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










Publication year range
1.
Methods Mol Biol ; 1889: 169-183, 2019.
Article in English | MEDLINE | ID: mdl-30367414

ABSTRACT

Skeletal muscle tissue engineering aims at creating functional skeletal muscle in vitro. Human muscle organoids can be used for potential applications in regenerative medicine, but also as an in vitro model for myogenesis or myopathology. However, the thickness of constructs is limited due to passive diffusion of nutrients and oxygen. Introduction of a vascular network in vitro may solve this limitation. Here, we describe tissue engineering of in vitro skeletal muscle consisting of human aligned myofibers with interspersed endothelial networks. To create bio-artificial muscle (BAM), human muscle progenitor cells are cocultured with human umbilical vein endothelial cells (HUVECs) in a fibrin hydrogel. The cell-gel mix is cast into silicone molds with end attachment sites and cultured in endothelial growth medium (EGM-2) for 1 week. The passive forces generated in the contracted hydrogel align the myogenic cells parallel to the long axis of the contracted gel such that they fuse into aligned multinucleated myofibers. This results in the formation of a 2 cm long and ~1.5 mm tick human BAM construct with endothelial networks.


Subject(s)
Coculture Techniques , Endothelial Cells/metabolism , Muscle, Skeletal/metabolism , Tissue Engineering , Biopsy , Cells, Cultured , Endothelial Cells/cytology , Human Umbilical Vein Endothelial Cells/cytology , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Muscle Development , Muscle, Skeletal/cytology , Myoblasts/cytology , Myoblasts/metabolism
2.
Front Physiol ; 9: 1076, 2018.
Article in English | MEDLINE | ID: mdl-30177884

ABSTRACT

Adult skeletal muscle progenitor cells can be embedded in an extracellular matrix (ECM) and tissue-engineered to form bio-artificial muscles (BAMs), composed of aligned post-mitotic myofibers. The ECM proteins which have been used most commonly are collagen type I and fibrin. Fibrin allows for in vitro vasculogenesis, however, high concentrations of fibrinolysis inhibitors are needed to inhibit degradation of the ECM and subsequent loss of BAM tissue structure. For in vivo implantation, fibrinolysis inhibition may prove difficult or even harmful to the host. Therefore, we adapted in vitro culture conditions to enhance the deposition of de novo synthesized collagen type I gradually replacing the degrading fibrin ECM. The in vitro viscoelastic properties of the fibrin BAMs and deposition of collagen were characterized. BAMs engineered with the addition of proline, hydroxyproline, and ascorbic acid in the tissue culture medium had a twofold increase in Young's Modulus, a 2.5-fold decrease in maximum strain, and a 1.6-fold increase in collagen deposition. Lowering the fibrin content of the BAMs also increased Young's Modulus, decreased maximum strain, and increased collagen deposition. Tissue engineering of BAMs with autologous ECM may allow for prolonged in vivo survival.

3.
Mol Ther ; 22(8): 1441-1449, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24769909

ABSTRACT

Repair of injured skeletal muscle by cell therapies has been limited by poor survival of injected cells. Use of a carrier scaffold delivering cells locally, may enhance in vivo cell survival, and promote skeletal muscle regeneration. Biomaterial scaffolds are often implanted into muscle tissue through invasive surgeries, which can result in trauma that delays healing. Minimally invasive approaches to scaffold implantation are thought to minimize these adverse effects. This hypothesis was addressed in the context of a severe mouse skeletal muscle injury model. A degradable, shape-memory alginate scaffold that was highly porous and compressible was delivered by minimally invasive surgical techniques to injured tibialis anterior muscle. The scaffold controlled was quickly rehydrated in situ with autologous myoblasts and growth factors (either insulin-like growth factor-1 (IGF-1) alone or IGF-1 with vascular endothelial growth factor (VEGF)). The implanted scaffolds delivering myoblasts and IGF-1 significantly reduced scar formation, enhanced cell engraftment, and improved muscle contractile function. The addition of VEGF to the scaffold further improved functional recovery likely through increased angiogenesis. Thus, the delivery of myoblasts and dual local release of VEGF and IGF-1 from degradable scaffolds implanted through a minimally invasive procedure effectively promoted the functional regeneration of injured skeletal muscle.


