Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Nat Biomed Eng ; 2(12): 942-954, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30581652

ABSTRACT

Adult skeletal muscle has a robust capacity for self-repair, owing to synergies between muscle satellite cells and the immune system. In vitro models of muscle self-repair would facilitate the basic understanding of muscle regeneration and the screening of therapies for muscle disease. Here, we show that the incorporation of macrophages into muscle tissues engineered from adult-rat myogenic cells enables near-complete structural and functional repair after cardiotoxic injury in vitro. First, we show that-in contrast with injured neonatal-derived engineered muscle-adult-derived engineered muscle fails to properly self-repair after injury, even when treated with pro-regenerative cytokines. We then show that rat bone-marrow-derived macrophages or human blood-derived macrophages resident within the in vitro engineered tissues stimulate muscle satellite cell-mediated myogenesis while significantly limiting myofibre apoptosis and degeneration. Moreover, bone-marrow-derived macrophages within engineered tissues implanted in a mouse dorsal window-chamber model augmented blood vessel ingrowth, cell survival, muscle regeneration and contractile function.

2.
Nat Commun ; 8(1): 1825, 2017 11 28.
Article in English | MEDLINE | ID: mdl-29184059

ABSTRACT

Despite increased use of human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) for drug development and disease modeling studies, methods to generate large, functional heart tissues for human therapy are lacking. Here we present a "Cardiopatch" platform for 3D culture and maturation of hiPSC-CMs that after 5 weeks of differentiation show robust electromechanical coupling, consistent H-zones, I-bands, and evidence for T-tubules and M-bands. Cardiopatch maturation markers and functional output increase during culture, approaching values of adult myocardium. Cardiopatches can be scaled up to clinically relevant dimensions, while preserving spatially uniform properties with high conduction velocities and contractile stresses. Within window chambers in nude mice, cardiopatches undergo vascularization by host vessels and continue to fire Ca2+ transients. When implanted onto rat hearts, cardiopatches robustly engraft, maintain pre-implantation electrical function, and do not increase the incidence of arrhythmias. These studies provide enabling technology for future use of hiPSC-CM tissues in human heart repair.


Subject(s)
Induced Pluripotent Stem Cells/transplantation , Myocytes, Cardiac/transplantation , Pluripotent Stem Cells/transplantation , Tissue Engineering/methods , Animals , Arrhythmias, Cardiac/therapy , Calcium/metabolism , Cell Differentiation/physiology , Cell Line , Cells, Cultured , Disease Models, Animal , Heterografts , Humans , Induced Pluripotent Stem Cells/physiology , Male , Mice , Mice, Nude , Myocardial Contraction/physiology , Myocardial Infarction/pathology , Myocardial Infarction/surgery , Myocardium/cytology , Myocardium/metabolism , Rats , Sarcomeres
3.
ACS Synth Biol ; 6(11): 2003-2013, 2017 11 17.
Article in English | MEDLINE | ID: mdl-28793186

ABSTRACT

The precise spatial and temporal control of gene expression, cell differentiation, and tissue morphogenesis has widespread application in regenerative medicine and the study of tissue development. In this work, we applied optogenetics to control cell differentiation and new tissue formation. Specifically, we engineered an optogenetic "on" switch that provides permanent transgene expression following a transient dose of blue light illumination. To demonstrate its utility in controlling cell differentiation and reprogramming, we incorporated an engineered form of the master myogenic factor MyoD into this system in multipotent cells. Illumination of cells with blue light activated myogenic differentiation, including upregulation of myogenic markers and fusion into multinucleated myotubes. Cell differentiation was spatially patterned by illumination of cell cultures through a photomask. To demonstrate the application of the system to controlling in vivo tissue development, the light inducible switch was used to control the expression of VEGF and angiopoietin-1, which induced angiogenic sprouting in a mouse dorsal window chamber model. Live intravital microscopy showed illumination-dependent increases in blood-perfused microvasculature. This optogenetic switch is broadly useful for applications in which sustained and patterned gene expression is desired following transient induction, including tissue engineering, gene therapy, synthetic biology, and fundamental studies of morphogenesis.


