51
|
Aydin O, Passaro AP, Elhebeary M, Pagan-Diaz GJ, Fan A, Nuethong S, Bashir R, Stice SL, Saif MTA. Development of 3D neuromuscular bioactuators. APL Bioeng 2020; 4:016107. [PMID: 32161837 PMCID: PMC7064368 DOI: 10.1063/1.5134477] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Accepted: 02/07/2020] [Indexed: 01/01/2023] Open
Abstract
Neuronal control of skeletal muscle bioactuators represents a critical milestone toward the realization of future biohybrid machines that may generate complex motor patterns and autonomously navigate through their environment. Animals achieve these feats using neural networks that generate robust firing patterns and coordinate muscle activity through neuromuscular units. Here, we designed a versatile 3D neuron-muscle co-culture platform to serve as a test-bed for neuromuscular bioactuators. We used our platform in conjunction with microelectrode array electrophysiology to study the roles of synergistic interactions in the co-development of neural networks and muscle tissues. Our platform design enables co-culture of a neuronal cluster with up to four target muscle actuators, as well as quantification of muscle contraction forces. Using engineered muscle tissue targets, we first demonstrated the formation of functional neuromuscular bioactuators. We then investigated possible roles of long-range interactions in neuronal outgrowth patterns and observed preferential outgrowth toward muscles compared to the acellular matrix or fibroblasts, indicating muscle-specific chemotactic cues acting on motor neurons. Next, we showed that co-cultured muscle strips exhibited significantly higher spontaneous contractility as well as improved sarcomere assembly compared to muscles cultured alone. Finally, we performed microelectrode array measurements on neuronal cultures, which revealed that muscle-conditioned medium enhances overall neural firing rates and the emergence of synchronous bursting patterns. Overall, our study illustrates the significance of neuron-muscle cross talk for the in vitro development of neuromuscular bioactuators.
Collapse
Affiliation(s)
- Onur Aydin
- Department of Mechanical Science and Engineering, University
of Illinois at Urbana-Champaign, Urbana, Illinois 61801,
USA
| | | | - Mohamed Elhebeary
- Department of Mechanical Science and Engineering, University
of Illinois at Urbana-Champaign, Urbana, Illinois 61801,
USA
| | - Gelson J. Pagan-Diaz
- Department of Bioengineering, University of Illinois at
Urbana-Champaign, Urbana, Illinois 61801, USA
| | - Anthony Fan
- Department of Mechanical Science and Engineering, University
of Illinois at Urbana-Champaign, Urbana, Illinois 61801,
USA
| | - Sittinon Nuethong
- Department of Mechanical Science and Engineering, University
of Illinois at Urbana-Champaign, Urbana, Illinois 61801,
USA
| | - Rashid Bashir
- Department of Bioengineering, University of Illinois at
Urbana-Champaign, Urbana, Illinois 61801, USA
| | | | - M. Taher A. Saif
- Department of Mechanical Science and Engineering, University
of Illinois at Urbana-Champaign, Urbana, Illinois 61801,
USA
| |
Collapse
|
52
|
Arrigoni C, Lopa S, Candrian C, Moretti M. Organs-on-a-chip as model systems for multifactorial musculoskeletal diseases. Curr Opin Biotechnol 2020; 63:79-88. [PMID: 31927146 DOI: 10.1016/j.copbio.2019.12.006] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 12/04/2019] [Accepted: 12/08/2019] [Indexed: 12/11/2022]
Abstract
Multifactorial diseases affecting musculoskeletal tissues are characterized by the interactions between multiple tissues, such as muscle and nerves in neuromuscular diseases, or multiple cellular components in a tissue, as in the case of bone tumors, interacting with bone cells. For these diseases also the influence of different biophysical and biochemical stimuli, such as mechanical overload and inflammatory molecules in osteoarthritis, play a key role. To investigate these complex phenomena, organ-on-a-chip systems have been developed, taking into account specific disease characteristics such as being directly derived from patients, the presence of specifically mutated cells, or a combination of relevant biophysical and/or biochemical stimuli. Depending on the envisaged application, different issues remain to be addressed. In particular, improving automation and output sensors are key for drug screening applications, while refining model microarchitecture to enhance physiological fidelity is needed for more basic science studies.
Collapse
Affiliation(s)
- Chiara Arrigoni
- Regenerative Medicine Technologies Laboratory, Ente Ospedaliero Cantonale (EOC), via Tesserete 46, 6900 Lugano, Switzerland
| | - Silvia Lopa
- IRCCS Istituto Ortopedico Galeazzi, Cell and Tissue Engineering Laboratory, via R. Galeazzi 4, 20161 Milano, Italy
| | - Christian Candrian
- Unità di Ortopedia e Traumatologia, Ospedale Regionale di Lugano, Ente Ospedaliero Cantonale (EOC), via Tesserete 46, 6900 Lugano, Switzerland
| | - Matteo Moretti
- Regenerative Medicine Technologies Laboratory, Ente Ospedaliero Cantonale (EOC), via Tesserete 46, 6900 Lugano, Switzerland; IRCCS Istituto Ortopedico Galeazzi, Cell and Tissue Engineering Laboratory, via R. Galeazzi 4, 20161 Milano, Italy
| |
Collapse
|
53
|
|
54
|
Vila OF, Qu Y, Vunjak-Novakovic G. In vitro models of neuromuscular junctions and their potential for novel drug discovery and development. Expert Opin Drug Discov 2019; 15:307-317. [PMID: 31846349 DOI: 10.1080/17460441.2020.1700225] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Introduction: Neuromuscular Junctions (NMJs) are the synapses between motor neurons and skeletal muscle fibers, and they are responsible for voluntary motor function. NMJs are affected at early stages of numerous neurodegenerative and neuroimmunological diseases. Due to the difficulty of systematically studying and manipulating NMJs in live subjects, in vitro systems with human tissue models would provide a powerful complement to simple cell cultures and animal models for mechanistic and drug development studies.Areas covered: The authors review the latest advances in in vitro models of NMJs, from traditional cell co-culture systems to novel tissue culture approaches, with focus on disease modeling and drug testing.Expert opinion: In recent years, more sophisticated in vitro models of human NMJs have been established. The combination of human stem cell technology with advanced tissue culture systems has resulted in systems that better recapitulate the human NMJ structure and function, and thereby allow for high-throughput quantitative functional measurements under both healthy and diseased conditions. Although they still have limitations, these advanced systems are increasingly demonstrating their utility for evaluating new therapies for motoneuron and autoimmune neuromuscular diseases, and we expect them to become an integral part of the drug discovery process in the near future.
Collapse
Affiliation(s)
- Olaia F Vila
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Yihuai Qu
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | | |
Collapse
|
55
|
Altman T, Geller D, Kleeblatt E, Gradus-Perry T, Perlson E. An in vitro compartmental system underlines the contribution of mitochondrial immobility to the ATP supply in the NMJ. J Cell Sci 2019; 132:jcs.234492. [PMID: 31722980 DOI: 10.1242/jcs.234492] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Accepted: 11/06/2019] [Indexed: 12/13/2022] Open
Abstract
The neuromuscular junction (NMJ) is the largest, most-complex synapse in the human body. Motor neuron (MN) diseases, such as amyotrophic lateral sclerosis (ALS), specifically target MNs and the NMJs. However, little is known about the reasons for MN-selective neuronal and synaptic vulnerability in MN diseases. Here, utilizing a compartmental microfluidic in vitro co-culture system, we provide a possible explanation for why the NMJ, other than its unusual dimensions, differs from other synapses. By using live-imaging techniques, we discovered that cultured MNs display higher axonal and synaptic mitochondrial immobility compared with sympathetic neurons (SNs), leading to a profound enrichment of mitochondria only in the MN NMJ. Furthermore, by employing a synaptic ATP sensor, we show that mitochondrial respiration is the key contributor to ATP production in MN NMJs but not in SN synapses. Taken together, our data suggest that mitochondrial localization underlies the unique and specific qualities of MN NMJs. Our findings shed light on the role of mitochondria in MN and NMJ maintenance, and possibly indicate how mitochondria may serve as a source for selective MN vulnerability in neurodegenerative diseases.This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Topaz Altman
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Ramat Aviv 69978, Tel Aviv, Israel
| | - Danielle Geller
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Ramat Aviv 69978, Tel Aviv, Israel.,Sagol School of Neuroscience, Tel Aviv University, Ramat Aviv 69978, Tel Aviv, Israel
| | - Elisabeth Kleeblatt
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Ramat Aviv 69978, Tel Aviv, Israel
| | - Tal Gradus-Perry
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Ramat Aviv 69978, Tel Aviv, Israel
| | - Eran Perlson
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Ramat Aviv 69978, Tel Aviv, Israel .,Sagol School of Neuroscience, Tel Aviv University, Ramat Aviv 69978, Tel Aviv, Israel
| |
Collapse
|
56
|
Soucy JR, Bindas AJ, Koppes AN, Koppes RA. Instrumented Microphysiological Systems for Real-Time Measurement and Manipulation of Cellular Electrochemical Processes. iScience 2019; 21:521-548. [PMID: 31715497 PMCID: PMC6849363 DOI: 10.1016/j.isci.2019.10.052] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Revised: 10/17/2019] [Accepted: 10/24/2019] [Indexed: 12/17/2022] Open
Abstract
Recent advancements in electronic materials and subsequent surface modifications have facilitated real-time measurements of cellular processes far beyond traditional passive recordings of neurons and muscle cells. Specifically, the functionalization of conductive materials with ligand-binding aptamers has permitted the utilization of traditional electronic materials for bioelectronic sensing. Further, microfabrication techniques have better allowed microfluidic devices to recapitulate the physiological and pathological conditions of complex tissues and organs in vitro or microphysiological systems (MPS). The convergence of these models with advances in biological/biomedical microelectromechanical systems (BioMEMS) instrumentation has rapidly bolstered a wide array of bioelectronic platforms for real-time cellular analytics. In this review, we provide an overview of the sensing techniques that are relevant to MPS development and highlight the different organ systems to integrate instrumentation for measurement and manipulation of cellular function. Special attention is given to how instrumented MPS can disrupt the drug development and fundamental mechanistic discovery processes.
Collapse
Affiliation(s)
- Jonathan R Soucy
- Department of Chemical Engineering, Northeastern University, Boston, MA 02115, USA
| | - Adam J Bindas
- Department of Chemical Engineering, Northeastern University, Boston, MA 02115, USA
| | - Abigail N Koppes
- Department of Chemical Engineering, Northeastern University, Boston, MA 02115, USA; Department of Biology, Northeastern University, Boston, MA 02115, USA
| | - Ryan A Koppes
- Department of Chemical Engineering, Northeastern University, Boston, MA 02115, USA.
| |
Collapse
|
57
|
A customizable microfluidic platform for medium-throughput modeling of neuromuscular circuits. Biomaterials 2019; 225:119537. [PMID: 31614290 PMCID: PMC7294901 DOI: 10.1016/j.biomaterials.2019.119537] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Revised: 10/02/2019] [Accepted: 10/07/2019] [Indexed: 01/27/2023]
Abstract
Neuromuscular circuits (NMCs) are vital for voluntary movement, and effective models of NMCs are needed to understand the pathogenesis of, as well as to identify effective treatments for, multiple diseases, including Duchenne’s muscular dystrophy and amyotrophic lateral sclerosis. Microfluidics are ideal for recapitulating the central and peripheral compartments of NMCs, but myotubes often detach before functional NMCs are formed. In addition, microfluidic systems are often limited to a single experimental unit, which significantly limits their application in disease modeling and drug discovery. Here, we developed a microfluidic platform (MFP) containing over 100 experimental units, making it suitable for medium-throughput applications. To overcome detachment, we incorporated a reactive polymer surface allowing customization of the environment to culture different cell types. Using this approach, we identified conditions that enable long-term co-culture of human motor neurons and myotubes differentiated from human induced pluripotent stem cells inside our MFP. Optogenetics demonstrated the formation of functional NMCs. Furthermore, we developed a novel application of the rabies tracing assay to efficiently identify NMCs in our MFP. Therefore, our MFP enables large-scale generation and quantification of functional NMCs for disease modeling and pharmacological drug targeting.