Subject(s)
Muscle, Skeletal/injuries , Muscle, Skeletal/surgery , Myoblasts, Skeletal/transplantation , Alginates/chemistry , Animals , Biocompatible Materials , Cell Proliferation , Cell Survival , Disease Models, Animal , Glucuronic Acid/chemistry , Hexuronic Acids/chemistry , Insulin-Like Growth Factor I/metabolism , Male , Mice , Mice, Inbred C3H , Muscle, Skeletal/physiopathology , Myoblasts, Skeletal/metabolism , Soft Tissue Injuries/therapy , Tissue Scaffolds/chemistry , Vascular Endothelial Growth Factor A/metabolism
4.
Mol Neurobiol ; 48(3): 397-403, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23371342

ABSTRACT

Dysfunction of the neuromuscular junction is involved in a wide range of muscular diseases. The development of neuromuscular junction through which skeletal muscle is innervated requires the functional modulation of acetylcholine receptor (AchR) clustering on myofibers. However, studies on AchR clustering in vitro are mostly done on monolayer muscle cell culture, which lacks a three-dimensional (3D) structure, a prominent limitation of the two-dimensional (2D) system. To enable a better understanding on the structure-function correlation underlying skeletal muscle innervation, a muscle system with a well-defined geometry mimicking the in vivo muscular setting is needed. Here, we report a 3D bio-artificial muscle (BAM) bioengineered from green fluorescent protein-transduced C3H murine myoblasts as a novel in vitro tissue-based model for muscle innervation studies. Our cell biological and molecular analysis showed that this BAM is structurally similar to in vivo muscle tissue and can reach the perinatal differentiation stage, higher than does 2D culture. Effective clustering and morphological maturation of AchRs on BAMs induced by agrin and laminin indicate the functional activity and plasticity of this BAM system toward innervation. Taken together, our results show that the BAM provides a favorable 3D environment that at least partially recapitulates real physiological skeletal muscle with regard to innervation. With a convenience of fabrication and manipulation, this 3D in vitro system offers a novel model for studying mechanisms underlying skeletal muscle innervation and testing therapeutic strategies for relevant nervous and muscular diseases.


Subject(s)
Bioartificial Organs , Muscles/metabolism , Receptors, Cholinergic/metabolism , Agrin/metabolism , Animals , Cell Differentiation/drug effects , Cell Shape/drug effects , Gene Expression Regulation/drug effects , Humans , Laminin/pharmacology , Mice , Muscle, Skeletal/cytology , Muscles/drug effects , Myosin Heavy Chains/genetics , Myosin Heavy Chains/metabolism , Protein Isoforms/genetics , Protein Isoforms/metabolism
5.
Tissue Eng Part A ; 18(19-20): 2000-7, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22646518

ABSTRACT

The successful use of transplanted cells and/or growth factors for tissue repair is limited by a significant cell loss and/or rapid growth factor diffusion soon after implantation. Highly porous alginate scaffolds formed with covalent crosslinking have been used to improve cell survival and growth factor release kinetics, but require open-wound surgical procedures for insertion and have not previously been designed to readily degrade in vivo. In this study, a biodegradable, partially crosslinked alginate scaffold with shape-memory properties was fabricated for minimally invasive surgical applications. A mixture of high and low molecular weight partially oxidized alginate modified with RGD peptides was covalently crosslinked using carbodiimide chemistry. The scaffold was compressible 11-fold and returned to its original shape when rehydrated. Scaffold degradation properties in vitro indicated ~85% mass loss by 28 days. The greater than 90% porous scaffolds released the recombinant growth factor insulin-like growth factor-1 over several days in vitro and allowed skeletal muscle cell survival, proliferation, and migration from the scaffold over a 28-day period. The compressible scaffold thus has the potential to be delivered by a minimally invasive technique, and when rehydrated in vivo with cells and/or growth factors, could serve as a temporary delivery vehicle for tissue repair.


