104 results on '"David L. Mack"'
Search Results
2. An automated platform for simultaneous, longitudinal analysis of engineered neuromuscular tissues for applications in neurotoxin potency testing
- Author
-
Jacob W. Fleming, Molly C. McCloskey, Kevin Gray, David R. Nash, Vincent Leung, Christos Michas, Shawn M. Luttrell, Christopher Cavanaugh, Julie Mathieu, Shawn Mcquire, Mark Bothwell, David L. Mack, Nicholas A. Geisse, and Alec S.T. Smith
- Subjects
Neuromuscular junction ,Engineered skeletal muscle ,Engineered tissue models ,Potency assay ,High-throughput model ,Toxicology. Poisons ,RA1190-1270 - Abstract
Animal models of the neuromuscular junction (NMJ) have been widely studied but exhibit critical differences from human biology limiting utility in drug and disease modelling. Challenges with scarcity, scalability, throughput, and ethical considerations further limit the suitability of animal models for preclinical screening. Engineered models have emerged as alternatives for studying NMJ functionality in response to genetic and/or pharmacological challenge. However, these models have faced challenges associated with their poorly scalable creation, sourcing suitable cells, and the extraction of reliable, quantifiable metrics. We present a turnkey iPSC-based model of the NMJ employing channelrhodopsin-2 expression within the motor neuron (MN) population driving muscle contraction in response to blue light. MNs co-cultured with engineered skeletal muscle tissues produced twitch forces of 34.7 ± 22.7 µN in response to blue light, with a response fidelity > 92 %. Histological analysis revealed characteristic punctate acetylcholine receptor staining co-localized with the presynaptic marker synaptic vesicle protein-2. Dose-response studies using botulinum neurotoxin showed loss of function in a dose- and time-dependent manner (EC50 − 0.11 ± 0.015 µg). Variability of the EC50 values between 2 different iPSC differentiations of both cell types and 2 users was less than 2 %. Further testing with the acute neurotoxins acetylcholine mustard and d-tubocurarine validated the biological relevance of the postsynaptic machinery of the model. This model marks a meaningful progression of 3D engineered models of the NMJ, providing engineered tissues at a throughput relevant to potency and screening applications with an abundant iPSC cell source and standardized hardware-software ecosystem allowing technology transfer across laboratories.
- Published
- 2025
- Full Text
- View/download PDF
3. High mobility group box 1 (HMGB1) is a potential disease biomarker in cell and mouse models of Duchenne muscular dystrophy
- Author
-
Rebecca A. Slick, Jessica Sutton, Margaret Haberman, Benjamin S. O'Brien, Jennifer A. Tinklenberg, Aashay Mardikar, Mariah J. Prom, Margaret Beatka, Melanie Gartz, Mark A. Vanden Avond, Emily Siebers, David L. Mack, J. Patrick Gonzalez, Allison D. Ebert, Kanneboyina Nagaraju, and Michael W. Lawlor
- Subjects
biomarker ,duchenne muscular dystrophy ,hmgb1 ,muscle differentiation ,rna sequencing ,Science ,Biology (General) ,QH301-705.5 - Published
- 2024
- Full Text
- View/download PDF
4. Corrigendum: Calcium handling maturation and adaptation to increased substrate stiffness in human iPSC-derived cardiomyocytes: the impact of full-length dystrophin deficiency
- Author
-
Josè Manuel Pioner, Lorenzo Santini, Chiara Palandri, Marianna Langione, Bruno Grandinetti, Silvia Querceto, Daniele Martella, Costanza Mazzantini, Beatrice Scellini, Lucrezia Giammarino, Flavia Lupi, Francesco Mazzarotto, Aoife Gowran, Davide Rovina, Rosaria Santoro, Giulio Pompilio, Chiara Tesi, Camilla Parmeggiani, Michael Regnier, Elisabetta Cerbai, David L. Mack, Corrado Poggesi, Cecilia Ferrantini, and Raffaele Coppini
- Subjects
human iPSC derived cardiomyocytes ,dystrophin (DMD) ,substrate stiffness ,calcium handing ,duchenne muscular dystrophy (DMD) ,Physiology ,QP1-981 - Published
- 2023
- Full Text
- View/download PDF
5. Calcium handling maturation and adaptation to increased substrate stiffness in human iPSC-derived cardiomyocytes: The impact of full-length dystrophin deficiency
- Author
-
Josè Manuel Pioner, Lorenzo Santini, Chiara Palandri, Marianna Langione, Bruno Grandinetti, Silvia Querceto, Daniele Martella, Costanza Mazzantini, Beatrice Scellini, Lucrezia Giammarino, Flavia Lupi, Francesco Mazzarotto, Aoife Gowran, Davide Rovina, Rosaria Santoro, Giulio Pompilio, Chiara Tesi, Camilla Parmeggiani, Michael Regnier, Elisabetta Cerbai, David L. Mack, Corrado Poggesi, Cecilia Ferrantini, and Raffaele Coppini
- Subjects
human iPSC derived cardiomyocytes ,dystrophin (DMD) ,substrate stiffness ,calcium handing ,duchenne muscular dystrophy (DMD) ,Physiology ,QP1-981 - Abstract
Cardiomyocytes differentiated from human induced Pluripotent Stem Cells (hiPSC- CMs) are a unique source for modelling inherited cardiomyopathies. In particular, the possibility of observing maturation processes in a simple culture dish opens novel perspectives in the study of early-disease defects caused by genetic mutations before the onset of clinical manifestations. For instance, calcium handling abnormalities are considered as a leading cause of cardiomyocyte dysfunction in several genetic-based dilated cardiomyopathies, including rare types such as Duchenne Muscular Dystrophy (DMD)-associated cardiomyopathy. To better define the maturation of calcium handling we simultaneously measured action potential and calcium transients (Ca-Ts) using fluorescent indicators at specific time points. We combined micropatterned substrates with long-term cultures to improve maturation of hiPSC-CMs (60, 75 or 90 days post-differentiation). Control-(hiPSC)-CMs displayed increased maturation over time (90 vs 60 days), with longer action potential duration (APD), increased Ca-T amplitude, faster Ca-T rise (time to peak) and Ca-T decay (RT50). The progressively increased contribution of the SR to Ca release (estimated by post-rest potentiation or Caffeine-induced Ca-Ts) appeared as the main determinant of the progressive rise of Ca-T amplitude during maturation. As an example of severe cardiomyopathy with early onset, we compared hiPSC-CMs generated from a DMD patient (DMD-ΔExon50) and a CRISPR-Cas9 genome edited cell line isogenic to the healthy control with deletion of a G base at position 263 of the DMD gene (c.263delG-CMs). In DMD-hiPSC-CMs, changes of Ca-Ts during maturation were less pronounced: indeed, DMD cells at 90 days showed reduced Ca-T amplitude and faster Ca-T rise and RT50, as compared with control hiPSC-CMs. Caffeine-Ca-T was reduced in amplitude and had a slower time course, suggesting lower SR calcium content and NCX function in DMD vs control cells. Nonetheless, the inotropic and lusitropic responses to forskolin were preserved. CRISPR-induced c.263delG-CM line recapitulated the same developmental calcium handling alterations observed in DMD-CMs. We then tested the effects of micropatterned substrates with higher stiffness. In control hiPSC-CMs, higher stiffness leads to higher amplitude of Ca-T with faster decay kinetics. In hiPSC-CMs lacking full-length dystrophin, however, stiffer substrates did not modify Ca-Ts but only led to higher SR Ca content. These findings highlighted the inability of dystrophin-deficient cardiomyocytes to adjust their calcium homeostasis in response to increases of extracellular matrix stiffness, which suggests a mechanism occurring during the physiological and pathological development (i.e. fibrosis).
- Published
- 2022
- Full Text
- View/download PDF
6. Editorial: Modeling neuromuscular diseases to determine molecular drivers of pathology and for drug discovery
- Author
-
Alec S. T. Smith, Megan L. McCain, Mark Bothwell, and David L. Mack
- Subjects
neuromuscular disease (NMD) ,disease modeling ,iPSC (induced pluripotent stem cell) ,cell line (C2C12) ,therapy screening ,in vitro versus in vivo ,Biology (General) ,QH301-705.5 - Published
- 2022
- Full Text
- View/download PDF
7. Voluntary wheel running complements microdystrophin gene therapy to improve muscle function in mdx mice
- Author
-
Shelby E. Hamm, Daniel D. Fathalikhani, Katherine E. Bukovec, Adele K. Addington, Haiyan Zhang, Justin B. Perry, Ryan P. McMillan, Michael W. Lawlor, Mariah J. Prom, Mark A. Vanden Avond, Suresh N. Kumar, Kirsten E. Coleman, J.B. Dupont, David L. Mack, David A. Brown, Carl A. Morris, J. Patrick Gonzalez, and Robert W. Grange
- Subjects
duchenne muscular dystrophy ,muscle physiology ,muscle pathology ,dystrophic grade ,voluntary exercise ,endurance ,Genetics ,QH426-470 ,Cytology ,QH573-671 - Abstract
We tested the hypothesis that voluntary wheel running would complement microdystrophin gene therapy to improve muscle function in young mdx mice, a model of Duchenne muscular dystrophy. mdx mice injected with a single dose of AAV9-CK8-microdystrophin or vehicle at age 7 weeks were assigned to three groups: mdxRGT (run, gene therapy), mdxGT (no run, gene therapy), or mdx (no run, no gene therapy). Wild-type (WT) mice were assigned to WTR (run) and WT (no run) groups. WTR and mdxRGT performed voluntary wheel running for 21 weeks; remaining groups were cage active. Robust expression of microdystrophin occurred in heart and limb muscles of treated mice. mdxRGT versus mdxGT mice showed increased microdystrophin in quadriceps but decreased levels in diaphragm. mdx final treadmill fatigue time was depressed compared to all groups, improved in mdxGT, and highest in mdxRGT. Both weekly running distance (km) and final treadmill fatigue time for mdxRGT and WTR were similar. Remarkably, mdxRGT diaphragm power was only rescued to 60% of WT, suggesting a negative impact of running. However, potential changes in fiber type distribution in mdxRGT diaphragms could indicate an adaptation to trade power for endurance. Post-treatment in vivo maximal plantar flexor torque relative to baseline values was greater for mdxGT and mdxRGT versus all other groups. Mitochondrial respiration rates from red quadriceps fibers were significantly improved in mdxGT animals, but the greatest bioenergetic benefit was observed in the mdxRGT group. Additional assessments revealed partial to full functional restoration in mdxGT and mdxRGT muscles relative to WT. These data demonstrate that voluntary wheel running combined with microdystrophin gene therapy in young mdx mice improved whole-body performance, affected muscle function differentially, mitigated energetic deficits, but also revealed some detrimental effects of exercise. With microdystrophin gene therapy currently in clinical trials, these data may help us understand the potential impact of exercise in treated patients.
