Instructive Biomaterials EngineeringMERLN Institute for Technology‐Inspired Regenerative MedicineMaastricht UniversityUniversiteitssingel 40Maastricht6229ERThe Netherlands
| |
Collapse
|
43
|
Rimington RP, Fleming JW, Capel AJ, Wheeler PC, Lewis MP. Bioengineered model of the human motor unit with physiologically functional neuromuscular junctions. Sci Rep 2021; 11:11695. [PMID: 34083648 PMCID: PMC8175425 DOI: 10.1038/s41598-021-91203-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Accepted: 05/24/2021] [Indexed: 02/04/2023] Open
Abstract
Investigations of the human neuromuscular junction (NMJ) have predominately utilised experimental animals, model organisms, or monolayer cell cultures that fail to represent the physiological complexity of the synapse. Consequently, there remains a paucity of data regarding the development of the human NMJ and a lack of systems that enable investigation of the motor unit. This work addresses this need, providing the methodologies to bioengineer 3D models of the human motor unit. Spheroid culture of iPSC derived motor neuron progenitors augmented the transcription of OLIG2, ISLET1 and SMI32 motor neuron mRNAs ~ 400, ~ 150 and ~ 200-fold respectively compared to monolayer equivalents. Axon projections of adhered spheroids exceeded 1000 μm in monolayer, with transcription of SMI32 and VACHT mRNAs further enhanced by addition to 3D extracellular matrices in a type I collagen concentration dependent manner. Bioengineered skeletal muscles produced functional tetanic and twitch profiles, demonstrated increased acetylcholine receptor (AChR) clustering and transcription of MUSK and LRP4 mRNAs, indicating enhanced organisation of the post-synaptic membrane. The number of motor neuron spheroids, or motor pool, required to functionally innervate 3D muscle tissues was then determined, generating functional human NMJs that evidence pre- and post-synaptic membrane and motor nerve axon co-localisation. Spontaneous firing was significantly elevated in 3D motor units, confirmed to be driven by the motor nerve via antagonistic inhibition of the AChR. Functional analysis outlined decreased time to peak twitch and half relaxation times, indicating enhanced physiology of excitation contraction coupling in innervated motor units. Our findings provide the methods to maximise the maturity of both iPSC motor neurons and primary human skeletal muscle, utilising cell type specific extracellular matrices and developmental timelines to bioengineer the human motor unit for the study of neuromuscular junction physiology.
Collapse
Affiliation(s)
- Rowan P. Rimington
- grid.6571.50000 0004 1936 8542National Centre for Sport and Exercise Medicine, School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, LE11 3TU Leicestershire UK
| | - Jacob W. Fleming
- grid.6571.50000 0004 1936 8542National Centre for Sport and Exercise Medicine, School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, LE11 3TU Leicestershire UK
| | - Andrew J. Capel
- grid.6571.50000 0004 1936 8542National Centre for Sport and Exercise Medicine, School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, LE11 3TU Leicestershire UK
| | - Patrick C. Wheeler
- grid.6571.50000 0004 1936 8542National Centre for Sport and Exercise Medicine, School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, LE11 3TU Leicestershire UK
| | - Mark P. Lewis
- grid.6571.50000 0004 1936 8542National Centre for Sport and Exercise Medicine, School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, LE11 3TU Leicestershire UK
| |
Collapse
|
44
|
Yamamoto K, Yamaoka N, Imaizumi Y, Nagashima T, Furutani T, Ito T, Okada Y, Honda H, Shimizu K. Development of a human neuromuscular tissue-on-a-chip model on a 24-well-plate-format compartmentalized microfluidic device. LAB ON A CHIP 2021; 21:1897-1907. [PMID: 34008665 DOI: 10.1039/d1lc00048a] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Engineered three-dimensional models of neuromuscular tissues are promising for use in mimicking their disorder states in vitro. Although several models have been developed, it is still challenging to mimic the physically separated structures of motor neurons (MNs) and skeletal muscle (SkM) fibers in the motor units in vivo. In this study, we aimed to develop microdevices for precisely compartmentalized coculturing of MNs and engineered SkM tissues. The developed microdevices, which fit a well of 24 well plates, had a chamber for MNs and chamber for SkM tissues. The two chambers were connected by microtunnels for axons, permissive to axons but not to cell bodies. Human iPSC (hiPSC)-derived MN spheroids in one chamber elongated their axons into microtunnels, which reached the tissue-engineered human SkM in the SkM chamber, and formed functional neuromuscular junctions with the muscle fibers. The cocultured SkM tissues with MNs on the device contracted spontaneously in response to spontaneous firing of MNs. The addition of a neurotransmitter, glutamate, into the MN chamber induced contraction of the cocultured SkM tissues. Selective addition of tetrodotoxin or vecuronium bromide into either chamber induced SkM tissue relaxation, which could be explained by the inhibitory mechanisms. We also demonstrated the application of chemical or mechanical stimuli to the middle of the axons of cocultured tissues on the device. Thus, compartmentalized neuromuscular tissue models fabricated on the device could be used for phenotypic screening to evaluate the cellular type specific efficacy of drug candidates and would be a useful tool in fundamental research and drug development for neuromuscular disorders.