Collapse
|
58
|
Vysokov N, McMahon SB, Raouf R. The role of Na V channels in synaptic transmission after axotomy in a microfluidic culture platform. Sci Rep 2019; 9:12915. [PMID: 31501450 PMCID: PMC6733904 DOI: 10.1038/s41598-019-49214-w] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Accepted: 08/20/2019] [Indexed: 01/07/2023] Open
Abstract
Voltage gated sodium channels are key players in aberrant pain signaling and sensitization of nociceptors after peripheral nerve injury. The extent to which sodium channel activity after injury contributes to synaptic transmission at the first pain synapse however remains unclear. To investigate the effect of axotomy on synaptic transmission between dorsal root ganglia neurons and dorsal horn neurons, we reconstructed the first pain synapse in a novel microfluidic based compartmentalized cell culture system, which recapitulates the connectivity of peripheral pain signaling. We show that following axotomy of the distal axons, inhibition of NaV1.7 and NaV1.8 sodium channels in incoming presynaptic DRG axons is no longer sufficient to block activation of these axons and the resulting synaptic transmission to dorsal horn neurons. We found that blockade of NaV1.6 activity is highly effective in reducing activation of incoming axons contributing to synaptic transmission after axotomy of DRG neurons. The microfluidic culture system described here offers an in vitro platform to recapitulate and study the first pain synapse.
Collapse
Affiliation(s)
- Nickolai Vysokov
- Wolfson Centre for Age-Related Diseases, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, SE1 1UL, United Kingdom
| | - Stephen B McMahon
- Wolfson Centre for Age-Related Diseases, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, SE1 1UL, United Kingdom
| | - Ramin Raouf
- Wolfson Centre for Age-Related Diseases, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, SE1 1UL, United Kingdom.
| |
Collapse
|
59
|
Natarajan A, Sethumadhavan A, Krishnan UM. Toward Building the Neuromuscular Junction: In Vitro Models To Study Synaptogenesis and Neurodegeneration. ACS OMEGA 2019; 4:12969-12977. [PMID: 31460423 PMCID: PMC6682064 DOI: 10.1021/acsomega.9b00973] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Accepted: 07/04/2019] [Indexed: 06/10/2023]
Abstract
The neuromuscular junction (NMJ) is a unique, specialized chemical synapse that plays a crucial role in transmitting and amplifying information from spinal motor neurons to skeletal muscles. NMJ complexity ensures closely intertwined interactions between numerous synaptic vesicles, signaling molecules, ion channels, motor neurons, glia, and muscle fibers, making it difficult to dissect the underlying mechanisms and factors affecting neurodegeneration and muscle loss. Muscle fiber or motor neuron cell death followed by rapid axonal degeneration due to injury or disease has a debilitating effect on movement and behavior, which adversely affects the quality of life. It thus becomes imperative to study the synapse and intercellular signaling processes that regulate plasticity at the NMJ and elucidate mechanisms and pathways at the cellular level. Studies using in vitro 2D cell cultures have allowed us to gain a fundamental understanding of how the NMJ functions. However, they do not provide information on the intricate signaling networks that exist between NMJs and the biological environment. The advent of 3D cell cultures and microfluidic lab-on-a-chip technologies has opened whole new avenues to explore the NMJ. In this perspective, we look at the challenges involved in building a functional NMJ and the progress made in generating models for studying the NMJ, highlighting the current and future applications of these models.
Collapse
Affiliation(s)
- Anupama Natarajan
- Centre
for Nanotechnology & Advanced Biomaterials (CeNTAB), School of Chemical
& Biotechnology, and School of Arts, Science & Humanities, SASTRA Deemed University, Thanjavur 613 401, India
| | - Anjali Sethumadhavan
- Centre
for Nanotechnology & Advanced Biomaterials (CeNTAB), School of Chemical
& Biotechnology, and School of Arts, Science & Humanities, SASTRA Deemed University, Thanjavur 613 401, India
| | - Uma Maheswari Krishnan
- Centre
for Nanotechnology & Advanced Biomaterials (CeNTAB), School of Chemical
& Biotechnology, and School of Arts, Science & Humanities, SASTRA Deemed University, Thanjavur 613 401, India
| |
Collapse
|
60
|
Mella J, Pérez V, Zelada D, Moreno N, Ionescu A, Perlson E, Henríquez JP. Efficient gene transfer into primary muscle cells to analyze nerve-independent postsynaptic organization in vitro. Neuromuscul Disord 2019; 29:533-542. [DOI: 10.1016/j.nmd.2019.05.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 04/23/2019] [Accepted: 05/17/2019] [Indexed: 01/08/2023]
|
61
|
Machado CB, Pluchon P, Harley P, Rigby M, Gonzalez Sabater V, Stevenson DC, Hynes S, Lowe A, Burrone J, Viasnoff V, Lieberam I. In Vitro Modelling of Nerve-Muscle Connectivity in a Compartmentalised Tissue Culture Device. ADVANCED BIOSYSTEMS 2019; 3:1800307. [PMID: 31428672 PMCID: PMC6699992 DOI: 10.1002/adbi.201800307] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/17/2018] [Indexed: 01/02/2023]
Abstract
Motor neurons project axons from the hindbrain and spinal cord to muscle, where they induce myofibre contractions through neurotransmitter release at neuromuscular junctions. Studies of neuromuscular junction formation and homeostasis have been largely confined to in vivo models. In this study we have merged three powerful tools - pluripotent stem cells, optogenetics and microfabrication - and designed an open microdevice in which motor axons grow from a neural compartment containing embryonic stem cell-derived motor neurons and astrocytes through microchannels to form functional neuromuscular junctions with contractile myofibers in a separate compartment. Optogenetic entrainment of motor neurons in this reductionist neuromuscular circuit enhanced neuromuscular junction formation more than two-fold, mirroring the activity-dependence of synapse development in vivo. We incorporated an established motor neuron disease model into our system and found that coculture of motor neurons with SOD1G93A astrocytes resulted in denervation of the central compartment and diminished myofiber contractions, a phenotype which was rescued by the Receptor Interacting Serine/Threonine Kinase 1 (RIPK1) inhibitor Necrostatin. This coculture system replicates key aspects of nerve-muscle connectivity in vivo and represents a rapid and scalable alternative to animal models of neuromuscular function and disease.
Collapse
Affiliation(s)
- Carolina Barcellos Machado
- Centre for Stem Cells and Regenerative Medicine, King’s
College London, London SE1 9RT, UK; Centre for Developmental
Neurobiology/MRC Centre for Neurodevelopmental Disorders, King’s
College London, London SE1 1UL, UK
| | - Perrine Pluchon
- Centre for Stem Cells and Regenerative Medicine, King’s
College London, London SE1 9RT, UK; Centre for Developmental
Neurobiology/MRC Centre for Neurodevelopmental Disorders, King’s
College London, London SE1 1UL, UK; Mechanobiology Institute, National
University of Singapore, Singapore 117411
| | - Peter Harley
- Centre for Stem Cells and Regenerative Medicine, King’s College London, London SE1 9RT, UK; Centre for Developmental Neurobiology/MRC Centre for Neurodevelopmental Disorders, King’s College London, London SE1 1UL, UK
| | | | - Victoria Gonzalez Sabater
- Centre for Developmental Neurobiology/MRC Centre for Neurodevelopmental Disorders, King’s College London, London SE1 1UL, UK
| | | | - Stephanie Hynes
- Centre for Stem Cells and Regenerative Medicine, King’s College London, London SE1 9RT, UK; Centre for Developmental Neurobiology/MRC Centre for Neurodevelopmental Disorders, King’s College London, London SE1 1UL, UK
| | - Andrew Lowe
- Centre for Developmental Neurobiology, King’s College London, London SE1 1UL, UK
| | - Juan Burrone
- Centre for Developmental Neurobiology/MRC Centre for Neurodevelopmental Disorders, King’s College London, London SE1 1UL, UK
| | - Virgile Viasnoff
- Mechanobiology Institute, National University of Singapore,
Singapore 117411
| | - Ivo Lieberam
- Centre for Stem Cells and Regenerative Medicine, King’s
College London, London SE1 9RT, UK; Centre for Developmental
Neurobiology/MRC Centre for Neurodevelopmental Disorders, King’s
College London, London SE1 1UL, UK
| |
Collapse
|
62
|
Arrigoni C, Petta D, Bersini S, Mironov V, Candrian C, Moretti M. Engineering complex muscle-tissue interfaces through microfabrication. Biofabrication 2019; 11:032004. [PMID: 31042682 DOI: 10.1088/1758-5090/ab1e7c] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Skeletal muscle is a tissue with a complex and hierarchical architecture that influences its functional properties. In order to exert its contractile function, muscle tissue is connected to neural, vascular and connective compartments, comprising finely structured interfaces which are orchestrated by multiple signalling pathways. Pathological conditions such as dystrophies and trauma, or physiological situations such as exercise and aging, modify the architectural organization of these structures, hence affecting muscle functionality. To overcome current limitations of in vivo and standard in vitro models, microfluidics and biofabrication techniques have been applied to better reproduce the microarchitecture and physicochemical environment of human skeletal muscle tissue. In the present review, we aim to critically discuss the role of those techniques, taken individually or in combination, in the generation of models that mimic the complex interfaces between muscle tissue and neural/vascular/tendon compartments. The exploitation of either microfluidics or biofabrication to model different muscle interfaces has led to the development of constructs with an improved spatial organization, thus presenting a better functionality as compared to standard models. However, the achievement of models replicating muscle-tissue interfaces with adequate architecture, presence of fundamental proteins and recapitulation of signalling pathways is still far from being achieved. Increased integration between microfluidics and biofabrication, providing the possibility to pattern cells in predetermined structures with higher resolution, will help to reproduce the hierarchical and heterogeneous structure of skeletal muscle interfaces. Such strategies will further improve the functionality of these techniques, providing a key contribution towards the study of skeletal muscle functions in physiology and pathology.
Collapse
Affiliation(s)
- Chiara Arrigoni
- Regenerative Medicine Technologies Lab, Ente Ospedaliero Cantonale (EOC), Via Tesserete 46, 6900 Lugano, Switzerland
| | | | | | | | | | | |
Collapse
|
63
|
Heiney K, Mateus JC, Lopes CDF, Neto E, Lamghari M, Aguiar P. µSpikeHunter: An advanced computational tool for the analysis of neuronal communication and action potential propagation in microfluidic platforms. Sci Rep 2019; 9:5777. [PMID: 30962522 PMCID: PMC6453950 DOI: 10.1038/s41598-019-42148-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Accepted: 02/26/2019] [Indexed: 12/11/2022] Open
Abstract
Understanding neuronal communication is fundamental in neuroscience, but there are few methodologies offering detailed analysis for well-controlled conditions. By interfacing microElectrode arrays with microFluidics (μEF devices), it is possible to compartmentalize neuronal cultures with a specified alignment of axons and microelectrodes. This setup allows the extracellular recording of spike propagation with a high signal-to-noise ratio over the course of several weeks. Addressing these μEF devices, we developed an advanced yet easy-to-use publically available computational tool, μSpikeHunter, which provides a detailed quantification of several communication-related properties such as propagation velocity, conduction failure, spike timings, and coding mechanisms. The combination of μEF devices and μSpikeHunter can be used in the context of standard neuronal cultures or with co-culture configurations where, for example, communication between sensory neurons and other cell types is monitored and assessed. The ability to analyze axonal signals (in a user-friendly, time-efficient, high-throughput manner) opens the door to new approaches in studies of peripheral innervation, neural coding, and neuroregeneration, among many others. We demonstrate the use of μSpikeHunter in dorsal root ganglion neurons where we analyze the presence of both anterograde and retrograde signals in μEF devices. A fully functional version of µSpikeHunter is publically available for download from https://github.com/uSpikeHunter .