- Published
- 2021
- Full Text
- View/download PDF
8. Incorporation of sensing modalities into de novo designed fluorescence-activating proteins
- Author
-
Jason C. Klima, Lindsey A. Doyle, Justin Daho Lee, Michael Rappleye, Lauren A. Gagnon, Min Yen Lee, Emilia P. Barros, Anastassia A. Vorobieva, Jiayi Dou, Samantha Bremner, Jacob S. Quon, Cameron M. Chow, Lauren Carter, David L. Mack, Rommie E. Amaro, Joshua C. Vaughan, Andre Berndt, Barry L. Stoddard, and David Baker
- Subjects
Science - Abstract
Fluorescent protein reporters based on GFP exist, but have intrinsic disadvantages. Here the authors incorporate pH, Ca2+ and protein–protein interaction sensing modalities into de novo designed mini-fluorescence-activating proteins (mFAPs), with increased photostability and smaller size, which bind a range of DFHBI chromophore variants.
- Published
- 2021
- Full Text
- View/download PDF
9. rAAV-related therapy fully rescues myonuclear and myofilament function in X-linked myotubular myopathy
- Author
-
Jacob A. Ross, Hichem Tasfaout, Yotam Levy, Jennifer Morgan, Belinda S. Cowling, Jocelyn Laporte, Edmar Zanoteli, Norma B. Romero, Dawn A. Lowe, Heinz Jungbluth, Michael W. Lawlor, David L. Mack, and Julien Ochala
- Subjects
Skeletal muscle ,Congenital myopathy ,Myotubularin ,Myonuclear domain ,Myofilament ,Force production ,Neurology. Diseases of the nervous system ,RC346-429 - Abstract
Abstract X-linked myotubular myopathy (XLMTM) is a life-threatening skeletal muscle disease caused by mutations in the MTM1 gene. XLMTM fibres display a population of nuclei mispositioned in the centre. In the present study, we aimed to explore whether positioning and overall distribution of nuclei affects cellular organization and contractile function, thereby contributing to muscle weakness in this disease. We also assessed whether gene therapy alters nuclear arrangement and function. We used tissue from human patients and animal models, including XLMTM dogs that had received increasing doses of recombinant AAV8 vector restoring MTM1 expression (rAAV8-cMTM1). We then used single isolated muscle fibres to analyze nuclear organization and contractile function. In addition to the expected mislocalization of nuclei in the centre of muscle fibres, a novel form of nuclear mispositioning was observed: irregular spacing between those located at the fibre periphery, and an overall increased number of nuclei, leading to dramatically smaller and inconsistent myonuclear domains. Nuclear mislocalization was associated with decreases in global nuclear synthetic activity, contractile protein content and intrinsic myofilament force production. A contractile deficit originating at the myofilaments, rather than mechanical interference by centrally positioned nuclei, was supported by experiments in regenerated mouse muscle. Systemic administration of rAAV8-cMTM1 at doses higher than 2.5 × 1013 vg kg−1 allowed a full rescue of all these cellular defects in XLMTM dogs. Altogether, these findings identify previously unrecognized pathological mechanisms in human and animal XLMTM, associated with myonuclear defects and contractile filament function. These defects can be reversed by gene therapy restoring MTM1 expression in dogs with XLMTM.
- Published
- 2020
- Full Text
- View/download PDF
10. Differentiation Capacity of Human Urine-Derived Stem Cells to Retain Telomerase Activity
- Author
-
Yingai Shi, Guihua Liu, Rongpei Wu, David L. Mack, Xiuzhi Susan Sun, Joshua Maxwell, Xuan Guan, Anthony Atala, and Yuanyuan Zhang
- Subjects
telomerase ,urine-derived stem cells ,longevity ,tissue regeneration ,differentiation ,Biology (General) ,QH301-705.5 - Abstract
Telomerase activity is essential for the self-renewal and potential of embryonic, induced pluripotent, and cancer stem cells, as well as a few somatic stem cells, such as human urine-derived stem cells (USCs). However, it remains unclear how telomerase activity affects the regeneration potential of somatic stem cells. The objective of this study was to determine the regenerative significance of telomerase activity, particularly to retain cell surface marker expression, multipotent differentiation capability, chromosomal stability, and in vivo tumorigenic transformation, in each clonal population of human primary USCs. In total, 117 USC specimens from 10 healthy male adults (25–57 years of age) were obtained. Polymerase chain reaction amplification of a telomeric repeat was used to detect USCs with positive telomerase activity (USCsTA+). A total of 80 USCsTA+ (70.2%) were identified from 117 USC clones, but they were not detected in the paired normal bladder smooth muscle cell and bone marrow stromal cell specimens. In the 20–40 years age group, approximately 75% of USC clones displayed positive telomerase activity, whereas in the 50 years age group, 59.2% of the USC clones expressed positive telomerase activity. USCsTA+ extended to passage 16, underwent 62.0 ± 4.8 population doublings, produced more cells, and were superior for osteogenic, myogenic, and uroepithelial differentiation compared to USCsTA−. Importantly, USCs displayed normal chromosome and no oncological transformation after being implanted in vivo. Overall, as a safe cell source, telomerase-positive USCs have a robust regenerative potential in cell proliferation and multipotent differentiation capacity.
- Published
- 2022
- Full Text
- View/download PDF
11. Human Induced Pluripotent Stem Cell-Derived TDP-43 Mutant Neurons Exhibit Consistent Functional Phenotypes Across Multiple Gene Edited Lines Despite Transcriptomic and Splicing Discrepancies
- Author
-
Alec S. T. Smith, Changho Chun, Jennifer Hesson, Julie Mathieu, Paul N. Valdmanis, David L. Mack, Byung-Ok Choi, Deok-Ho Kim, and Mark Bothwell
- Subjects
ALS (amyotrophic lateral sclerosis) ,iPSC (induced pluripotent stem cell) ,transcriptomics ,electrophysiologic analysis ,disease model ,Biology (General) ,QH301-705.5 - Abstract
Gene editing technologies hold great potential to enhance our ability to model inheritable neurodegenerative diseases. Specifically, engineering multiple amyotrophic lateral sclerosis (ALS) mutations into isogenic cell populations facilitates determination of whether different causal mutations cause pathology via shared mechanisms, and provides the capacity to separate these mechanisms from genotype-specific effects. As gene-edited, cell-based models of human disease become more commonplace, there is an urgent need to verify that these models constitute consistent and accurate representations of native biology. Here, commercially sourced, induced pluripotent stem cell-derived motor neurons from Cellular Dynamics International, edited to express the ALS-relevant mutations TDP-43M337V and TDP-43Q331K were compared with in-house derived lines engineered to express the TDP-43Q331K mutation within the WTC11 background. Our results highlight electrophysiological and mitochondrial deficits in these edited cells that correlate with patient-derived cells, suggesting a consistent cellular phenotype arising from TDP-43 mutation. However, significant differences in the transcriptomic profiles and splicing behavior of the edited cells underscores the need for careful comparison of multiple lines when attempting to use these cells as a means to better understand the onset and progression of ALS in humans.
- Published
- 2021
- Full Text
- View/download PDF
12. A simplified approach for derivation of induced pluripotent stem cells from Epstein-Barr virus immortalized B-lymphoblastoid cell lines
- Author
-
Stephen J. Walker, Ashley L. Wagoner, Dana Leavitt, and David L. Mack
- Subjects
Lymphoblastoid cell lines ,Induced pluripotent stem cells ,Reprogramming ,Epstein-Barr virus ,Science (General) ,Q1-390 ,Social sciences (General) ,H1-99 - Abstract
Given the limited availability of tissue, especially brain tissue, for neurological diseases and disorders research, the development of alternative biological tools for investigations of underlying molecular and genetic mechanisms is imperative. One important resource for this task is the large repositories that bank immortalized blood cells (i.e. lymphoblastoid cell lines; LCLs) from affected individuals and their unaffected family members. These repositories document demographic, phenotypic, and, in some cases, genotypic information about the donors and thus provide a ready-made sample source for hypothesis testing. Importantly, patient-specific LCLs can be used to generate induced pluripotent stem cells (iPSC) that, in turn, can be used to create specific cell types for use in mechanistic studies. To investigate this concept further, LCLs from two males (proband and sibling) were obtained from one such repository, the Autism Genetics Resource Exchange (AGRE), and iPSCs were generated by transfection with Epi5 Episomal iPSC reprogramming plasmids. Characterization of the resultant cell lines by PCR, RT-PCR, immunocytochemistry, karyotyping, and the Taqman® human pluripotent stem cell Scorecard™ Panel, was used to provide evidence of endogenous pluripotency and then to evaluate the trilineage potential of four representative clones. Results indicated that all four iPSC lines were initially pluripotent and displayed the trilineage potential predictive for successful differentiation to mesoderm, endoderm, or ectoderm-derived cell types. Compared to other published protocols, this study details a somewhat simplified approach, used here specifically for the generation and characterization of induced pluripotent stem cells from well-characterized and banked LCLs.
- Published
- 2021
- Full Text
- View/download PDF
13. Decoupling absorption and emission processes in super-resolution localization of emitters in a plasmonic hotspot
- Author
-
David L. Mack, Emiliano Cortés, Vincenzo Giannini, Peter Török, Tyler Roschuk, and Stefan A. Maier
- Subjects
Science - Abstract
Reporting the position of molecules and the electromagnetic enhancement in a plasmonic hotspot is difficult. Here Macket al. use a large Stokes-shifted molecule to spectrally decouple the emission process of the dye from the plasmonic system, keeping the absorption on resonance with the plasmon resonance of the antenna.
- Published
- 2017
- Full Text
- View/download PDF
14. Isolation and Mechanical Measurements of Myofibrils from Human Induced Pluripotent Stem Cell-Derived Cardiomyocytes
- Author
-
Josè Manuel Pioner, Alice W. Racca, Jordan M. Klaiman, Kai-Chun Yang, Xuan Guan, Lil Pabon, Veronica Muskheli, Rebecca Zaunbrecher, Jesse Macadangdang, Mark Y. Jeong, David L. Mack, Martin K. Childers, Deok-Ho Kim, Chiara Tesi, Corrado Poggesi, Charles E. Murry, and Michael Regnier
- Subjects
Medicine (General) ,R5-920 ,Biology (General) ,QH301-705.5 - Abstract
Summary: Tension production and contractile properties are poorly characterized aspects of excitation-contraction coupling of human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs). Previous approaches have been limited due to the small size and structural immaturity of early-stage hiPSC-CMs. We developed a substrate nanopatterning approach to produce hiPSC-CMs in culture with adult-like dimensions, T-tubule-like structures, and aligned myofibrils. We then isolated myofibrils from hiPSC-CMs and measured the tension and kinetics of activation and relaxation using a custom-built apparatus with fast solution switching. The contractile properties and ultrastructure of myofibrils more closely resembled human fetal myofibrils of similar gestational age than adult preparations. We also demonstrated the ability to study the development of contractile dysfunction of myofibrils from a patient-derived hiPSC-CM cell line carrying the familial cardiomyopathy MYH7 mutation (E848G). These methods can bring new insights to understanding cardiomyocyte maturation and developmental mechanical dysfunction of hiPSC-CMs with cardiomyopathic mutations. : In this article, Pioner and colleagues reported contractile properties of isolated myofibrils from hiPSC-CMs with highly mature morphology. This approach permits quantitative assessment of maturation and contractile properties of hiPSC-CMs and can be used to study the development of contractile dysfunction in genetically based cardiac diseases. The authors present a patient-derived cell line carrying a novel familial cardiomyopathy MYH7 mutation (E848G).
