1
|
Badu-Mensah A, Guo X, Mendez R, Parsaud H, Hickman JJ. The Effect of Skeletal Muscle-Specific Creatine Treatment on ALS NMJ Integrity and Function. Int J Mol Sci 2023; 24:13519. [PMID: 37686322 PMCID: PMC10487911 DOI: 10.3390/ijms241713519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 08/16/2023] [Accepted: 08/19/2023] [Indexed: 09/10/2023] Open
Abstract
Although skeletal muscle (hSKM) has been proven to be actively involved in Amyotrophic Lateral Sclerosis (ALS) neuromuscular junction (NMJ) dysfunction, it is rarely considered as a pharmacological target in preclinical drug discovery. This project investigated how improving ALS hSKM viability and function effects NMJ integrity. Phenotypic ALS NMJ human-on-a-chip models developed from patient-derived induced pluripotent stem cells (iPSCs) were used to study the effect of hSKM-specific creatine treatment on clinically relevant functional ALS NMJ parameters, such as NMJ numbers, fidelity, stability, and fatigue index. Results indicated comparatively enhanced NMJ numbers, fidelity, and stability, as well as reduced fatigue index, across all hSKM-specific creatine-treated systems. Immunocytochemical analysis of the NMJs also revealed improved post-synaptic nicotinic Acetylcholine receptor (AChR) clustering and cluster size in systems supplemented with creatine relative to the un-dosed control. This work strongly suggests hSKM as a therapeutic target in ALS drug discovery. It also demonstrates the need to consider all tissues involved in multi-systemic diseases, such as ALS, in drug discovery efforts. Finally, this work further establishes the BioMEMs NMJ platform as an effective means of performing mutation-specific drug screening, which is a step towards personalized medicine for rare diseases.
Collapse
Affiliation(s)
- Agnes Badu-Mensah
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway, Suite 400, Orlando, FL 32826, USA; (A.B.-M.); (X.G.); (R.M.); (H.P.)
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32816, USA
| | - Xiufang Guo
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway, Suite 400, Orlando, FL 32826, USA; (A.B.-M.); (X.G.); (R.M.); (H.P.)
| | - Roxana Mendez
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway, Suite 400, Orlando, FL 32826, USA; (A.B.-M.); (X.G.); (R.M.); (H.P.)
| | - Hemant Parsaud
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway, Suite 400, Orlando, FL 32826, USA; (A.B.-M.); (X.G.); (R.M.); (H.P.)
| | - James J. Hickman
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway, Suite 400, Orlando, FL 32826, USA; (A.B.-M.); (X.G.); (R.M.); (H.P.)
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32816, USA
| |
Collapse
|
2
|
Jangir H, Hickman JJ. Mimicking the Tendon Microenvironment to Enhance Skeletal Muscle Adhesion and Longevity in a Functional Microcantilever Platform. ACS Biomater Sci Eng 2023; 9:4698-4708. [PMID: 37462389 PMCID: PMC10430766 DOI: 10.1021/acsbiomaterials.3c00235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Accepted: 06/23/2023] [Indexed: 08/15/2023]
Abstract
Microcantilever platforms are functional models for studying skeletal muscle force dynamics in vitro. However, the contractile force generated by the myotubes can cause them to detach from the cantilevers, especially during long-term experiments, thus impeding the chronic investigations of skeletal muscles for drug efficacy and toxicity. To improve the integration of myotubes with microcantilevers, we drew inspiration from the elastomeric proteins, elastin and resilin, that are present in the animal and insect worlds, respectively. The spring action of these proteins plays a critical role in force dampening in vivo. In animals, elastin is present in the collagenous matrix of the tendon which is the attachment point of muscles to bones. The tendon microenvironment consists of elastin, collagen, and an aqueous jelly-like mass of proteoglycans. In an attempt to mimic this tendon microenvironment, elastin, collagen, heparan sulfate proteoglycan, and hyaluronic acid were deposited on a positively charged silane substrate. This enabled the long-term survival of mechanically active myotubes on glass and silicon microcantilevers for over 28 days. The skeletal muscle cultures were derived from both primary and induced pluripotent stem cell (iPSC)-derived human skeletal muscles. Both types of myoblasts formed myotubes which survived for five weeks. Primary skeletal muscles and iPSC-derived skeletal muscles also showed a similar trend in fatigue index values. Upon integration with the microcantilever system, the primary muscle and iPSC-derived myotubes were tested successively over a one month period, thus paving the way for long-term chronic experiments on these systems for both drug efficacy and toxicity studies.
