Alternative fat: redefining adipocytes for biomanufacturing cultivated meat

Alternative fat: redefining adipocytes for biomanufacturing cultivated meat

Source Node: 1892368
    • Swartz B.
    • Bomkamp C.

    The Science of Cultivated Meat.

    Good Food Institute, 2022

    • Post M.J.
    • et al.

    Scientific, sustainability and regulatory challenges of cultured meat.

    Nat. Food. 2020; 1: 403-415

    • Teng P.M.
    • et al.

    The Evolving Singapore Agrifood Ecosystem.

    NTS Insight. 2019; ()

  • Requirements for the Safety Assessment of Novel Foods and Novel Food Ingredients.

    Singapore Food Agency (SFA), 2022

  • Will cell-based meat ever be a dinner staple?.

    Nature. 2020; 588: S64-S67

  • Club-goers take first bites of lab-made chicken.

    Nat. Biotechnol. 2021; 39: 257-258

    • Keast R.S.J.
    • Costanzo A.

    Is fat the sixth taste primary? Evidence and implications.

    Flavour. 2015; 4: 5

    • Liu D.
    • et al.

    Mechanism of fat taste perception: association with diet and obesity.

    Prog. Lipid Res. 2016; 63: 41-49

    • Pepino M.Y.
    • et al.

    The fatty acid translocase gene CD36 and lingual lipase influence oral sensitivity to fat in obese subjects.

    J. Lipid Res. 2012; 53: 561-566

    • Iida F.
    • et al.

    Effect of fat content on sensory characteristics of marbled beef from Japanese Black steers.

    Anim. Sci. J. 2015; 86: 707-715

    • Savell J.W.
    • Cross H.R.

    The role of fat in the palatability of beef, pork, and lamb.

    in: Designing Foods: Animal Product Options in the Marketplace. National Academies Press (US), 1988

    • Frank D.
    • et al.

    Effect of marbling on volatile generation, oral breakdown and in mouth flavor release of grilled beef.

    Meat Sci. 2017; 133: 61-68

    • Bermingham E.N.
    • et al.

    Assessment of atherogenic index, long-chain omega-3 fatty acid and phospholipid content of prime beef: a survey of commercially sourced New Zealand Wagyu and Angus beef cattle.

    Anim. Prod. Sci. 2021; 61: 179-190

    • Therkildsen M.
    • et al.

    Collagen, intramuscular fat and proteolysis affect Warner–Bratzler shear-force of muscles from Bos taurus breed types differently at weaning, after backgrounding on pasture, and after feedlotting.

    Anim. Prod. Sci. 2021; 61: 432-443

    • Dreher J.
    • et al.

    Formation and characterization of plant-based emulsified and crosslinked fat crystal networks to mimic animal fat tissue.

    J. Food Sci. 2020; 85: 421-431

    • Yashini M.
    • et al.

    Protein-based fat replacers – a review of recent advances.

    Food Rev. Int. 2021; 37: 197-223

    • Burnett D.D.
    • et al.

    Biology, strategies, and fresh meat consequences of manipulating the fatty acid composition of meat.

    J. Anim. Sci. 2020; 98: skaa033

    • Guan X.
    • et al.

    Bioprocessing technology of muscle stem cells: implications for cultured meat.

    Trends Biotechnol. 2022; 40: 721-734

    • Warner R.D.

    Review: analysis of the process and drivers for cellular meat production.

    Animal. 2019; 13: 3041-3058

    • Maljaars J.
    • et al.

    Effect of fat saturation on satiety, hormone release, and food intake.

    Am. J. Clin. Nutr. 2009; 89: 1019-1024

    • Samra R.A.

    Fats and satiety.

    in: Montmayeur J.P. le Coutre J. Fat Detection: Taste, Texture, and Post Ingestive Effects. Ch. 15, CRC Press/Taylor & Francis, 2010

    • Potter G.
    • et al.

    A more open approach is needed to develop cell-based fish technology: it starts with zebrafish.

    One Earth. 2020; 3: 54-64

    • Mehta F.
    • et al.

    Adipogenesis from bovine precursors.

    in: Rønning S.B. Myogenesis: Methods and Protocols. Springer, New York2019: 111-125

    • 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

    • Dohmen R.G.J.
    • et al.

    Muscle-derived fibro-adipogenic progenitor cells for production of cultured bovine adipose tissue.