- Published
- 2016
- Full Text
- View/download PDF
15. Dystrophin-deficient cardiomyocytes derived from human urine: New biologic reagents for drug discovery
- Author
-
Xuan Guan, David L. Mack, Claudia M. Moreno, Jennifer L. Strande, Julie Mathieu, Yingai Shi, Chad D. Markert, Zejing Wang, Guihua Liu, Michael W. Lawlor, Emily C. Moorefield, Tara N. Jones, James A. Fugate, Mark E. Furth, Charles E. Murry, Hannele Ruohola-Baker, Yuanyuan Zhang, Luis F. Santana, and Martin K. Childers
- Subjects
Biology (General) ,QH301-705.5 - Abstract
The ability to extract somatic cells from a patient and reprogram them to pluripotency opens up new possibilities for personalized medicine. Induced pluripotent stem cells (iPSCs) have been employed to generate beating cardiomyocytes from a patient's skin or blood cells. Here, iPSC methods were used to generate cardiomyocytes starting from the urine of a patient with Duchenne muscular dystrophy (DMD). Urine was chosen as a starting material because it contains adult stem cells called urine-derived stem cells (USCs). USCs express the canonical reprogramming factors c-myc and klf4, and possess high telomerase activity. Pluripotency of urine-derived iPSC clones was confirmed by immunocytochemistry, RT-PCR and teratoma formation. Urine-derived iPSC clones generated from healthy volunteers and a DMD patient were differentiated into beating cardiomyocytes using a series of small molecules in monolayer culture. Results indicate that cardiomyocytes retain the DMD patient's dystrophin mutation. Physiological assays suggest that dystrophin-deficient cardiomyocytes possess phenotypic differences from normal cardiomyocytes. These results demonstrate the feasibility of generating cardiomyocytes from a urine sample and that urine-derived cardiomyocytes retain characteristic features that might be further exploited for mechanistic studies and drug discovery.
- Published
- 2014
- Full Text
- View/download PDF
16. Interferon-γ Elicits Pathological Hallmarks of ALS in Human Motor Neurons
- Author
-
Changho Chun, Jung Hyun Lee, Alec S.T. Smith, David L. Mack, Mark Bothwell, and Paul Nghiem
- Abstract
Neuroinflammation is an established factor contributing to amyotrophic lateral sclerosis (ALS) pathology, implicating the possible detrimental effects of inflammatory cytokines on motor neurons. The RNA/DNA-binding protein TDP-43 has emerged as a pivotal actor in ALS, because TDP-43 mutations cause familial ALS and loss of nuclear TDP-43, associated with its redistribution into cytoplasmic aggregates (TDP-43 proteinopathy) in motor neurons occurs in 97% of ALS cases. However, mechanisms linking neuroinflammation to TDP-43 mislocalization have not been described. Programmed death-ligand 1 (PD-L1) is an immune-modulatory protein, highly expressed on cell surfaces following acute inflammatory stress. To determine which inflammatory cytokines might impact motor neuron function, seven cytokines known to be elevated in ALS patients’ cerebrospinal fluid were tested for their effects on PD-L1 expression in human iPSC-derived motor neurons. Among the tested cytokines, only interferon-γ (IFN-γ) was found to strongly promote PD-L1 expression. Thus, we hypothesized that excessive exposure to IFN-γ may contribute to motor neuron degeneration in ALS. We observed that neuronal populations exposed to IFN-γ exhibited severe TDP-43 cytoplasmic aggregation and excitotoxic behavior correlated with impaired neural firing activity, hallmarks of ALS pathology, in both normal and ALS mutant (TARDB1K+/-) neurons. Single-cell RNA sequencing revealed possible mechanisms for these effects. Motor neurons exposed to IFN-γ exhibited an extensive shift of their gene expression profile toward a neurodegenerative phenotype. Notably, IFN-γ treatment induced aberrant expression levels for 70 genes that are listed in the recent literature as being dysregulated in various ALS subtypes. Additionally, we found that genes related to neuronal electrophysiology, protein aggregation, and TDP-43 misregulation were abnormally expressed in IFN-γ treated cells. Moreover, IFN-γ induced a significant reduction in the expression of genes that encode indispensable proteins for neuromuscular synapse development and maintenance, implying that the continuous cytokine exposure could directly impair signal transmission between motor axons and muscle membranes. Our findings suggest that IFN-γ could be a potent upstream pathogenic driver of ALS and provide potential candidates for future therapeutic targets to treat sporadic forms of ALS, which account for roughly 90% of reported cases.
- Published
- 2022
- Full Text
- View/download PDF
17. Creating stem cell‐derived neuromuscular junctions in vitro
- Author
-
David L. Mack, Alec S.T. Smith, and Shawn M. Luttrell
- Subjects
Motor Neurons ,Arc (protein) ,Tissue Engineering ,Physiology ,Drug discovery ,Stem Cells ,Induced Pluripotent Stem Cells ,Neuromuscular Junction ,Skeletal muscle ,Biology ,Neuromuscular junction ,In vitro ,Synapse ,Cellular and Molecular Neuroscience ,medicine.anatomical_structure ,Lab-On-A-Chip Devices ,Physiology (medical) ,medicine ,Animals ,Humans ,Neurology (clinical) ,Stem cell ,Induced pluripotent stem cell ,Neuroscience - Abstract
Recent development of novel therapies has improved mobility and quality of life for people suffering from inheritable neuromuscular disorders. Despite this progress, the majority of neuromuscular disorders are still incurable, in part due to a lack of predictive models of neuromuscular junction (NMJ) breakdown. Improvement of predictive models of a human NMJ would be transformative in terms of expanding our understanding of the mechanisms that underpin development, maintenance, and disease, and as a testbed with which to evaluate novel therapeutics. Induced pluripotent stem cells (iPSCs) are emerging as a clinically relevant and non-invasive cell source to create human NMJs to study synaptic development and maturation, as well as disease modeling and drug discovery. This review will highlight the recent advances and remaining challenges to generating an NMJ capable of eliciting contraction of stem cell-derived skeletal muscle in vitro. We explore the advantages and shortcomings of traditional NMJ culturing platforms, as well as the pioneering technologies and novel, biomimetic culturing systems currently in use to guide development and maturation of the neuromuscular synapse and extracellular microenvironment. Then, we will explore how this NMJ-in-a-dish can be used to study normal assembly and function of the efferent portion of the neuromuscular arc, and how neuromuscular disease-causing mutations disrupt structure, signaling, and function.
- Published
- 2021
- Full Text
- View/download PDF
18. Increased tissue stiffness triggers contractile dysfunction and telomere shortening in dystrophic cardiomyocytes
- Author
-
Edward L. LaGory, Colin Holbrook, Sang-Ging Ong, Chris Denning, Andrew H. Chang, John W. Day, Martin K. Childers, Alexandre J.S. Ribeiro, Honghui Wang, Alex C.Y. Chang, Gaspard Pardon, Joseph C. Wu, Kassie Koleckar, Sara Ancel, Helen M. Blau, David L. Mack, John Ramunas, Amato J. Giaccia, Asuka Eguchi, Haodi Wu, and Beth L. Pruitt
- Subjects
0301 basic medicine ,Duchenne muscular dystrophy ,Cardiomyopathy ,Fluorescent Antibody Technique ,Gene Expression ,Cardiovascular ,Biochemistry ,Muscular Dystrophies ,hiPSC-CM ,0302 clinical medicine ,Fibrosis ,2.1 Biological and endogenous factors ,Myocytes, Cardiac ,Muscular Dystrophy ,Cells, Cultured ,Telomere Shortening ,Pediatric ,telomere ,Stem Cell Research - Induced Pluripotent Stem Cell - Human ,Cell Differentiation ,Cell biology ,Cellular Microenvironment ,Mechanosensitive channels ,Cardiomyopathies ,Duchenne/ Becker Muscular Dystrophy ,musculoskeletal diseases ,DNA damage ,Intellectual and Developmental Disabilities (IDD) ,Telomere Capping ,Clinical Sciences ,Induced Pluripotent Stem Cells ,Bioengineering ,Biology ,Article ,Immunophenotyping ,03 medical and health sciences ,Rare Diseases ,Downregulation and upregulation ,DMD ,Genetics ,medicine ,Humans ,Mechanical Phenomena ,Stem Cell Research - Induced Pluripotent Stem Cell ,fibrosis ,Cell Biology ,Stem Cell Research ,medicine.disease ,Myocardial Contraction ,Brain Disorders ,Telomere ,dilated cardiomyopathy ,Muscular Dystrophy, Duchenne ,Orphan Drug ,030104 developmental biology ,Musculoskeletal ,Culture Media, Conditioned ,Biochemistry and Cell Biology ,030217 neurology & neurosurgery ,Biomarkers ,Developmental Biology - Abstract
Summary Duchenne muscular dystrophy (DMD) is a rare X-linked recessive disease that is associated with severe progressive muscle degeneration culminating in death due to cardiorespiratory failure. We previously observed an unexpected proliferation-independent telomere shortening in cardiomyocytes of a DMD mouse model. Here, we provide mechanistic insights using human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs). Using traction force microscopy, we show that DMD hiPSC-CMs exhibit deficits in force generation on fibrotic-like bioengineered hydrogels, aberrant calcium handling, and increased reactive oxygen species levels. Furthermore, we observed a progressive post-mitotic telomere shortening in DMD hiPSC-CMs coincident with downregulation of shelterin complex, telomere capping proteins, and activation of the p53 DNA damage response. This telomere shortening is blocked by blebbistatin, which inhibits contraction in DMD cardiomyocytes. Our studies underscore the role of fibrotic stiffening in the etiology of DMD cardiomyopathy. In addition, our data indicate that telomere shortening is progressive, contraction dependent, and mechanosensitive, and suggest points of therapeutic intervention., Highlights • DMD hiPSC-CMs exhibit aberrant calcium handling and defective force generation • DMD hiPSC-CMs undergo proliferation-independent telomere shortening • Telomere shortening activates the p53 DNA damage pathway • Telomere shortening in DMD hiPSC-CMs is contraction dependent, In this article, Chang, Blau, and colleagues show that Duchenne muscular dystrophy (DMD) iPSC-derived cardiomyocytes exhibit proliferation-independent telomere shortening, p53 activation and mitochondrial dysfunction.
- Published
- 2021
19. Perpendicular Shear Stresses Drive Transmural Helical Remodeling in Engineered Human Ventricular Models
- Author
-
Nisa P. Williams, Kevin M. Beussman, John R. Foster, Marcus Rhodehamel, Charles A. Williams, Jonathan H. Tsui, Alec S.T. Smith, David L. Mack, Charles E. Murry, Nathan J. Sniadecki, and Deok-Ho Kim
- Abstract
Tissue engineering with human induced pluripotent stem cell-derived cardiomyocytes enables unique opportunities for creating physiological models of the heart in vitro. However, there are few approaches available that can recapitulate the complex structure-function relationships that govern cardiac function at the macroscopic organ level. Here, we report a down-scaled, conical human 3D ventricular model with controllable cellular organization using multilayered, patterned cardiac sheets. Tissue engineered ventricles whose cardiomyocytes were pre-aligned parallel or perpendicular to the long axis outperformed those whose cardiomyocytes were angled or randomly oriented. Notably, the inner layers of perpendicular cardiac sheets realigned over 4 days into a parallel orientation, creating a helical transmural architecture, whereas minimal remodeling occurred in the parallel or angled sheets. Finite element analysis of engineered ventricles demonstrated that circumferential alignment leads to maximal perpendicular shear stress at the inner layer, whereas longitudinal orientation leads to maximal parallel stress. We hypothesize that cellular remodeling occurs to reduce perpendicular shear stresses in myocardium. This advanced platform provides evidence that physical forces such as shear stress drive self-organization of cardiac architecture.