Subject(s)
Angiopoietin-1 , Cell Differentiation , Gene Expression Regulation , MyoD Protein , Optogenetics/methods , Vascular Endothelial Growth Factor A , Angiopoietin-1/biosynthesis , Angiopoietin-1/genetics , Animals , Cell Line , Mice , MyoD Protein/genetics , MyoD Protein/metabolism , Vascular Endothelial Growth Factor A/biosynthesis , Vascular Endothelial Growth Factor A/genetics
4.
Methods ; 99: 81-90, 2016 Apr 15.
Article in English | MEDLINE | ID: mdl-26455485

ABSTRACT

For over two decades, research groups have been developing methods to engineer three-dimensional skeletal muscle tissues. These tissues hold great promise for use in disease modeling and pre-clinical drug development, and have potential to serve as therapeutic grafts for functional muscle repair. Recent advances in the field have resulted in the engineering of regenerative muscle constructs capable of survival, vascularization, and functional maturation in vivo as well as the first-time creation of functional human engineered muscles for screening of therapeutics in vitro. In this review, we will discuss the methodologies that have progressed work in the muscle tissue engineering field to its current state. The emphasis will be placed on the existing procedures to generate myogenic cell sources and form highly functional muscle tissues in vitro, techniques to monitor and evaluate muscle maturation and performance in vitro and in vivo, and surgical strategies to both create diseased environments and ensure implant survival and rapid integration into the host. Finally, we will suggest the most promising methodologies that will enable continued progress in the field.


Subject(s)
Muscle, Skeletal/cytology , Tissue Engineering , Animals , Cell Culture Techniques , Cells, Cultured , Humans , Muscle, Skeletal/physiology , Regeneration , Regenerative Medicine , Satellite Cells, Skeletal Muscle/physiology , Stem Cell Niche
5.
Annu Rev Biomed Eng ; 17: 217-42, 2015.
Article in English | MEDLINE | ID: mdl-26643021

ABSTRACT

Although skeletal muscle is one of the most regenerative organs in our body, various genetic defects, alterations in extrinsic signaling, or substantial tissue damage can impair muscle function and the capacity for self-repair. The diversity and complexity of muscle disorders have attracted much interest from both cell biologists and, more recently, bioengineers, leading to concentrated efforts to better understand muscle pathology and develop more efficient therapies. This review describes the biological underpinnings of muscle development, repair, and disease, and discusses recent bioengineering efforts to design and control myomimetic environments, both to study muscle biology and function and to aid in the development of new drug, cell, and gene therapies for muscle disorders. The synergy between engineering-aided biological discovery and biology-inspired engineering solutions will be the path forward for translating laboratory results into clinical practice.


Subject(s)
Models, Biological , Muscle, Skeletal/injuries , Animals , Biomedical Engineering , Biomimetics , Humans , Muscle Development , Muscle, Skeletal/pathology , Muscle, Skeletal/physiopathology , Muscular Diseases/pathology , Muscular Diseases/physiopathology , Muscular Diseases/therapy , Regeneration , Satellite Cells, Skeletal Muscle/cytology , Satellite Cells, Skeletal Muscle/physiology
6.
Elife ; 4: e04885, 2015 Jan 09.
Article in English | MEDLINE | ID: mdl-25575180

ABSTRACT

Existing in vitro models of human skeletal muscle cannot recapitulate the organization and function of native muscle, limiting their use in physiological and pharmacological studies. Here, we demonstrate engineering of electrically and chemically responsive, contractile human muscle tissues ('myobundles') using primary myogenic cells. These biomimetic constructs exhibit aligned architecture, multinucleated and striated myofibers, and a Pax7(+) cell pool. They contract spontaneously and respond to electrical stimuli with twitch and tetanic contractions. Positive correlation between contractile force and GCaMP6-reported calcium responses enables non-invasive tracking of myobundle function and drug response. During culture, myobundles maintain functional acetylcholine receptors and structurally and functionally mature, evidenced by increased myofiber diameter and improved calcium handling and contractile strength. In response to diversely acting drugs, myobundles undergo dose-dependent hypertrophy or toxic myopathy similar to clinical outcomes. Human myobundles provide an enabling platform for predictive drug and toxicology screening and development of novel therapeutics for muscle-related disorders.