Collapse
Affiliation(s)
- Kazuki Yamamoto
- Department of Biomolecular Engineering, Graduate School of Engineering, Nagoya University, Nagoya, Aichi, Japan.
| | - Nao Yamaoka
- Department of Biomolecular Engineering, Graduate School of Engineering, Nagoya University, Nagoya, Aichi, Japan.
| | - Yu Imaizumi
- Department of Biomolecular Engineering, Graduate School of Engineering, Nagoya University, Nagoya, Aichi, Japan.
| | - Takunori Nagashima
- Department of Biomolecular Engineering, Graduate School of Engineering, Nagoya University, Nagoya, Aichi, Japan.
| | - Taiki Furutani
- Department of Biomolecular Engineering, Graduate School of Engineering, Nagoya University, Nagoya, Aichi, Japan.
| | - Takuji Ito
- Department of Neurology, Aichi Medical University School of Medicine, Nagakute, Aichi, Japan
| | - Yohei Okada
- Department of Neurology, Aichi Medical University School of Medicine, Nagakute, Aichi, Japan
| | - Hiroyuki Honda
- Department of Biomolecular Engineering, Graduate School of Engineering, Nagoya University, Nagoya, Aichi, Japan.
| | - Kazunori Shimizu
- Department of Biomolecular Engineering, Graduate School of Engineering, Nagoya University, Nagoya, Aichi, Japan.
| |
Collapse
|
45
|
Stoklund Dittlau K, Krasnow EN, Fumagalli L, Vandoorne T, Baatsen P, Kerstens A, Giacomazzi G, Pavie B, Rossaert E, Beckers J, Sampaolesi M, Van Damme P, Van Den Bosch L. Human motor units in microfluidic devices are impaired by FUS mutations and improved by HDAC6 inhibition. Stem Cell Reports 2021; 16:2213-2227. [PMID: 33891869 PMCID: PMC8452598 DOI: 10.1016/j.stemcr.2021.03.029] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 03/23/2021] [Accepted: 03/25/2021] [Indexed: 12/13/2022] Open
Abstract
Neuromuscular junctions (NMJs) ensure communication between motor neurons (MNs) and muscle; however, in MN disorders, such as amyotrophic lateral sclerosis (ALS), NMJs degenerate resulting in muscle atrophy. The aim of this study was to establish a versatile and reproducible in vitro model of a human motor unit to investigate the effects of ALS-causing mutations. Therefore, we generated a co-culture of human induced pluripotent stem cell (iPSC)-derived MNs and human primary mesoangioblast-derived myotubes in microfluidic devices. A chemotactic and volumetric gradient facilitated the growth of MN neurites through microgrooves resulting in the interaction with myotubes and the formation of NMJs. We observed that ALS-causing FUS mutations resulted in reduced neurite outgrowth as well as an impaired neurite regrowth upon axotomy. NMJ numbers were likewise reduced in the FUS-ALS model. Interestingly, the selective HDAC6 inhibitor, Tubastatin A, improved the neurite outgrowth, regrowth, and NMJ morphology, prompting HDAC6 inhibition as a potential therapeutic strategy for ALS. Human motor units with functional NMJs can be generated using microfluidic devices FUS-ALS motor units display impaired neurite regrowth, outgrowth and NMJ numbers HDAC6 inhibition alleviate FUS-ALS motor unit pathology in vitro