Collapse
Affiliation(s)
- Kristine Heiney
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135, Porto, Portugal.,INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135, Porto, Portugal
| | - José C Mateus
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135, Porto, Portugal.,INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135, Porto, Portugal.,ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Rua Jorge de Viterbo Ferreira, 4050-313, Porto, Portugal
| | - Cátia D F Lopes
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135, Porto, Portugal.,INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135, Porto, Portugal
| | - Estrela Neto
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135, Porto, Portugal.,INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135, Porto, Portugal
| | - Meriem Lamghari
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135, Porto, Portugal.,INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135, Porto, Portugal
| | - Paulo Aguiar
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135, Porto, Portugal. .,INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135, Porto, Portugal.
| |
Collapse
|
64
|
Mobini S, Song YH, McCrary MW, Schmidt CE. Advances in ex vivo models and lab-on-a-chip devices for neural tissue engineering. Biomaterials 2019; 198:146-166. [PMID: 29880219 PMCID: PMC6957334 DOI: 10.1016/j.biomaterials.2018.05.012] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Revised: 04/25/2018] [Accepted: 05/07/2018] [Indexed: 02/08/2023]
Abstract
The technologies related to ex vivo models and lab-on-a-chip devices for studying the regeneration of brain, spinal cord, and peripheral nerve tissues are essential tools for neural tissue engineering and regenerative medicine research. The need for ex vivo systems, lab-on-a-chip technologies and disease models for neural tissue engineering applications are emerging to overcome the shortages and drawbacks of traditional in vitro systems and animal models. Ex vivo models have evolved from traditional 2D cell culture models to 3D tissue-engineered scaffold systems, bioreactors, and recently organoid test beds. In addition to ex vivo model systems, we discuss lab-on-a-chip devices and technologies specifically for neural tissue engineering applications. Finally, we review current commercial products that mimic diseased and normal neural tissues, and discuss the future directions in this field.
Collapse
Affiliation(s)
- Sahba Mobini
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA
| | - Young Hye Song
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA
| | - Michaela W McCrary
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA
| | - Christine E Schmidt
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
65
|
Ko E, Yu SJ, Pagan‐Diaz GJ, Mahmassani Z, Boppart MD, Im SG, Bashir R, Kong H. Matrix Topography Regulates Synaptic Transmission at the Neuromuscular Junction. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2019; 6:1801521. [PMID: 30937256 PMCID: PMC6425454 DOI: 10.1002/advs.201801521] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Revised: 10/26/2018] [Indexed: 05/19/2023]
Abstract
Recreation of a muscle that can be controlled by the nervous system would provide a major breakthrough for treatments of injury and diseases. However, the underlying basis of how neuron-muscle interfaces are formed is still not understood sufficiently. Here, it is hypothesized that substrate topography regulates neural innervation and synaptic transmission by mediating the cross-talk between neurons and muscles. This hypothesis is examined by differentiating neural stem cells on the myotubes, formed on the substrate with controlled groove width. The substrate with the groove width of 1600 nm, a similar size to the myofibril diameter, serves to produce larger and aligned myotubes than the flat substrate. The myotubes formed on the grooved substrate display increases in the acetylcholine receptor expression. Reciprocally, motor neuron progenitor cells differentiated from neural stem cells innervate the larger and aligned myotubes more actively than randomly oriented myotubes. As a consequence, mature and aligned myotubes respond to glutamate (i.e., an excitatory neurotransmitter) and curare (i.e., a neuromuscular antagonist) more rapidly and homogeneously than randomly oriented myotubes. The results of this study will be broadly useful for improving the quality of engineered muscle used in a series of applications including drug screening, regeneration therapies, and biological machinery assembly.
Collapse
Affiliation(s)
- Eunkyung Ko
- Department of BioengineeringUniversity of Illinois at Urbana–ChampaignUrbanaIL61801USA
- Department of BioengineeringMicro and Nanotechnology LaboratoryUniversity of Illinois at Urbana–ChampaignUrbanaIL61801USA
| | - Seung Jung Yu
- Department of Chemical and Biomolecular Engineering and KI for the Nano CenturyKorea Advanced Institute of Science and Technology (KAIST)Daejeon305‐701Republic of Korea
| | - Gelson J. Pagan‐Diaz
- Department of BioengineeringMicro and Nanotechnology LaboratoryUniversity of Illinois at Urbana–ChampaignUrbanaIL61801USA
| | - Ziad Mahmassani
- Department of Kinesiology and Community HealthBeckman Institute for Advanced Science and TechnologyUniversity of Illinois at Urbana–ChampaignUrbanaIL61801USA
| | - Marni D. Boppart
- Department of Kinesiology and Community HealthBeckman Institute for Advanced Science and TechnologyUniversity of Illinois at Urbana–ChampaignUrbanaIL61801USA
| | - Sung Gap Im
- Department of Chemical and Biomolecular Engineering and KI for the Nano CenturyKorea Advanced Institute of Science and Technology (KAIST)Daejeon305‐701Republic of Korea
| | - Rashid Bashir
- Department of BioengineeringMicro and Nanotechnology LaboratoryUniversity of Illinois at Urbana–ChampaignUrbanaIL61801USA
- Carl R. Woese Institute for Genomic Biology and Beckman Institute for Advanced Science and TechnologyUniversity of Illinois at Urbana–ChampaignUrbanaIL61801USA
- Carle Illinois College of MedicineUniversity of Illinois at Urbana–ChampaignUrbanaIL61801USA
| | - Hyunjoon Kong
- Department of BioengineeringUniversity of Illinois at Urbana–ChampaignUrbanaIL61801USA
- Carl R. Woese Institute for Genomic Biology and Beckman Institute for Advanced Science and TechnologyUniversity of Illinois at Urbana–ChampaignUrbanaIL61801USA
- Carle Illinois College of MedicineUniversity of Illinois at Urbana–ChampaignUrbanaIL61801USA
- Department of Chemical and Biomolecular EngineeringUniversity of Illinois at Urbana–ChampaignUrbanaIL61801USA
| |
Collapse
|
66
|
Ionescu A, Gradus T, Altman T, Maimon R, Saraf Avraham N, Geva M, Hayden M, Perlson E. Targeting the Sigma-1 Receptor via Pridopidine Ameliorates Central Features of ALS Pathology in a SOD1 G93A Model. Cell Death Dis 2019; 10:210. [PMID: 30824685 PMCID: PMC6397200 DOI: 10.1038/s41419-019-1451-2] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Revised: 11/16/2018] [Accepted: 02/12/2019] [Indexed: 12/29/2022]
Abstract
Amyotrophic Lateral Sclerosis (ALS) is a fatal neurodegenerative disease affecting both the upper and lower motor neurons (MNs), with no effective treatment currently available. Early pathological events in ALS include perturbations in axonal transport (AT), formation of toxic protein aggregates and Neuromuscular Junction (NMJ) disruption, which all lead to axonal degeneration and motor neuron death. Pridopidine is a small molecule that has been clinically developed for Huntington disease. Here we tested the efficacy of pridopidine for ALS using in vitro and in vivo models. Pridopidine beneficially modulates AT deficits and diminishes NMJ disruption, as well as motor neuron death in SOD1G93A MNs and in neuromuscular co-cultures. Furthermore, we demonstrate that pridopidine activates the ERK pathway and mediates its beneficial effects through the sigma-1 receptor (S1R). Strikingly, in vivo evaluation of pridopidine in SOD1G93A mice reveals a profound reduction in mutant SOD1 aggregation in the spinal cord, and attenuation of NMJ disruption, as well as subsequent muscle wasting. Taken together, we demonstrate for the first time that pridopidine improves several cellular and histological hallmark pathologies of ALS through the S1R.
Collapse
Affiliation(s)
- Ariel Ionescu
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, 69978, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, 69978, Israel
| | - Tal Gradus
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, 69978, Israel
| | - Topaz Altman
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, 69978, Israel
| | - Roy Maimon
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, 69978, Israel
| | - Noi Saraf Avraham
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, 69978, Israel
| | - Michal Geva
- Teva Pharmaceuticals Ltd, Petah Tikva, Israel
- Prilenia Therapeutics, Herzliya, Israel
| | - Michael Hayden
- Teva Pharmaceuticals Ltd, Petah Tikva, Israel
- Prilenia Therapeutics, Herzliya, Israel
| | - Eran Perlson
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, 69978, Israel.
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, 69978, Israel.
| |
Collapse
|
67
|
Singh T, Vazquez M. Time-Dependent Addition of Neuronal and Schwann Cells Increase Myotube Viability and Length in an In Vitro Tri-culture Model of the Neuromuscular Junction. REGENERATIVE ENGINEERING AND TRANSLATIONAL MEDICINE 2019. [DOI: 10.1007/s40883-019-00095-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
68
|
Atmaramani R, Black BJ, Lam KH, Sheth VM, Pancrazio JJ, Schmidtke DW, Alsmadi NZ. The Effect of Microfluidic Geometry on Myoblast Migration. MICROMACHINES 2019; 10:E143. [PMID: 30795574 PMCID: PMC6412509 DOI: 10.3390/mi10020143] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Revised: 02/04/2019] [Accepted: 02/19/2019] [Indexed: 11/23/2022]
Abstract
In vitro systems comprised of wells interconnected by microchannels have emerged as a platform for the study of cell migration or multicellular models. In the present study, we systematically evaluated the effect of microchannel width on spontaneous myoblast migration across these microchannels-from the proximal to the distal chamber. Myoblast migration was examined in microfluidic devices with varying microchannel widths of 1.5⁻20 µm, and in chips with uniform microchannel widths over time spans that are relevant for myoblast-to-myofiber differentiation in vitro. We found that the likelihood of spontaneous myoblast migration was microchannel width dependent and that a width of 3 µm was necessary to limit spontaneous migration below 5% of cells in the seeded well after 48 h. These results inform the future design of Polydimethylsiloxane (PDMS) microchannel-based co-culture platforms as well as future in vitro studies of myoblast migration.
Collapse
Affiliation(s)
- Rahul Atmaramani
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX 75080, USA.
| | - Bryan J Black
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX 75080, USA.
| | - Kevin H Lam
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX 75080, USA.
| | - Vinit M Sheth
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX 75080, USA.
| | - Joseph J Pancrazio
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX 75080, USA.
| | - David W Schmidtke
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX 75080, USA.
| | | |
Collapse
|
69
|
Chan T, Williams E, Cohen O, Eliceiri BP, Baird A, Costantini TW. CHRFAM7A alters binding to the neuronal alpha-7 nicotinic acetylcholine receptor. Neurosci Lett 2019; 690:126-131. [PMID: 30308236 PMCID: PMC6320298 DOI: 10.1016/j.neulet.2018.10.010] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Revised: 10/04/2018] [Accepted: 10/07/2018] [Indexed: 01/14/2023]
Abstract
INTRODUCTION CHRFAM7A is a uniquely-human gene that encodes a human-specific variant of the alpha-7 nicotinic acetylcholine receptor (α7nAchR). While the homopentameric α7nAChR consists of 5 equal subunits, previous studies demonstrated that CHRFAM7A expression disrupts the formation of α7nAChR homopentamers. Here we use a rat neuronal cell line expressing CHRFAM7A and a transgenic mouse expressing CHRFAM7A to define the alpha-bungarotoxin (α-BTX) binding in vitro and in vivo. METHODS Rat PC12 cells were stably transfected with human CHRFAM7A. α-BTX, a protein that irreversibly binds the α7nAchR, was utilized to assess the capacity for CHRFAM7A to interfere with α 7AchR subunits using immunohistochemistry and flow cytometry. To evaluate the effects of CHRFAM7A on α7nAchR at the neuromuscular junction in vivo, transgenic mice were engineered to express the uniquely human gene CHRFAM7A under the control of the EF1-α promoter. Using this model, muscle was harvested and CHRFAM7A and CHRNA7 gene expression evaluated by PCR. Binding of α-BTX to the α7nAchR in muscle was compared in sibling-matched wild-type C57 mice by immunostaining the neuromuscular junction using α-BTX and neurofilament antibodies. RESULTS Expression of CHRFAM7A in transfected, but not vector cells, was confirmed by PCR and by immunoblotting using an antibody we raised to a peptide sequence unique to CHRFAM7A. CHRFAM7A decreased α-BTX binding as detected by immunohistochemistry and flow cytometry. In vivo, α-BTX co-stained with neurofilament at the neuromuscular junction in wild-type mice, however, α-BTX staining was decreased at the neuromuscular junction of CHRFAM7A transgenic mice. CONCLUSION CHRFAM7A expression interferes with the binding of α7nAchR to α-BTX. Understanding the contribution of this uniquely human gene to human disease will be important in the identification of potential therapeutic targets.