Subject(s)
Alginates/chemistry , Biocompatible Materials/chemistry , Tissue Engineering/methods , Tissue Scaffolds/chemistry , Animals , Cell Movement/physiology , Cell Proliferation , Cell Survival/physiology , Cells, Cultured , Glucuronic Acid/chemistry , Hexuronic Acids/chemistry , Intercellular Signaling Peptides and Proteins/administration & dosage , Microscopy, Electron, Scanning
6.
Endocrinology ; 152(1): 193-206, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21084444

ABSTRACT

Testosterone (T) supplementation increases skeletal muscle mass, circulating GH, IGF-I, and im IGF-I expression, but the role of GH and IGF-I in mediating T's effects on the skeletal muscle remains poorly understood. Here, we show that T administration increased body weight and the mass of the androgen-dependent levator ani muscle in hypophysectomized as well as castrated plus hypophysectomized adult male rats. T stimulated the proliferation of primary human skeletal muscle cells (hSKMCs) in vitro, an effect blocked by transfecting hSKMCs with small interference RNA targeting human IGF-I receptor (IGF-IR). In differentiation conditions, T promoted the fusion of hSKMCs into larger myotubes, an effect attenuated by small interference RNA targeting human IGF-IR. Notably, MKR mice, which express a dominant negative form of the IGF-IR in skeletal muscle fibers, treated with a GnRH antagonist (acyline) to suppress endogenous T, responded to T administration by an attenuated increase in the levator ani muscle mass. In conclusion, circulating GH and IGF-I are not essential for mediating T's effects on an androgen-responsive skeletal muscle. IGF-I signaling plays an important role in mediating T's effects on skeletal muscle progenitor cell growth and differentiation in vitro. However, IGF-IR signaling in skeletal muscle fibers does not appear to be obligatory for mediating the anabolic effects of T on the mass of androgen-responsive skeletal muscles in mice.


Subject(s)
Gene Expression Regulation/drug effects , Growth Hormone/metabolism , Insulin-Like Growth Factor I/metabolism , Muscle, Skeletal/drug effects , Testosterone/pharmacology , Animals , Body Weight , Cells, Cultured , Gene Expression Regulation/physiology , Growth Hormone/genetics , Humans , Hypophysectomy , Insulin-Like Growth Factor I/genetics , Male , Mice , Muscle, Skeletal/cytology , Muscle, Skeletal/metabolism , Orchiectomy , RNA, Small Interfering , Rats , Rats, Sprague-Dawley , Receptor, IGF Type 1/antagonists & inhibitors
7.
FASEB J ; 23(10): 3325-34, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19487307

ABSTRACT

Identification of factors that improve muscle function in boys with Duchenne muscular dystrophy (DMD) could lead to an improved quality of life. To establish a functional in vitro assay for muscle strength, mdx murine myoblasts, the genetic homologue of DMD, were tissue engineered in 96-microwell plates into 3-dimensional muscle constructs with parallel arrays of striated muscle fibers. When electrically stimulated, they generated tetanic forces measured with an automated motion tracking system. Thirty-one compounds of interest as potential treatments for patients with DMD were tested at 3 to 6 concentrations. Eleven of the compounds (insulin-like growth factor-1, creatine, beta-hydroxy-beta-methylbutyrate, trichostatin A, lisinopril, and 6 from the glucocorticoid family) significantly increased tetanic force relative to placebo-treated controls. The glucocorticoids methylprednisolone, deflazacort, and prednisone increased tetanic forces at low doses (EC(50) of 6, 19, and 56 nM, respectively), indicating a direct muscle mechanism by which they may be benefitting DMD patients. The tetanic force assay also identified beneficial compound interactions (arginine plus deflazacort and prednisone plus creatine) as well as deleterious interactions (prednisone plus creatine inhibited by pentoxifylline) of combinatorial therapies taken by some DMD patients. Since mdx muscle in vivo and DMD patients respond in a similar manner to many of these compounds, the in vitro assay will be a useful tool for the rapid identification of new potential treatments for muscle weakness in DMD and other muscle disorders.