- Published
- 2022
- Full Text
- View/download PDF
20. Role of Topographic Cues in Engineering the Muscle Niche
- Author
-
Jesse Macadangdang, Christian Mandrycky, Changho Chun, Nicholas A. Geisse, David L. Mack, and Alec S. T. Smith
- Abstract
Cells are exquisitely receptive to the physical cues present within their native microenvironment. Manipulation of substrate topography is therefore a simple strategy to promote the development of cells in vitro toward a phenotype that is more representative of their in vivo counterparts. In the cases of cardiac and skeletal muscle, substrate topographies have been used to promote uniaxial alignment, myofibrillar development, and cytoskeletal organization in cultured cells for downstream applications in basic biological studies, disease modelling, and drug screening. In this chapter, we review the advantages conferred on muscle cultures by topographic patterns, discuss methods for producing patterns of different dimensions, and provide a perspective on the role these technologies could play in enhancing the predictive power of next generation preclinical assays.
- Published
- 2022
- Full Text
- View/download PDF
21. Incorporation of sensing modalities into de novo designed fluorescence-activating proteins
- Author
-
Samantha Bremner, Lindsey Doyle, Andre Berndt, Jason C. Klima, David L. Mack, Jiayi Dou, Lauren Carter, Justin Daho Lee, Emilia P. Barros, Joshua C. Vaughan, Cameron M. Chow, David Baker, Min Yen Lee, Michael Rappleye, Rommie E. Amaro, Jacob S. Quon, Lauren A. Gagnon, Anastassia A. Vorobieva, Barry L. Stoddard, and Department of Bio-engineering Sciences
- Subjects
Models, Molecular ,0301 basic medicine ,Chemistry(all) ,Science ,Green Fluorescent Proteins ,Gfp reporter ,General Physics and Astronomy ,Bioengineering ,Physics and Astronomy(all) ,01 natural sciences ,Article ,Fluorescence ,General Biochemistry, Genetics and Molecular Biology ,Green fluorescent protein ,03 medical and health sciences ,Models ,Chlorocebus aethiops ,0103 physical sciences ,Animals ,Humans ,Fluorescent protein ,Acetylcholine metabolism ,X-ray crystallography ,Fluorescent Dyes ,Multidisciplinary ,010304 chemical physics ,Biochemistry, Genetics and Molecular Biology(all) ,Extramural ,Chemistry ,Molecular ,General Chemistry ,Fluorescent proteins ,Hydrogen-Ion Concentration ,Acetylcholine ,Luminescent Proteins ,Fluorescence intensity ,Wide-field fluorescence microscopy ,HEK293 Cells ,030104 developmental biology ,COS Cells ,Biophysics ,Calcium ,Generic health relevance ,Protein design - Abstract
Through the efforts of many groups, a wide range of fluorescent protein reporters and sensors based on green fluorescent protein and its relatives have been engineered in recent years. Here we explore the incorporation of sensing modalities into de novo designed fluorescence-activating proteins, called mini-fluorescence-activating proteins (mFAPs), that bind and stabilize the fluorescent cis-planar state of the fluorogenic compound DFHBI. We show through further design that the fluorescence intensity and specificity of mFAPs for different chromophores can be tuned, and the fluorescence made sensitive to pH and Ca2+ for real-time fluorescence reporting. Bipartite split mFAPs enable real-time monitoring of protein–protein association and (unlike widely used split GFP reporter systems) are fully reversible, allowing direct readout of association and dissociation events. The relative ease with which sensing modalities can be incorporated and advantages in smaller size and photostability make de novo designed fluorescence-activating proteins attractive candidates for optical sensor engineering., Fluorescent protein reporters based on GFP exist, but have intrinsic disadvantages. Here the authors incorporate pH, Ca2+ and protein–protein interaction sensing modalities into de novo designed mini-fluorescence-activating proteins (mFAPs), with increased photostability and smaller size, which bind a range of DFHBI chromophore variants.
- Published
- 2021
22. High-throughput, real-time monitoring of engineered skeletal muscle function using magnetic sensing
- Author
-
Alec ST Smith, Shawn M Luttrell, Jean-Baptiste Dupont, Kevin Gray, Daniel Lih, Jacob W Fleming, Nathan J Cunningham, Sofia Jepson, Jennifer Hesson, Julie Mathieu, Lisa Maves, Bonnie J Berry, Elliot C Fisher, Nathan J Sniadecki, Nicholas A Geisse, and David L Mack
- Subjects
Biomaterials ,Biomedical Engineering ,Medicine (miscellaneous) - Abstract
Engineered muscle tissues represent powerful tools for examining tissue level contractile properties of skeletal muscle. However, limitations in the throughput associated with standard analysis methods limit their utility for longitudinal study, high throughput drug screens, and disease modeling. Here we present a method for integrating 3D engineered skeletal muscles with a magnetic sensing system to facilitate non-invasive, longitudinal analysis of developing contraction kinetics. Using this platform, we show that engineered skeletal muscle tissues derived from both induced pluripotent stem cell and primary sources undergo improvements in contractile output over time in culture. We demonstrate how magnetic sensing of contractility can be employed for simultaneous assessment of multiple tissues subjected to different doses of known skeletal muscle inotropes as well as the stratification of healthy versus diseased functional profiles in normal and dystrophic muscle cells. Based on these data, this combined culture system and magnet-based contractility platform greatly broadens the potential for 3D engineered skeletal muscle tissues to impact the translation of novel therapies from the lab to the clinic.
- Published
- 2022
23. ACTA1 H40Y mutant iPSC-derived skeletal myocytes display mitochondrial defects in an in vitro model of nemaline myopathy
- Author
-
Melanie Gartz, Margaret Haberman, Jessica Sutton, Rebecca A. Slick, Shawn M. Luttrell, David L. Mack, and Michael W. Lawlor
- Subjects
Cell Biology - Published
- 2023
- Full Text
- View/download PDF
24. A Change of Heart: Human Cardiac Tissue Engineering as a Platform for Drug Development
- Author
-
Samantha B. Bremner, Karen S. Gaffney, Nathan J. Sniadecki, and David L. Mack
- Subjects
Pluripotent Stem Cells ,Drug Development ,Tissue Engineering ,Induced Pluripotent Stem Cells ,Humans ,Arrhythmias, Cardiac ,Cell Differentiation ,Myocytes, Cardiac ,Cardiology and Cardiovascular Medicine - Abstract
Purpose of Review Human cardiac tissue engineering holds great promise for early detection of drug-related cardiac toxicity and arrhythmogenicity during drug discovery and development. We describe shortcomings of the current drug development pathway, recent advances in the development of cardiac tissue constructs as drug testing platforms, and the challenges remaining in their widespread adoption. Recent Findings Human pluripotent stem cell-derived cardiomyocytes (hPSC-CMs) have been used to develop a variety of constructs including cardiac spheroids, microtissues, strips, rings, and chambers. Several ambitious studies have used these constructs to test a significant number of drugs, and while most have shown proper negative inotropic and arrhythmogenic responses, few have been able to demonstrate positive inotropy, indicative of relative hPSC-CM immaturity. Summary Several engineered human cardiac tissue platforms have demonstrated native cardiac physiology and proper drug responses. Future studies addressing hPSC-CM immaturity and inclusion of patient-specific cell lines will further advance the utility of such models for in vitro drug development.
- Published
- 2022
25. Full-Length Dystrophin Deficiency Leads to Contractile and Calcium Transient Defects in Human Engineered Heart Tissues
- Author
-
Samantha B Bremner, Christian J Mandrycky, Andrea Leonard, Ruby M Padgett, Alan R Levinson, Ethan S Rehn, J Manuel Pioner, Nathan J Sniadecki, and David L Mack
- Subjects
Biomaterials ,History ,Polymers and Plastics ,Biomedical Engineering ,Medicine (miscellaneous) ,Business and International Management ,Industrial and Manufacturing Engineering - Abstract
Cardiomyopathy is currently the leading cause of death for patients with Duchenne muscular dystrophy (DMD), a severe neuromuscular disorder affecting young boys. Animal models have provided insight into the mechanisms by which dystrophin protein deficiency causes cardiomyopathy, but there remains a need to develop human models of DMD to validate pathogenic mechanisms and identify therapeutic targets. Here, we have developed human engineered heart tissues (EHTs) from CRISPR-edited, human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) expressing a truncated dystrophin protein lacking part of the actin-binding domain. The 3D EHT platform enables direct measurement of contractile force, simultaneous monitoring of Ca2+ transients, and assessment of myofibril structure. Dystrophin-mutant EHTs produced less contractile force as well as delayed kinetics of force generation and relaxation, as compared to isogenic controls. Contractile dysfunction was accompanied by reduced sarcomere length, increased resting cytosolic Ca2+ levels, delayed Ca2+ release and reuptake, and increased beat rate irregularity. Transcriptomic analysis revealed clear differences between dystrophin-deficient and control EHTs, including downregulation of genes related to Ca2+ homeostasis and extracellular matrix organization, and upregulation of genes related to regulation of membrane potential, cardiac muscle development, and heart contraction. These findings indicate that the EHT platform provides the cues necessary to expose the clinically-relevant, functional phenotype of force production as well as mechanistic insights into the role of Ca2+ handling and transcriptomic dysregulation in dystrophic cardiac function, ultimately providing a powerful platform for further studies in disease modeling and drug discovery.