Collapse
Affiliation(s)
- Katarina Stoklund Dittlau
- KU Leuven - University of Leuven, Department of Neurosciences, Experimental Neurology, and Leuven Brain Institute, Leuven, Belgium; VIB, Center for Brain & Disease Research, Laboratory of Neurobiology, Leuven, Belgium
| | - Emily N Krasnow
- KU Leuven - University of Leuven, Department of Neurosciences, Experimental Neurology, and Leuven Brain Institute, Leuven, Belgium; VIB, Center for Brain & Disease Research, Laboratory of Neurobiology, Leuven, Belgium
| | - Laura Fumagalli
- KU Leuven - University of Leuven, Department of Neurosciences, Experimental Neurology, and Leuven Brain Institute, Leuven, Belgium; VIB, Center for Brain & Disease Research, Laboratory of Neurobiology, Leuven, Belgium
| | - Tijs Vandoorne
- KU Leuven - University of Leuven, Department of Neurosciences, Experimental Neurology, and Leuven Brain Institute, Leuven, Belgium; VIB, Center for Brain & Disease Research, Laboratory of Neurobiology, Leuven, Belgium
| | - Pieter Baatsen
- VIB, Center for Brain & Disease Research, Research Group Molecular Neurobiology, Leuven, Belgium; KU Leuven - University of Leuven, VIB Bio Imaging Core, Leuven, Belgium
| | - Axelle Kerstens
- VIB, Center for Brain & Disease Research, Research Group Molecular Neurobiology, Leuven, Belgium; KU Leuven - University of Leuven, VIB Bio Imaging Core, Leuven, Belgium
| | - Giorgia Giacomazzi
- KU Leuven - University of Leuven, Department of Development and Regeneration, Stem Cell and Developmental Biology, Leuven, Belgium
| | - Benjamin Pavie
- VIB, Center for Brain & Disease Research, Research Group Molecular Neurobiology, Leuven, Belgium; KU Leuven - University of Leuven, VIB Bio Imaging Core, Leuven, Belgium
| | - Elisabeth Rossaert
- KU Leuven - University of Leuven, Department of Neurosciences, Experimental Neurology, and Leuven Brain Institute, Leuven, Belgium; VIB, Center for Brain & Disease Research, Laboratory of Neurobiology, Leuven, Belgium
| | - Jimmy Beckers
- KU Leuven - University of Leuven, Department of Neurosciences, Experimental Neurology, and Leuven Brain Institute, Leuven, Belgium; VIB, Center for Brain & Disease Research, Laboratory of Neurobiology, Leuven, Belgium
| | - Maurilio Sampaolesi
- KU Leuven - University of Leuven, Department of Development and Regeneration, Stem Cell and Developmental Biology, Leuven, Belgium
| | - Philip Van Damme
- KU Leuven - University of Leuven, Department of Neurosciences, Experimental Neurology, and Leuven Brain Institute, Leuven, Belgium; VIB, Center for Brain & Disease Research, Laboratory of Neurobiology, Leuven, Belgium; University Hospitals Leuven, Department of Neurology, Leuven, Belgium
| | - Ludo Van Den Bosch
- KU Leuven - University of Leuven, Department of Neurosciences, Experimental Neurology, and Leuven Brain Institute, Leuven, Belgium; VIB, Center for Brain & Disease Research, Laboratory of Neurobiology, Leuven, Belgium.
