Preclinical research studies for treating severe muscular injuries: focus on tissue-engineered strategies

Preclinical research studies for treating severe muscular injuries: focus on tissue-engineered strategies

Source Node: 1851517
    • Vandusen K.W.
    • et al.

    Engineered skeletal muscle units for repair of volumetric muscle loss in the tibialis anterior muscle of a rat.

    Tissue Eng. A. 2014; 20: 2920-2930

    • Mertens J.P.
    • et al.

    Engineering muscle constructs for the creation of functional engineered musculoskeletal tissue.

    Regen. Med. 2014; 9: 89-100

    • Li M.T.A.
    • et al.

    Functional analysis of limb recovery following autograft treatment of volumetric muscle loss in the quadriceps femoris.

    J. Biomech. 2014; 47: 2013-2021

    • Kasprzycka P.
    • et al.

    The factors present in regenerating muscles impact bone marrow-derived mesenchymal stromal/stem cell fusion with myoblasts.

    Stem Cell Res Ther. 2019; 10: 1-17

    • Smoak M.M.
    • Mikos A.G.

    Advances in biomaterials for skeletal muscle engineering and obstacles still to overcome.

    Mater. Today Bio. 2020; 7100069

    • Grasman J.M.
    • et al.

    Biomimetic scaffolds for regeneration of volumetric muscle loss in skeletal muscle injuries.

    Acta Biomater. 2015; 25: 2-15

    • Cezar C.A.
    • Mooney D.J.

    Biomaterial-based delivery for skeletal muscle repair.

    Adv. Drug Deliv. Rev. 2015; 84: 188-197

    • Juhas M.
    • et al.

    Design, evaluation, and application of engineered skeletal muscle.

    Methods. 2016; 99: 81-90

    • Dessauge F.
    • et al.

    3D in vitro models of skeletal muscle: myopshere, myobundle and bioprinted muscle construct.

    Vet. Res. 2021; 52: 1-12

    • Kho A.T.
    • et al.

    Transcriptome-scale similarities between mouse and human skeletal muscles with normal and myopathic phenotypes.

    BMC Musculoskelet. Disord. 2006; 7: 1-9

    • Ziermann J.M.
    • et al.

    Anatomical comparison across heads, fore- and hindlimbs in mammals using network models.

    J. Anat. 2021; 239: 12-31

    • Halsey L.G.
    • White C.R.

    Terrestrial locomotion energy costs vary considerably between species: no evidence that this is explained by rate of leg force production or ecology.

    Sci. Reports. 2019; 9: 1-13

    • Bonetto A.
    • et al.

    Assessment of muscle mass and strength in mice.

    Bonekey Rep. 2015; 4: 732

    • Kin S.
    • et al.

    Regeneration of skeletal muscle using in situ tissue engineering on an acellular collagen sponge scaffold in a rabbit model.

    ASAIO J. 2007; 53: 506-513

    • Ward C.L.
    • et al.

    Autologous minced muscle grafts improve muscle strength in a porcine model of volumetric muscle loss injury.

    J. Orthop. Trauma. 2016; 30: e396-e403

    • Turner N.J.
    • et al.

    Biologic scaffold remodeling in a dog model of complex musculoskeletal injury.

    J. Surg. Res. 2012; 176: 490-502

    • Musci R.V.
    • et al.

    The Dunkin Hartley guinea pig is a model of primary osteoarthritis that also exhibits early onset myofiber remodeling that resembles human musculoskeletal aging.

    Front. Physiol. 2020; 11: 1412

    • Nimphius S.
    • et al.

    Comparison of quadriceps and hamstring muscle activity during an isometric squat between strength-matched men and women.

    J. Sports Sci. Med. 2019; 18: 101

    • Hardy D.
    • et al.

    Comparative study of injury models for studying muscle regeneration in mice.

    PLoS ONE. 2016; 11e0147198

    • Le G.
    • et al.

    Freeze injury of the tibialis anterior muscle.

    Methods Mol. Biol. 2016; 1460: 33-41

    • Alheib O.
    • et al.

    Injectable laminin-biofunctionalized gellan gum hydrogels loaded with myoblasts for skeletal muscle regeneration.

    Acta Biomater. 2022; 143: 282-294

    • Ge J.
    • et al.

    Gold and gold-silver alloy nanoparticles enhance the myogenic differentiation of myoblasts through p38 MAPK signaling pathway and promote in vivo skeletal muscle regeneration.

    Biomaterials. 2018; 175: 19-29

    • Guo B.
    • et al.

    Degradable conductive self-healing hydrogels based on dextran-graft-tetraaniline and N-carboxyethyl chitosan as injectable carriers for myoblast cell therapy and muscle regeneration.