Collapse
Affiliation(s)
- Himanshi Jangir
- Nanoscience Technology Center, University of Central Florida, 12424 Research Pkwy, Orlando, Florida 32826, United States
| | - James J. Hickman
- Nanoscience Technology Center, University of Central Florida, 12424 Research Pkwy, Orlando, Florida 32826, United States
| |
Collapse
|
3
|
Vesga-Castro C, Aldazabal J, Vallejo-Illarramendi A, Paredes J. Contractile force assessment methods for in vitro skeletal muscle tissues. eLife 2022; 11:e77204. [PMID: 35604384 PMCID: PMC9126583 DOI: 10.7554/elife.77204] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Accepted: 04/27/2022] [Indexed: 02/06/2023] Open
Abstract
Over the last few years, there has been growing interest in measuring the contractile force (CF) of engineered muscle tissues to evaluate their functionality. However, there are still no standards available for selecting the most suitable experimental platform, measuring system, culture protocol, or stimulation patterns. Consequently, the high variability of published data hinders any comparison between different studies. We have identified that cantilever deflection, post deflection, and force transducers are the most commonly used configurations for CF assessment in 2D and 3D models. Additionally, we have discussed the most relevant emerging technologies that would greatly complement CF evaluation with intracellular and localized analysis. This review provides a comprehensive analysis of the most significant advances in CF evaluation and its critical parameters. In order to compare contractile performance across experimental platforms, we have used the specific force (sF, kN/m2), CF normalized to the calculated cross-sectional area (CSA). However, this parameter presents a high variability throughout the different studies, which indicates the need to identify additional parameters and complementary analysis suitable for proper comparison. We propose that future contractility studies in skeletal muscle constructs report detailed information about construct size, contractile area, maturity level, sarcomere length, and, ideally, the tetanus-to-twitch ratio. These studies will hopefully shed light on the relative impact of these variables on muscle force performance of engineered muscle constructs. Prospective advances in muscle tissue engineering, particularly in muscle disease models, will require a joint effort to develop standardized methodologies for assessing CF of engineered muscle tissues.
Collapse
Affiliation(s)
- Camila Vesga-Castro
- University of Navarra, Tecnun School of Engineering, Manuel de LardizábalSan SebastianSpain
- University of Navarra, Biomedical Engineering Center, Campus UniversitarioPamplonaSpain
- Group of Neurosciences, Department of Pediatrics, UPV/EHU, Hospital Donostia - IIS BiodonostiaSan SebastianSpain
| | - Javier Aldazabal
- University of Navarra, Tecnun School of Engineering, Manuel de LardizábalSan SebastianSpain
- University of Navarra, Biomedical Engineering Center, Campus UniversitarioPamplonaSpain
| | - Ainara Vallejo-Illarramendi
- Group of Neurosciences, Department of Pediatrics, UPV/EHU, Hospital Donostia - IIS BiodonostiaSan SebastianSpain
- CIBERNED, Instituto de Salud Carlos III, Ministry of Science, Innovation, and UniversitiesMadridSpain
| | - Jacobo Paredes
- University of Navarra, Tecnun School of Engineering, Manuel de LardizábalSan SebastianSpain
- University of Navarra, Biomedical Engineering Center, Campus UniversitarioPamplonaSpain
| |
Collapse
|
4
|
Badu-Mensah A, Guo X, McAleer CW, Rumsey JW, Hickman JJ. Functional skeletal muscle model derived from SOD1-mutant ALS patient iPSCs recapitulates hallmarks of disease progression. Sci Rep 2020; 10:14302. [PMID: 32868812 PMCID: PMC7459299 DOI: 10.1038/s41598-020-70510-3] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Accepted: 07/30/2020] [Indexed: 12/12/2022] Open
Abstract
Recent findings suggest a pathologic role of skeletal muscle in amyotrophic lateral sclerosis (ALS) onset and progression. However, the exact mechanism by which this occurs remains elusive due to limited human-based studies. To this end, phenotypic ALS skeletal muscle models were developed from induced pluripotent stem cells (iPSCs) derived from healthy individuals (WT) and ALS patients harboring mutations in the superoxide dismutase 1 (SOD1) gene. Although proliferative, SOD1 myoblasts demonstrated delayed and reduced fusion efficiency compared to WT. Additionally, SOD1 myotubes exhibited significantly reduced length and cross-section. Also, SOD1 myotubes had loosely arranged myosin heavy chain and reduced acetylcholine receptor expression per immunocytochemical analysis. Functional analysis indicated considerably reduced contractile force and synchrony in SOD1 myotubes. Mitochondrial assessment indicated reduced inner mitochondrial membrane potential (ΔΨm) and metabolic plasticity in the SOD1-iPSC derived myotubes. This work presents the first well-characterized in vitro iPSC-derived muscle model that demonstrates SOD1 toxicity effects on human muscle regeneration, contractility and metabolic function in ALS. Current findings align with previous ALS patient biopsy studies and suggest an active contribution of skeletal muscle in NMJ dysfunction. Further, the results validate this model as a human-relevant platform for ALS research and drug discovery studies.