- Published
- 2022
- Full Text
- View/download PDF
26. Characterizing the functional impact of embryonic myosin mutations in stem cell-derived skeletal muscle
- Author
-
Christian Mandrycky, Saffie Mohran, Matthew C. Childers, Alec S.T. Smith, Michael Regnier, and David L. Mack
- Subjects
Biophysics - Published
- 2023
- Full Text
- View/download PDF
27. Absence of full-length dystrophin impairs normal maturation and contraction of cardiomyocytes derived from human-induced pluripotent stem cells
- Author
-
Corrado Poggesi, Chiara Tesi, Alice Ward Racca, Michael R. Hoopmann, Cecilia Ferrantini, J. Manuel Pioner, Lil Pabon, Jordan M. Klaiman, Veronica Muskheli, Michael Regnier, Xuan Guan, Martin K. Childers, Deok Ho Kim, David L. Mack, Charles E. Murry, Robert L. Moritz, and Jesse Macadangdang
- Subjects
musculoskeletal diseases ,Physiology ,Duchenne muscular dystrophy ,Cellular differentiation ,Induced Pluripotent Stem Cells ,Cardiomyopathy ,Cell Line ,Dystrophin ,03 medical and health sciences ,0302 clinical medicine ,Myofibrils ,Physiology (medical) ,medicine ,Humans ,Myocytes, Cardiac ,Calcium Signaling ,Muscular dystrophy ,Induced pluripotent stem cell ,030304 developmental biology ,0303 health sciences ,biology ,Cell Differentiation ,Original Articles ,medicine.disease ,Myocardial Contraction ,Cell biology ,Muscular Dystrophy, Duchenne ,Kinetics ,Cell culture ,Case-Control Studies ,biology.protein ,CRISPR-Cas9 genome editing ,Human iPSC-cardiomyocytes ,Muscular Dystrophy ,dystrophin ,myofibrils ,Cardiomyopathies ,Cardiology and Cardiovascular Medicine ,Myofibril ,030217 neurology & neurosurgery - Abstract
Aims Heart failure invariably affects patients with various forms of muscular dystrophy (MD), but the onset and molecular sequelae of altered structure and function resulting from full-length dystrophin (Dp427) deficiency in MD heart tissue are poorly understood. To better understand the role of dystrophin in cardiomyocyte development and the earliest phase of Duchenne muscular dystrophy (DMD) cardiomyopathy, we studied human cardiomyocytes differentiated from induced pluripotent stem cells (hiPSC-CMs) obtained from the urine of a DMD patient. Methods and results The contractile properties of patient-specific hiPSC-CMs, with no detectable dystrophin (DMD-CMs with a deletion of exon 50), were compared to CMs containing a CRISPR-Cas9 mediated deletion of a single G base at position 263 of the dystrophin gene (c.263delG-CMs) isogenic to the parental line of hiPSC-CMs from a healthy individual. We hypothesized that the absence of a dystrophin-actin linkage would adversely affect myofibril and cardiomyocyte structure and function. Cardiomyocyte maturation was driven by culturing long-term (80–100 days) on a nanopatterned surface, which resulted in hiPSC-CMs with adult-like dimensions and aligned myofibrils. Conclusions Our data demonstrate that lack of Dp427 results in reduced myofibril contractile tension, slower relaxation kinetics, and to Ca2+ handling abnormalities, similar to DMD cells, suggesting either retarded or altered maturation of cardiomyocyte structures associated with these functions. This study offers new insights into the functional consequences of Dp427 deficiency at an early stage of cardiomyocyte development in both patient-derived and CRISPR-generated models of dystrophin deficiency.
- Published
- 2019
- Full Text
- View/download PDF
28. The Role of Extracellular Vesicles in the Progression of ALS and Their Potential as Biomarkers and Therapeutic Agents with Which to Combat the Disease
- Author
-
Mark Bothwell, Changho Chun, David L. Mack, and Alec S.T. Smith
- Subjects
business.industry ,InformationSystems_INFORMATIONSTORAGEANDRETRIEVAL ,Cancer research ,Medicine ,Disease ,business ,GeneralLiterature_REFERENCE(e.g.,dictionaries,encyclopedias,glossaries) ,Extracellular vesicles - Published
- 2020
- Full Text
- View/download PDF
29. Voluntary wheel running complements microdystrophin gene therapy to improve muscle function in mdx mice
- Author
-
Jean-Baptiste Dupont, Daniel D. Fathalikhani, Haiyan Zhang, Michael W. Lawlor, Justin B. Perry, Ryan P. McMillan, Shelby E. Hamm, Carl Morris, David Brown, J. Patrick Gonzalez, Robert W. Grange, Mark A. Vanden Avond, Katherine E. Bukovec, Kirsten E. Coleman, Suresh Kumar, David L. Mack, Mariah J. Prom, and Adele K. Addington
- Subjects
0301 basic medicine ,medicine.medical_specialty ,muscle pathology ,Duchenne muscular dystrophy ,Genetic enhancement ,QH426-470 ,03 medical and health sciences ,myosin heavy chain ,0302 clinical medicine ,Internal medicine ,mitochondrial respiration ,Myosin ,medicine ,Genetics ,Treadmill ,Molecular Biology ,duchenne muscular dystrophy ,dystrophic grade ,endurance ,muscle power ,QH573-671 ,business.industry ,muscle physiology ,Correction ,medicine.disease ,Mitochondrial respiration ,Diaphragm (structural system) ,030104 developmental biology ,Endocrinology ,Turnover ,030220 oncology & carcinogenesis ,Wheel running ,Molecular Medicine ,durability ,voluntary exercise ,Original Article ,business ,Cytology - Abstract
We tested the hypothesis that voluntary wheel running would complement microdystrophin gene therapy to improve muscle function in young mdx mice, a model of Duchenne muscular dystrophy. mdx mice injected with a single dose of AAV9-CK8-microdystrophin or vehicle at age 7 weeks were assigned to three groups: mdxRGT (run, gene therapy), mdxGT (no run, gene therapy), or mdx (no run, no gene therapy). Wild-type (WT) mice were assigned to WTR (run) and WT (no run) groups. WTR and mdxRGT performed voluntary wheel running for 21 weeks; remaining groups were cage active. Robust expression of microdystrophin occurred in heart and limb muscles of treated mice. mdxRGT versus mdxGT mice showed increased microdystrophin in quadriceps but decreased levels in diaphragm. mdx final treadmill fatigue time was depressed compared to all groups, improved in mdxGT, and highest in mdxRGT. Both weekly running distance (km) and final treadmill fatigue time for mdxRGT and WTR were similar. Remarkably, mdxRGT diaphragm power was only rescued to 60% of WT, suggesting a negative impact of running. However, potential changes in fiber type distribution in mdxRGT diaphragms could indicate an adaptation to trade power for endurance. Post-treatment in vivo maximal plantar flexor torque relative to baseline values was greater for mdxGT and mdxRGT versus all other groups. Mitochondrial respiration rates from red quadriceps fibers were significantly improved in mdxGT animals, but the greatest bioenergetic benefit was observed in the mdxRGT group. Additional assessments revealed partial to full functional restoration in mdxGT and mdxRGT muscles relative to WT. These data demonstrate that voluntary wheel running combined with microdystrophin gene therapy in young mdx mice improved whole-body performance, affected muscle function differentially, mitigated energetic deficits, but also revealed some detrimental effects of exercise. With microdystrophin gene therapy currently in clinical trials, these data may help us understand the potential impact of exercise in treated patients., Graphical abstract, We report that voluntary exercise complements microdystrophin gene therapy in the mdx mouse, a model of Duchenne muscular dystrophy (DMD), by increasing running endurance, muscle strength, and energy production. These and other findings may help us understand how exercise in DMD boys may influence microdystrophin gene therapy.
- Published
- 2020
30. Embryonic myosin mutations T187I and R672C result in mechanical and structural dysfunction in hiPSC derived skeletal myotubes
- Author
-
Saffie Mohran, Matthew C. Childers, Christian Mandrycky, Shawn M. Luttrell, Kati Buckingham, Michael Bamshad, David L. Mack, and Michael Regnier
- Subjects
Biophysics - Published
- 2022
- Full Text
- View/download PDF
31. Advances and Current Challenges Associated with the Use of Human Induced Pluripotent Stem Cells in Modeling Neurodegenerative Disease
- Author
-
David L. Mack, Bonnie J. Berry, Alec S.T. Smith, and Jessica E. Young
- Subjects
Animal Use Alternatives ,Neurons ,0301 basic medicine ,Histology ,Computer science ,business.industry ,Drug discovery ,Induced Pluripotent Stem Cells ,Cell Culture Techniques ,Cell Differentiation ,Neurodegenerative Diseases ,Disease ,Epigenesis, Genetic ,03 medical and health sciences ,030104 developmental biology ,Humans ,Identification (biology) ,Epigenetics ,Personalized medicine ,Anatomy ,Stem cell ,Human Induced Pluripotent Stem Cells ,business ,Induced pluripotent stem cell ,Neuroscience - Abstract
One of the most profound advances in the last decade of biomedical research has been the development of human induced pluripotent stem cell (hiPSC) models for identification of disease mechanisms and drug discovery. Human iPSC technology has the capacity to revolutionize healthcare and the realization of personalized medicine, but differentiated tissues derived from stem cells come with major criticisms compared to native tissue, including variability in genetic backgrounds, a lack of functional maturity, and differences in epigenetic profiles. It is widely believed that increasing complexity will lead to improved clinical relevance, so methods are being developed that go from a single cell type to various levels of 2-D coculturing and 3-D organoids. As this inevitable trend continues, it will be essential to thoroughly understand the strengths and weaknesses of more complex models and to develop criteria for assessing biological relevance. We believe the payoff of robust, high-throughput, clinically meaningful human stem cell models could be the elimination of often inadequate animal models. To facilitate this transition, we will look at the challenges and strategies of complex model development through the lens of neurodegeneration to encapsulate where the disease-in-a-dish field currently is and where it needs to go to improve.
- Published
- 2018
- Full Text
- View/download PDF
32. Solubilized Amnion Membrane Hyaluronic Acid Hydrogel Accelerates Full‐Thickness Wound Healing
- Author
-
Sean V. Murphy, John D. Jackson, Lujie Song, Khiry Sutton, Anthony Atala, Aleksander Skardal, David L. Mack, Shay Soker, and Rebecca Haug
- Subjects
0301 basic medicine ,Keratinocytes ,Male ,Cryopreservation ,030207 dermatology & venereal diseases ,chemistry.chemical_compound ,Wound care ,Mice ,0302 clinical medicine ,Translational Research Articles and Reviews ,Re-Epithelialization ,Hyaluronic acid ,Hyaluronic Acid ,Cells, Cultured ,Skin ,Amnion ,integumentary system ,Tissue Scaffolds ,Chemistry ,Biomaterial ,Hydrogels ,General Medicine ,Membrane ,medicine.anatomical_structure ,Cytokines ,Intercellular Signaling Peptides and Proteins ,Female ,Stem cell ,medicine.medical_specialty ,Wound healing ,Mice, Nude ,Amniotic Stem Cells ,03 medical and health sciences ,Tissue Engineering and Regenerative Medicine ,medicine ,Animals ,Humans ,Tissue Engineering ,Cell Biology ,Fibroblasts ,Surgery ,Wound Healing / Fibrosis ,Hydrogel ,030104 developmental biology ,Solubilized amnion membrane ,Developmental Biology ,Biomedical engineering - Abstract
The early and effective treatment of wounds is vital to ensure proper wound closure and healing with appropriate functional and cosmetic outcomes. The use of human amnion membranes for wound care has been shown to be safe and effective. However, the difficulty in handling and placing thin sheets of membrane, and the high costs associated with the use of living cellularized tissue has limited the clinical application of amniotic membrane wound healing products. Here, we describe a novel amnion membrane-derived product, processed to result in a cell-free solution, while maintaining high concentrations of cell-derived cytokines and growth factors. The solubilized amnion membrane (SAM) combined with the carrier hyaluronic acid (HA) hydrogel (HA-SAM) is easy to produce, store, and apply to wounds. We demonstrated the efficacy of HA-SAM as a wound treatment using a full-thickness murine wound model. HA-SAM significantly accelerated wound closure through re-epithelialization and prevented wound contraction. HA-SAM-treated wounds had thicker regenerated skin, increased total number of blood vessels, and greater numbers of proliferating keratinocytes within the epidermis. Overall, this study confirms the efficacy of the amnion membrane as a wound treatment/dressing, and overcomes many of the limitations associated with using fresh, cryopreserved, or dehydrated tissue by providing a hydrogel delivery system for SAM.