Collapse
Affiliation(s)
- Theresa Chan
- Division of Trauma, Surgical Critical Care, Burns and Acute Care Surgery, Department of Surgery, University of California San Diego Health, 200 W. Arbor Drive #8896, San Diego, CA, 92103, USA.
| | - Elliot Williams
- Division of Trauma, Surgical Critical Care, Burns and Acute Care Surgery, Department of Surgery, University of California San Diego Health, 200 W. Arbor Drive #8896, San Diego, CA, 92103, USA.
| | - Olga Cohen
- Division of Trauma, Surgical Critical Care, Burns and Acute Care Surgery, Department of Surgery, University of California San Diego Health, 200 W. Arbor Drive #8896, San Diego, CA, 92103, USA.
| | - Brian P Eliceiri
- Division of Trauma, Surgical Critical Care, Burns and Acute Care Surgery, Department of Surgery, University of California San Diego Health, 200 W. Arbor Drive #8896, San Diego, CA, 92103, USA.
| | - Andrew Baird
- Division of Trauma, Surgical Critical Care, Burns and Acute Care Surgery, Department of Surgery, University of California San Diego Health, 200 W. Arbor Drive #8896, San Diego, CA, 92103, USA.
| | - Todd W Costantini
- Division of Trauma, Surgical Critical Care, Burns and Acute Care Surgery, Department of Surgery, University of California San Diego Health, 200 W. Arbor Drive #8896, San Diego, CA, 92103, USA.
| |
Collapse
|
70
|
Badiola-Mateos M, Hervera A, Del Río JA, Samitier J. Challenges and Future Prospects on 3D in-vitro Modeling of the Neuromuscular Circuit. Front Bioeng Biotechnol 2018; 6:194. [PMID: 30622944 PMCID: PMC6297173 DOI: 10.3389/fbioe.2018.00194] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Accepted: 11/27/2018] [Indexed: 12/18/2022] Open
Abstract
Movement of skeletal-muscle fibers is generated by the coordinated action of several cells taking part within the locomotion circuit (motoneurons, sensory-neurons, Schwann cells, astrocytes, microglia, and muscle-cells). Failures in any part of this circuit could impede or hinder coordinated muscle movement and cause a neuromuscular disease (NMD) or determine its severity. Studying fragments of the circuit cannot provide a comprehensive and complete view of the pathological process. We trace the historic developments of studies focused on in-vitro modeling of the spinal-locomotion circuit and how bioengineered innovative technologies show advantages for an accurate mimicking of physiological conditions of spinal-locomotion circuit. New developments on compartmentalized microfluidic culture systems (cμFCS), the use of human induced pluripotent stem cells (hiPSCs) and 3D cell-cultures are analyzed. We finally address limitations of current study models and three main challenges on neuromuscular studies: (i) mimic the whole spinal-locomotion circuit including all cell-types involved and the evaluation of independent and interdependent roles of each one; (ii) mimic the neurodegenerative response of mature neurons in-vitro as it occurs in-vivo; and (iii) develop, tune, implement, and combine cμFCS, hiPSC, and 3D-culture technologies to ultimately create patient-specific complete, translational, and reliable NMD in-vitro model. Overcoming these challenges would significantly facilitate understanding the events taking place in NMDs and accelerate the process of finding new therapies.
Collapse
Affiliation(s)
- Maider Badiola-Mateos
- Institute for Bioengineering of Catalonia-Barcelona Institute of Science and Technology, Barcelona, Spain.,Department of Electronics and Biomedical Engineering, Faculty of Physics, Universitat de Barcelona, Barcelona, Spain
| | - Arnau Hervera
- Institute for Bioengineering of Catalonia-Barcelona Institute of Science and Technology, Barcelona, Spain.,Department of Cell Biology, Physiology and Immunology, Faculty of Biology, Universitat de Barcelona, Barcelona, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas, Barcelona, Spain.,Institut de Neurociències de la Universitat de Barcelona, Barcelona, Spain
| | - José Antonio Del Río
- Institute for Bioengineering of Catalonia-Barcelona Institute of Science and Technology, Barcelona, Spain.,Department of Cell Biology, Physiology and Immunology, Faculty of Biology, Universitat de Barcelona, Barcelona, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas, Barcelona, Spain.,Institut de Neurociències de la Universitat de Barcelona, Barcelona, Spain
| | - Josep Samitier
- Institute for Bioengineering of Catalonia-Barcelona Institute of Science and Technology, Barcelona, Spain.,Department of Electronics and Biomedical Engineering, Faculty of Physics, Universitat de Barcelona, Barcelona, Spain.,Centro de Investigación Biomédica en Red en Bioingeniería, Biomateriales y Nanomedicina, Madrid, Spain
| |
Collapse
|
71
|
Abd Al Samid M, McPhee JS, Saini J, McKay TR, Fitzpatrick LM, Mamchaoui K, Bigot A, Mouly V, Butler-Browne G, Al-Shanti N. A functional human motor unit platform engineered from human embryonic stem cells and immortalized skeletal myoblasts. STEM CELLS AND CLONING-ADVANCES AND APPLICATIONS 2018; 11:85-93. [PMID: 30519053 PMCID: PMC6233953 DOI: 10.2147/sccaa.s178562] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Background Although considerable research on neuromuscular junctions (NMJs) has been conducted, the prospect of in vivo NMJ studies is limited and these studies are challenging to implement. Therefore, there is a clear unmet need to develop a feasible, robust, and physiologically relevant in vitro NMJ model. Objective We aimed to establish a novel functional human NMJs platform, which is serum and neural complex media/neural growth factor-free, using human immortalized myoblasts and human embryonic stem cells (hESCs)-derived neural progenitor cells (NPCs) that can be used to understand the mechanisms of NMJ development and degeneration. Methods Immortalized human myoblasts were co-cultured with hESCs derived committed NPCs. Over the course of the 7 days myoblasts differentiated into myotubes and NPCs differentiated into motor neurons. Results Neuronal axon sprouting branched to form multiple NMJ innervation sites along the myotubes and the myotubes showed extensive, spontaneous contractile activity. Choline acetyltransferase and βIII-tubulin immunostaining confirmed that the NPCs had matured into cholinergic motor neurons. Postsynaptic site of NMJs was further characterized by staining dihydropyridine receptors, ryanodine receptors, and acetylcholine receptors by α-bungarotoxin. Conclusion We established a functional human motor unit platform for in vitro investigations. Thus, this co-culture system can be used as a novel platform for 1) drug discovery in the treatment of neuromuscular disorders, 2) deciphering vital features of NMJ formation, regulation, maintenance, and repair, and 3) exploring neuromuscular diseases, age-associated degeneration of the NMJ, muscle aging, and diabetic neuropathy and myopathy.
Collapse
Affiliation(s)
- Marwah Abd Al Samid
- Healthcare Science Research Institute, School of Healthcare Science, Manchester Metropolitan University, Manchester, UK,
| | - Jamie S McPhee
- Department of Sport and Exercise Science, Manchester Metropolitan University, Manchester, UK
| | - Jasdeep Saini
- Healthcare Science Research Institute, School of Healthcare Science, Manchester Metropolitan University, Manchester, UK,
| | - Tristan R McKay
- Healthcare Science Research Institute, School of Healthcare Science, Manchester Metropolitan University, Manchester, UK,
| | - Lorna M Fitzpatrick
- Healthcare Science Research Institute, School of Healthcare Science, Manchester Metropolitan University, Manchester, UK,
| | - Kamel Mamchaoui
- Center for Research in Myology, Sorbonne Université-INSERM, Paris, France
| | - Anne Bigot
- Center for Research in Myology, Sorbonne Université-INSERM, Paris, France
| | - Vincent Mouly
- Center for Research in Myology, Sorbonne Université-INSERM, Paris, France
| | | | - Nasser Al-Shanti
- Healthcare Science Research Institute, School of Healthcare Science, Manchester Metropolitan University, Manchester, UK,
| |
Collapse
|
72
|
Al Samid MA, Al-Shanti N, Odeh M. Motor Neuron-Skeletal Muscle Co Culture Model: A Potential Novel in Vitro and Computaional Platform to Investigate Cancer Cachexia. 2018 1ST INTERNATIONAL CONFERENCE ON CANCER CARE INFORMATICS (CCI) 2018. [DOI: 10.1109/cancercare.2018.8618261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/02/2023]
|
73
|
Labroo P, Hilgart D, Davis B, Lambert C, Sant H, Gale B, Shea JE, Agarwal J. Drug-delivering nerve conduit improves regeneration in a critical-sized gap. Biotechnol Bioeng 2018; 116:143-154. [PMID: 30229866 DOI: 10.1002/bit.26837] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Revised: 09/04/2018] [Accepted: 09/05/2018] [Indexed: 01/03/2023]
Abstract
Autologous nerve grafts are the current "gold standard" for repairing large nerve gaps. However, they cause morbidity at the donor nerve site and only a limited amount of nerve can be harvested. Nerve conduits are a promising alternative to autografts and can act as guidance cues for the regenerating axons, without the need to harvest donor nerve. Separately, it has been shown that localized delivery of GDNF can enhance axon growth and motor recovery. FK506, an FDA approved small molecule, has also been shown to enhance peripheral nerve regeneration. This paper describes the design of a novel hole-based drug delivery apparatus integrated with a polytetrafluoroethylene (PTFE) nerve conduit for controlled local delivery of a protein such as GDNF or a small molecule such as FK506. The PTFE devices were tested in a diffusion chamber, and the bioactivity of the released media was evaluated by measuring neurite growth of dorsal root ganglions (DRGs) exposed to the released drugs. The drug delivering nerve guide was able to release bioactive concentrations of FK506 or GDNF. Following these tests, optimized drug releasing nerve conduits were implanted across 10 mm sciatic nerve gaps in a BL6 yellow fluorescent protein (YFP) mouse model, where they demonstrated significant improvement in muscle mass, compound muscle action potential, and axon myelination in vivo as compared with nerve conduits without the drug. The drug delivery nerve guide could release drug for extended periods of time and enhance axon growth in vitro and in vivo.
Collapse
Affiliation(s)
- Pratima Labroo
- Department of Mechanical Engineering, University of Utah, Salt Lake City, Utah
| | - David Hilgart
- Department of Bioengineering, University of Utah, Salt Lake City, Utah
| | - Brett Davis
- Department of Bioengineering, University of Utah, Salt Lake City, Utah
| | - Christopher Lambert
- Department of Mechanical Engineering, University of Utah, Salt Lake City, Utah
| | - Himanshu Sant
- Department of Mechanical Engineering, University of Utah, Salt Lake City, Utah
| | - Bruce Gale
- Department of Mechanical Engineering, University of Utah, Salt Lake City, Utah
| | - Jill E Shea
- Department of Surgery, University of Utah, Salt Lake City, Utah
| | - Jayant Agarwal
- Department of Surgery, University of Utah, Salt Lake City, Utah
| |
Collapse
|
74
|
Gershoni-Emek N, Altman T, Ionescu A, Costa CJ, Gradus-Pery T, Willis DE, Perlson E. Localization of RNAi Machinery to Axonal Branch Points and Growth Cones Is Facilitated by Mitochondria and Is Disrupted in ALS. Front Mol Neurosci 2018; 11:311. [PMID: 30233312 PMCID: PMC6134038 DOI: 10.3389/fnmol.2018.00311] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Accepted: 08/15/2018] [Indexed: 12/19/2022] Open
Abstract
Local protein synthesis in neuronal axons plays an important role in essential spatiotemporal signaling processes; however, the molecular basis for the post-transcriptional regulation controlling this process in axons is still not fully understood. Here we studied the axonal mechanisms underlying the transport and localization of microRNA (miRNA) and the RNAi machinery along the axon. We first identified miRNAs, Dicer, and Argonaute-2 (Ago2) in motor neuron (MN) axons. We then studied the localization of RNAi machinery and demonstrated that mitochondria associate with miR-124 and RNAi proteins in axons. Importantly, this co-localization occurs primarily at axonal branch points and growth cones. Moreover, using live cell imaging of a functional Cy3-tagged miR-124, we revealed that this miRNA is actively transported with acidic compartments in axons, and associates with stalled mitochondria at growth cones and axonal branch points. Finally, we observed enhanced retrograde transport of miR-124-Cy3, and a reduction in its localization to static mitochondria in MNs expressing the ALS causative gene hSOD1G93A. Taken together, our data suggest that mitochondria participate in the axonal localization and transport of RNAi machinery, and further imply that alterations in this mechanism may be associated with neurodegeneration in ALS.