Collapse
Affiliation(s)
- Agnes Badu-Mensah
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway, Suite 400, Orlando, FL, 32826, USA.,College of Medicine, Burnett School of Biomedical Sciences, University of Central Florida, Orlando, FL, 32816, USA
| | - Xiufang Guo
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway, Suite 400, Orlando, FL, 32826, USA
| | | | - John W Rumsey
- Hesperos Inc., 12501 Research Pkwy, Suite 100, 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., 12501 Research Pkwy, Suite 100, Orlando, FL, 32826, USA.
| |
Collapse
|
5
|
Sasserath T, Rumsey JW, McAleer CW, Bridges LR, Long CJ, Elbrecht D, Schuler F, Roth A, Bertinetti‐LaPatki C, Shuler ML, Hickman JJ. Differential Monocyte Actuation in a Three-Organ Functional Innate Immune System-on-a-Chip. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2020; 7:2000323. [PMID: 32670763 PMCID: PMC7341107 DOI: 10.1002/advs.202000323] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Revised: 04/09/2020] [Indexed: 05/24/2023]
Abstract
A functional, human, multiorgan, pumpless, immune system-on-a-chip featuring recirculating THP-1 immune cells with cardiomyocytes, skeletal muscle, and liver in separate compartments in a serum-free medium is developed. This in vitro platform can emulate both a targeted immune response to tissue-specific damage, and holistic proinflammatory immune response to proinflammatory compound exposure. The targeted response features fluorescently labeled THP-1 monocytes selectively infiltrating into an amiodarone-damaged cardiac module and changes in contractile force measurements without immune-activated damage to the other organ modules. In contrast to the targeted immune response, general proinflammatory treatment of immune human-on-a-chip systems with lipopolysaccharide (LPS) and interferon-γ (IFN-γ) causes nonselective damage to cells in all three-organ compartments. Biomarker analysis indicates upregulation of the proinflammation cytokines TNF-α, IL-6, IL-10, MIP-1, MCP-1, and RANTES in response to LPS + IFN-γ treatment indicative of the M1 macrophage phenotype, whereas amiodarone treatment only leads to an increase in the restorative cytokine IL-6 which is a marker for the M2 phenotype. This system can be used as an alternative to humanized animal models to determine direct immunological effects of biological therapeutics including monoclonal antibodies, vaccines, and gene therapies, and the indirect effects caused by cytokine release from target tissues in response to a drug's pharmacokinetics (PK)/pharmacodynamics (PD) profile.
Collapse
Affiliation(s)
- Trevor Sasserath
- Hesperos, Inc.12501 Research Parkway, Suite 100OrlandoFL32826USA
| | - John W. Rumsey
- Hesperos, Inc.12501 Research Parkway, Suite 100OrlandoFL32826USA
| | | | | | | | - Daniel Elbrecht
- Hesperos, Inc.12501 Research Parkway, Suite 100OrlandoFL32826USA
| | - Franz Schuler
- Hoffmann‐La RochePharmaceuticals DivisionBldg 73, Rm 117bBasel4070Switzerland
| | - Adrian Roth
- Hoffmann‐La RochePharmaceuticals DivisionBldg 73, Rm 117bBasel4070Switzerland
| | | | | | - James J. Hickman
- Hesperos, Inc.12501 Research Parkway, Suite 100OrlandoFL32826USA
- NanoScience Technology Center, University of Central Florida12424 Research Parkway, Suite 400OrlandoFL32826USA
| |
Collapse
|
6
|
Colón A, Badu-Mensah A, Guo X, Goswami A, Hickman JJ. Differentiation of Intrafusal Fibers from Human Induced Pluripotent Stem Cells. ACS Chem Neurosci 2020; 11:1085-1092. [PMID: 32159941 DOI: 10.1021/acschemneuro.0c00055] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Human-based "body-on-a-chip" technology provides powerful platforms in developing models for drug evaluation and disease evaluations in phenotypic models. Induced pluripotent stem cells (iPSCs) are ideal cell sources for generating different cell types for these in vitro functional systems and recapitulation of the neuromuscular reflex arc would allow for the study of patient specific neuromuscular diseases. Regarding relevant afferent (intrafusal fibers, sensory neurons) and efferent (extrafusal fibers, motoneurons) cells, in vitro differentiation of intrafusal fiber from human iPSCs has not been established. This work demonstrates a protocol for inducing an enrichment of intrafusal bag fibers from iPSCs using morphological analysis and immunocytochemistry. Phosphorylation of the ErbB2 receptors and S46 staining indicated a 3-fold increase of total intrafusal fibers further confirming the efficiency of the protocol. Integration of induced intrafusal fibers would enable more accurate reflex arc models and application of this protocol on patient iPSCs would allow for patient-specific disease modeling.