- Published
- 2017
33. Taking the Next Steps in Regenerative Rehabilitation: Establishment of a New Interdisciplinary Field
- Author
-
Christopher L. Dearth, David L. Mack, Kenton W. Gregory, George J. Christ, Joseph A. Roche, Carmelo Chisari, Paul F. Pasquina, Tomoki Aoyama, Steven L. Wolf, Nick J. Willett, Kimberly S. Topp, Joseph P. Hart, Martin J. Stoddart, Marzia Bedoni, William R. Wagner, Michael L. Boninger, Gerard A. Malanga, Hua Xie, Alice Gualerzi, Lloyd F. Rose, Laura J. Miller, Trevor A. Dyson-Hudson, Stuart J. Warden, M. Terry Loghmani, Luis M. Alvarez, Junichi Tajino, Fabrisia Ambrosio, Thomas A. Rando, Kelley Ann Brix, Carmen M. Terzic, Stephen M. Goldman, Akira Ito, Linda J. Noble-Haeusslein, Hiroshi Kuroki, and Christopher H. Evans
- Subjects
030506 rehabilitation ,Certification ,medicine.medical_treatment ,education ,MEDLINE ,Physical Therapy, Sports Therapy and Rehabilitation ,Regenerative Medicine ,Regenerative medicine ,Article ,Food and drug administration ,03 medical and health sciences ,0302 clinical medicine ,Rehabilitation ,Tissue engineering ,Congresses as Topic ,Curriculum ,Fellowships and Scholarships ,Humans ,medicine ,Interdisciplinarity ,Clinical Practice ,Engineering ethics ,0305 other medical science ,Psychology ,030217 neurology & neurosurgery - Abstract
The growing field of regenerative rehabilitation has great potential to improve clinical outcomes for individuals with disabilities. However, the science to elucidate the specific biological underpinnings of regenerative rehabilitation-based approaches is still in its infancy and critical questions regarding clinical translation and implementation still exist. In a recent roundtable discussion from International Consortium for Regenerative Rehabilitation stakeholders, key challenges to progress in the field were identified. The goal of this article is to summarize those discussions and to initiate a broader discussion among clinicians and scientists across the fields of regenerative medicine and rehabilitation science to ultimately progress regenerative rehabilitation from an emerging field to an established interdisciplinary one. Strategies and case studies from consortium institutions-including interdisciplinary research centers, formalized courses, degree programs, international symposia, and collaborative grants-are presented. We propose that these strategic directions have the potential to engage and train clinical practitioners and basic scientists, transform clinical practice, and, ultimately, optimize patient outcomes.
- Published
- 2020
34. Astrocyte-derived extracellular vesicles enhance the survival and electrophysiological function of human cortical neurons in vitro
- Author
-
Jung Hyun Lee, Hye-Jin Kim, Jong Bum Lee, Deok Ho Kim, Mark Bothwell, Changho Chun, Dana S. Kamenz, David L. Mack, Alec S.T. Smith, and Claire D. Clelland
- Subjects
Apolipoprotein E ,Nervous system ,Proteomics ,Population ,Induced Pluripotent Stem Cells ,Biophysics ,Bioengineering ,02 engineering and technology ,Neuroprotection ,Article ,Biomaterials ,03 medical and health sciences ,Extracellular Vesicles ,Heat shock protein ,medicine ,Humans ,education ,Cells, Cultured ,030304 developmental biology ,Neurons ,0303 health sciences ,education.field_of_study ,Chemistry ,Peripheral Nervous System Diseases ,021001 nanoscience & nanotechnology ,LRP1 ,Cell biology ,Electrophysiology ,medicine.anatomical_structure ,Mechanics of Materials ,Astrocytes ,Ceramics and Composites ,0210 nano-technology ,Astrocyte - Abstract
Neurons derived from human induced pluripotent stem cells (hiPSCs) are powerful tools for modeling neural pathophysiology and preclinical efficacy/toxicity screening of novel therapeutic compounds. However, human neurons cultured in vitro typically do not fully recapitulate the physiology of the human nervous system, especially in terms of exhibiting morphological maturation, longevity, and electrochemical signaling ability comparable to that of adult human neurons. In this study, we investigated the potential for astrocyte-derived extracellular vesicles (EVs) to modulate survival and electrophysiological function of human neurons in vitro. Specifically, we demonstrate that EVs obtained from human astrocytes promote enhanced single cell electrophysiological function and anti-apoptotic behavior in a homogeneous population of human iPSC-derived cortical neurons. Furthermore, EV-proteomic analysis was performed to identify cargo proteins with the potential to promote the physiological enhancement observed. EV cargos were found to include neuroprotective proteins such as heat shock proteins, alpha-synuclein, and lipoprotein receptor-related protein 1 (LRP1), as well as apolipoprotein E (APOE), which negatively regulates neuronal apoptosis, and a peroxidasin homolog that supports neuronal oxidative stress management. Proteins that positively regulate neuronal excitability and synaptic development were also detected, such as potassium channel tetramerization domain containing 12 (KCTD12), glucose-6- phosphate dehydrogenase (G6PD), kinesin family member 5B (KIF5B), spectrin-alpha non-erythrocytic1 (SPTAN1). The remarkable improvements in electrophysiological function and evident inhibition of apoptotic signaling in cultured neurons exposed to these cargos may hold significance for improving preclinical in vitro screening modalities. In addition, our collected data highlight the potential for EV-based therapeutics as a potential class of future clinical treatment for tackling inveterate central and peripheral neuropathies.
- Published
- 2020
35. A More Open Approach Is Needed to Develop Cell-Based Fish Technology: It Starts with Zebrafish
- Author
-
David L. Mack, Lisa Maves, Alec S.T. Smith, Jeanot Muster, Wendy W. Weston, Greg Potter, Alain Rostain, Nguyen T.K. Vo, and Alessandro Bertero
- Subjects
protein retention ,Natural resource economics ,Fish farming ,animal protein ,cell-based meat ,cellular agriculture ,cultivated meat ,lean fish ,ocean health ,protein ,sustainability ,zebrafish ,Earth and Planetary Sciences (miscellaneous) ,Environmental impact assessment ,Protein retention ,Zebrafish ,General Environmental Science ,biology ,food and beverages ,biology.organism_classification ,Natural resource ,Animal protein ,Sustainability ,Business ,Cell based - Abstract
The global demand for fish is rising and projected to increase for years to come. However, there is uncertainty whether this increased demand can be met by the conventional approaches of capture fisheries and fish farming because of wild stock depletion, natural resource requirements, and environmental impact concerns. One proposed complementary solution is to manufacture the same meat directly from fish cells, as cell-based fish. More than 30 ventures are competing to commercialize cell-based meat broadly, but the field lacks a foundation of shared scientific knowledge, which threatens to delay progress. Here, we recommend taking a research-focused, more open and collaborative approach to cell-based fish meat development that targets lean fish and an unlikely but very attractive candidate for accelerating research and development, the zebrafish. Although substantial work lies ahead, cell-based meat technology could prove to be a more efficient, less resource-intensive method of producing lean fish meat.
- Published
- 2020
36. Nicorandil, a Nitric Oxide Donor and ATP-Sensitive Potassium Channel Opener, Protects Against Dystrophin-Deficient Cardiomyopathy
- Author
-
Zhi-Dong Ge, Martin K. Childers, Muhammad Afzal, David L. Mack, Allison D. Ebert, Jered V. McGivern, Xuan Guan, Courtney Gastonguay, Melanie Reiter, and Jennifer L. Strande
- Subjects
Male ,0301 basic medicine ,Cardiomyopathy ,Mitochondrion ,Pharmacology ,Mitochondria, Heart ,Ventricular Function, Left ,chemistry.chemical_compound ,KATP Channels ,Myocytes, Cardiac ,Pharmacology (medical) ,Nicorandil ,Heart metabolism ,Cardioprotection ,biology ,Cardiology ,Cardiomyopathies ,Cardiology and Cardiovascular Medicine ,Dystrophin ,Signal Transduction ,medicine.drug ,musculoskeletal diseases ,Xanthine Oxidase ,medicine.medical_specialty ,ATP-sensitive potassium channel ,Induced Pluripotent Stem Cells ,Myocardial Reperfusion Injury ,Nitric Oxide ,Article ,Cell Line ,Nitric oxide ,03 medical and health sciences ,Internal medicine ,medicine ,Animals ,Humans ,Nitric Oxide Donors ,Dose-Response Relationship, Drug ,business.industry ,Recovery of Function ,Muscular Dystrophy, Animal ,medicine.disease ,Disease Models, Animal ,Oxidative Stress ,030104 developmental biology ,chemistry ,Mice, Inbred mdx ,biology.protein ,Reactive Oxygen Species ,business - Abstract
Background: Dystrophin-deficient cardiomyopathy is a growing clinical problem without targeted treatments. We investigated whether nicorandil promotes cardioprotection in human dystrophin-deficient induced pluripotent stem cell (iPSC)-derived cardiomyocytes and the muscular dystrophy mdx mouse heart. Methods and Results: Dystrophin-deficient iPSC-derived cardiomyocytes had decreased levels of endothelial nitric oxide synthase and neuronal nitric oxide synthase. The dystrophin-deficient cardiomyocytes had increased cell injury and death after 2 hours of stress and recovery. This was associated with increased levels of reactive oxygen species and dissipation of the mitochondrial membrane potential. Nicorandil pretreatment was able to abolish these stress-induced changes through a mechanism that involved the nitric oxide–cyclic guanosine monophosphate pathway and mitochondrial adenosine triphosphate-sensitive potassium channels. The increased reactive oxygen species levels in the dystrophin-deficient cardiomyocytes were associated with diminished expression of select antioxidant genes and increased activity of xanthine oxidase. Furthermore, nicorandil was found to improve the restoration of cardiac function after ischemia and reperfusion in the isolated mdx mouse heart. Conclusion: Nicorandil protects against stress-induced cell death in dystrophin-deficient cardiomyocytes and preserves cardiac function in the mdx mouse heart subjected to ischemia and reperfusion injury. This suggests a potential therapeutic role for nicorandil in dystrophin-deficient cardiomyopathy.
- Published
- 2016
- Full Text
- View/download PDF
37. A tunable hydrogel system for long‐term release of cell‐secreted cytokines and bioprinted in situ wound cell delivery
- Author
-
Aleksander Skardal, Kathryn Crowell, Sean V. Murphy, David L. Mack, Shay Soker, and Anthony Atala
- Subjects
0301 basic medicine ,In situ ,Materials science ,Cell ,Biomedical Engineering ,Mice, Nude ,02 engineering and technology ,Article ,Biomaterials ,Extracellular matrix ,Mice ,03 medical and health sciences ,chemistry.chemical_compound ,In vivo ,Hyaluronic acid ,medicine ,Animals ,Humans ,Secretion ,Amnion ,Hyaluronic Acid ,Heparin ,Stem Cells ,Hydrogels ,Cells, Immobilized ,021001 nanoscience & nanotechnology ,Cell delivery ,Cell biology ,030104 developmental biology ,medicine.anatomical_structure ,chemistry ,Immunology ,Cytokines ,Heterografts ,Wounds and Injuries ,0210 nano-technology ,Wound healing ,Stem Cell Transplantation - Abstract
For many cellular therapies being evaluated in preclinical and clinical trials, the mechanisms behind their therapeutic effects appear to be the secretion of growth factors and cytokines, also known as paracrine activity. Often, delivered cells are transient, and half-lives of the growth factors that they secrete are short, limiting their long-term effectiveness. The goal of this study was to optimize a hydrogel system capable of in situ cell delivery that could sequester and release growth factors secreted from those cells after the cells were no longer present. Here, we demonstrate the use of a fast photocross-linkable heparin-conjugated hyaluronic acid (HA-HP) hydrogel as a cell delivery vehicle for sustained growth factor release, which extends paracrine activity. The hydrogel could be modulated through cross-linking geometries and heparinization to support sustained release proteins and heparin-binding growth factors. To test the hydrogel in vivo, we used it to deliver amniotic fluid-derived stem (AFS) cells, which are known to secrete cytokines and growth factors, in full thickness skin wounds in a nu/nu murine model. Despite transience of the AFS cells in vivo, the HA-HP hydrogel with AFS cells improved wound closure and reepithelialization and increased vascularization and production of extracellular matrix in vivo. These results suggest that HA-HP hydrogel has the potential to prolong the paracrine activity of cells, thereby increasing their therapeutic effectiveness in wound healing. © 2016 Wiley Periodicals, Inc. J Biomed Mater Res Part B: Appl Biomater, 105B: 1986-2000, 2017.
