Zephyrnet Logo

Bioprocessing technology of muscle stem cells: implications for cultured meat

Date:

    • Mukund K.
    • Subramaniam S.

    Skeletal muscle: a review of molecular structure and function, in health and disease.

    Rev. Syst. Biol. Med. 2020; 12: 46

    • Post M.J.
    • et al.

    Scientific, sustainability and regulatory challenges of cultured meat.

    Nat. Food. 2020; 1: 403-415

    • Guan X.
    • et al.

    Trends and ideas in technology, regulation and public acceptance of cultured meat.

    Future Foods. 2021; 3100032

  • Cultured beef: medical technology to produce food.

    J. Sci. Food Agric. 2014; 94: 1039-1041

    • Feige P.
    • et al.

    Orienting muscle stem cells for regeneration in homeostasis, aging, and disease.

    Cell Stem Cell. 2018; 23: 653-664

    • Relaix F.
    • et al.

    Perspectives on skeletal muscle stem cells.

    Nat. Commun. 2021; 12: 692

    • Gonzalez M.L.
    • et al.

    Satellite cells and their regulation in livestock.

    J. Anim. Sci. 2020; 98skaa081

    • Tierney M.T.
    • Sacco A.

    Satellite cell heterogeneity in skeletal muscle homeostasis.

    Trends Cell Biol. 2016; 26: 434-444

    • 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

    • Yin H.
    • et al.

    Satellite cells and the muscle stem cell niche.

    Physiol. Rev. 2013; 93: 23-67

    • Ding S.
    • et al.

    Characterization and isolation of highly purified porcine satellite cells.

    Cell Death Discov. 2017; 3: 17003

    • Metzger K.
    • et al.

    Establishment and validation of cell pools using primary muscle cells derived from satellite cells of pig skeletal muscle.

    In Vitro Cell Dev. Biol. Anim. 2020; 56: 193-199

    • Choi K.-H.
    • et al.

    Muscle stem cell isolation and in vitro culture for meat production: a methodological review.

    Compr. Rev. Food Sci. Food Saf. 2020; 20: 429-457

    • Matsuyoshi Y.
    • et al.

    Isolation and purification of satellite cells from young rats by Percoll density gradient centrifugation.

    Methods Mol. Biol. 2019; 1889: 81-93

    • Evano B.
    • Tajbakhsh S.

    Skeletal muscle stem cells in comfort and stress.

    NPJ Regen. Med. 2018; 3: 24

    • Benedetti A.
    • et al.

    A novel approach for the isolation and long-term expansion of pure satellite cells based on ice-cold treatment.

    Skelet. Muscle. 2021; 11: 7

    • Hoang V.T.
    • et al.

    Standardized xeno- and serum-free culture platform enables large-scale expansion of high-quality mesenchymal stem/stromal cells from perinatal and adult tissue sources.

    Cytotherapy. 2021; 23: 88-99

    • Solis-Castro O.O.
    • et al.

    Establishment and neural differentiation of neural crest-derived stem cells (NCSCS) from human dental pulp in serum-free conditions.

    Stem Cells Transl. Med. 2020; 9: 1462-1476

    • Kolkmann A.M.
    • et al.

    Serum-free media for the growth of primary bovine myoblasts.

    Cytotechnology. 2020; 72: 111-120

    • Zhang X.
    • et al.

    A novel chemically defined serum- and feeder-free medium for undifferentiated growth of porcine pluripotent stem cells.

    J. Cell. Physiol. 2019; 234: 15380-15394

    • Wilkinson A.C.
    • et al.

    Long-term ex vivo haematopoietic-stem-cell expansion allows nonconditioned transplantation.

    Nature. 2019; 571: 117-121

    • Fu X.
    • et al.

    Combination of inflammation-related cytokines promotes long-term muscle stem cell expansion.

    Cell Res. 2015; 25: 655-673

    • Garcia S.M.
    • et al.

    High-yield purification, preservation, and serial transplantation of human satellite cells.

    Stem Cell Rep. 2018; 10: 1160-1174

    • Xu C.
    • et al.

    A zebrafish embryo culture system defines factors that promote vertebrate myogenesis across species.

    Cell. 2013; 155: 909-921

    • Gilbert P.M.
    • et al.

    Substrate elasticity regulates skeletal muscle stem cell self-renewal in culture.

    Science. 2010; 329: 1078-1081

    • Arandel L.
    • et al.

    Immortalized human myotonic dystrophy muscle cell lines to assess therapeutic compounds.