Collapse
Affiliation(s)
- Alisha Colón
- Hybrid Systems Lab, NanoScience Technology Center, University of Central Florida, 12424 Research Parkway, Suite 400, Orlando, Florida 32826, United States
| | - Agnes Badu-Mensah
- Hybrid Systems Lab, NanoScience Technology Center, University of Central Florida, 12424 Research Parkway, Suite 400, Orlando, Florida 32826, United States
| | - Xiufang Guo
- Hybrid Systems Lab, NanoScience Technology Center, University of Central Florida, 12424 Research Parkway, Suite 400, Orlando, Florida 32826, United States
| | - Arindom Goswami
- Hybrid Systems Lab, NanoScience Technology Center, University of Central Florida, 12424 Research Parkway, Suite 400, Orlando, Florida 32826, United States
| | - James J. Hickman
- Hybrid Systems Lab, NanoScience Technology Center, University of Central Florida, 12424 Research Parkway, Suite 400, Orlando, Florida 32826, United States
| |
Collapse
|
7
|
Najjar SA, Smith AST, Long CJ, McAleer CW, Cai Y, Srinivasan B, Martin C, Vandenburgh HH, Hickman JJ. A multiplexed in vitro assay system for evaluating human skeletal muscle functionality in response to drug treatment. Biotechnol Bioeng 2019; 117:736-747. [PMID: 31758543 DOI: 10.1002/bit.27231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Revised: 10/29/2019] [Accepted: 11/19/2019] [Indexed: 11/07/2022]
Abstract
In vitro systems that mimic organ functionality have become increasingly important tools in drug development studies. Systems that measure the functional properties of skeletal muscle are beneficial to compound screening studies and also for integration into multiorgan devices. To date, no studies have investigated human skeletal muscle responses to drug treatments at the single myotube level in vitro. This report details a microscale cantilever chip-based assay system for culturing individual human myotubes. The cantilevers, along with a laser and photo-detector system, enable measurement of myotube contractions in response to broad-field electrical stimulation. This system was used to obtain baseline functional parameters for untreated human myotubes, including peak contractile force and time-to-fatigue data. The cultured myotubes were then treated with known myotoxic compounds and the resulting functional changes were compared to baseline measurements as well as known physiological responses in vivo. The collected data demonstrate the system's capacity for screening direct effects of compound action on individual human skeletal myotubes in a reliable, reproducible, and noninvasive manner. Furthermore, it has the potential to be utilized for high-content screening, disease modeling, and exercise studies of human skeletal muscle performance utilizing iPSCs derived from specific patient populations such as the muscular dystrophies.
Collapse
Affiliation(s)
- Sarah A Najjar
- NanoScience Technology Center, University of Central Florida, Orlando, Florida
| | - Alexander S T Smith
- NanoScience Technology Center, University of Central Florida, Orlando, Florida
| | - Christopher J Long
- NanoScience Technology Center, University of Central Florida, Orlando, Florida
| | | | - Yunqing Cai
- NanoScience Technology Center, University of Central Florida, Orlando, Florida
| | - Balaji Srinivasan
- NanoScience Technology Center, University of Central Florida, Orlando, Florida
| | - Candace Martin
- NanoScience Technology Center, University of Central Florida, Orlando, Florida
| | - Herman H Vandenburgh
- Department of Pathology and Laboratory Medicine, Brown University, Providence, Rhode Island
| | - James J Hickman
- NanoScience Technology Center, University of Central Florida, Orlando, Florida
| |
Collapse
|
8
|
Coln EA, Colon A, Long CJ, Sriram NN, Esch M, Prot JM, Elbrecht DH, Wang Y, Jackson M, Shuler ML, Hickman JJ. Piezoelectric BioMEMS Cantilever for Measurement of Muscle Contraction and for Actuation of Mechanosensitive Cells. MRS COMMUNICATIONS 2019; 9:1186-1192. [PMID: 33777497 PMCID: PMC7995331 DOI: 10.1557/mrc.2019.129] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Accepted: 09/06/2019] [Indexed: 06/12/2023]
Abstract
A piezoelectric biomedical microelectromechanical system (bioMEMS) cantilever device was designed and fabricated to act as either a sensing element for muscle tissue contraction or as an actuator to apply mechanical force to cells. The sensing ability of the piezoelectric cantilevers was shown by monitoring the electrical signal generated from the piezoelectric aluminum nitride in response to the contraction of iPSC-derived cardiomyocytes cultured on the piezoelectric cantilevers. Actuation was demonstrated by applying electrical pulses to the piezoelectric cantilever and observing bending via an optical detection method. This piezoelectric cantilever device was designed to be incorporated into body-on-a-chip systems.