- Published
- 2016
- Full Text
- View/download PDF
38. Embryonic Myosin Mutations T178I and R672C Develop Mechanical Dysfunction When Expressed in Hipsc Derived Skeletal Myotubes
- Author
-
Michael Regnier, Kati J. Buckingham, David L. Mack, Alec S.T. Smith, Michael J. Bamshad, Saffie Mohran, and Shawn M. Luttrell
- Subjects
Myogenesis ,Myosin ,Biophysics ,Biology ,Embryonic stem cell ,Cell biology - Published
- 2021
- Full Text
- View/download PDF
39. The Progression of Dystrophin-Deficient Dilated Cardiomyopathy through Mechanical and Ca2+ Dysfunction in a Duchenne Muscular Dystrophy Rat Model
- Author
-
Saffie Mohran, Michael Regnier, David L. Mack, Kristina B. Kooiker, Shawn M. Luttrell, and Timothy S. McMillen
- Subjects
Pathology ,medicine.medical_specialty ,biology ,business.industry ,Duchenne muscular dystrophy ,Rat model ,Biophysics ,medicine ,biology.protein ,Dilated cardiomyopathy ,medicine.disease ,Dystrophin ,business - Published
- 2021
- Full Text
- View/download PDF
40. Reversion to embryonic transcriptional splicing patterns may underlie diabetic myopathy
- Author
-
David L. Mack
- Subjects
0301 basic medicine ,medicine.medical_specialty ,Physiology ,Reversion ,Bioinformatics ,Article ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,Muscular Diseases ,Physiology (medical) ,Diabetes mellitus ,Internal medicine ,Diabetes Mellitus ,medicine ,Humans ,Myopathy ,business.industry ,Alternative splicing ,medicine.disease ,Embryonic stem cell ,Alternative Splicing ,030104 developmental biology ,Endocrinology ,RNA splicing ,Neurology (clinical) ,medicine.symptom ,business - Published
- 2017
- Full Text
- View/download PDF
41. LATE BREAKING NEWS E-POSTER PRESENTATION
- Author
-
Bradley P Morgan, Eva R. Chin, Martin K. Childers, SiWei Luo, Darren Hwee, Robert W. Grange, David L. Mack, Fady I. Malik, and Jordan M. Klaiman
- Subjects
Presentation ,History ,Neurology ,media_common.quotation_subject ,Pediatrics, Perinatology and Child Health ,Neurology (clinical) ,Genetics (clinical) ,media_common ,Visual arts - Published
- 2020
- Full Text
- View/download PDF
42. Use of Cells from Remotely Collected Urine to Generate Human Induced Pluripotent Stem Cells and Myofibers that Recapitulate Unconventional Myopathies
- Author
-
Michael J. Bamshad, Michael Regnier, David L. Mack, Saffie Mohran, Kati J. Buckingham, and Shawn M. Luttrell
- Subjects
Biophysics ,Urine ,Human Induced Pluripotent Stem Cells ,Biology ,Cell biology - Published
- 2020
- Full Text
- View/download PDF
43. Development of Mechanical and Structural Dysfunction in Skeletal Muscle from a Duchene Muscular Dystrophy Rat Model
- Author
-
Shawn M. Luttrell, Thomas C. Irving, Michael Regnier, Saffie Mohran, Chen-Ching Yuan, David L. Mack, and Weikang Ma
- Subjects
Pathology ,medicine.medical_specialty ,medicine.anatomical_structure ,business.industry ,Rat model ,Biophysics ,Medicine ,Skeletal muscle ,STRUCTURAL DYSFUNCTION ,Muscular dystrophy ,business ,medicine.disease - Published
- 2020
- Full Text
- View/download PDF
44. AAV-mediated gene transfer restores a normal muscle transcriptome in a canine model of X-linked myotubular myopathy
- Author
-
David L. Mack, Jean-Baptiste Dupont, Ana Buj-Bello, Michael W. Lawlor, Robert W. Grange, John T. Gray, Jianjun Guo, and Martin K. Childers
- Subjects
0303 health sciences ,Pathology ,medicine.medical_specialty ,Gene transfer ,Spinal muscular atrophy ,Biology ,medicine.disease ,X-linked myotubular myopathy ,Biceps ,3. Good health ,law.invention ,Transcriptome ,03 medical and health sciences ,0302 clinical medicine ,law ,Gene expression ,medicine ,Recombinant DNA ,Gene ,030217 neurology & neurosurgery ,030304 developmental biology - Abstract
Multiple clinical trials employing recombinant adeno-associated viral (rAAV) vectors have been initiated for neuromuscular disorders, including Duchenne and limb-girdle muscular dystrophies, spinal muscular atrophy, and recently X-linked myotubular myopathy (XLMTM). Previous work from our laboratory on a canine model of XLMTM showed that a single rAAV8-cMTM1 systemic infusion corrects structural abnormalities within the muscle and restores contractile function, with affected dogs surviving more than four years post injection. This exceptional therapeutic efficacy presents a unique opportunity to identify the downstream molecular drivers of XLMTM pathology, and to what extent the whole muscle transcriptome is restored to normal after gene transfer. Herein, RNA-sequencing was used to examine the transcriptomes of the Biceps femoris and Vastus lateralis in a previously-described canine cohort showing dose-dependent clinical improvements after rAAV8-cMTM1 gene transfer. Our analysis confirmed several dysregulated genes previously observed in XLMTM mice, but also identified new transcripts linked to XLMTM pathology. We demonstrated XLMTM transcriptome remodeling and dose-dependent normalization of gene expression after gene transfer and created new metrics to pinpoint potential biomarkers of disease progression and correction.
- Published
- 2018
- Full Text
- View/download PDF
45. Engineered developmental niche enables predictive phenotypic screening in human dystrophic cardiomyopathy
- Author
-
Xuan Guan, Eunpyo Choi, Yak-Nam Wang, David L. Mack, Michael Regnier, Dayae Kim, Smith As, Leung W, Hannele Ruohola-Baker, Max R. Salick, Macadangdang, Martin K. Childers, Su-In Lee, and Jason W. Miklas
- Subjects
0303 health sciences ,Phenotypic screening ,Mutant ,030204 cardiovascular system & hematology ,Biology ,Sarcomere ,Cell biology ,Transcriptome ,03 medical and health sciences ,0302 clinical medicine ,Directed differentiation ,Nitrendipine ,medicine ,biology.protein ,Induced pluripotent stem cell ,Dystrophin ,030304 developmental biology ,medicine.drug - Abstract
Directed differentiation of human pluripotent stem cells (hPSCs) into cardiomyocytes typically produces cells with structural, functional, and biochemical properties that most closely resemble those present in the fetal heart. Here we establish an in vitro engineered developmental cardiac niche to produce matured hPSC-derived cardiomyocytes (hPSC-CMs) with enhanced sarcomere development, electrophysiology, contractile function, mitochondrial capacity, and a more mature transcriptome. When this developmental cardiac niche was applied to dystrophin mutant hPSC-CMs, a robust disease phenotype emerged, which was not observed in non-matured diseased hPSC-CMs. Matured dystrophin mutant hPSC-CMs exhibited a greater propensity for arrhythmia as measured via beat rate variability, most likely due to higher resting cytosolic calcium content. Using a custom nanopatterned microelectrode array platform to screen functional output in hPSC-CMs exposed to our engineered developmental cardiac niche, we identified calcium channel blocker, nitrendipine, mitigated hPSC-CM arrhythmogenic behavior and correctly identified sildenafil as a false positive. Taken together, we demonstrate our developmental cardiac niche platform enables robust hPSC-CM maturation allowing for more accurate disease modeling and predictive drug screening.
- Published
- 2018
- Full Text
- View/download PDF
46. Nanopatterned Human iPSC-Based Model of a Dystrophin-Null Cardiomyopathic Phenotype
- Author
-
Rachel Lucero, Martin K. Childers, Xuan Guan, Jesse Macadangdang, David L. Mack, Alec S.T. Smith, Deok Ho Kim, and Stefan Czerniecki
- Subjects
biology ,Duchenne muscular dystrophy ,Cardiomyopathy ,Fluorescence recovery after photobleaching ,medicine.disease ,Actin cytoskeleton ,Bioinformatics ,Phenotype ,Article ,General Biochemistry, Genetics and Molecular Biology ,Cell biology ,Modeling and Simulation ,medicine ,biology.protein ,Nanotopography ,Dystrophin ,Induced pluripotent stem cell - Abstract
Human induced pluripotent stem cell derived cardiomyocytes (hiPSC-CMs) offer unprecedented opportunities to study inherited heart conditions in vitro, but are phenotypically immature, limiting their ability to effectively model adult-onset diseases. Cardiomyopathy is becoming the leading cause of death in patients with Duchenne muscular dystrophy (DMD), but the pathogenesis of this disease phenotype is not fully understood. Therefore, we aimed to test whether biomimetic nanotopography could further stratify the disease phenotype of DMD hiPSC-CMs to create more translationally relevant cardiomyocytes for disease modeling applications. We found that anisotropic nanotopography was necessary to distinguish structural differences between normal and DMD hiPSC-CMs, as these differences were masked on conventional flat substrates. DMD hiPSC-CMs exhibited a diminished structural and functional response to the underlying nanotopography compared to normal cardiomyocytes at both the macroscopic and subcellular levels. This blunted response may be due to a lower level of actin cytoskeleton turnover as measured by fluorescence recovery after photobleaching. Taken together these data suggest that DMD hiPSC-CMs are less adaptable to changes in their extracellular environment, and highlight the utility of nanotopographic substrates for effectively stratifying normal and structural cardiac disease phenotypes in vitro.