Collapse
Affiliation(s)
- Noga Gershoni-Emek
- Sagol School of Neuroscience and Department of Physiology and Pharmacology, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Topaz Altman
- Sagol School of Neuroscience and Department of Physiology and Pharmacology, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Ariel Ionescu
- Sagol School of Neuroscience and Department of Physiology and Pharmacology, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | | | - Tal Gradus-Pery
- Sagol School of Neuroscience and Department of Physiology and Pharmacology, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Dianna E Willis
- Burke Neurological Institute, White Plains, NY, United States.,Brain & Mind Research Institute, Weill Cornell Medicine, New York, NY, United States
| | - Eran Perlson
- Sagol School of Neuroscience and Department of Physiology and Pharmacology, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
75
|
Smoak MM, Pearce HA, Mikos AG. Microfluidic devices for disease modeling in muscle tissue. Biomaterials 2018; 198:250-258. [PMID: 30193908 DOI: 10.1016/j.biomaterials.2018.08.059] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Revised: 07/30/2018] [Accepted: 08/27/2018] [Indexed: 12/19/2022]
Abstract
Microfluidic devices have advanced significantly in recent years and are a promising technology for the field of tissue engineering. Highly sophisticated microfabrication techniques have paved the way for the development of complex ex vivo models capable of incorporating and measuring the real-time response of multiple cell types interacting together in a single system. Muscle-on-a-chip technology has drastically improved and serves as a drug screening platform for many muscular diseases such as muscular dystrophy, tendinosis, fibromyalgia, mitochondrial myopathy, and myasthenia gravis. This review seeks to communicate the gaps in knowledge of current muscular disease models and highlight the power of microfluidic devices in enabling researchers to better understand disease pathology and provide high throughput screening of therapeutics for muscular myopathies.
Collapse
Affiliation(s)
- Mollie M Smoak
- Department of Bioengineering, Rice University, Houston, TX 77030, USA
| | - Hannah A Pearce
- Department of Bioengineering, Rice University, Houston, TX 77030, USA
| | - Antonios G Mikos
- Department of Bioengineering, Rice University, Houston, TX 77030, USA.
| |
Collapse
|
76
|
Tillmaand EG, Sweedler JV. Integrating Mass Spectrometry with Microphysiological Systems for Improved Neurochemical Studies. ACTA ACUST UNITED AC 2018; 2. [PMID: 30148282 DOI: 10.21037/mps.2018.05.01] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Microphysiological systems, often referred to as "organs-on-chips", are in vitro platforms designed to model the spatial, chemical, structural, and physiological elements of in vivo cellular environments. They enhance the evaluation of complex engineered biological systems and are a step between traditional cell culture and in vivo experimentation. As neurochemists and measurement scientists studying the molecules involved in intercellular communication in the nervous system, we focus here on recent advances in neuroscience using microneurological systems and their potential to interface with mass spectrometry. We discuss a number of examples - microfluidic devices, spheroid cultures, hydrogels, scaffolds, and fibers - highlighting those that would benefit from mass spectrometric technologies to obtain improved chemical information.
Collapse
Affiliation(s)
- Emily G Tillmaand
- Department of Chemistry, the Neuroscience Program and the Beckman Institute, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Jonathan V Sweedler
- Department of Chemistry, the Neuroscience Program and the Beckman Institute, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
| |
Collapse
|
77
|
Development of Microplatforms to Mimic the In Vivo Architecture of CNS and PNS Physiology and Their Diseases. Genes (Basel) 2018; 9:genes9060285. [PMID: 29882823 PMCID: PMC6027402 DOI: 10.3390/genes9060285] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Revised: 05/28/2018] [Accepted: 05/31/2018] [Indexed: 12/16/2022] Open
Abstract
Understanding the mechanisms that govern nervous tissues function remains a challenge. In vitro two-dimensional (2D) cell culture systems provide a simplistic platform to evaluate systematic investigations but often result in unreliable responses that cannot be translated to pathophysiological settings. Recently, microplatforms have emerged to provide a better approximation of the in vivo scenario with better control over the microenvironment, stimuli and structure. Advances in biomaterials enable the construction of three-dimensional (3D) scaffolds, which combined with microfabrication, allow enhanced biomimicry through precise control of the architecture, cell positioning, fluid flows and electrochemical stimuli. This manuscript reviews, compares and contrasts advances in nervous tissues-on-a-chip models and their applications in neural physiology and disease. Microplatforms used for neuro-glia interactions, neuromuscular junctions (NMJs), blood-brain barrier (BBB) and studies on brain cancer, metastasis and neurodegenerative diseases are addressed. Finally, we highlight challenges that can be addressed with interdisciplinary efforts to achieve a higher degree of biomimicry. Nervous tissue microplatforms provide a powerful tool that is destined to provide a better understanding of neural health and disease.
Collapse
|
78
|
Santhanam N, Kumanchik L, Guo X, Sommerhage F, Cai Y, Jackson M, Martin C, Saad G, McAleer CW, Wang Y, Lavado A, Long CJ, Hickman JJ. Stem cell derived phenotypic human neuromuscular junction model for dose response evaluation of therapeutics. Biomaterials 2018; 166:64-78. [PMID: 29547745 PMCID: PMC5866791 DOI: 10.1016/j.biomaterials.2018.02.047] [Citation(s) in RCA: 87] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Revised: 02/20/2018] [Accepted: 02/24/2018] [Indexed: 01/01/2023]
Abstract
There are currently no functional neuromuscular junction (hNMJ) systems composed of human cells that could be used for drug evaluations or toxicity testing in vitro. These systems are needed to evaluate NMJs for diseases such as amyotrophic lateral sclerosis, spinal muscular atrophy or other neurodegenerative diseases or injury states. There are certainly no model systems, animal or human, that allows for isolated treatment of motoneurons or muscle capable of generating dose response curves to evaluate pharmacological activity of these highly specialized functional units. A system was developed in which human myotubes and motoneurons derived from stem cells were cultured in a serum-free medium in a BioMEMS construct. The system is composed of two chambers linked by microtunnels to enable axonal outgrowth to the muscle chamber that allows separate stimulation of each component and physiological NMJ function and MN stimulated tetanus. The muscle's contractions, induced by motoneuron activation or direct electrical stimulation, were monitored by image subtraction video recording for both frequency and amplitude. Bungarotoxin, BOTOX® and curare dose response curves were generated to demonstrate pharmacological relevance of the phenotypic screening device. This quantifiable functional hNMJ system establishes a platform for generating patient-specific NMJ models by including patient-derived iPSCs.
Collapse
Affiliation(s)
- Navaneetha Santhanam
- Hybrid Systems Lab, NanoScience Technology Center, University of Central Florida, 12424 Research Parkway, Suite 400, Orlando, FL 32826, USA
| | - Lee Kumanchik
- Hybrid Systems Lab, NanoScience Technology Center, University of Central Florida, 12424 Research Parkway, Suite 400, Orlando, FL 32826, USA
| | - Xiufang Guo
- Hybrid Systems Lab, NanoScience Technology Center, University of Central Florida, 12424 Research Parkway, Suite 400, Orlando, FL 32826, USA
| | - Frank Sommerhage
- Hybrid Systems Lab, NanoScience Technology Center, University of Central Florida, 12424 Research Parkway, Suite 400, Orlando, FL 32826, USA
| | - Yunqing Cai
- Hybrid Systems Lab, NanoScience Technology Center, University of Central Florida, 12424 Research Parkway, Suite 400, Orlando, FL 32826, USA
| | - Max Jackson
- Hybrid Systems Lab, NanoScience Technology Center, University of Central Florida, 12424 Research Parkway, Suite 400, Orlando, FL 32826, USA
| | - Candace Martin
- Hybrid Systems Lab, NanoScience Technology Center, University of Central Florida, 12424 Research Parkway, Suite 400, Orlando, FL 32826, USA
| | - George Saad
- Hybrid Systems Lab, NanoScience Technology Center, University of Central Florida, 12424 Research Parkway, Suite 400, Orlando, FL 32826, USA
| | - Christopher W. McAleer
- Hybrid Systems Lab, NanoScience Technology Center, University of Central Florida, 12424 Research Parkway, Suite 400, Orlando, FL 32826, USA
| | - Ying Wang
- Hybrid Systems Lab, NanoScience Technology Center, University of Central Florida, 12424 Research Parkway, Suite 400, Orlando, FL 32826, USA,Department of Biomedical Engineering, 305 Weill Hall, Cornell University, Ithaca, NY, 14853, USA
| | - Andrea Lavado
- Hybrid Systems Lab, NanoScience Technology Center, University of Central Florida, 12424 Research Parkway, Suite 400, Orlando, FL 32826, USA
| | - Christopher J. Long
- Hybrid Systems Lab, NanoScience Technology Center, University of Central Florida, 12424 Research Parkway, Suite 400, Orlando, FL 32826, USA
| | - James J. Hickman
- Hybrid Systems Lab, NanoScience Technology Center, University of Central Florida, 12424 Research Parkway, Suite 400, Orlando, FL 32826, USA,correspondence:
| |
Collapse
|
79
|
miR126-5p Downregulation Facilitates Axon Degeneration and NMJ Disruption via a Non-Cell-Autonomous Mechanism in ALS. J Neurosci 2018; 38:5478-5494. [PMID: 29773756 PMCID: PMC6001038 DOI: 10.1523/jneurosci.3037-17.2018] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2017] [Revised: 04/15/2018] [Accepted: 04/23/2018] [Indexed: 01/06/2023] Open
Abstract
Axon degeneration and disruption of neuromuscular junctions (NMJs) are key events in amyotrophic lateral sclerosis (ALS) pathology. Although the disease's etiology is not fully understood, it is thought to involve a non-cell-autonomous mechanism and alterations in RNA metabolism. Here, we identified reduced levels of miR126-5p in presymptomatic ALS male mice models, and an increase in its targets: axon destabilizing Type 3 Semaphorins and their coreceptor Neuropilins. Using compartmentalized in vitro cocultures, we demonstrated that myocytes expressing diverse ALS-causing mutations promote axon degeneration and NMJ dysfunction, which were inhibited by applying Neuropilin1 blocking antibody. Finally, overexpressing miR126-5p is sufficient to transiently rescue axon degeneration and NMJ disruption both in vitro and in vivo Thus, we demonstrate a novel mechanism underlying ALS pathology, in which alterations in miR126-5p facilitate a non-cell-autonomous mechanism of motor neuron degeneration in ALS.SIGNIFICANCE STATEMENT Despite some progress, currently no effective treatment is available for amyotrophic lateral sclerosis (ALS). We suggest a novel regulatory role for miR126-5p in ALS and demonstrate, for the first time, a mechanism by which alterations in miR126-5p contribute to axon degeneration and NMJ disruption observed in ALS. We show that miR126-5p is altered in ALS models and that it can modulate Sema3 and NRP protein expression. Furthermore, NRP1 elevations in motor neurons and muscle secretion of Sema3A contribute to axon degeneration and NMJ disruption in ALS. Finally, overexpressing miR126-5p is sufficient to transiently rescue NMJ disruption and axon degeneration both in vitro and in vivo.
Collapse
|
80
|
Negro S, Lessi F, Duregotti E, Aretini P, La Ferla M, Franceschi S, Menicagli M, Bergamin E, Radice E, Thelen M, Megighian A, Pirazzini M, Mazzanti CM, Rigoni M, Montecucco C. CXCL12α/SDF-1 from perisynaptic Schwann cells promotes regeneration of injured motor axon terminals. EMBO Mol Med 2018; 9:1000-1010. [PMID: 28559442 PMCID: PMC5538331 DOI: 10.15252/emmm.201607257] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
The neuromuscular junction has retained through evolution the capacity to regenerate after damage, but little is known on the inter-cellular signals involved in its functional recovery from trauma, autoimmune attacks, or neurotoxins. We report here that CXCL12α, also abbreviated as stromal-derived factor-1 (SDF-1), is produced specifically by perisynaptic Schwann cells following motor axon terminal degeneration induced by α-latrotoxin. CXCL12α acts via binding to the neuronal CXCR4 receptor. A CXCL12α-neutralizing antibody or a specific CXCR4 inhibitor strongly delays recovery from motor neuron degeneration in vivo Recombinant CXCL12α in vivo accelerates neurotransmission rescue upon damage and very effectively stimulates the axon growth of spinal cord motor neurons in vitro These findings indicate that the CXCL12α-CXCR4 axis plays an important role in the regeneration of the neuromuscular junction after motor axon injury. The present results have important implications in the effort to find therapeutics and protocols to improve recovery of function after different forms of motor axon terminal damage.