Collapse
Affiliation(s)
- Elizabeth A. Coln
- Hybrid Systems Laboratory, University of Central Florida, NanoScience Technology Center, 12424 Research Parkway, Suite 400, Orlando, FL 32826
- Department of Electrical & Computer Engineering, University of Central Florida, 4328 Scorpius St., Orlando, FL 32816
| | - Alisha Colon
- Hybrid Systems Laboratory, University of Central Florida, NanoScience Technology Center, 12424 Research Parkway, Suite 400, Orlando, FL 32826
| | | | | | - Mandy Esch
- Department of Biomedical Engineering, Cornell University, Weill Hall, Ithaca, NY 14853
| | - Jean-Matthieu Prot
- Department of Biomedical Engineering, Cornell University, Weill Hall, Ithaca, NY 14853
| | - Daniel H. Elbrecht
- Hybrid Systems Laboratory, University of Central Florida, NanoScience Technology Center, 12424 Research Parkway, Suite 400, Orlando, FL 32826
| | - Ying Wang
- Department of Biomedical Engineering, Cornell University, Weill Hall, Ithaca, NY 14853
| | - Max Jackson
- Hesperos, Inc., 3259 Progress Drive #158, Orlando, FL 32826
| | - Michael L. Shuler
- Hesperos, Inc., 3259 Progress Drive #158, Orlando, FL 32826
- Department of Biomedical Engineering, Cornell University, Weill Hall, Ithaca, NY 14853
| | - James J. Hickman
- Hybrid Systems Laboratory, University of Central Florida, NanoScience Technology Center, 12424 Research Parkway, Suite 400, Orlando, FL 32826
- Department of Electrical & Computer Engineering, University of Central Florida, 4328 Scorpius St., Orlando, FL 32816
- Hesperos, Inc., 3259 Progress Drive #158, Orlando, FL 32826
| |
Collapse
|
9
|
McAleer CW, Pointon A, Long CJ, Brighton RL, Wilkin BD, Bridges LR, Narasimhan Sriram N, Fabre K, McDougall R, Muse VP, Mettetal JT, Srivastava A, Williams D, Schnepper MT, Roles JL, Shuler ML, Hickman JJ, Ewart L. On the potential of in vitro organ-chip models to define temporal pharmacokinetic-pharmacodynamic relationships. Sci Rep 2019; 9:9619. [PMID: 31270362 PMCID: PMC6610665 DOI: 10.1038/s41598-019-45656-4] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Accepted: 06/07/2019] [Indexed: 12/20/2022] Open
Abstract
Functional human-on-a-chip systems hold great promise to enable quantitative translation to in vivo outcomes. Here, we explored this concept using a pumpless heart only and heart:liver system to evaluate the temporal pharmacokinetic/pharmacodynamic (PKPD) relationship for terfenadine. There was a time dependent drug-induced increase in field potential duration in the cardiac compartment in response to terfenadine and that response was modulated using a metabolically competent liver module that converted terfenadine to fexofenadine. Using this data, a mathematical model was developed to predict the effect of terfenadine in preclinical species. Developing confidence that microphysiological models could have a transformative effect on drug discovery, we also tested a previously discovered proprietary AstraZeneca small molecule and correctly determined the cardiotoxic response to its metabolite in the heart:liver system. Overall our findings serve as a guiding principle to future investigations of temporal concentration response relationships in these innovative in vitro models, especially, if validated across multiple time frames, with additional pharmacological mechanisms and molecules representing a broad chemical diversity.
Collapse
Affiliation(s)
| | - Amy Pointon
- Drug Safety and Metabolism, IMED Biotech Unit, AstraZeneca, Cambridge, UK
| | - Christopher J Long
- Hesperos, Inc., 3259 Progress Dr., Room 158, Orlando, FL, 32826-3230, USA
| | - Rocky L Brighton
- Hesperos, Inc., 3259 Progress Dr., Room 158, Orlando, FL, 32826-3230, USA
| | - Benjamin D Wilkin
- Hesperos, Inc., 3259 Progress Dr., Room 158, Orlando, FL, 32826-3230, USA
| | - L Richard Bridges
- Hesperos, Inc., 3259 Progress Dr., Room 158, Orlando, FL, 32826-3230, USA
| | | | - Kristin Fabre
- Drug Safety and Metabolism, IMED Biotech Unit, AstraZeneca, Waltham, USA
| | - Robin McDougall
- Drug Safety and Metabolism, IMED Biotech Unit, AstraZeneca, Waltham, USA
| | - Victorine P Muse
- Drug Safety and Metabolism, IMED Biotech Unit, AstraZeneca, Waltham, USA
| | - Jerome T Mettetal
- Drug Safety and Metabolism, IMED Biotech Unit, AstraZeneca, Waltham, USA
| | | | - Dominic Williams
- Drug Safety and Metabolism, IMED Biotech Unit, AstraZeneca, Cambridge, UK
| | - Mark T Schnepper
- NanoScience Technology Center, 12424 Research Parkway, Suite 400, Orlando, FL, 32826, USA
| | - Jeff L Roles
- Hesperos, Inc., 3259 Progress Dr., Room 158, Orlando, FL, 32826-3230, USA
| | - Michael L Shuler
- Hesperos, Inc., 3259 Progress Dr., Room 158, Orlando, FL, 32826-3230, USA
| | - James J Hickman
- Hesperos, Inc., 3259 Progress Dr., Room 158, Orlando, FL, 32826-3230, USA.