    NPJ Sci. Food. 2022; 6: 6

    • Sugii S.
    • et al.

    Reassessment of adipocyte technology for cellular agriculture of alternative fat.

    Compr. Rev. Food Sci. Food Saf. 2022; ()

    • Torii S.-I.
    • et al.

    Thiazolidinedione induces the adipose differentiation of fibroblast-like cells resident within bovine skeletal muscle.

    Cell Biol. Int. 1998; 22: 421-427

    • Wosczyna M.N.
    • et al.

    Targeting microRNA-mediated gene repression limits adipogenic conversion of skeletal muscle mesenchymal stromal cells.

    Cell Stem Cell. 2021; 28: 1323-1334.e1328

    • Aguiari P.
    • et al.

    High glucose induces adipogenic differentiation of muscle-derived stem cells.

    Proc. Natl. Acad. Sci. U. S. A. 2008; 105: 1226-1231

    • Liu Y.
    • et al.

    Cloning and characterization of adipogenin and its overexpression enhances fat accumulation of bovine myosatellite cells.

    Gene. 2017; 601: 27-35

    • Li X.Z.
    • et al.

    Oleic acid in the absence of a PPARγ agonist increases adipogenic gene expression in bovine muscle satellite cells1.

    J. Anim. Sci. 2019; 97: 4114-4123

    • Gu H.
    • et al.

    Targeted overexpression of PPARγ in skeletal muscle by random insertion and CRISPR/Cas9 transgenic pig cloning enhances oxidative fiber formation and intramuscular fat deposition.

    FASEB J. 2021; 35e21308

    • Zhang L.
    • et al.

    Ectopical expression of FABP4 gene can induce bovine muscle-derived stem cells adipogenesis.

    Biochem. Biophys. Res. Commun. 2017; 482: 352-358

    • Ong W.K.
    • et al.

    Adipose tissue: understanding the heterogeneity of stem cells for regenerative medicine.

    Biomolecules. 2021; 11: 918

    • Oki Y.
    • et al.

    Effect of volatile fatty acids on adipocyte differentiation in bovine dedifferentiated fat (DFAT) cells in vitro.

    Genes Cells. 2022; 27: 5-13

    • Hassan A.
    • et al.

    Selenium promotes adipogenic determination and differentiation of chicken embryonic fibroblasts with regulation of genes involved in fatty acid uptake, triacylglycerol synthesis and lipolysis.

    J. Nutr. Biochem. 2014; 25: 858-867

    • Kim D.H.
    • et al.

    Adipogenic and myogenic potentials of chicken embryonic fibroblasts in vitro: combination of fatty acids and insulin induces adipogenesis.

    Lipids. 2020; 55: 163-171

    • Lee J.
    • et al.

    Research note: potential usage of DF-1 cell line as a new cell model for avian adipogenesis.

    Poult. Sci. 2021; 100101057

    • Liu S.
    • et al.

    Transdifferentiation of fibroblasts into adipocyte-like cells by chicken adipogenic transcription factors.

    Comp. Biochem. Physiol. A Mol. Integr. Physiol. 2010; 156: 502-508

    • Yin J.
    • et al.

    In vitro myogenic and adipogenic differentiation model of genetically engineered bovine embryonic fibroblast cell lines.

    Biotechnol. Lett. 2010; 32: 195-202

    • Navarro M.
    • et al.

    Livestock pluripotency is finally captured in vitro.

    Reprod. Fertil. Dev. 2019; 32: 11-39

    • Yuen J.S.K. Jr
    • et al.

    Perspectives on scaling production of adipose tissue for food applications.

    Biomaterials. 2022; 280121273

    • Hong N.
    • et al.

    Fish stem cell cultures.

    Int. J. Biol. Sci. 2011; 7: 392-402

    • Li H.
    • et al.

    Defining the pluripotent marker genes for identification of teleost fish cell pluripotency during reprogramming.

    Front. Genet. 2022; 13819682

    • Su Y.
    • et al.

    Induced pluripotent stem cells from farm animals.

    J. Anim. Sci. 2020; 98: skaa343

    • Ahfeldt T.
    • et al.

    Programming human pluripotent stem cells into white and brown adipocytes.

    Nat. Cell Biol. 2012; 14: 209-219

    • Mohsen-Kanson T.
    • et al.

    Differentiation of human induced pluripotent stem cells into brown and white adipocytes: role of Pax3.