| |
Collapse
|
46
|
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: 1.3] [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
|
47
|
de Jongh R, Spijkers XM, Pasteuning-Vuhman S, Vulto P, Pasterkamp RJ. Neuromuscular junction-on-a-chip: ALS disease modeling and read-out development in microfluidic devices. J Neurochem 2021; 157:393-412. [PMID: 33382092 DOI: 10.1111/jnc.15289] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 12/24/2020] [Accepted: 12/28/2020] [Indexed: 12/21/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal and progressive neurodegenerative disease affecting upper and lower motor neurons with no cure available. Clinical and animal studies reveal that the neuromuscular junction (NMJ), a synaptic connection between motor neurons and skeletal muscle fibers, is highly vulnerable in ALS and suggest that NMJ defects may occur at the early stages of the disease. However, mechanistic insight into how NMJ dysfunction relates to the onset and progression of ALS is incomplete, which hampers therapy development. This is, in part, caused by a lack of robust in vitro models. The ability to combine microfluidic and induced pluripotent stem cell (iPSC) technologies has opened up new avenues for studying molecular and cellular ALS phenotypes in vitro. Microfluidic devices offer several advantages over traditional culture approaches when modeling the NMJ, such as the spatial separation of different cell types and increased control over the cellular microenvironment. Moreover, they are compatible with 3D cell culture, which enhances NMJ functionality and maturity. Here, we review how microfluidic technology is currently being employed to develop more reliable in vitro NMJ models. To validate and phenotype such models, various morphological and functional read-outs have been developed. We describe and discuss the relevance of these read-outs and specifically illustrate how these read-outs have enhanced our understanding of NMJ pathology in ALS. Finally, we share our view on potential future directions and challenges.
Collapse
Affiliation(s)
- Rianne de Jongh
- Department of Translational Neuroscience, University Medical Center Utrecht Brain Center, Utrecht University, Utrecht, The Netherlands
| | - Xandor M Spijkers
- Department of Translational Neuroscience, University Medical Center Utrecht Brain Center, Utrecht University, Utrecht, The Netherlands.,Mimetas B.V., Organ-on-a-chip Company, Leiden, The Netherlands
| | - Svetlana Pasteuning-Vuhman
- Department of Translational Neuroscience, University Medical Center Utrecht Brain Center, Utrecht University, Utrecht, The Netherlands
| | - Paul Vulto
- Mimetas B.V., Organ-on-a-chip Company, Leiden, The Netherlands
| | - R Jeroen Pasterkamp
- Department of Translational Neuroscience, University Medical Center Utrecht Brain Center, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
48
|
Lamotte JDD, Roqueviere S, Gautier H, Raban E, Bouré C, Fonfria E, Krupp J, Nicoleau C. hiPSC-Derived Neurons Provide a Robust and Physiologically Relevant In Vitro Platform to Test Botulinum Neurotoxins. Front Pharmacol 2021; 11:617867. [PMID: 33519485 PMCID: PMC7840483 DOI: 10.3389/fphar.2020.617867] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Accepted: 11/26/2020] [Indexed: 12/16/2022] Open
Abstract
Botulinum neurotoxins (BoNTs) are zinc metalloproteases that block neurotransmitter release at the neuromuscular junction (NMJ). Their high affinity for motor neurons combined with a high potency have made them extremely effective drugs for the treatment of a variety of neurological diseases as well as for aesthetic applications. Current in vitro assays used for testing and developing BoNT therapeutics include primary rodent cells and immortalized cell lines. Both models have limitations concerning accuracy and physiological relevance. In order to improve the translational value of preclinical data there is a clear need to use more accurate models such as human induced Pluripotent Stem Cells (hiPSC)-derived neuronal models. In this study we have assessed the potential of four different human iPSC-derived neuronal models including Motor Neurons for BoNT testing. We have characterized these models in detail and found that all models express all proteins needed for BoNT intoxication and showed that all four hiPSC-derived neuronal models are sensitive to both serotype A and E BoNT with Motor Neurons being the most sensitive. We showed that hiPSC-derived Motor Neurons expressed authentic markers after only 7 days of culture, are functional and able to form active synapses. When cultivated with myotubes, we demonstrated that they can innervate myotubes and induce contraction, generating an in vitro model of NMJ showing dose-responsive sensitivity BoNT intoxication. Together, these data demonstrate the promise of hiPSC-derived neurons, especially Motor Neurons, for pharmaceutical BoNT testing and development.