- NanoScience Technology Center, 12424 Research Parkway, Suite 400, Orlando, FL, 32826, USA.
| | - Lorna Ewart
- Drug Safety and Metabolism, IMED Biotech Unit, AstraZeneca, Cambridge, UK.
| |
Collapse
|
10
|
McAleer CW, Long CJ, Elbrecht D, Sasserath T, Bridges LR, Rumsey JW, Martin C, Schnepper M, Wang Y, Schuler F, Roth AB, Funk C, Shuler ML, Hickman JJ. Multi-organ system for the evaluation of efficacy and off-target toxicity of anticancer therapeutics. Sci Transl Med 2019; 11:eaav1386. [PMID: 31217335 DOI: 10.1126/scitranslmed.aav1386] [Citation(s) in RCA: 91] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Revised: 11/16/2018] [Accepted: 05/17/2019] [Indexed: 12/14/2022]
Abstract
A pumpless, reconfigurable, multi-organ-on-a-chip system containing recirculating serum-free medium can be used to predict preclinical on-target efficacy, metabolic conversion, and measurement of off-target toxicity of drugs using functional biological microelectromechanical systems. In the first configuration of the system, primary human hepatocytes were cultured with two cancer-derived human bone marrow cell lines for antileukemia drug analysis in which diclofenac and imatinib demonstrated a cytostatic effect on bone marrow cancer proliferation. Liver viability was not affected by imatinib; however, diclofenac reduced liver viability by 30%. The second configuration housed a multidrug-resistant vulva cancer line, a non-multidrug-resistant breast cancer line, primary hepatocytes, and induced pluripotent stem cell-derived cardiomyocytes. Tamoxifen reduced viability of the breast cancer cells only after metabolite generation but did not affect the vulva cancer cells except when coadministered with verapamil, a permeability glycoprotein inhibitor. Both tamoxifen alone and coadministration with verapamil produced off-target cardiac effects as indicated by a reduction of contractile force, beat frequency, and conduction velocity but did not affect viability. These systems demonstrate the utility of a human cell-based in vitro culture system to evaluate both on-target efficacy and off-target toxicity for parent drugs and their metabolites; these systems can augment and reduce the use of animals and increase the efficiency of drug evaluations in preclinical studies.
Collapse
Affiliation(s)
| | | | - Daniel Elbrecht
- Hesperos Inc., 3259 Progress Drive, Room 158, Orlando, FL 32826, USA
| | - Trevor Sasserath
- Hesperos Inc., 3259 Progress Drive, Room 158, Orlando, FL 32826, USA
| | - L Richard Bridges
- Hesperos Inc., 3259 Progress Drive, Room 158, Orlando, FL 32826, USA
| | - John W Rumsey
- Hesperos Inc., 3259 Progress Drive, Room 158, Orlando, FL 32826, USA
| | - Candace Martin
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway, Suite 400, Orlando, FL 32826, USA
| | - Mark Schnepper
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway, Suite 400, Orlando, FL 32826, USA
| | - Ying Wang
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway, Suite 400, Orlando, FL 32826, USA
| | - Franz Schuler
- Roche Pharma Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Grenzacherstrasse 124, CH-4070 Basel, Switzerland
| | - Adrian B Roth
- Roche Pharma Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Grenzacherstrasse 124, CH-4070 Basel, Switzerland
| | - Christoph Funk
- Roche Pharma Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Grenzacherstrasse 124, CH-4070 Basel, Switzerland
| | - Michael L Shuler
- Hesperos Inc., 3259 Progress Drive, Room 158, Orlando, FL 32826, USA
| | - James J Hickman
- Hesperos Inc., 3259 Progress Drive, Room 158, Orlando, FL 32826, USA.
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway, Suite 400, Orlando, FL 32826, USA
| |
Collapse
|
11
|
Shimizu K, Genma R, Gotou Y, Nagasaka S, Honda H. Three-Dimensional Culture Model of Skeletal Muscle Tissue with Atrophy Induced by Dexamethasone. Bioengineering (Basel) 2017; 4:E56. [PMID: 28952535 PMCID: PMC5590463 DOI: 10.3390/bioengineering4020056] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Revised: 06/12/2017] [Accepted: 06/12/2017] [Indexed: 12/30/2022] Open
Abstract
Drug screening systems for muscle atrophy based on the contractile force of cultured skeletal muscle tissues are required for the development of preventive or therapeutic drugs for atrophy. This study aims to develop a muscle atrophy model by inducing atrophy in normal muscle tissues constructed on microdevices capable of measuring the contractile force and to verify if this model is suitable for drug screening using the contractile force as an index. Tissue engineered skeletal muscles containing striated myotubes were prepared on the microdevices for the study. The addition of 100 µM dexamethasone (Dex), which is used as a muscle atrophy inducer, for 24 h reduced the contractile force significantly. An increase in the expression of Atrogin-1 and MuRF-1 in the tissues treated with Dex was established. A decrease in the number of striated myotubes was also observed in the tissues treated with Dex. Treatment with 8 ng/mL Insulin-like Growth Factor (IGF-I) for 24 h significantly increased the contractile force of the Dex-induced atrophic tissues. The same treatment, though, had no impact on the force of the normal tissues. Thus, it is envisaged that the atrophic skeletal muscle tissues induced by Dex can be used for drug screening against atrophy.