Collapse
Affiliation(s)
- Samuele Negro
- Department of Biomedical Sciences, University of Padua, Padua, Italy
| | - Francesca Lessi
- Laboratory of Genomics, Pisa Science Foundation, Pisa, Italy
| | - Elisa Duregotti
- Department of Biomedical Sciences, University of Padua, Padua, Italy
| | - Paolo Aretini
- Laboratory of Genomics, Pisa Science Foundation, Pisa, Italy
| | - Marco La Ferla
- Laboratory of Genomics, Pisa Science Foundation, Pisa, Italy
| | - Sara Franceschi
- Laboratory of Genomics, Pisa Science Foundation, Pisa, Italy
| | | | - Elisanna Bergamin
- Department of Biomedical Sciences, University of Padua, Padua, Italy
| | - Egle Radice
- Institute for Research in Biomedicine, Università della Svizzera Italiana, Bellinzona, Switzerland
| | - Marcus Thelen
- Institute for Research in Biomedicine, Università della Svizzera Italiana, Bellinzona, Switzerland
| | - Aram Megighian
- Department of Biomedical Sciences, University of Padua, Padua, Italy
| | - Marco Pirazzini
- Department of Biomedical Sciences, University of Padua, Padua, Italy
| | | | - Michela Rigoni
- Department of Biomedical Sciences, University of Padua, Padua, Italy
| | - Cesare Montecucco
- Department of Biomedical Sciences, University of Padua, Padua, Italy .,CNR Institute of Neuroscience, Padua, Italy
| |
Collapse
|
81
|
Bucchia M, Merwin SJ, Re DB, Kariya S. Limitations and Challenges in Modeling Diseases Involving Spinal Motor Neuron Degeneration in Vitro. Front Cell Neurosci 2018; 12:61. [PMID: 29559895 PMCID: PMC5845677 DOI: 10.3389/fncel.2018.00061] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Accepted: 02/20/2018] [Indexed: 12/12/2022] Open
Abstract
Pathogenic conditions involving degeneration of spinal motor neurons (MNs), such as amyotrophic lateral sclerosis, sarcopenia, and spinal cord injury, mostly occur in individuals whose spinal MNs are fully mature. There is currently no effective treatment to prevent death or promote axonal regeneration of the spinal MNs affected in these patients. To increase our understanding and find a cure for such conditions, easily controllable and monitorable cell culture models allow for a better dissection of certain molecular and cellular events that cannot be teased apart in whole organism models. To date, various types of spinal MN cultures have been described. Yet these models are all based on the use of immature neurons or neurons uncharacterized for their degree of maturity after being isolated and cultured. Additionally, studying only MNs cannot give a comprehensive and complete view of the neurodegenerative processes usually involving other cell types. To date, there is no confirmed in vitro model faithfully emulating disease or injury of the mature spinal MNs. In this review, we summarize the different limitations of currently available culture models, and discuss the challenges that have to be overcome for developing more reliable and translational platforms for the in vitro study of spinal MN degeneration.
Collapse
Affiliation(s)
- Monica Bucchia
- Department of Neurology, Columbia University Medical Center, New York, NY, United States
| | - Samantha J Merwin
- Department of Environmental Health Sciences, Columbia University Medical Center, New York, NY, United States
| | - Diane B Re
- Department of Environmental Health Sciences, Columbia University Medical Center, New York, NY, United States
| | - Shingo Kariya
- Department of Neurology, Columbia University Medical Center, New York, NY, United States
| |
Collapse
|
82
|
Prox J, Smith T, Holl C, Chehade N, Guo L. Integrated biocircuits: engineering functional multicellular circuits and devices. J Neural Eng 2018; 15:023001. [DOI: 10.1088/1741-2552/aaa906] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
|
83
|
Affiliation(s)
- Lei Li
- Department of Neuroscience, School of Medicine, Case Western Reserve University, Cleveland, Ohio 44106, USA
| | - Wen-Cheng Xiong
- Department of Neuroscience, School of Medicine, Case Western Reserve University, Cleveland, Ohio 44106, USA
- Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, Ohio 44106, USA
| | - Lin Mei
- Department of Neuroscience, School of Medicine, Case Western Reserve University, Cleveland, Ohio 44106, USA
- Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, Ohio 44106, USA
| |
Collapse
|
84
|
Rothbauer M, Zirath H, Ertl P. Recent advances in microfluidic technologies for cell-to-cell interaction studies. LAB ON A CHIP 2018; 18:249-270. [PMID: 29143053 DOI: 10.1039/c7lc00815e] [Citation(s) in RCA: 193] [Impact Index Per Article: 27.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/12/2023]
Abstract
Microfluidic cell cultures are ideally positioned to become the next generation of in vitro diagnostic tools for biomedical research, where key biological processes such as cell signalling and dynamic cell-to-cell interactions can be reliably analysed under reproducible physiological cell culture conditions. In the last decade, a large number of microfluidic cell analysis systems have been developed for a variety of applications including drug target optimization, drug screening and toxicological testing. More recently, advanced in vitro microfluidic cell culture systems have emerged that are capable of replicating the complex three-dimensional architectures of tissues and organs and thus represent valid biological models for investigating the mechanism and function of human tissue structures, as well as studying the onset and progression of diseases such as cancer. In this review, we present the most important developments in single-cell, 2D and 3D microfluidic cell culture systems for studying cell-to-cell interactions published over the last 6 years, with a focus on cancer research and immunotherapy, vascular models and neuroscience. In addition, the current technological development of microdevices with more advanced physiological cell microenvironments that integrate multiple organ models, namely, the so-called body-, human- and multi-organ-on-a-chip, is reviewed.
Collapse
Affiliation(s)
- Mario Rothbauer
- Vienna University of Technology, Faculty of Technical Chemistry, Institute of Applied Synthetic Chemistry, Getreidemarkt 9, 1060 Vienna, Austria.
| | | | | |
Collapse
|
85
|
Structuring a multi-nodal neural network in vitro within a novel design microfluidic chip. Biomed Microdevices 2018; 20:9. [DOI: 10.1007/s10544-017-0254-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
86
|
Wang YI, Carmona C, Hickman JJ, Shuler ML. Multiorgan Microphysiological Systems for Drug Development: Strategies, Advances, and Challenges. Adv Healthc Mater 2018; 7:10.1002/adhm.201701000. [PMID: 29205920 PMCID: PMC5805562 DOI: 10.1002/adhm.201701000] [Citation(s) in RCA: 91] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Revised: 09/18/2017] [Indexed: 12/19/2022]
Abstract
Traditional cell culture and animal models utilized for preclinical drug screening have led to high attrition rates of drug candidates in clinical trials due to their low predictive power for human response. Alternative models using human cells to build in vitro biomimetics of the human body with physiologically relevant organ-organ interactions hold great potential to act as "human surrogates" and provide more accurate prediction of drug effects in humans. This review is a comprehensive investigation into the development of tissue-engineered human cell-based microscale multiorgan models, or multiorgan microphysiological systems for drug testing. The evolution from traditional models to macro- and microscale multiorgan systems is discussed in regards to the rationale for recent global efforts in multiorgan microphysiological systems. Current advances in integrating cell culture and on-chip analytical technologies, as well as proof-of-concept applications for these multiorgan microsystems are discussed. Major challenges for the field, such as reproducibility and physiological relevance, are discussed with comparisons of the strengths and weaknesses of various systems to solve these challenges. Conclusions focus on the current development stage of multiorgan microphysiological systems and new trends in the field.
Collapse
Affiliation(s)
- Ying I Wang
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Carlos Carmona
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway Suite 400, Orlando, FL 32826, USA
| | - James J Hickman
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway Suite 400, Orlando, FL 32826, USA
- Hesperos, Inc., 3259 Progress Dr, Room 158, Orlando, FL 32826
| | - Michael L Shuler
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA
- Hesperos, Inc., 3259 Progress Dr, Room 158, Orlando, FL 32826
- Robert Frederick Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, NY 14853, USA
| |
Collapse
|
87
|
Clarkson BDS, Patel MS, LaFrance-Corey RG, Howe CL. Retrograde interferon-gamma signaling induces major histocompatibility class I expression in human-induced pluripotent stem cell-derived neurons. Ann Clin Transl Neurol 2017; 5:172-185. [PMID: 29468178 PMCID: PMC5817842 DOI: 10.1002/acn3.516] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Revised: 11/28/2017] [Accepted: 11/30/2017] [Indexed: 02/06/2023] Open
Abstract
Objective Injury-associated axon-intrinsic signals are thought to underlie pathogenesis and progression in many neuroinflammatory and neurodegenerative diseases, including multiple sclerosis (MS). Retrograde interferon gamma (IFN γ) signals are known to induce expression of major histocompatibility class I (MHC I) genes in murine axons, thereby increasing the susceptibility of these axons to attack by antigen-specific CD8+ T cells. We sought to determine whether the same is true in human neurons. Methods A novel microisolation chamber design was used to physically isolate and manipulate axons from human skin fibroblast-derived induced pluripotent stem cell (iPSC)-derived neuron-enriched neural aggregates. Fluorescent retrobeads were used to assess the fraction of neurons with projections to the distal chamber. Axons were treated with IFN γ for 72 h and expression of MHC class I and antigen presentation genes were evaluated by RT-PCR and immunofluorescence. Results Human iPSC-derived neural stem cells maintained as 3D aggregate cultures in the cell body chamber of polymer microisolation chambers extended dense axonal projections into the fluidically isolated distal chamber. Treatment of these axons with IFN γ resulted in upregulation of MHC class I and antigen processing genes in the neuron cell bodies. IFN γ-induced MHC class I molecules were also anterogradely transported into the distal axon. Interpretation These results provide conclusive evidence that human axons are competent to express MHC class I molecules, suggesting that inflammatory factors enriched in demyelinated lesions may render axons vulnerable to attack by autoreactive CD8+ T cells in patients with MS. Future work will be aimed at identifying pathogenic anti-axonal T cells in these patients.
Collapse
Affiliation(s)
| | - Misha S Patel
- Department of Neurology Mayo Clinic Rochester Minnesota
| | | | - Charles L Howe
- Department of Neurology Mayo Clinic Rochester Minnesota.,Department of Neuroscience Mayo Clinic Rochester Minnesota.,Department of Immunology Mayo Clinic Rochester Minnesota.,Center for Multiple Sclerosis and Autoimmune Neurology Mayo Clinic Rochester Minnesota
| |
Collapse
|
88
|
Terenzio M, Schiavo G, Fainzilber M. Compartmentalized Signaling in Neurons: From Cell Biology to Neuroscience. Neuron 2017; 96:667-679. [PMID: 29096079 DOI: 10.1016/j.neuron.2017.10.015] [Citation(s) in RCA: 92] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Revised: 09/27/2017] [Accepted: 10/09/2017] [Indexed: 12/18/2022]
Abstract
Neurons are the largest known cells, with complex and highly polarized morphologies. As such, neuronal signaling is highly compartmentalized, requiring sophisticated transfer mechanisms to convey and integrate information within and between sub-neuronal compartments. Here, we survey different modes of compartmentalized signaling in neurons, highlighting examples wherein the fundamental cell biological processes of protein synthesis and degradation, membrane trafficking, and organelle transport are employed to enable the encoding and integration of information, locally and globally within a neuron. Comparisons to other cell types indicate that neurons accentuate widely shared mechanisms, providing invaluable models for the compartmentalization and transfer mechanisms required and used by most eukaryotic cells.
Collapse
Affiliation(s)
- Marco Terenzio
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Giampietro Schiavo
- Sobell Department of Motor Neuroscience and Movement Disorders, Institute of Neurology, University College London, London WC1N 3BG, UK; Discoveries Centre for Regenerative and Precision Medicine at UCL, London WC1N 3BG, UK; UK Dementia Research Institute at UCL, London WC1E 6BT, UK
| | - Mike Fainzilber
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot 76100, Israel.
| |
Collapse
|
89
|
Mills R, Taylor-Weiner H, Correia JC, Agudelo LZ, Allodi I, Kolonelou C, Martinez-Redondo V, Ferreira DMS, Nichterwitz S, Comley LH, Lundin V, Hedlund E, Ruas JL, Teixeira AI. Neurturin is a PGC-1α1-controlled myokine that promotes motor neuron recruitment and neuromuscular junction formation. Mol Metab 2017; 7:12-22. [PMID: 29157948 PMCID: PMC5784328 DOI: 10.1016/j.molmet.2017.11.001] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Revised: 10/31/2017] [Accepted: 11/01/2017] [Indexed: 02/07/2023] Open
Abstract
OBJECTIVE We examined whether skeletal muscle overexpression of PGC-1α1 or PGC-1α4 affected myokine secretion and neuromuscular junction (NMJ) formation. METHODS A microfluidic device was used to model endocrine signaling and NMJ formation between primary mouse myoblast-derived myotubes and embryonic stem cell-derived motor neurons. Differences in hydrostatic pressure allowed for fluidic isolation of either cell type or unidirectional signaling in the fluid phase. Myotubes were transduced to overexpress PGC-1α1 or PGC-1α4, and myokine secretion was quantified using a proximity extension assay. Morphological and functional changes in NMJs were measured by fluorescent microscopy and by monitoring muscle contraction upon motor neuron stimulation. RESULTS Skeletal muscle transduction with PGC-1α1, but not PGC-1α4, increased NMJ formation and size. PGC-1α1 increased muscle secretion of neurturin, which was sufficient and necessary for the effects of muscle PGC-1α1 on NMJ formation. CONCLUSIONS Our findings indicate that neurturin is a mediator of PGC-1α1-dependent retrograde signaling from muscle to motor neurons.