    Acta Biomater. 2019; 84: 180-193

    • Davoudi S.
    • et al.

    Muscle stem cell intramuscular delivery within hyaluronan methylcellulose improves engraftment efficiency and dispersion.

    Biomaterials. 2018; 173: 34-46

    • Rossi C.A.
    • et al.

    In vivo tissue engineering of functional skeletal muscle by freshly isolated satellite cells embedded in a photopolymerizable hydrogel.

    FASEB J. 2011; 25: 2296-2304

    • Gilbert-Honick J.
    • et al.

    Adipose-derived stem/stromal cells on electrospun fibrin microfiber bundles enable moderate muscle reconstruction in a volumetric muscle loss model.

    Cell Transplant. 2018; 27: 1644-1656

    • Nakayama K.H.
    • et al.

    Treatment of volumetric muscle loss in mice using nanofibrillar scaffolds enhances vascular organization and integration.

    Commun. Biol. 2019; 2: 170

    • Gilbert-Honick J.
    • et al.

    Engineering functional and histological regeneration of vascularized skeletal muscle.

    Biomaterials. 2018; 164: 70-79

    • Yang H.S.
    • et al.

    Nanopatterned muscle cell patches for enhanced myogenesis and dystrophin expression in a mouse model of muscular dystrophy.

    Biomaterials. 2014; 35: 1478-1486

    • Levenberg S.
    • et al.

    Engineering vascularized skeletal muscle tissue.

    Nat. Biotechnol. 2005; 23: 879-884

    • Lee E.M.
    • et al.

    Therapeutic effects of mouse adipose-derived stem cells and losartan in the skeletal muscle of injured mdx mice.

    Cell Transplant. 2015; 24: 939-953

    • Yucel N.
    • et al.

    Humanizing the mdx mouse model of DMD: the long and the short of it.

    npj Regen. Med. 2018; 3: 1-11

    • Laumonier T.
    • Menetrey J.

    Muscle injuries and strategies for improving their repair.

    J. Exp. Orthop. 2016; 3: 15

    • Hwang J.H.
    • et al.

    Combination therapy of human adipose-derived stem cells and basic fibroblast growth factor hydrogel in muscle regeneration.

    Biomaterials. 2013; 34: 6037-6045

    • Takanari K.
    • et al.

    Skeletal muscle derived stem cells microintegrated into a biodegradable elastomer for reconstruction of the abdominal wall.

    Biomaterials. 2017; 113: 31-41

    • Okano T.
    • Matsuda T.

    Muscular tissue engineering: capillary-incorporated hybrid muscular tissues in vivo tissue culture.

    Cell Transplant. 1998; 7: 435-442

    • Ding K.
    • et al.

    Injectable thermosensitive chitosan/b-glycerophosphate/collagen hydrogel maintains the plasticity of skeletal muscle satellite cells and supports their in vivo viability.

    Cell Biol. Int. 2013; 37: 977-987

    • Gorecka A.
    • et al.

    Autologous transplantation of adipose-derived stem cells improves functional recovery of skeletal muscle without direct participation in new myofiber formation.

    Stem Cell Res Ther. 2018; 9: 195

    • Costantini M.
    • et al.

    Microfluidic-enhanced 3D bioprinting of aligned myoblast-laden hydrogels leads to functionally organized myofibers in vitro and in vivo.

    Biomaterials. 2017; 131: 98-110

    • Ko I.K.
    • et al.

    The effect of in vitro formation of acetylcholine receptor (AChR) clusters in engineered muscle fibers on subsequent innervation of constructs in vivo.

    Biomaterials. 2013; 34: 3246-3255

    • Beier J.P.
    • et al.

    Tissue engineering of injectable muscle: three-dimensional myoblast-fibrin injection in the syngeneic rat animal model.

    Plast. Reconstr. Surg. 2006; 118: 1113-1121

    • Juhas M.
    • et al.

    Biomimetic engineered muscle with capacity for vascular integration and functional maturation in vivo.

    Proc. Natl. Acad. Sci. U. S. A. 2014; 111: 5508-5513

    • Cimenci C.E.
    • et al.

    Laminin mimetic peptide nanofibers regenerate acute muscle defect.

    Acta Biomater. 2017; 60: 190-200

    • Pumberger M.
    • et al.

    Synthetic niche to modulate regenerative potential of MSCs and enhance skeletal muscle regeneration.

    Biomaterials. 2016; 99: 95-108

    • Casella A.
    • et al.

    Endogenous electric signaling as a blueprint for conductive materials in tissue engineering.