Collapse
Affiliation(s)
- Kazunori Shimizu
- Department of Biomolecular Engineering, Graduate School of Engineering, Nagoya University, Nagoya 464-8603, Japan.
| | - Riho Genma
- Department of Biomolecular Engineering, Graduate School of Engineering, Nagoya University, Nagoya 464-8603, Japan.
| | - Yuuki Gotou
- Department of Biomolecular Engineering, Graduate School of Engineering, Nagoya University, Nagoya 464-8603, Japan.
| | - Sumire Nagasaka
- Department of Biomolecular Engineering, Graduate School of Engineering, Nagoya University, Nagoya 464-8603, Japan.
| | - Hiroyuki Honda
- Department of Biomolecular Engineering, Graduate School of Engineering, Nagoya University, Nagoya 464-8603, Japan.
- Innovative Research Center for Preventive Medical Engineering, Nagoya University, Nagoya 464-8601, Japan.
| |
Collapse
|
12
|
Wang YI, Oleaga C, Long CJ, Esch MB, McAleer CW, Miller PG, Hickman JJ, Shuler ML. Self-contained, low-cost Body-on-a-Chip systems for drug development. Exp Biol Med (Maywood) 2017; 242:1701-1713. [PMID: 29065797 DOI: 10.1177/1535370217694101] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Integrated multi-organ microphysiological systems are an evolving tool for preclinical evaluation of the potential toxicity and efficacy of drug candidates. Such systems, also known as Body-on-a-Chip devices, have a great potential to increase the successful conversion of drug candidates entering clinical trials into approved drugs. Systems, to be attractive for commercial adoption, need to be inexpensive, easy to operate, and give reproducible results. Further, the ability to measure functional responses, such as electrical activity, force generation, and barrier integrity of organ surrogates, enhances the ability to monitor response to drugs. The ability to operate a system for significant periods of time (up to 28 d) will provide potential to estimate chronic as well as acute responses of the human body. Here we review progress towards a self-contained low-cost microphysiological system with functional measurements of physiological responses. Impact statement Multi-organ microphysiological systems are promising devices to improve the drug development process. The development of a pumpless system represents the ability to build multi-organ systems that are of low cost, high reliability, and self-contained. These features, coupled with the ability to measure electrical and mechanical response in addition to chemical or metabolic changes, provides an attractive system for incorporation into the drug development process. This will be the most complete review of the pumpless platform with recirculation yet written.
Collapse
Affiliation(s)
- Ying I Wang
- 1 Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Carlota Oleaga
- 2 NanoScience Technology Center, University of Central Florida, Orlando, FL 32826, USA
| | - Christopher J Long
- 2 NanoScience Technology Center, University of Central Florida, Orlando, FL 32826, USA.,3 Hesperos, Inc., Orlando, FL 32826, USA
| | - Mandy B Esch
- 4 Center for Nanoscale Science and Technology, National Institute of Standards and Technology, Gaithersburg, MD 20899, USA
| | - Christopher W McAleer
- 2 NanoScience Technology Center, University of Central Florida, Orlando, FL 32826, USA.,3 Hesperos, Inc., Orlando, FL 32826, USA
| | - Paula G Miller
- 1 Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA
| | - James J Hickman
- 2 NanoScience Technology Center, University of Central Florida, Orlando, FL 32826, USA.,3 Hesperos, Inc., Orlando, FL 32826, USA
| | - Michael L Shuler
- 1 Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA.,3 Hesperos, Inc., Orlando, FL 32826, USA
| |
Collapse
|
13
|
Uzel SGM, Platt RJ, Subramanian V, Pearl TM, Rowlands CJ, Chan V, Boyer LA, So PTC, Kamm RD. Microfluidic device for the formation of optically excitable, three-dimensional, compartmentalized motor units. SCIENCE ADVANCES 2016; 2:e1501429. [PMID: 27493991 PMCID: PMC4972469 DOI: 10.1126/sciadv.1501429] [Citation(s) in RCA: 146] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/12/2015] [Accepted: 07/06/2016] [Indexed: 05/21/2023]
Abstract
Motor units are the fundamental elements responsible for muscle movement. They are formed by lower motor neurons and their muscle targets, synapsed via neuromuscular junctions (NMJs). The loss of NMJs in neurodegenerative disorders (such as amyotrophic lateral sclerosis or spinal muscle atrophy) or as a result of traumatic injuries affects millions of lives each year. Developing in vitro assays that closely recapitulate the physiology of neuromuscular tissues is crucial to understand the formation and maturation of NMJs, as well as to help unravel the mechanisms leading to their degeneration and repair. We present a microfluidic platform designed to coculture myoblast-derived muscle strips and motor neurons differentiated from mouse embryonic stem cells (ESCs) within a three-dimensional (3D) hydrogel. The device geometry mimics the spinal cord-limb physical separation by compartmentalizing the two cell types, which also facilitates the observation of 3D neurite outgrowth and remote muscle innervation. Moreover, the use of compliant pillars as anchors for muscle strips provides a quantitative functional readout of force generation. Finally, photosensitizing the ESC provides a pool of source cells that can be differentiated into optically excitable motor neurons, allowing for spatiodynamic, versatile, and noninvasive in vitro control of the motor units.