Subject(s)
Drug Evaluation, Preclinical/methods , Muscle Contraction/drug effects , Muscular Dystrophy, Duchenne/drug therapy , Myoblasts/drug effects , Animals , Electric Stimulation , Male , Mice , Mice, Inbred mdx , Muscle Contraction/physiology , Myoblasts/physiology , Tissue Engineering
8.
Muscle Nerve ; 37(4): 438-47, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18236465

ABSTRACT

A tissue-based approach to in vitro drug screening allows for determination of the cumulative positive and negative effects of a drug at the tissue rather than the cellular or subcellular level. Skeletal muscle myoblasts were tissue-engineered into three-dimensional muscle with parallel myofibers generating directed forces. When grown attached to two flexible micro-posts (mu posts) acting as artificial tendons in a 96-well plate format, the miniature bioartificial muscles (mBAMs) generated tetanic (active) forces upon electrical stimulation measured with a novel image-based motion detection system. mBAM myofiber hypertrophy and active force increased in response to insulin-like growth factor 1. In contrast, mBAM deterioration and weakness was observed with a cholesterol-lowering statin. The results described in this study demonstrate the integration of tissue engineering and biomechanical testing into a single platform for the screening of compounds affecting muscle strength.


Subject(s)
Biological Assay/methods , Drug Evaluation, Preclinical/methods , Muscle Contraction/drug effects , Muscular Atrophy/drug therapy , Tissue Engineering/methods , Animals , Atorvastatin , Biological Assay/instrumentation , Cells, Cultured , Drug Evaluation, Preclinical/instrumentation , Electric Stimulation , Heptanoic Acids/pharmacology , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Insulin-Like Growth Factor I/pharmacology , Male , Mice , Mice, Inbred C3H , Muscular Atrophy/physiopathology , Myoblasts/cytology , Myoblasts/drug effects , Pyrroles/pharmacology , Tissue Engineering/instrumentation
9.
Biomaterials ; 29(1): 75-84, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17928049

ABSTRACT

Skeletal muscle transplantation strategies for muscle repair or gene therapy involve either the injection of proliferating myoblasts followed by fusion with host myofibers or implantation of ex vivo differentiated myofibers; however, both implant procedures are associated with significant cell loss. Biodegradable porous, gas-foamed poly-lactide-co-glycolide (PLG) scaffolds have desirable characteristics for cell transfer and were used to study attachment, growth, differentiation and survival of human myogenic cells. Primary human myoblasts suspended in clinical grade extracellular matrixes (ECMs) and adhered to PLG scaffolds differentiated in vitro into high-density tropomyosin positive myofibers. An immunodeficient non-obese diabetic/severe combined immunodeficiency (NOD/SCID) mouse implant model was used to study the transfer and in vivo survival of differentiated human myofibers on these scaffolds. Scaffold rigidity allowed the myofibers to be maintained under tension in vitro and following subcutaneous transplantation in vivo. Following implantation, myofiber density on the PLG scaffolds decreased linearly by 78% over a 4-week period. ECM composed of either Tisseel fibrin or Zyderm collagen type I did not significantly affect in vivo cell viability over the 4-week period. Varying PLG scaffold microsphere content (10-100%) also had little effect on cell survival in vivo. In contrast, when the residual NK cell population in the immunodeficient NOD/SCID mouse model was depleted with anti-asialo GM1 (ASGM1) antiserum, in vivo cell survival significantly increased from 22% to 34% after 4 weeks. With further improvements in cell survival, PLG scaffolds may prove useful for the implantation of primary human myofibers in future clinical applications.