    Bioelectricity. 2021; 3: 27-41

    • Dong R.
    • et al.

    Conductive biomaterials for muscle tissue engineering.

    Biomaterials. 2020; 229119584

    • da Silva L.P.
    • et al.

    Electric phenomenon: a disregarded tool in tissue engineering and regenerative medicine.

    Trends Biotechnol. 2020; 38: 24-49

    • Srisuk P.
    • et al.

    Electroactive gellan gum/polyaniline spongy-like hydrogels.

    ACS Biomater. Sci. Eng. 2018; 4: 1779-1787

    • Du Y.
    • et al.

    Biomimetic elastomeric, conductive and biodegradable polycitrate-based nanocomposites for guiding myogenic differentiation and skeletal muscle regeneration.

    Biomaterials. 2018; 157: 40-50

    • Kim M.H.
    • et al.

    The effect of VEGF on the myogenic differentiation of adipose tissue derived stem cells within thermosensitive hydrogel matrices.

    Biomaterials. 2010; 31: 1213-1218

    • Garg K.
    • et al.

    Losartan administration reduces fibrosis but hinders functional recovery after volumetric muscle loss injury.

    J. Appl. Physiol. 2014; 117: 1120-1131

    • Fakhfakh R.
    • et al.

    Losartan enhances the success of myoblast transplantation.

    Cell Transplant. 2012; 21: 139-152

    • Park J.K.
    • et al.

    Losartan improves adipose tissue-derived stem cell niche by inhibiting transforming growth factor-β and fibrosis in skeletal muscle injury.

    Cell Transplant. 2012; 21: 2407-2424

    • Sharma P.
    • et al.

    Stem cells and growth factors-based delivery approaches for chronic wound repair and regeneration: a promise to heal from within.

    Life Sci. 2021; 268118932

    • Syverud B.C.
    • et al.

    Isolation and purification of satellite cells for skeletal muscle tissue engineering.

    J. Regen. Med. 2014; 3: 117

    • Kozakowska M.
    • et al.

    Myoblast-conditioned media improve regeneration and revascularization of ischemic muscles in diabetic mice.

    Stem Cell Res Ther. 2015; 6: 61

    • Wolff T.
    • et al.

    FACS-purified myoblasts producing controlled VEGF levels induce safe and stable angiogenesis in chronic hind limb ischemia.

    J. Cell. Mol. Med. 2012; 16: 107-117

    • Elhussieny A.
    • et al.

    Mesenchymal stem cells for regenerative medicine for Duchenne muscular dystrophy.

    in: Gaina G. Muscular Dystrophy: Research Updates and Therapeutic Strategies. IntechOpen, 2020https://doi.org/10.5772/intechopen.92824

    • Sun Z.
    • et al.

    In vitro differentiation of rat mesenchymal stem cells into skeletal muscle cells induced by myoblast differentiation factor and 5-azacytidine.

    Chinese J. Reparative Reconstr. Surg. 2007; 21: 1371-1375

    • Kapur S.K.
    • Katz A.J.

    Review of the adipose derived stem cell secretome.

    Biochimie. 2013; 95: 2222-2228

    • Salgado A.J.
    • et al.

    Adipose tissue derived stem cells secretome: soluble factors and their roles in regenerative medicine.

    Curr. Stem Cell Res. Ther. 2010; 5: 103-110

    • Darabi R.
    • et al.

    Human ES- and iPS-derived myogenic progenitors restore DYSTROPHIN and improve contractility upon transplantation in dystrophic mice.

    Cell Stem Cell. 2012; 10: 610-619

    • Rao L.
    • et al.

    Engineering human pluripotent stem cells into a functional skeletal muscle tissue.

    Nat. Commun. 2018; 9: 126

    • Miyagoe-Suzuki Y.
    • Takeda S.

    Skeletal muscle generated from induced pluripotent stem cells – induction and application.

    World J. Stem Cells. 2017; 9: 8997

    • Deinsberger J.
    • et al.

    Global trends in clinical trials involving pluripotent stem cells: a systematic multi-database analysis.

    NPJ Regen. Med. 2020; 5: 15

    • Blaisdell F.

    The pathophysiology of skeletal muscle ischemia and the reperfusion syndrome: a review.

    Cardiovasc. Surg. 2002; 10: 620-630

    • Criswell T.L.
    • et al.

    The role of endothelial cells in myofiber differentiation and the vascularization and innervation of bioengineered muscle tissue in vivo.

    Biomaterials. 2013; 34: 140-149

    • Portalska K.J.
    • et al.

    Endothelial differentiation of mesenchymal stromal cells.