Collapse
Affiliation(s)
- Sebastien G. M. Uzel
- Department of Mechanical Engineering, Massachusetts Institute of Technology (MIT), Cambridge, MA 02139, USA
| | - Randall J. Platt
- Department of Biological Engineering, MIT, Cambridge, MA 02139, USA
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | | | - Taylor M. Pearl
- Department of Biological Engineering, MIT, Cambridge, MA 02139, USA
| | | | - Vincent Chan
- Department of Mechanical Engineering, Massachusetts Institute of Technology (MIT), Cambridge, MA 02139, USA
| | | | - Peter T. C. So
- Department of Mechanical Engineering, Massachusetts Institute of Technology (MIT), Cambridge, MA 02139, USA
- Department of Biological Engineering, MIT, Cambridge, MA 02139, USA
- BioSystems and Micromechanics (BioSyM) IRG, Singapore-MIT Alliance for Research and Technology, Singapore, Singapore
| | - Roger D. Kamm
- Department of Mechanical Engineering, Massachusetts Institute of Technology (MIT), Cambridge, MA 02139, USA
- Department of Biological Engineering, MIT, Cambridge, MA 02139, USA
- BioSystems and Micromechanics (BioSyM) IRG, Singapore-MIT Alliance for Research and Technology, Singapore, Singapore
- Corresponding author.
| |
Collapse
|
14
|
Smith AST, Davis J, Lee G, Mack DL, Kim DH. Muscular dystrophy in a dish: engineered human skeletal muscle mimetics for disease modeling and drug discovery. Drug Discov Today 2016; 21:1387-1398. [PMID: 27109386 DOI: 10.1016/j.drudis.2016.04.013] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Revised: 03/24/2016] [Accepted: 04/15/2016] [Indexed: 01/16/2023]
Abstract
Engineered in vitro models using human cells, particularly patient-derived induced pluripotent stem cells (iPSCs), offer a potential solution to issues associated with the use of animals for studying disease pathology and drug efficacy. Given the prevalence of muscle diseases in human populations, an engineered tissue model of human skeletal muscle could provide a biologically accurate platform to study basic muscle physiology, disease progression, and drug efficacy and/or toxicity. Such platforms could be used as phenotypic drug screens to identify compounds capable of alleviating or reversing congenital myopathies, such as Duchene muscular dystrophy (DMD). Here, we review current skeletal muscle modeling technologies with a specific focus on efforts to generate biomimetic systems for investigating the pathophysiology of dystrophic muscle.
Collapse
Affiliation(s)
- Alec S T Smith
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA
| | - Jennifer Davis
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA; Department of Pathology, University of Washington, Seattle, WA 98195, USA; Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98109, USA; Center for Cardiovascular Biology, University of Washington, Seattle, WA 98109, USA
| | - Gabsang Lee
- Institute for Cell Engineering, Department of Neurology, The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - David L Mack
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98109, USA; Department of Rehabilitation Medicine, University of Washington, Seattle, WA 98195, USA
| | - Deok-Ho Kim
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA; Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98109, USA; Center for Cardiovascular Biology, University of Washington, Seattle, WA 98109, USA.
| |
Collapse
|
15
|
McAleer CW, Rumsey JW, Stancescu M, Hickman JJ. Functional myotube formation from adult rat satellite cells in a defined serum-free system. Biotechnol Prog 2015; 31:997-1003. [PMID: 25683642 PMCID: PMC5015122 DOI: 10.1002/btpr.2063] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Revised: 01/28/2015] [Indexed: 12/28/2022]
Abstract
This manuscript describes the development of a culture system whereby mature contracting myotubes were formed from adult rat derived satellite cells. Satellite cells, extracted from the Tibialis Anterior of adult rats, were grown in defined serum-free growth and differentiation media, on a nonbiological substrate, N-1[3-trimethoxysilyl propyl] diethylenetriamine. Myotubes were evaluated morphologically and immunocytochemically, using MyHC specific antibodies, as well as functionally using patch clamp electrophysiology to measure ion channel activity. Results indicated the establishment of the rapid expression of adult myosin isoforms that contrasts to their slow development in embryonic cultures. This culture system has applications in the understanding and treatment of age-related muscle myopathy, muscular dystrophy, and for skeletal muscle engineering by providing a more relevant phenotype for both in vitro and in vivo applications.
Collapse
Affiliation(s)
- Christopher W McAleer
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway, Suite 400, Orlando, FL, 32826
| | - John W Rumsey
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway, Suite 400, Orlando, FL, 32826
| | - Maria Stancescu
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway, Suite 400, Orlando, FL, 32826
| | - James J Hickman
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway, Suite 400, Orlando, FL, 32826
| |
Collapse
|