Subject(s)
Acetylcholine/pharmacology , Bioengineering/methods , Caffeine/pharmacology , Muscle, Skeletal/drug effects , Muscle, Skeletal/physiology , Biomechanical Phenomena/drug effects , Calcium/metabolism , Calcium Signaling/drug effects , Genes, Reporter , Humans , Muscle Contraction/physiology , Reproducibility of Results
7.
Biomaterials ; 35(35): 9438-46, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25154662

ABSTRACT

Highly functional engineered skeletal muscle constructs could serve as physiological models of muscle function and regeneration and have utility in therapeutic replacement of damaged or diseased muscle tissue. In this study, we examined the roles of different myogenic cell fractions and culturing conditions in the generation of highly functional engineered muscle. Fibrin-based muscle bundles were fabricated using either freshly-isolated myogenic cells or their adherent fraction pre-cultured for 36 h. Muscle bundles made of these cells were cultured in both static and dynamic conditions and systematically characterized with respect to early myogenic events and contractile function. Following 2 weeks of culture, we observed both individual and synergistic benefits of using the adherent cell fraction and dynamic culture on muscle formation and function. In particular, optimal culture conditions resulted in significant increase in the total cross-sectional muscle area (- 3-fold), myofiber size (- 1.6-fold), myonuclei density (- 1.2-fold), and force generation (- 9-fold) compared to traditional use of freshly-isolated cells and static culture. Curiously, we observed that only a simultaneous use of the adherent cell fraction and dynamic culture resulted in accelerated formation of differentiated myofibers which were critical for providing a niche-like environment for maintenance of a satellite cell pool early during culture. Our study identifies key parameters for engineering large-size, highly functional skeletal muscle tissues with improved ability for retention of functional satellite cells.


Subject(s)
Muscle Fibers, Skeletal/cytology , Satellite Cells, Skeletal Muscle/cytology , Animals , Cell Adhesion , Cell Culture Techniques , Cell Differentiation , Cells, Cultured , Muscle Fibers, Skeletal/metabolism , Rats , Rats, Sprague-Dawley , Tissue Engineering/methods
8.
Proc Natl Acad Sci U S A ; 111(15): 5508-13, 2014 Apr 15.
Article in English | MEDLINE | ID: mdl-24706792

ABSTRACT

Tissue-engineered skeletal muscle can serve as a physiological model of natural muscle and a potential therapeutic vehicle for rapid repair of severe muscle loss and injury. Here, we describe a platform for engineering and testing highly functional biomimetic muscle tissues with a resident satellite cell niche and capacity for robust myogenesis and self-regeneration in vitro. Using a mouse dorsal window implantation model and transduction with fluorescent intracellular calcium indicator, GCaMP3, we nondestructively monitored, in real time, vascular integration and the functional state of engineered muscle in vivo. During a 2-wk period, implanted engineered muscle exhibited a steady ingrowth of blood-perfused microvasculature along with an increase in amplitude of calcium transients and force of contraction. We also demonstrated superior structural organization, vascularization, and contractile function of fully differentiated vs. undifferentiated engineered muscle implants. The described in vitro and in vivo models of biomimetic engineered muscle represent enabling technology for novel studies of skeletal muscle function and regeneration.


Subject(s)
Biomimetics/methods , Muscle Development/physiology , Muscle, Skeletal/blood supply , Muscle, Skeletal/growth & development , Tissue Engineering/methods , Animals , Cobra Cardiotoxin Proteins/toxicity , Mice , Mice, Nude , Microvessels/growth & development , Muscle Contraction/physiology , Muscle, Skeletal/drug effects
9.
Curr Opin Biotechnol ; 24(5): 880-6, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23711735

ABSTRACT

Healthy skeletal muscle has a remarkable capacity for regeneration. Even at a mature age, muscle tissue can undergo a robust rebuilding process that involves the formation of new muscle cells and extracellular matrix and the re-establishment of vascular and neural networks. Understanding and reverse-engineering components of this process is essential for our ability to restore loss of muscle mass and function in cases where the natural ability of muscle for self-repair is exhausted or impaired. In this article, we will describe current approaches to restore the function of diseased or injured muscle through combined use of myogenic stem cells, biomaterials, and functional tissue-engineered muscle. Furthermore, we will discuss possibilities for expanding the future use of human cell sources toward the development of cell-based clinical therapies and in vitro models of human muscle disease.


Subject(s)
Muscle, Skeletal/physiology , Regeneration , Tissue Engineering , Animals , Humans , Models, Biological , Satellite Cells, Skeletal Muscle/cytology , Satellite Cells, Skeletal Muscle/physiology , Wound Healing
10.
Tissue Eng Part A ; 18(9-10): 957-67, 2012 May.
Article in English | MEDLINE | ID: mdl-22115339