    PLoS ONE. 2012; 7e46842

    • Marelli-Berg F.M.
    • et al.

    Isolation of endothelial cells from murine tissue.

    J. Immunol. Methods. 2000; 244: 205-215

    • Tremblay J.P.
    • et al.

    Results of a triple blind clinical study of myoblast transplantations without immunosuppressive treatment in young boys with Duchenne muscular dystrophy.

    Cell Transplant. 1993; 2: 99-112

    • Tremblay J.P.
    • et al.

    Myoblast transplantation between monozygotic twin girl carriers of Duchenne muscular dystrophy.

    Neuromuscul. Disord. 1993; 3: 583-592

    • Walker P.A.
    • et al.

    Effect of needle diameter and flow rate on rat and human mesenchymal stromal cell characterization and viability.

    Tissue Eng. C Methods. 2010; 16: 989-997

    • Potts M.B.
    • et al.

    Devices for cell transplantation into the central nervous system: design considerations and emerging technologies.

    Surg. Neurol. Int. 2013; 4: 22

    • Li P.
    • et al.

    Transplantation of human umbilical cord-derived mesenchymal stems cells for the treatment of Becker muscular dystrophy in affected pedigree members.

    Int. J. Mol. Med. 2015; 35: 1051-1057

    • Dai A.
    • et al.

    Efficacy of stem cell therapy in ambulatory and nonambulatory children with Duchenne muscular dystrophy – Phase I-II.

    Degener. Neurol. Neuromuscul. Dis. 2018; 8: 63-77

    • Winkler T.
    • et al.

    Immunomodulatory placental-expanded, mesenchymal stromal cells improve muscle function following hip arthroplasty.

    J. Cachexia. Sarcopenia Muscle. 2018; 9: 880-897

    • Buckingham M.
    • et al.

    The formation of skeletal muscle: from somite to limb.

    J. Anat. 2003; 202: 59-68

    • Tajbakhsh S.

    Stem cells to tissue: molecular, cellular and anatomical heterogeneity in skeletal muscle.

    Curr. Opin. Genet. Dev. 2003; 13: 413-422

    • Kadi F.
    • et al.

    The behaviour of satellite cells in response to exercise: what have we learned from human studies?.

    Pflugers Arch. 2005; 451: 319-327

    • Seale P.
    • et al.

    Pax7 is required for the specification of myogenic satellite cells.

    Cell. 2000; 102: 777-786

    • Soleimani V.D.
    • et al.

    Transcriptional dominance of Pax7 in adult myogenesis is due to high-affinity recognition of homeodomain motifs.

    Dev. Cell. 2012; 22: 1208-1220

    • Hernández-Hernández J.M.
    • et al.

    The myogenic regulatory factors, determinants of muscle development, cell identity and regeneration.

    Semin. Cell Dev. Biol. 2017; 72: 10-18

    • Hinterberger T.J.
    • et al.

    Expression of the muscle regulatory factor MRF4 during somite and skeletal myofiber development.

    Dev. Biol. 1991; 147: 144-156

    • Wells L.
    • et al.

    Myosin heavy chain isoforms regulate muscle function but not myofibril assembly.

    EMBO J. 1996; 15: 4454-4459

    • Toumi H.
    • Best T.M.

    The inflammatory response: friend or enemy for muscle injury?.

    Br. J. Sports Med. 2003; 37: 284-286

    • Tidball J.G.

    Inflammatory cell response to acute muscle injury.

    Med. Sci. Sports Exerc. 1995; 27: 1022-1032

    • Lehto M.
    • et al.

    Skeletal muscle injury—molecular changes in the collagen during healing.

    Res. Exp. Med. 1985; 185: 95-106

    • Mann C.J.
    • et al.

    Aberrant repair and fibrosis development in skeletal muscle.

    Skelet. Muscle. 2011; 1: 1-20

    • Li Y.
    • Huard J.

    Differentiation of muscle-derived cells into myofibroblasts in injured skeletal muscle.

    Am. J. Pathol. 2002; 161: 895-907

    • Petrof B.J.
    • et al.

    Dystrophin protects the sarcolemma from stresses developed during muscle contraction.

    Proc. Natl. Acad. Sci. U. S. A. 1993; 90: 3710-3714

    • Colognato H.
    • et al.

    Laminin polymerization induces a receptor-cytoskeleton network.

    J. Cell Biol. 1999; 145: 619-631

    • Nomizu M.
    • et al.

    Active peptides from the carboxyl-terminal globular domain of laminin α2 and Drosophila α chains.

    FEBS Lett. 1996; 396: 37-42

    • Suzuki N.
    • et al.