Collapse
Affiliation(s)
- Richard Mills
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Scheeles väg 2, 171 77, Stockholm, Sweden
| | - Hermes Taylor-Weiner
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Scheeles väg 2, 171 77, Stockholm, Sweden
| | - Jorge C Correia
- Molecular and Cellular Exercise Physiology, Department of Physiology and Pharmacology, Karolinska Institutet, von Eulers väg 8, 171 77, Stockholm, Sweden
| | - Leandro Z Agudelo
- Molecular and Cellular Exercise Physiology, Department of Physiology and Pharmacology, Karolinska Institutet, von Eulers väg 8, 171 77, Stockholm, Sweden
| | - Ilary Allodi
- Department of Neuroscience, Karolinska Institutet, Retzius väg 8, 17177, Stockholm, Sweden
| | - Christina Kolonelou
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Scheeles väg 2, 171 77, Stockholm, Sweden
| | - Vicente Martinez-Redondo
- Molecular and Cellular Exercise Physiology, Department of Physiology and Pharmacology, Karolinska Institutet, von Eulers väg 8, 171 77, Stockholm, Sweden
| | - Duarte M S Ferreira
- Molecular and Cellular Exercise Physiology, Department of Physiology and Pharmacology, Karolinska Institutet, von Eulers väg 8, 171 77, Stockholm, Sweden
| | - Susanne Nichterwitz
- Department of Neuroscience, Karolinska Institutet, Retzius väg 8, 17177, Stockholm, Sweden
| | - Laura H Comley
- Department of Neuroscience, Karolinska Institutet, Retzius väg 8, 17177, Stockholm, Sweden
| | - Vanessa Lundin
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Scheeles väg 2, 171 77, Stockholm, Sweden
| | - Eva Hedlund
- Department of Neuroscience, Karolinska Institutet, Retzius väg 8, 17177, Stockholm, Sweden
| | - Jorge L Ruas
- Molecular and Cellular Exercise Physiology, Department of Physiology and Pharmacology, Karolinska Institutet, von Eulers väg 8, 171 77, Stockholm, Sweden.
| | - Ana I Teixeira
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Scheeles väg 2, 171 77, Stockholm, Sweden.
| |
Collapse
|
90
|
Black BJ, Atmaramani R, Pancrazio JJ. Spontaneous and Evoked Activity from Murine Ventral Horn Cultures on Microelectrode Arrays. Front Cell Neurosci 2017; 11:304. [PMID: 29033792 PMCID: PMC5626830 DOI: 10.3389/fncel.2017.00304] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Accepted: 09/12/2017] [Indexed: 12/11/2022] Open
Abstract
Motor neurons are the site of action for several neurological disorders and paralytic toxins, with cell bodies located in the ventral horn (VH) of the spinal cord along with interneurons and support cells. Microelectrode arrays (MEAs) have emerged as a high content assay platform for mechanistic studies and drug discovery. Here, we explored the spontaneous and evoked electrical activity of VH cultures derived from embryonic mouse spinal cord on multi-well plates of MEAs. Primary VH cultures from embryonic day 15–16 mice were characterized by expression of choline acetyltransferase (ChAT) by immunocytochemistry. Well resolved, all-or-nothing spontaneous spikes with profiles consistent with extracellular action potentials were observed after 3 days in vitro, persisting with consistent firing rates until at least day in vitro 19. The majority of the spontaneous activity consisted of tonic firing interspersed with coordinated bursting across the network. After 5 days in vitro, spike activity was readily evoked by voltage pulses where a minimum amplitude and duration required for excitation was 300 mV and 100 μs/phase, respectively. We characterized the sensitivity of spontaneous and evoked activity to a host of pharmacological agents including AP5, CNQX, strychnine, ω-agatoxin IVA, and botulinum neurotoxin serotype A (BoNT/A). These experiments revealed sensitivity of the cultured VH to both agonist and antagonist compounds in a manner consistent with mature tissue derived from slices. In the case of BoNT/A, we also demonstrated intoxication persistence over an 18-day period, followed by partial intoxication recovery induced by N- and P/Q-type calcium channel agonist GV-58. In total, our findings suggest that VH cultures on multi-well MEA plates may represent a moderate throughput, high content assay for performing mechanistic studies and for screening potential therapeutics pertaining to paralytic toxins and neurological disorders.
Collapse
Affiliation(s)
- Bryan J Black
- Neuronal Networks and Interfaces Laboratory, Department of Bioengineering, The University of Texas at Dallas, Richardson, TX, United States
| | - Rahul Atmaramani
- Neuronal Networks and Interfaces Laboratory, Department of Bioengineering, The University of Texas at Dallas, Richardson, TX, United States
| | - Joseph J Pancrazio
- Neuronal Networks and Interfaces Laboratory, Department of Bioengineering, The University of Texas at Dallas, Richardson, TX, United States
| |
Collapse
|
91
|
Mis K, Grubic Z, Lorenzon P, Sciancalepore M, Mars T, Pirkmajer S. In Vitro Innervation as an Experimental Model to Study the Expression and Functions of Acetylcholinesterase and Agrin in Human Skeletal Muscle. Molecules 2017; 22:molecules22091418. [PMID: 28846617 PMCID: PMC6151842 DOI: 10.3390/molecules22091418] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Revised: 08/18/2017] [Accepted: 08/23/2017] [Indexed: 12/19/2022] Open
Abstract
Acetylcholinesterase (AChE) and agrin, a heparan-sulfate proteoglycan, reside in the basal lamina of the neuromuscular junction (NMJ) and play key roles in cholinergic transmission and synaptogenesis. Unlike most NMJ components, AChE and agrin are expressed in skeletal muscle and α-motor neurons. AChE and agrin are also expressed in various other types of cells, where they have important alternative functions that are not related to their classical roles in NMJ. In this review, we first focus on co-cultures of embryonic rat spinal cord explants with human skeletal muscle cells as an experimental model to study functional innervation in vitro. We describe how this heterologous rat-human model, which enables experimentation on highly developed contracting human myotubes, offers unique opportunities for AChE and agrin research. We then highlight innovative approaches that were used to address salient questions regarding expression and alternative functions of AChE and agrin in developing human skeletal muscle. Results obtained in co-cultures are compared with those obtained in other models in the context of general advances in the field of AChE and agrin neurobiology.
Collapse
Affiliation(s)
- Katarina Mis
- Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Zaloška 4, SI-1000 Ljubljana, Slovenia.
| | - Zoran Grubic
- Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Zaloška 4, SI-1000 Ljubljana, Slovenia.
| | - Paola Lorenzon
- Department of Life Sciences, University of Trieste, via A. Fleming 22, I-34127 Trieste, Italy.
| | - Marina Sciancalepore
- Department of Life Sciences, University of Trieste, via A. Fleming 22, I-34127 Trieste, Italy.
| | - Tomaz Mars
- Department of Life Sciences, University of Trieste, via A. Fleming 22, I-34127 Trieste, Italy.
| | - Sergej Pirkmajer
- Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Zaloška 4, SI-1000 Ljubljana, Slovenia.
| |
Collapse
|
92
|
Park S, Choi Y, Kwak G, Hong YB, Jung N, Kim J, Choi BO, Jung SC. Application of differentiated human tonsil-derived stem cells to trembler-J mice. Muscle Nerve 2017; 57:478-486. [PMID: 28796340 DOI: 10.1002/mus.25763] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Revised: 08/03/2017] [Accepted: 08/05/2017] [Indexed: 12/13/2022]
Abstract
INTRODUCTION Mesenchymal stem cells (MSCs) can differentiate into various cell types. METHODS In this study we investigated the potential of human tonsil-derived MSCs (T-MSCs) for neuromuscular regeneration in trembler-J (Tr-J) mice, a model for Charcot-Marie-Tooth disease type 1A (CMT1A). RESULTS T-MSCs differentiated toward skeletal myocytes with increased expression of skeletal muscle-related markers (including troponin I type 1, and myogenin), and the formation of myotubes in vitro. In-situ transplantation of T-MSC-derived myocytes (T-MSC myocytes) into the gastrocnemius muscle in Tr-J mice enhanced motor function, with recovery of compound muscle action potential amplitudes. Morphology of the sciatic nerve and skeletal muscle recovered without the formation of teratomas, and the expression levels of nerve growth factor and glial-cell-line-derived neurotrophic factor were increased significantly in T-MSC myocytes compared with T-MSCs in vitro. DISCUSSION Transplantation of T-MSC myocytes could enable neuromuscular regeneration in patients with CMT1A. Muscle Nerve 57: 478-486, 2018.
Collapse
Affiliation(s)
- Saeyoung Park
- Department of Biochemistry, College of Medicine, Ewha Womans University, 1071 Anyangcheon-Ro, Yangcheon-Gu, Seoul, 07985, Republic of Korea
| | - Yoonyoung Choi
- Department of Biochemistry, College of Medicine, Ewha Womans University, 1071 Anyangcheon-Ro, Yangcheon-Gu, Seoul, 07985, Republic of Korea
| | - Geon Kwak
- Department of Health Sciences and Technology, Sungkyunkwan University, Seoul, Republic of Korea
| | - Young Bin Hong
- Stem Cell & Regenerative Medicine Institute, Samsung Medical Center, Seoul, Republic of Korea
| | - Namhee Jung
- Department of Biochemistry, College of Medicine, Ewha Womans University, 1071 Anyangcheon-Ro, Yangcheon-Gu, Seoul, 07985, Republic of Korea
| | - Jieun Kim
- Department of Biochemistry, College of Medicine, Ewha Womans University, 1071 Anyangcheon-Ro, Yangcheon-Gu, Seoul, 07985, Republic of Korea
| | - Byung-Ok Choi
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Sung-Chul Jung
- Department of Biochemistry, College of Medicine, Ewha Womans University, 1071 Anyangcheon-Ro, Yangcheon-Gu, Seoul, 07985, Republic of Korea
| |
Collapse
|
93
|
Cortés D, Carballo-Molina OA, Castellanos-Montiel MJ, Velasco I. The Non-Survival Effects of Glial Cell Line-Derived Neurotrophic Factor on Neural Cells. Front Mol Neurosci 2017; 10:258. [PMID: 28878618 PMCID: PMC5572274 DOI: 10.3389/fnmol.2017.00258] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2017] [Accepted: 07/31/2017] [Indexed: 01/23/2023] Open
Abstract
Glial cell line-derived neurotrophic factor (GDNF) was first characterized as a survival-promoting molecule for dopaminergic neurons (DANs). Afterwards, other cells were also discovered to respond to GDNF not only as a survival factor but also as a protein supporting other cellular functions, such as proliferation, differentiation, maturation, neurite outgrowth and other phenomena that have been less studied than survival and are now more extendedly described here in this review article. During development, GDNF favors the commitment of neural precursors towards dopaminergic, motor, enteric and adrenal neurons; in addition, it enhances the axonal growth of some of these neurons. GDNF also induces the acquisition of a dopaminergic phenotype by increasing the expression of Tyrosine Hydroxylase (TH), Nurr1 and other proteins that confer this identity and promote further dendritic and electrical maturation. In motor neurons (MNs), GDNF not only promotes proliferation and maturation but also participates in regenerating damaged axons and modulates the neuromuscular junction (NMJ) at both presynaptic and postsynaptic levels. Moreover, GDNF modulates the rate of neuroblastoma (NB) and glioblastoma cancer cell proliferation. Additionally, the presence or absence of GDNF has been correlated with conditions such as depression, pain, muscular soreness, etc. Although, the precise role of GDNF is unknown, it extends beyond a survival effect. The understanding of the complete range of properties of this trophic molecule will allow us to investigate its broad mechanisms of action to accelerate and/or improve therapies for the aforementioned pathological conditions.