    Dis. Model. Mech. 2017; 10: 487

    • Massenet J.
    • et al.

    Derivation and characterization of immortalized human muscle satellite cell clones from muscular dystrophy patients and healthy individuals.

    Cells. 2020; 9: 1780

    • Xiong Y.L.

    Muscle proteins.

    in: Yada R.Y. Proteins in food processing. (2nd edn). Woodhead, 2018: 127-148

    • Kang D.-H.
    • et al.

    Engineered whole cut meat-like tissue by the assembly of cell fibers using tendon-gel integrated bioprinting.

    Nat. Commun. 2021; 12: 5059

    • Reza M.M.
    • et al.

    Irisin is a pro-myogenic factor that induces skeletal muscle hypertrophy and rescues denervation-induced atrophy.

    Nat. Commun. 2017; 8: 1104

    • Rodríguez-Fdez S.
    • et al.

    Vav2 catalysis-dependent pathways contribute to skeletal muscle growth and metabolic homeostasis.

    Nat. Commun. 2020; 11: 5808

  • Bioenergetics mechanisms regulating muscle stem cell self-renewal commitment and function.

    Biomed. Pharmacother. 2018; 103: 463-472

    • Bhattacharya D.
    • Scimè A.

    Mitochondrial function in muscle stem cell fates.

    Front. Cell Dev. Biol. 2020; 8: 480

    • Handschin C.
    • Spiegelman B.M.

    PGC-1 coactivators and the regulation of skeletal muscle fiber-type determination.

    Cell Metab. 2011; 13: 351

    • Yamaguchi T.
    • et al.

    Continuous mild heat stress induces differentiation of mammalian myoblasts, shifting fiber type from fast to slow.

    Am. J. Physiol. Cell Physiol. 2010; 298: C140-C148

    • Akimoto T.
    • et al.

    Exercise stimulates PGC-1α transcription in skeletal muscle through activation of the p38 MAPK pathway.

    J. Biol. Chem. 2005; 280: 19587-19593

  • A review on the genetic regulation of myogenesis and muscle development.

    Am. J. Biochem. Biotechnol. 2019; 15: 1-12

    • Allan S.J.
    • et al.

    Bioprocess design considerations for cultured meat production with a focus on the expansion bioreactor.

    Front. Sustain. Food Syst. 2019; 3: 9

    • Zhang G.
    • et al.

    Challenges and possibilities for bio-manufacturing cultured meat.

    Trends Food Sci. Technol. 2020; 97: 443-450

    • Li X.
    • et al.

    A conceptual air-lift reactor design for large scale animal cell cultivation in the context of in vitro meat production.

    Chem. Eng. Sci. 2020; 211115269

    • Simaria A.S.
    • et al.

    Allogeneic cell therapy bioprocess economics and optimization: single-use cell expansion technologies.

    Biotechnol. Bioeng. 2014; 111: 69-83

    • Xing Z.Z.
    • et al.

    Scale-up analysis for a CHO cell culture process in large-scale bioreactors.

    Biotechnol. Bioeng. 2009; 103: 733-746

    • Stephenson M.
    • Grayson W.

    Recent advances in bioreactors for cell-based therapies.

    F1000Res. 2018; 7: 517

    • Grein T.A.
    • et al.

    Multiphase mixing characteristics in a microcarrier-based stirred tank bioreactor suitable for human mesenchymal stem cell expansion.

    Process Biochem. 2016; 51: 1109-1119

    • Wei Y.
    • et al.

    Human skeletal muscle-derived stem cells retain stem cell properties after expansion in myosphere culture.

    Exp. Cell Res. 2011; 317: 1016-1027

    • Bodiou V.
    • et al.

    Microcarriers for upscaling cultured meat production.

    Front. Nutr. 2020; 7: 10

    • Vergeer R.
    • et al.

    Tea of cultivated meat – future projections of different scenarios.

    CE Delft, 2021

    • Rangarajan S.
    • et al.

    Use of flow, electrical, and mechanical stimulation to promote engineering of striated muscles.

    Ann. Biomed. Eng. 2014; 42: 1391-1405

    • Kang H.W.
    • et al.

    A 3D bioprinting system to produce human-scale tissue constructs with structural integrity.

    Nat. Biotechnol. 2016; 34: 312-319

    • Wang J.
    • et al.

    The construction and application of three-dimensional biomaterials.

