Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
CULTURED MEAT COMPOSITIONS
Document Type and Number:
WIPO Patent Application WO/2019/016795
Kind Code:
A1
Abstract:
The invention is directed to a method for producing an edible composition, comprising incubating a three-dimensional porous scaffold and a plurality of cell types comprising: myoblasts or progenitor cells thereof, at least one type of extracellular (ECM)-secreting cell and endothelial cells or progenitor cells thereof, and inducing myoblasts differentiation into myotubes.

Inventors:
BEN-ARYE TOM (IL)
LEVENBERG SHULAMIT (IL)
Application Number:
PCT/IL2018/050776
Publication Date:
January 24, 2019
Filing Date:
July 15, 2018
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
TECHNION RES & DEV FOUNDATION (IL)
International Classes:
C12N5/07; A23L13/00; A61L27/56
Foreign References:
US2682466A1954-06-29
US3142571A1964-07-28
US3488770A1970-01-06
US3498794A1970-03-03
US3759715A1973-09-18
US3778522A1973-12-11
US3794731A1974-02-26
US3814823A1974-06-04
US24858172A1972-04-28
US3840679A1974-10-08
US4666828A1987-05-19
US4683202A1987-07-28
US4801531A1989-01-31
US5192659A1993-03-09
US5272057A1993-12-21
Other References:
MATHENY, JG.: "In Vitro-Cultured Meat Production", TISSUE ENGINEERING, vol. 11, no. 5/6, 3 May 2005 (2005-05-03), pages 659 - 662, XP009519007, DOI: 10.1089/ten.2005.11.659
LEVENBERG, SHULAMIT ET AL.: "Engineering vascularized skeletal muscle tissue", NATURE BIOTECHNOLOGY, vol. 23.7, 19 June 2005 (2005-06-19), pages 879 - 884, XP002677366
BHAT, ZUHAIB FAYAZ ET AL.: "Prospectus of cultured meat-advancing meat alternatives", JOURNAL OF FOOD SCIENCE AND TECHNOLOGY, vol. 48.2, 30 December 2010 (2010-12-30), pages 125 - 140, XP055571196
KOFFLER, JACOB ET AL.: "Improved vascular organization enhances functional integration of engineered skeletal muscle grafts", PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES, vol. 108, no. 36, 30 August 2011 (2011-08-30), pages 14789 - 14794, XP055571232
XU, HELAN ET AL.: "Intrinsically water-stable electrospun three-dimensional ultrafine fibrous soy protein scaffolds for soft tissue engineering using adipose derived mesenchymal stem cells", RSC ADVANCES 4.30 (2014, vol. 4, no. 30, 8 March 2014 (2014-03-08), pages 15451 - 15457, XP055571239
SAMBROOK ET AL.: "Current Protocols in Molecular Biology", 1989, JOHN WILEY AND SONS
FRESHNEY: "Culture of Animal Cells - A Manual of Basic Technique", vol. I-III, 1994, APPLETON & LANGE
PERBAL: "A Practical Guide to Molecular Cloning", 1988, JOHN WILEY & SONS
WATSON ET AL.: "Recombinant DNA", SCIENTIFIC AMERICAN
"Genome Analysis: A Laboratory Manual Series", vol. 1-4, 1998, COLD SPRING HARBOR LABORATORY PRESS
"Strategies for Protein Purification and Characterization - A Laboratory Course Manual", 1996, CSHL PRESS
"Bacteriophage Methods and Protocols", vol. 1, article "Isolation, Characterization, and Interactions"
See also references of EP 3728560A4
Attorney, Agent or Firm:
KESTEN, Dov et al. (IL)
Download PDF:
Claims:
CLAIMS

1. A method for producing an edible composition, the method comprising the steps of: a. incubating a three-dimensional porous scaffold and a plurality of cell types comprising: (i) myoblasts or progenitor cells thereof; and at least one of: (ii) at least one type of extracellular matrix (ECM)-secreting cell; or (iii) endothelial cells or progenitor cells thereof, wherein said myoblasts or progenitor cells thereof and said endothelial cells or progenitor cells thereof are incubated at a ratio ranging between 10: 1 to 1:10; and b. inducing differentiation of myoblasts or progenitor cells thereof into myotubes, thereby producing the edible composition.

2. The method of claim 1, wherein said plurality of cell types comprises myoblasts or progenitor cells thereof, at least one type of extracellular matrix (ECM) -secreting cell, and endothelial cells or progenitor cells thereof.

3. The method of any one of claims 1 or 2, wherein said ECM-secreting cell is selected from the group consisting of: stromal cells, fibroblasts, pericytes, smooth muscle cells and progenitor cells thereof.