Collapse
|
49
|
Advances in 3D neuronal microphysiological systems: towards a functional nervous system on a chip. In Vitro Cell Dev Biol Anim 2021; 57:191-206. [PMID: 33438114 PMCID: PMC7802613 DOI: 10.1007/s11626-020-00532-8] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Accepted: 11/16/2020] [Indexed: 12/18/2022]
Abstract
Microphysiological systems (MPS) designed to study the complexities of the peripheral and central nervous systems have made marked improvements over the years and have allowed researchers to assess in two and three dimensions the functional interconnectivity of neuronal tissues. The recent generation of brain organoids has further propelled the field into the nascent recapitulation of structural, functional, and effective connectivities which are found within the native human nervous system. Herein, we will review advances in culture methodologies, focused especially on those of human tissues, which seek to bridge the gap from 2D cultures to hierarchical and defined 3D MPS with the end goal of developing a robust nervous system-on-a-chip platform. These advances have far-reaching implications within basic science, pharmaceutical development, and translational medicine disciplines.
Collapse
|
50
|
Gupta D, Santoso JW, McCain ML. Characterization of Gelatin Hydrogels Cross-Linked with Microbial Transglutaminase as Engineered Skeletal Muscle Substrates. Bioengineering (Basel) 2021; 8:bioengineering8010006. [PMID: 33418892 PMCID: PMC7825108 DOI: 10.3390/bioengineering8010006] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Revised: 12/18/2020] [Accepted: 12/30/2020] [Indexed: 12/12/2022] Open
Abstract
Engineered in vitro models of skeletal muscle are essential for efficiently screening drug safety and efficacy. However, conventional culture substrates poorly replicate physical features of native muscle and do not support long-term culture, which limits tissue maturity. Micromolded gelatin hydrogels cross-linked with microbial transglutaminase (gelatin-MTG hydrogels) have previously been shown to induce C21C2 myotube alignment and improve culture longevity. However, several properties of gelatin-MTG hydrogels have not been systematically characterized, such as changes in elastic modulus during incubation in culture-like conditions and their ability to support sarcomere maturation. In this study, various gelatin-MTG hydrogels were fabricated and incubated in ambient or culture-like conditions. Elastic modulus, mass, and transmittance were measured over a one- or two-week period. Compared to hydrogels in phosphate buffered saline (PBS) or ambient air, hydrogels in Dulbecco’s Modified Eagle Medium (DMEM) and 5% CO2 demonstrated the most stable elastic modulus. A subset of gelatin-MTG hydrogels was micromolded and seeded with C2C12 or primary chick myoblasts, which aligned and fused into multinucleated myotubes with relatively mature sarcomeres. These data are important for fabricating gelatin-MTG hydrogels with predictable and stable mechanical properties and highlight their advantages as culture substrates for engineering relatively mature and stable muscle tissues.
Collapse
Affiliation(s)
- Divya Gupta
- Laboratory for Living Systems Engineering, Department of Biomedical Engineering, Viterbi School of Engineering, University of Southern California, 1042 Downey Way, DRB 140, Los Angeles, CA 90089, USA; (D.G.); (J.W.S.)
| | - Jeffrey W. Santoso
- Laboratory for Living Systems Engineering, Department of Biomedical Engineering, Viterbi School of Engineering, University of Southern California, 1042 Downey Way, DRB 140, Los Angeles, CA 90089, USA; (D.G.); (J.W.S.)
| | - Megan L. McCain
- Laboratory for Living Systems Engineering, Department of Biomedical Engineering, Viterbi School of Engineering, University of Southern California, 1042 Downey Way, DRB 140, Los Angeles, CA 90089, USA; (D.G.); (J.W.S.)
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, 1975 Zonal Ave, Los Angeles, CA 90033, USA
- Correspondence:
| |
Collapse
|