    Adv. Biosyst. 2020; 4e1900238

    • MacQueen L.A.
    • et al.

    Muscle tissue engineering in fibrous gelatin: implications for meat analogs.

    NPJ Sci. Food. 2019; 3: 20

    • Jiao Y.
    • et al.

    Construction and application of the textile-based tissue engineering scaffold: a review.

    Biomater. Sci. 2020; 8: 3574-3600

    • Ben Arye T.
    • et al.

    Textured soy protein scaffolds enable the generation of three-dimensional bovine skeletal muscle tissue for cell-based meat.

    Nat. Food. 2020; 1: 210-220

    • Verbruggen S.
    • et al.

    Bovine myoblast cell production in a microcarriers-based system.

    Cytotechnology. 2018; 70: 503-512

    • Shoba E.
    • et al.

    Strategic design of cardiac mimetic core-shell nanofibrous scaffold impregnated with salvianolic acid B and magnesium L-ascorbic acid 2 phosphate for myoblast differentiation.

    Mater. Sci. Eng. C. 2018; 90: 131-147

    • Sadeghi A.
    • et al.

    Surface modification of electrospun PLGA scaffold with collagen for bioengineered skin substitutes.

    Mater. Sci. Eng. C. 2016; 66: 130-137

    • Hocquette J.F.
    • et al.

    Intramuscular fat content in meat-producing animals: development, genetic and nutritional control, and identification of putative markers.

    Animal. 2010; 4: 303-319

    • Venter C.
    • Niesler C.

    A triple co-culture method to investigate the effect of macrophages and fibroblasts on myoblast proliferation and migration.

    Biotechniques. 2018; 64: 52-58

    • Simsa R.
    • et al.

    Extracellular heme proteins influence bovine myosatellite cell proliferation and the color of cell-based meat.

    Foods. 2019; 8: 521

    • Stephens N.
    • et al.

    Bringing cultured meat to market: technical, socio-political, and regulatory challenges in cellular agriculture.

    Trends Food Sci. Technol. 2018; 78: 155-166

    • Yu Z.
    • et al.

    The effects of algal extracellular substances on algal growth, metabolism and long-term medium recycle, and inhibition alleviation through ultrasonication.

    Bioresour. Technol. 2018; 267: 192-200

    • Mohorcich J.
    • Reese J.

    Cell-cultured meat: lessons from GMO adoption and resistance.

    Appetite. 2019; 143: 9

    • Lee H.J.
    • et al.

    Status of meat alternatives and their potential role in the future meat market – a review.

    Asian Australas. J. Anim. Sci. 2020; 33: 1533-1543

    • Kuppusamy P.
    • et al.

    Adipose and muscle cell co-culture system: a novel in vitro tool to mimic the in vivo cellular environment.

    Biology. 2020; 10: 6

    • Krieger J.
    • et al.

    3D skeletal muscle fascicle engineering is improved with TGF-β1 treatment of myogenic cells and their co-culture with myofibroblasts.

    PeerJ. 2018; 6e4939

    • Evano B.
    • et al.

    Dynamics of asymmetric and symmetric divisions of muscle stem cells in vivo and on artificial niches.

    Cell Rep. 2020; 30: 3195-3206

    • Dumont N.A.
    • et al.

    Intrinsic and extrinsic mechanisms regulating satellite cell function.

    Development. 2015; 142: 1572-1581

    • Rose L.
    • Gönczy P.

    Polarity establishment, asymmetric division and segregation of fate determinants in early C. elegans embryos.

    WormBook. 2014; ()

    • Carmena A.

    The case of the scribble polarity module in asymmetric neuroblast division in development and tumorigenesis.

    Int. J. Mol. Sci. 2020; 21: 2865

    • Chen C.
    • et al.

    The ins(ide) and outs(ide) of asymmetric stem cell division.

    Curr. Opin. Cell Biol. 2016; 43: 1-6

    • Segalés J.
    • et al.

    Regulation of muscle stem cell functions: a focus on the p38 MAPK signaling pathway.

    Front. Cell Dev. Biol. 2016; 4: 91

    • Wei Q.
    • Paterson B.M.

    Regulation of MyoD function in the dividing myoblast.

    FEBS Lett. 2001; 490: 171-178

    • Borodinsky
    • et al.

    Calcium signaling in skeletal muscle development, maintenance and regeneration.

    Cell Calcium. 2016; 59: 91-97

    • Saxton R.A.
    • Sabatini D.M.

    mTOR signaling in growth, metabolism, and disease.