    Stem Cells. 2014; 32: 1459-1467

    • Mohorčich J.
    • Reese J.

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

    Appetite. 2019; 143104408

    • O’Neill E.N.
    • et al.

    Considerations for the development of cost-effective cell culture media for cultivated meat production.

    Compr. Rev. Food Sci. Food Saf. 2021; 20: 686-709

    • Messmer T.
    • et al.

    A serum-free media formulation for cultured meat production supports bovine satellite cell differentiation in the absence of serum starvation.

    Nat. Food. 2022; 3: 74-85

    • Stout A.J.
    • et al.

    Simple and effective serum-free medium for sustained expansion of bovine satellite cells for cell cultured meat.

    Commun. Biol. 2022; 5: 466

    • Kolkmann A.M.
    • et al.

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

    Cytotechnology. 2020; 72: 111-120

    • Skubis A.
    • et al.

    Impact of antibiotics on the proliferation and differentiation of human adipose-derived mesenchymal stem cells.

    Int. J. Mol. Sci. 2017; 18: 2522

    • Dufau J.
    • et al.

    In vitro and ex vivo models of adipocytes.

    Am. J. Phys. Cell Phys. 2021; 320: C822-C841

    • Nematbakhsh S.
    • et al.

    Molecular regulation of lipogenesis, adipogenesis and fat deposition in chicken.

    Genes (Basel). 2021; 12: 414

    • Cheng B.
    • et al.

    Cocktail supplement with rosiglitazone: a novel inducer for chicken preadipocyte differentiation in vitro.

    Biosci. Rep. 2016; 36: PMC5293590

    • Salmeron C.

    Adipogenesis in fish.

    J. Exp. Biol. 2018; 221: jeb161588

    • Volz A.-C.
    • Kluger P.J.

    Completely serum-free and chemically defined adipocyte development and maintenance.

    Cytotherapy. 2018; 20: 576-588

    • Sandhu M.A.
    • et al.

    Influence of bovine serum lipids and fetal bovine serum on the expression of cell surface markers in cultured bovine preadipocytes.

    Cells Tissues Organs. 2017; 204: 13-24

    • Jurek S.
    • et al.

    Optimizing adipogenic transdifferentiation of bovine mesenchymal stem cells: a prominent role of ascorbic acid in FABP4 induction.

    Adipocyte. 2020; 9: 35-50

    • Boone C.
    • et al.

    Culture of porcine stromal-vascular cells in serum-free medium: differential action of various hormonal agents on adipose conversion.

    J. Anim. Sci. 2000; 78: 885-895

    • Hausman G.J.
    • Richardson R.L.

    Newly recruited and pre-existing preadipocytes in cultures of porcine stromal-vascular cells: morphology, expression of extracellular matrix components, and lipid accretion.

    J. Anim. Sci. 1998; 76: 48-60

    • Matsubara Y.
    • et al.

    Fatty acids but not dexamethasone are essential inducers for chick adipocyte differentiation in vitro.

    Comp. Biochem. Physiol. A Mol. Integr. Physiol. 2008; 151: 511-518

    • Kinkel A.D.
    • et al.

    Oil red-O stains non-adipogenic cells: a precautionary note.

    Cytotechnology. 2004; 46: 49-56

    • Yuan C.
    • et al.

    Fast adipogenesis tracking system (FATS) – a robust, high-throughput, automation-ready adipogenesis quantification technique.

    Stem Cell Res. Ther. 2019; 10: 38

    • Odegard I.
    • et al.

    TEA of cultivated meat. Future projections for different scenarios.

    • Risner D.
    • et al.

    Preliminary techno-economic assessment of animal cell-based meat.

    Foods. 2021; 10: 3

    • Tripathi N.K.
    • Shrivastava A.

    Recent developments in bioprocessing of recombinant proteins: expression hosts and process development.

    Front. Bioeng. Biotechnol. 2019; 7: 420

    • Ng J.Y.
    • et al.

    Chlorella vulgaris extract as a serum replacement that enhances mammalian cell growth and protein expression.

    Front. Bioeng. Biotechnol. 2020; 8564667

    • Ho Y.Y.
    • et al.

    Applications and analysis of hydrolysates in animal cell culture.

    Bioresour. Bioprocess. 2021; 8: 93

    • Okamoto Y.
    • et al.