ABSTRACT

The field of skeletal muscle tissue engineering is currently hampered by the lack of methods to form large muscle constructs composed of dense, aligned, and mature myofibers and limited understanding of structure-function relationships in developing muscle tissues. In our previous studies, engineered muscle sheets with elliptical pores ("muscle networks") were fabricated by casting cells and fibrin gel inside elastomeric tissue molds with staggered hexagonal posts. In these networks, alignment of cells around the elliptical pores followed the local distribution of tissue strains that were generated by cell-mediated compaction of fibrin gel against the hexagonal posts. The goal of this study was to assess how systematic variations in pore elongation affect the morphology and contractile function of muscle networks. We found that in muscle networks with more elongated pores the force production of individual myofibers was not altered, but the myofiber alignment and efficiency of myofiber formation were significantly increased yielding an increase in the total contractile force despite a decrease in the total tissue volume. Beyond a certain pore length, increase in generated contractile force was mainly contributed by more efficient myofiber formation rather than enhanced myofiber alignment. Collectively, these studies show that changes in local tissue geometry can exert both direct structural and indirect myogenic effects on the functional output of engineered muscle. Different hydrogel formulations and pore geometries will be explored in the future to further augment contractile function of engineered muscle networks and promote their use for basic structure-function studies in vitro and, eventually, for efficient muscle repair in vivo.


Subject(s)
Dimethylpolysiloxanes/chemistry , Myoblasts/metabolism , Tissue Engineering/methods , Animals , Cells, Cultured , Myoblasts/cytology , Rats , Rats, Sprague-Dawley
11.
Stud Health Technol Inform ; 163: 552-4, 2011.
Article in English | MEDLINE | ID: mdl-21335855

ABSTRACT

We are developing a simulator of peripheral nerve block utilizing a mixed-reality approach: the combination of a physical model, an MRI-derived virtual model, mechatronics and spatial tracking. Our design uses tangible (physical) interfaces to simulate surface anatomy, haptic feedback during needle insertion, mechatronic display of muscle twitch corresponding to the specific nerve stimulated, and visual and haptic feedback for the injection syringe. The twitch response is calculated incorporating the sensed output of a real neurostimulator. The virtual model is isomorphic with the physical model and is derived from segmented MRI data. This model provides the subsurface anatomy and, combined with electromagnetic tracking of a sham ultrasound probe and a standard nerve block needle, supports simulated ultrasound display and measurement of needle location and proximity to nerves and vessels. The needle tracking and virtual model also support objective performance metrics of needle targeting technique.


Subject(s)
Electric Stimulation/methods , Models, Biological , Nerve Block/instrumentation , Nerve Block/methods , Surgery, Computer-Assisted/methods , Ultrasonography, Interventional/methods , User-Computer Interface , Computer Simulation , Electric Stimulation/instrumentation , Equipment Design , Equipment Failure Analysis , Humans , Manikins , Surgery, Computer-Assisted/instrumentation , Systems Integration , Ultrasonography, Interventional/instrumentation
12.
Biomaterials ; 31(13): 3520-6, 2010 May.
Article in English | MEDLINE | ID: mdl-20144481

ABSTRACT

Tracheal injury is a rare but complex problem. Primary tracheal reconstructions are commonly performed, but complications such as tension and inadequate vascular supply limit the length of surgical resection. The objective of the present study was to determine whether a hydrated, decellularized porcine tracheal extracellular matrix showed the potential to serve as a functional tracheal replacement graft. Porcine tracheas were decellularized and evaluated to characterize their biochemical composition and biomechanical behavior. Hydrated decellularized tracheal matrix (HDTM) grafts (>5 cm) were implanted heterotopically beneath the strap muscle and wrapped in the omentum in a canine model for 2 and 8 weeks followed by histologic and mechanical analysis. HDTM patches (2 x 3 cm) were also used in a patch tracheoplasty model. The repair site was evaluated bronchoscopically and radiographically, and the grafts were analyzed by histologic methods to evaluate epithelialization and persistence of the cartilage rings. The present study showed that HDTM maintains mechanical characteristics necessary for function under physiologic loading conditions even after 8 weeks of heterotopic implantation. After orthotopic implantation, the grafts were shown to support development of a columnar, pseudostratified, ciliated epithelium, but the cartilage structures showed histologic evidence of degradation and limited new cartilage formation. The results of the study showed tracheal ECM scaffolds support the formation of site-specific epithelium and provide sufficient mechanical integrity withstand physiologic pressures in the short-term. However, for long-term success, it appears that pre-implantation to allow vascularization or preseeding of the graft with chondrocytes will be necessary.


Subject(s)
Tissue Engineering , Trachea , Animals , Cartilage/transplantation , Dogs , Swine
SELECTION OF CITATIONS
SEARCH DETAIL
...