Collapse
Affiliation(s)
- Daniel Cortés
- Instituto de Fisiología Celular—Neurociencias, Universidad Nacional Autónoma de MéxicoMéxico City, Mexico
- Laboratorio de Reprogramación Celular del IFC-UNAM, Instituto Nacional de Neurología y NeurologíaMéxico City, Mexico
| | - Oscar A. Carballo-Molina
- Instituto de Fisiología Celular—Neurociencias, Universidad Nacional Autónoma de MéxicoMéxico City, Mexico
- Laboratorio de Reprogramación Celular del IFC-UNAM, Instituto Nacional de Neurología y NeurologíaMéxico City, Mexico
| | - María José Castellanos-Montiel
- Instituto de Fisiología Celular—Neurociencias, Universidad Nacional Autónoma de MéxicoMéxico City, Mexico
- Laboratorio de Reprogramación Celular del IFC-UNAM, Instituto Nacional de Neurología y NeurologíaMéxico City, Mexico
| | - Iván Velasco
- Instituto de Fisiología Celular—Neurociencias, Universidad Nacional Autónoma de MéxicoMéxico City, Mexico
- Laboratorio de Reprogramación Celular del IFC-UNAM, Instituto Nacional de Neurología y NeurologíaMéxico City, Mexico
| |
Collapse
|
94
|
Compartmentalized Microfluidic Platforms: The Unrivaled Breakthrough of In Vitro Tools for Neurobiological Research. J Neurosci 2017; 36:11573-11584. [PMID: 27852766 DOI: 10.1523/jneurosci.1748-16.2016] [Citation(s) in RCA: 88] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Revised: 09/08/2016] [Accepted: 09/28/2016] [Indexed: 12/15/2022] Open
Abstract
Microfluidic technology has become a valuable tool to the scientific community, allowing researchers to study fine cellular mechanisms with higher variable control compared with conventional systems. It has evolved tremendously, and its applicability and flexibility made its usage grow exponentially and transversely to several research fields. This has been particularly noticeable in neuroscience research, where microfluidic platforms made it possible to address specific questions extending from axonal guidance, synapse formation, or axonal transport to the development of 3D models of the CNS to allow pharmacological testing and drug screening. Furthermore, the continuous upgrade of microfluidic platforms has allowed a deeper study of the communication occurring between different neuronal and glial cells or between neurons and other peripheral tissues, both in physiological and pathological conditions. Importantly, the evolution of microfluidic technology has always been accompanied by the development of new computational tools addressing data acquisition, analysis, and modeling.
Collapse
|
95
|
Haring AP, Sontheimer H, Johnson BN. Microphysiological Human Brain and Neural Systems-on-a-Chip: Potential Alternatives to Small Animal Models and Emerging Platforms for Drug Discovery and Personalized Medicine. Stem Cell Rev Rep 2017; 13:381-406. [PMID: 28488234 PMCID: PMC5534264 DOI: 10.1007/s12015-017-9738-0] [Citation(s) in RCA: 86] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Translational challenges associated with reductionist modeling approaches, as well as ethical concerns and economic implications of small animal testing, drive the need for developing microphysiological neural systems for modeling human neurological diseases, disorders, and injuries. Here, we provide a comprehensive review of microphysiological brain and neural systems-on-a-chip (NSCs) for modeling higher order trajectories in the human nervous system. Societal, economic, and national security impacts of neurological diseases, disorders, and injuries are highlighted to identify critical NSC application spaces. Hierarchical design and manufacturing of NSCs are discussed with distinction for surface- and bulk-based systems. Three broad NSC classes are identified and reviewed: microfluidic NSCs, compartmentalized NSCs, and hydrogel NSCs. Emerging areas and future directions are highlighted, including the application of 3D printing to design and manufacturing of next-generation NSCs, the use of stem cells for constructing patient-specific NSCs, and the application of human NSCs to 'personalized neurology'. Technical hurdles and remaining challenges are discussed. This review identifies the state-of-the-art design methodologies, manufacturing approaches, and performance capabilities of NSCs. This work suggests NSCs appear poised to revolutionize the modeling of human neurological diseases, disorders, and injuries.
Collapse
Affiliation(s)
- Alexander P Haring
- Department of Industrial and Systems Engineering, Virginia Tech, Blacksburg, VA, 24061, USA
- Macromolecules Innovation Institute, Virginia Tech, Blacksburg, VA, 24061, USA
| | - Harald Sontheimer
- Glial Biology in Health, Disease, and Cancer Center, Virginia Tech Carilion Research Institute, Roanoke, VA, 24016, USA
- School of Neuroscience, Virginia Tech, Blacksburg, VA, 24061, USA
| | - Blake N Johnson
- Department of Industrial and Systems Engineering, Virginia Tech, Blacksburg, VA, 24061, USA.
- Macromolecules Innovation Institute, Virginia Tech, Blacksburg, VA, 24061, USA.
- School of Neuroscience, Virginia Tech, Blacksburg, VA, 24061, USA.
- Department of Materials Science and Engineering, Virginia Tech, Blacksburg, VA, 24061, USA.
| |
Collapse
|
96
|
Zahavi EE, Maimon R, Perlson E. Spatial-specific functions in retrograde neuronal signalling. Traffic 2017; 18:415-424. [DOI: 10.1111/tra.12487] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Revised: 03/16/2017] [Accepted: 04/05/2017] [Indexed: 12/15/2022]
Affiliation(s)
- Eitan Erez Zahavi
- Department of Physiology and Pharmacology; Sackler Faculty of Medicine; Tel Aviv University; Tel Aviv Israel
| | - Roy Maimon
- Department of Physiology and Pharmacology; Sackler Faculty of Medicine; Tel Aviv University; Tel Aviv Israel
| | - Eran Perlson
- Department of Physiology and Pharmacology; Sackler Faculty of Medicine; Tel Aviv University; Tel Aviv Israel
| |
Collapse
|
97
|
Rotem N, Magen I, Ionescu A, Gershoni-Emek N, Altman T, Costa CJ, Gradus T, Pasmanik-Chor M, Willis DE, Ben-Dov IZ, Hornstein E, Perlson E. ALS Along the Axons - Expression of Coding and Noncoding RNA Differs in Axons of ALS models. Sci Rep 2017; 7:44500. [PMID: 28300211 PMCID: PMC5353576 DOI: 10.1038/srep44500] [Citation(s) in RCA: 76] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Accepted: 02/08/2017] [Indexed: 12/18/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a multifactorial lethal motor neuron disease with no known treatment. Although the basic mechanism of its degenerative pathogenesis remains poorly understood, a subcellular spatial alteration in RNA metabolism is thought to play a key role. The nature of these RNAs remains elusive, and a comprehensive characterization of the axonal RNAs involved in maintaining neuronal health has yet to be described. Here, using cultured spinal cord (SC) neurons grown using a compartmented platform followed by next-generation sequencing (NGS) technology, we find that RNA expression differs between the somatic and axonal compartments of the neuron, for both mRNA and microRNA (miRNA). Further, the introduction of SOD1G93A and TDP43A315T, established ALS-related mutations, changed the subcellular expression and localization of RNAs within the neurons, showing a spatial specificity to either the soma or the axon. Altogether, we provide here the first combined inclusive profile of mRNA and miRNA expression in two ALS models at the subcellular level. These data provide an important resource for studies on the roles of local protein synthesis and axon degeneration in ALS and can serve as a possible target pool for ALS treatment.
Collapse
Affiliation(s)
- Nimrod Rotem
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, and the Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 69978, Israel
| | - Iddo Magen
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Ariel Ionescu
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, and the Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 69978, Israel
| | - Noga Gershoni-Emek
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, and the Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 69978, Israel
| | - Topaz Altman
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, and the Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 69978, Israel
| | - Christopher J Costa
- Brain and Mind Research Institute, Weill Cornell Medicine, Burke Medical Research Institute, White Plains, NY, USA
| | - Tal Gradus
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, and the Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 69978, Israel
| | | | - Dianna E Willis
- Brain and Mind Research Institute, Weill Cornell Medicine, Burke Medical Research Institute, White Plains, NY, USA
| | - Iddo Z Ben-Dov
- Department of Nephrology and Hypertension, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Eran Hornstein
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Eran Perlson
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, and the Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 69978, Israel
| |
Collapse
|
98
|
Ibáñez CF, Andressoo JO. Biology of GDNF and its receptors — Relevance for disorders of the central nervous system. Neurobiol Dis 2017; 97:80-89. [DOI: 10.1016/j.nbd.2016.01.021] [Citation(s) in RCA: 165] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Revised: 01/14/2016] [Accepted: 01/25/2016] [Indexed: 01/15/2023] Open
|
99
|
Labroo P, Shea J, Sant H, Gale B, Agarwal J. Effect Of combining FK506 and neurotrophins on neurite branching and elongation. Muscle Nerve 2016; 55:570-581. [PMID: 27503321 DOI: 10.1002/mus.25370] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Revised: 07/28/2016] [Accepted: 08/05/2016] [Indexed: 12/21/2022]
Abstract
INTRODUCTION There is a clinical need to improve the outcomes of peripheral nerve regeneration and repair after injury. In addition to its immunosuppressive effects, FK506 (tacrolimus) has been shown to have neuroregenerative properties. To determine biologically relevant local FK506 and growth factor concentrations, we performed an in vitro bioassay using dorsal root ganglion (DRG) from chicken embryos. METHODS Neurite elongation and neurite branching were analyzed microscopically after addition of FK506, glial cell line-derived neurotrophic factor (GDNF), and nerve growth factor (NGF), each alone and in combination. RESULTS FK506 induced modest neurite elongation (∼500-800 µm) without improving neurite branching significantly. The combination of FK506 with NGF, GDNF, or both, exerted a potentiating or competitive effect on neurite elongation (∼700-1100 µm) based on dosage and competitive effect on neurite branching (∼0.2-0.4). CONCLUSIONS These results strongly suggest that the interaction of FK506 with GDNF and NGF mediates distinct enhancement of neurite growth. Muscle Nerve 55: 570-581, 2017.
Collapse
Affiliation(s)
- Pratima Labroo
- Department of Mechanical Engineering, University of Utah, Salt Lake City, Utah, USA
| | - Jill Shea
- Department of Surgery, University of Utah, 30 N 1900 E, 3b400, Salt Lake City, Utah, 84132, USA
| | - Himanshu Sant
- Department of Mechanical Engineering, University of Utah, Salt Lake City, Utah, USA
| | - Bruce Gale
- Department of Mechanical Engineering, University of Utah, Salt Lake City, Utah, USA
| | - Jayant Agarwal
- Department of Surgery, University of Utah, 30 N 1900 E, 3b400, Salt Lake City, Utah, 84132, USA
| |
Collapse
|
100
|
On-chip, multisite extracellular and intracellular recordings from primary cultured skeletal myotubes. Sci Rep 2016; 6:36498. [PMID: 27812002 PMCID: PMC5095645 DOI: 10.1038/srep36498] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2016] [Accepted: 10/17/2016] [Indexed: 02/07/2023] Open
Abstract
In contrast to the extensive use of microelectrode array (MEA) technology in electrophysiological studies of cultured neurons and cardiac muscles, the vast field of skeletal muscle research has yet to adopt the technology. Here we demonstrate an empowering MEA technology for high quality, multisite, long-term electrophysiological recordings from cultured skeletal myotubes. Individual rat skeletal myotubes cultured on micrometer sized gold mushroom-shaped microelectrode (gMμE) based MEA tightly engulf the gMμEs, forming a high seal resistance between the myotubes and the gMμEs. As a consequence, spontaneous action potentials generated by the contracting myotubes are recorded as extracellular field potentials with amplitudes of up to 10 mV for over 14 days. Application of a 10 ms, 0.5-0.9 V voltage pulse through the gMμEs electroporated the myotube membrane, and transiently converted the extracellular to intracellular recording mode for 10-30 min. In a fraction of the cultures stable attenuated intracellular recordings were spontaneously produced. In these cases or after electroporation, subthreshold spontaneous potentials were also recorded. The introduction of the gMμE-MEA as a simple-to-use, high-quality electrophysiological tool together with the progress made in the use of cultured human myotubes opens up new venues for basic and clinical skeletal muscle research, preclinical drug screening, and personalized medicine.
Collapse
|