    Proliferation and differentiation of primary bovine myoblasts using Chlorella vulgaris extract for sustainable production of cultured meat.

    Biotechnol. Prog. 2022; 38e3239

    • Casado-Diaz A.
    • et al.

    Influence of olive oil and its components on mesenchymal stem cell biology.

    World J. Stem Cells. 2019; 11: 1045-1064

    • Riera-Heredia N.
    • et al.

    Fatty acids from fish or vegetable oils promote the adipogenic fate of mesenchymal stem cells derived from gilthead sea bream bone potentially through different pathways.

    PLoS ONE. 2019; 14e0215926

    • 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: 44

    • Bellani C.F.
    • et al.

    Scale-up technologies for the manufacture of adherent cells.

    Front. Nutr. 2020; 7575146

    • Zhang G.
    • et al.

    Challenges and possibilities for bio-manufacturing cultured meat.

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

    • Hanga M.P.
    • et al.

    Bioprocess development for scalable production of cultivated meat.

    Biotechnol. Bioeng. 2020; 117: 3029-3039

    • Bodiou V.
    • et al.

    Microcarriers for upscaling cultured meat production.

    Front. Nutr. 2020; 7: 10

    • Muoio F.
    • et al.

    Human adipose stem cells (hASCs) grown on biodegradable microcarriers in serum- and xeno-free medium preserve their undifferentiated status.

    J. Funct. Biomater. 2021; 12: 25

    • Fish K.D.
    • et al.

    Prospects and challenges for cell-cultured fat as a novel food ingredient.

    Trends Food Sci. Technol. 2020; 98: 53-67

    • Ng S.
    • Kurisawa M.

    Integrating biomaterials and food biopolymers for cultured meat production.

    Acta Biomater. 2021; 124: 108-129

    • Murphy C.S.
    • et al.

    In vitro tissue-engineered adipose constructs for modeling disease.

    BMC Biomed. Eng. 2019; 1: 27

    • Volz A.-C.
    • et al.

    A cellulose-based material for vascularized adipose tissue engineering.

    J. Biomed. Mater. Res. B Appl. Biomater. 2019; 107: 1431-1439

    • Louis F.
    • et al.

    Bioprinted vascularized mature adipose tissue with collagen microfibers for soft tissue regeneration.

    Cyborg Bionic Syst. 2021; 2021: 1412542

    • Yang F.
    • et al.

    Optimization of co-culture conditions for a human vascularized adipose tissue model.

    Bioengineering. 2020; 7: 114

    • Klingelhutz A.J.
    • et al.

    Scaffold-free generation of uniform adipose spheroids for metabolism research and drug discovery.

    Sci. Rep. 2018; 8: 523

    • Ma Y.N.
    • et al.

    Three-dimensional spheroid culture of adipose stromal vascular cells for studying adipogenesis in beef cattle.

    Animal. 2018; 12: 2123-2129

    • Muller S.
    • et al.

    Human adipose stromal-vascular fraction self-organizes to form vascularized adipose tissue in 3D cultures.

    Sci. Rep. 2019; 9: 7250

    • Ianovici I.
    • et al.

    3D-printable plant protein-enriched scaffolds for cultivated meat development.

    Biomaterials. 2022; 284121487

    • Co E.D.
    • Marangoni A.G.

    Organogels: an alternative edible oil-structuring method.

    J. Am. Oil Chem. Soc. 2012; 89: 749-780

    • Wood J.D.
    • et al.

    Fat deposition, fatty acid composition and meat quality: a review.

    Meat Sci. 2008; 78: 343-358

    • Gesta S.
    • et al.

    Developmental origin of fat: tracking obesity to its source.

    Cell. 2007; 131: 242-256

    • Berry D.C.
    • et al.

    The developmental origins of adipose tissue.

    Development. 2013; 140: 3939-3949

    • Guo Y.
    • et al.

    Lipid droplets at a glance.

    J. Cell Sci. 2009; 122: 749-752

    • Sugii S.
    • Velan S.S.

    Physical and physiological properties of fat.

    in: Seiberlich N. Advances in Magnetic Resonance Technology and Applications (Chapter 25). Academic Press, 2020: 667-679

    • Ghaben A.L.
    • Scherer P.E.

    Adipogenesis and metabolic health.

    Nat. Rev. Mol. Cell Biol. 2019; 20: 242-258

  • Time Stamp:

    More from Biotechnology Trends