Subject(s)
Absorbable Implants , Cell Culture Techniques/methods , Cell Differentiation , Extracellular Matrix/metabolism , Musculoskeletal System/cytology , Animals , Cell Survival , Cell Transplantation , Cells, Cultured , Humans , Killer Cells, Natural , Mice
10.
Tissue Eng ; 12(7): 1833-41, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16889513

ABSTRACT

Bioengineered tissues transduced to secrete recombinant proteins may serve as a long-term delivery vehicle for therapeutic proteins when implanted in vivo. Insulin-like growth factor 1 (IGF1) is an anabolic growth factor for skeletal muscle that can stimulate myoblast proliferation and myofiber hypertrophy. To determine whether the release of IGF1 from an engineered bioartificial skeletal muscle (BAM) could stimulate the growth of skeletal muscle in a paracrine manner, we established an in vitro perfusion system for genetically engineered IGF1 BAMs. BAMs were bioengineered from C2C12 murine myoblasts stably transduced with a retroviral vector to synthesize and secrete IGF1 (C2-IGF1 BAMs). C2-IGF1 BAMs or nontransduced control C2 BAMs were cocultured with avian BAMS (ABAMs) in constantly perfused biochambers. During 11 days of perfusion, IGF1 levels in the C2-IGF1 BAM perfusion medium increased linearly from 1 to 20 ng/mL. The ABAMs maintained in biochambers with the C2-IGF1 BAMs had significantly more myofibers (69%, p < 0.005) and larger myofiber cross-sectional areas (40%, p < 0.001) compared to those cocultured with control C2 BAMs. These studies show that levels of IGF1 secreted from the C2-IGF1 BAMs are sufficient to produce an anabolic paracrine effect on nongenetically engineered BAMs, and the in vitro perfusion system provides a model for screening proteins effective in stimulating localized skeletal muscle growth.


Subject(s)
Bioreactors , Insulin-Like Growth Factor I/biosynthesis , Muscle, Skeletal/growth & development , Myoblasts, Skeletal/physiology , Paracrine Communication , Tissue Engineering , Animals , Chick Embryo , Insulin-Like Growth Factor I/genetics , Muscle, Skeletal/cytology , Myoblasts, Skeletal/cytology , Paracrine Communication/genetics , Transduction, Genetic
11.
Mol Ther ; 14(3): 442-51, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16750937

ABSTRACT

Human muscle progenitor cells transduced with lentiviral vectors secreted high levels of blood clotting factor IX (FIX). When bioengineered into postmitotic myofibers as human bioartificial muscles (HBAMs) and subcutaneously implanted into immunodeficient mice, they secreted FIX into the circulation for >3 months. The HBAM-derived FIX was biologically active, consistent with the cells' ability to conduct the necessary posttranslational modifications. These bioengineered muscle implants are retrievable, an inherent safety feature that distinguishes this "reversible" gene therapy approach from most other gene therapy strategies. When myofibers were bioengineered from human myoblasts expressing FIX and vascular endothelial growth factor, circulating FIX levels were increased and maintained long term within the therapeutic range, consistent with the generation of a vascular network around the HBAM. The present study implicates an important role for angiogenesis in the efficient delivery of therapeutic proteins using tissue engineered stem cell-based gene therapies.


Subject(s)
Factor IX/genetics , Genetic Therapy/methods , Hemophilia A/therapy , Myoblasts, Skeletal/metabolism , Myoblasts, Skeletal/transplantation , Neovascularization, Physiologic , Tissue Engineering , Animals , Genetic Vectors/genetics , Green Fluorescent Proteins/analysis , Green Fluorescent Proteins/genetics , Humans , Lentivirus/genetics , Mice , Mice, SCID , Neovascularization, Physiologic/genetics , Transduction, Genetic , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...