- Published
- 2015
- Full Text
- View/download PDF
47. Long-term effects of systemic gene therapy in a canine model of myotubular myopathy
- Author
-
Alan H. Beggs, Robert W. Grange, Ana Buj-Bello, Valerie E. Kelly, Matthew Elverman, Michael W. Lawlor, Martin K. Childers, Melissa A. Goddard, Anthony P. Marsh, Jessica M. Snyder, David L. Mack, Karine Poulard, Hui Meng, Approches génétiques intégrées et nouvelles thérapies pour les maladies rares (INTEGRARE), École pratique des hautes études (EPHE), Université Paris sciences et lettres (PSL)-Université Paris sciences et lettres (PSL)-Université d'Évry-Val-d'Essonne (UEVE)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Généthon, Genomics Program and Division of Genetics, Harvard Medical School [Boston] (HMS)-Boston Children's Hospital-The Manton Center for Orphan Disease Research, Généthon, Laboratoire d'Astrophysique de Marseille (LAM), Centre National de la Recherche Scientifique (CNRS)-Institut national des sciences de l'Univers (INSU - CNRS)-Aix Marseille Université (AMU)-Centre National d'Études Spatiales [Toulouse] (CNES), Wake Forest University, École pratique des hautes études (EPHE)-Université d'Évry-Val-d'Essonne (UEVE)-GENETHON 3-Institut National de la Santé et de la Recherche Médicale (INSERM), Généthon, Evry, and École Pratique des Hautes Études (EPHE)
- Subjects
0301 basic medicine ,Pathology ,Physiology ,Myotubularin ,muscle ,Genetic enhancement ,viruses ,medicine.disease_cause ,0302 clinical medicine ,Transduction, Genetic ,Longitudinal Studies ,Respiratory system ,Adeno-associated virus ,myotubular myopathy ,Glucuronidase ,Adenosine Triphosphatases ,Neurologic Examination ,[SDV.BA]Life Sciences [q-bio]/Animal biology ,Dependovirus ,Protein Tyrosine Phosphatases, Non-Receptor ,gene therapy ,3. Good health ,Female ,medicine.symptom ,Myopathies, Structural, Congenital ,medicine.medical_specialty ,Neuromuscular disease ,Transgene ,canine ,adeno-associated virus ,Biology ,gait ,Virus ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,Dogs ,Physiology (medical) ,medicine ,Animals ,Humans ,Muscle, Skeletal ,Gait Disorders, Neurologic ,[SDV.GEN]Life Sciences [q-bio]/Genetics ,Muscle weakness ,Genetic Therapy ,neuromuscular disease ,medicine.disease ,NAD ,Respiration Disorders ,Disease Models, Animal ,Microscopy, Electron ,030104 developmental biology ,Mutation ,Neurology (clinical) ,030217 neurology & neurosurgery - Abstract
Introduction X-linked myotubular myopathy (XLMTM), a devastating pediatric disease caused by the absence of the protein myotubularin, results from mutations in the MTM1 gene. While there is no cure for XLMTM, we previously reported effects of MTM1 gene therapy using adeno-associated virus (AAV) vector on muscle weakness and pathology in MTM1-mutant dogs. Here, we followed 2 AAV-infused dogs over 4 years. Methods We evaluated gait, strength, respiration, neurological function, muscle pathology, AAV vector copy number (VCN), and transgene expression. Results Four years following AAV-mediated gene therapy, gait, respiratory performance, neurological function and pathology in AAV-infused XLMTM dogs remained comparable to their healthy littermate controls despite a decline in VCN and muscle strength. Conclusions AAV-mediated gene transfer of MTM1 in young XLMTM dogs results in long-term expression of myotubularin transgene with normal muscular performance and neurological function in the absence of muscle pathology. These findings support a clinical trial in patients. Muscle Nerve 56: 943–953, 2017
- Published
- 2017
- Full Text
- View/download PDF
48. Gene Therapy for Inherited Muscle Diseases
- Author
-
David L. Mack, Zejing Wang, Martin K. Childers, and Robynne Braun
- Subjects
medicine.medical_specialty ,Weakness ,Genetic enhancement ,Duchenne muscular dystrophy ,Genetic Vectors ,Physical Therapy, Sports Therapy and Rehabilitation ,Disease ,Muscle disorder ,Regenerative Medicine ,Article ,Cohort Studies ,Dogs ,Physical medicine and rehabilitation ,Muscular Diseases ,medicine ,Animals ,Humans ,Muscular dystrophy ,Myopathy ,Genetics ,business.industry ,Rehabilitation ,Skeletal muscle ,Genetic Therapy ,Physical and Rehabilitation Medicine ,Prognosis ,medicine.disease ,Combined Modality Therapy ,Survival Analysis ,Muscular Dystrophy, Duchenne ,Disease Models, Animal ,Treatment Outcome ,medicine.anatomical_structure ,medicine.symptom ,business ,Forecasting ,Myopathies, Structural, Congenital - Abstract
The development of clinical vectors to correct genetic mutations that cause inherited myopathies and related disorders of skeletal muscle is advancing at an impressive rate. Adeno-associated virus vectors are attractive for clinical use because (1) adeno-associated viruses do not cause human disease and (2) these vectors are able to persist for years. New vectors are now becoming available as gene therapy delivery tools, and recent preclinical experiments have demonstrated the feasibility, safety, and efficacy of gene therapy with adeno-associated virus for long-term correction of muscle pathology and weakness in myotubularin-deficient canine and murine disease models. In this review, recent advances in the application of gene therapies to treat inherited muscle disorders are presented, including Duchenne muscular dystrophy and x-linked myotubular myopathy. Potential areas for therapeutic synergies between rehabilitation medicine and genetics are also discussed.
- Published
- 2014
- Full Text
- View/download PDF
49. Optical Investigation of Action Potential and Calcium Handling Maturation of hiPSC-Cardiomyocytes on Biomimetic Substrates
- Author
-
Corrado Poggesi, Chiara Tesi, Sara Landi, Cecilia Ferrantini, Camilla Parmeggiani, Marianna Langione, Bruno Grandinetti, Michael Regnier, Chiara Palandri, Laura Sartiani, Josè Manuel Pioner, Patrizia Benzoni, Andrea Barbuti, Flavia Lupi, David L. Mack, Raffaele Coppini, Luca Boarino, Daniele Martella, Elisabetta Cerbai, Federico Ferrarese Lupi, Valentina Balducci, Silvia Querceto, and Lorenzo Santini
- Subjects
0301 basic medicine ,Inotrope ,Cardiomyopathy ,Action Potentials ,cardiomyocytes ,Stimulation ,030204 cardiovascular system & hematology ,Substrate Specificity ,lcsh:Chemistry ,action potential ,0302 clinical medicine ,Biomimetics ,Myocytes, Cardiac ,lcsh:QH301-705.5 ,Cells, Cultured ,Excitation Contraction Coupling ,Spectroscopy ,Chemistry ,Cell Differentiation ,Hydrogels ,General Medicine ,3. Good health ,Computer Science Applications ,Cell biology ,Sarcoplasmic Reticulum ,fluorescence ,Cardiomyopathies ,Calcium handling ,Adrenergic beta-Antagonists ,Induced Pluripotent Stem Cells ,chemistry.chemical_element ,Calcium ,Article ,Catalysis ,Inorganic Chemistry ,03 medical and health sciences ,Action potential ,Cardiomyocytes ,Fluorescence ,Human induced pluripotent stem cells ,Long-term culture ,Maturation ,medicine ,Humans ,Physical and Theoretical Chemistry ,Molecular Biology ,maturation ,Endoplasmic reticulum ,Organic Chemistry ,medicine.disease ,human induced pluripotent stem cells ,long-term culture ,Electrophysiology ,030104 developmental biology ,lcsh:Biology (General) ,lcsh:QD1-999 ,calcium handling ,Function (biology) - Abstract
Cardiomyocytes from human induced pluripotent stem cells (hiPSC-CMs) are the most promising human source with preserved genetic background of healthy individuals or patients. This study aimed to establish a systematic procedure for exploring development of hiPSC-CM functional output to predict genetic cardiomyopathy outcomes and identify molecular targets for therapy. Biomimetic substrates with microtopography and physiological stiffness can overcome the immaturity of hiPSC-CM function. We have developed a custom-made apparatus for simultaneous optical measurements of hiPSC-CM action potential and calcium transients to correlate these parameters at specific time points (day 60, 75 and 90 post differentiation) and under inotropic interventions. In later-stages, single hiPSC-CMs revealed prolonged action potential duration, increased calcium transient amplitude and shorter duration that closely resembled those of human adult cardiomyocytes from fresh ventricular tissue of patients. Thus, the major contribution of sarcoplasmic reticulum and positive inotropic response to &beta, adrenergic stimulation are time-dependent events underlying excitation contraction coupling (ECC) maturation of hiPSC-CM, biomimetic substrates can promote calcium-handling regulation towards adult-like kinetics. Simultaneous optical recordings of long-term cultured hiPSC-CMs on biomimetic substrates favor high-throughput electrophysiological analysis aimed at testing (mechanistic hypothesis on) disease progression and pharmacological interventions in patient-derived hiPSC-CMs.
- Published
- 2019
- Full Text
- View/download PDF
50. Genetic Modification of Primate Amniotic Fluid-Derived Stem Cells Produces Pancreatic Progenitor Cells in vitro
- Author
-
So Young Chun, Diego Lorenzetti, Anthony Atala, Yu Zhou, Jun Wang, Sayed Hadi Mirmalek-Sani, Mark E. Furth, J. Koudy Williams, Emily C. Moorefield, David L. Mack, and Shay Soker
- Subjects
medicine.medical_specialty ,Maf Transcription Factors, Large ,Histology ,Cellular differentiation ,Gene Expression ,Nerve Tissue Proteins ,Biology ,Article ,Mice ,Internal medicine ,Basic Helix-Loop-Helix Transcription Factors ,medicine ,Animals ,Humans ,Progenitor cell ,Pancreas ,Homeodomain Proteins ,Stem Cells ,Nuclear Proteins ,Cell Differentiation ,Amniotic stem cells ,Amniotic Fluid ,Cell biology ,Macaca fascicularis ,Homeobox Protein Nkx-2.2 ,medicine.anatomical_structure ,Endocrinology ,Amniotic epithelial cells ,NEUROD1 ,Trans-Activators ,PDX1 ,Anatomy ,Stem cell ,Transcription Factors - Abstract
Insulin therapy for type 1 diabetes does not prevent serious long-term complications including vascular disease, neuropathy, retinopathy and renal failure. Stem cells, including amniotic fluid-derived stem (AFS) cells - highly expansive, multipotent and nontumorigenic cells - could serve as an appropriate stem cell source for β-cell differentiation. In the current study we tested whether nonhuman primate (nhp)AFS cells ectopically expressing key pancreatic transcription factors were capable of differentiating into a β-cell-like cell phenotype in vitro. nhpAFS cells were obtained from Cynomolgus monkey amniotic fluid by immunomagnetic selection for a CD117 (c-kit)-positive population. RT-PCR for endodermal and pancreatic lineage-specific markers was performed on AFS cells after adenovirally transduced expression of PDX1, NGN3 and MAFA. Expression of MAFA was sufficient to induce insulin mRNA expression in nhpAFS cell lines, whereas a combination of MAFA, PDX1 and NGN3 further induced insulin expression, and also induced the expression of other important endocrine cell genes such as glucagon, NEUROD1, NKX2.2, ISL1 and PCSK2. Higher induction of these and other important pancreatic genes was achieved by growing the triply infected AFS cells in media supplemented with a combination of B27, betacellulin and nicotinamide, as well as culturing the cells on extracellular matrix-coated plates. The expression of pancreatic genes such as NEUROD1, glucagon and insulin progressively decreased with the decline of adenovirally expressed PDX1, NGN3 and MAFA. Together, these experiments suggest that forced expression of pancreatic transcription factors in primate AFS cells induces them towards the pancreatic lineage.
- Published
- 2013
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.