    Identification of α-dystroglycan binding sequences in the laminin α2 chain LG4-5 module.

    Matrix Biol. 2010; 29: 143-151

  • Laminin enriched scaffolds for tissue engineering applications.

    Adv. Tissue Eng. Regen. Med. Open Access. 2017; 2: 00033

    • Sastry S.K.
    • et al.

    Integrin α subunit ratios, cytoplasmic domains, and growth factor synergy regulate muscle proliferation and differentiation.

    J. Cell Biol. 1996; 133: 169-184

    • Wang P.Y.
    • et al.

    The roles of RGD and grooved topography in the adhesion, morphology, and differentiation of C2C12 skeletal myoblasts.

    Biotechnol. Bioeng. 2012; 109: 2104-2115

    • Liu J.
    • et al.

    Increasing α7β1-integrin promotes muscle cell proliferation, adhesion, and resistance to apoptosis without changing gene expression.

    Am. J. Physiol. Cell Physiol. 2008; 294: C627-C640

    • Katagiri F.
    • et al.

    Screening of integrin-binding peptides in a laminin peptide library derived from the mouse laminin β chain short arm regions.

    Arch. Biochem. Biophys. 2014; 550: 33-41

    • Tashiro K.
    • et al.

    A synthetic peptide containing the IKVAV sequence from the A chain of laminin mediates cell attachment, migration, and neurite outgrowth.

    J. Biol. Chem. 1989; 264: 16174-16182

    • Koivunen E.
    • et al.

    Selection of peptides binding to the α5β1 integrin from phage display library.

    J. Biol. Chem. 1993; 268: 20205-20210

    • Miller K.J.
    • et al.

    Hepatocyte growth factor affects satellite cell activation and differentiation in regenerating skeletal muscle.

    Am. J. Phys. Cell Phys. 2000; 278: C174-C181

    • Ju Y.M.
    • et al.

    In situ regeneration of skeletal muscle tissue through host cell recruitment.

    Acta Biomater. 2014; 10: 4332-4339

    • Lefraucheur J.P.
    • Sebille A.

    Basic fibroblast growth factor promotes in vivo muscle regeneration in murine muscular dystrophy.

    Neurosci. Lett. 1995; 202: 121-124

    • Pawlikowski B.
    • et al.

    Regulation of skeletal muscle stem cells by fibroblast growth factors.

    Dev. Dyn. 2017; 246: 359-367

    • Vaidya T.B.
    • et al.

    Fibroblast growth factor and transforming growth factor beta repress transcription of the myogenic regulatory gene MyoD1.

    Mol. Cell. Biol. 1989; 9: 3576-3579

    • McFarlane C.
    • et al.

    Myostatin signals through Pax7 to regulate satellite cell self-renewal.

    Exp. Cell Res. 2008; 314: 317-329

    • Arsic N.
    • et al.

    Vascular endothelial growth factor stimulates skeletal muscle regeneration in Vivo.

    Mol. Ther. 2004; 10: 844-854

    • Wang X.
    • et al.

    Effects of interleukin-6, leukemia inhibitory factor, and ciliary neurotrophic factor on the proliferation and differentiation of adult human myoblasts.

    Cell. Mol. Neurobiol. 2008; 28: 113-124

    • AL, S.
    • et al.

    Interleukin-6 is an essential regulator of satellite cell-mediated skeletal muscle hypertrophy.

    Cell Metab. 2008; 7: 33-44

    • Milewska M.
    • et al.

    Interleukin-8 enhances myocilin expression, Akt-FoxO3 signaling and myogenic differentiation in rat skeletal muscle cells.

    J. Cell. Physiol. 2019; 234: 19675-19690

    • Moresi V.
    • et al.

    Tumor necrosis factor-α inhibition of skeletal muscle regeneration is mediated by a caspase-dependent stem cell response.

    Stem Cells. 2008; 26: 997-1008

    • Musarò A.
    • et al.

    IGF-1 induces skeletal myocyte hypertrophy through calcineurin in association with GATA-2 and NF-ATc1.

    Nature. 1999; 400: 581-585

    • Acharyya S.
    • et al.

    TNF inhibits notch-1 in skeletal muscle cells by Ezh2 and DNA methylation mediated repression: implications in Duchenne muscular dystrophy.

    PLoS ONE. 2010; 5e12479

    • Li G.
    • et al.

    IL-6 improves myogenesis in long-term skeletal muscle atrophy via the JAK/STAT3 signalling pathway.

    Int. J. Clin. Exp. Pathol. 2017; 10: 4023-4034

  • Time Stamp:

    More from Biotechnology Trends