    Cell. 2017; 169: 361-371

    • Popov D.V.
    • et al.

    Regulation of PGC-1α isoform expression in skeletal muscles.

    Acta Nat. 2015; 7: 48-59

    • Cui S.
    • et al.

    Β-Catenin is essential for differentiation of primary myoblasts via cooperation with MyoD and α-catenin.

    Development. 2019; 146dev167080

    • Ding S.
    • et al.

    Maintaining bovine satellite cells stemness through p38 pathway.

    Sci. Rep. 2018; 8: 10808

    • Anderson J.E.

    Hepatocyte growth factor and satellite cell activation.

    Adv. Exp. Med. Biol. 2016; 900: 1-25

    • Machida S.
    • Booth F.W.

    Insulin-like growth factor 1 and muscle growth: implication for satellite cell proliferation.

    Proc. Nutr. Soc. 2004; 63: 337-340

    • Golding J.P.
    • et al.

    Skeletal muscle stem cells express anti-apoptotic ErbB receptors during activation from quiescence.

    Exp. Cell Res. 2007; 313: 341-356

    • Asakura A.
    • Verma M.

    Muscle satellite cell cross-talk with a vascular niche maintains quiescence via VEGF and Notch signaling.

    Cell Stem Cell. 2018; 23: 530-543

    • Galluzzo P.
    • et al.

    17β-Estradiol regulates the first steps of skeletal muscle cell differentiation via ER-α-mediated signals.

    Am. J. Physiol. Cell Physiol. 2009; 297: C1249-C1262

    • Lacham-Kaplan O.
    • et al.

    Divergent regulation of myotube formation and gene expression by E2 and EPA during in-vitro differentiation of C2C12 myoblasts.

    Int. J. Mol. Sci. 2020; 21: 745

    • Wang X.J.
    • et al.

    Glucocorticoids retard skeletal muscle development and myoblast protein synthesis through a mechanistic target of rapamycin (MTOR)-signaling pathway in broilers (Gallus gallus domesticus).

    Stress. 2015; 18: 686-698

    • Zhao P.
    • Hoffman E.P.

    Embryonic myogenesis pathways in muscle regeneration.

    Dev. Dyn. 2004; 229: 380-392

    • Baghdadi M.B.
    • et al.

    Reciprocal signalling by Notch–collagen V–CALCR retains muscle stem cells in their niche.

    Nature. 2018; 557: 714-718

    • Li X.
    • et al.

    Effects of COL8a1 on the proliferation of muscle-derived satellite cells.

    Cell Biol. Int. 2018; 42: 1132-1140

    • Ishii K.
    • et al.

    Recapitulation of extracellular laminin environment maintains stemness of satellite cells in vitro.

    Stem Cell Rep. 2018; 10: 568-582

    • Charville G.W.
    • et al.

    Ex vivo expansion and in vivo self-renewal of human muscle stem cells.

    Stem Cell Rep. 2015; 5: 621-632

    • Flamini V.
    • et al.

    The satellite cell niche regulates the balance between myoblast differentiation and self-renewal via p53.

    Stem Cell Rep. 2018; 10: 970-983

    • Judson R.N.
    • et al.

    Inhibition of methyltransferase Setd7 allows the in vitro expansion of myogenic stem cells with improved therapeutic potential.

    Cell Stem Cell. 2018; 22: 177-190.e7

    • Price F.D.
    • et al.

    Inhibition of JAK–STAT signaling stimulates adult satellite cell function.

    Nat. Med. 2014; 20: 1174-1181

    • Zismanov V.
    • et al.

    Phosphorylation of EIF2α is a translational control mechanism regulating muscle stem cell quiescence and self-renewal.

    Cell Stem Cell. 2016; 18: 79-90

    • Monge C.
    • et al.

    Quiescence of human muscle stem cells is favored by culture on natural biopolymeric films.

    Stem Cell Res. Ther. 2017; 8: 104

    • Baghdadi M.B.
    • et al.

    Notch-induced miR-708 antagonizes satellite cell migration and maintains quiescence.

    Cell Stem Cell. 2018; 23: 859

    • Sato T.
    • et al.

    miR-195/497 induce postnatal quiescence of skeletal muscle stem cells.

    Nat. Commun. 2014; 5: 4597

  • Source: https://www.cell.com/trends/biotechnology/fulltext/S0167-7799(21)00265-1?rss=yes

    spot_img

    Latest Intelligence

    spot_img