4. The method of any one of claims 1 to 3, wherein said plurality of cell types comprises myoblasts, ECM-secreting cells and endothelial cells.

5. The method of any one of claims 1 to 4, wherein said a progenitor cell of a myoblast is a satellite cell.

6. The method of any one of claims 1 to 5, wherein said endothelial cells are selected from skeletal microvascular endothelial cells, aortic smooth muscle cells, or a combination thereof.

7. The method of any one of claims 1 to 6, wherein said plurality of cell types comprises satellite cells, ECM-secreting cells, and endothelial cells.

8. The method of claim 7, wherein said myoblasts or progenitor cells thereof and ECM- secreting cells are incubated at a ratio ranging between 10: 1 to 1: 1.

9. The method of claim 7, wherein said ECM-secreting cells and endothelial cells are incubated at a ratio ranging between 1 : 10 to 1: 1.

10. The method of any one of claims 7 to 9, wherein said satellite cells, said ECM-secreting cells, and said endothelial cells are incubated at a ratio ranging between 10: 1: 1 to 2: 1: 10.

11. The method of any one of claims 7 to 10, wherein said ECM- secreting cell is a fibroblast, a progenitor cell thereof, or a combination thereof.

12. The method of claim 1, wherein said three-dimensional porous scaffold is selected from the group consisting of: a textured protein, a non-textured protein, and a polysaccharide.

13. The method of claim 12, wherein said textured protein is textured soy protein.

14. The method of any one of claims 12 or 13, wherein said three dimensional porous scaffold comprises pores with an average diameter ranging from 20 to 1,000 micrometers.

15. The method of any one of claims 1 to 14, wherein said plurality of cell types are non- human cells.

16. The method of any one of claims 1 to 15, wherein said plurality of cells are derived from a livestock mammal.

17. The method of any one of claims 1 to 16, wherein said myoblasts or progenitor cells thereof and said three-dimensional porous scaffold are incubated at a ratio ranging from 103 to 107 of said myoblasts or progenitor cells thereof to 10 mg of said three- dimensional porous scaffold.

18. The method of any one of claims 1 to 17, wherein said three-dimensional porous scaffold further comprises an extracellular matrix.

19. A composition comprising: a. a three-dimensional porous scaffold; b. myotubes comprising 10,000-250,000 myotube nuclei per mm3 of said three- dimensional porous scaffold; and c. a plurality of cell types selected from the group consisting of: (i) myoblasts or progenitor cells thereof; and at least one of: (ii) at least one type of ECM- secreting cell; or (iii) endothelial cells or progenitor cells thereof, wherein said endothelial cells or progenitor cells thereof comprise less than 15% of said plurality of cells.

20. The composition of claim 19, wherein said plurality of cell types comprises myoblasts or progenitor cells thereof, at least one type of extracellular matrix (ECM)-secreting cells, and endothelial cells or progenitor cells thereof.

21. The composition of claim 19 or 20, wherein said a progenitor cell of a myoblast is a satellite cell.

22. The composition of any one of claims 19 to 21, wherein said plurality of cell types comprises satellite cells, ECM-secreting cells, and endothelial cells.

23. The composition of any one of claims 19 to 22, wherein said ECM-secreting cell is a fibroblast, a progenitor cell thereof, or a combination thereof.

24. The composition of any one of claims 19 to 23, wherein said three-dimensional porous scaffold is selected from the group consisting of: a textured protein, a non-textured protein, and a polysaccharide.

25. The composition of claim 24, wherein said textured protein is textured soy protein.

26. The composition of any one of claims 19 or 25, wherein said three dimensional porous scaffold comprises pores with an average diameter ranging from 20 to 1,000 micrometers.

27. The composition of any one of claims 19 to 26, wherein said plurality of cell types are non-human cells.

28. The composition of any one of claims 19 to 27, wherein said plurality of cells are derived from a livestock mammal.

29. The composition of any one of claims 19 to 28, wherein said three-dimensional porous scaffold further comprises an extracellular matrix.

30. The composition of any one of claims 19 to 29, wherein said composition is edible.

Description:
CULTURED MEAT COMPOSITIONS

CROSS REFERENCE

[001] This application claims the benefit of priority to U.S. Provisional Patent Application No. 62/532,998 filed July 15, 2017, the contents of which are incorporated herein by reference in their entirety.

FIELD OF INVENTION

[002] The present invention is directed to, inter alia, compositions of cultured meat and methods of producing the same.

BACKGROUND OF THE INVENTION

[003] Cultured meat, also known as engineered or clean meat, is produced from cell culture using tissue engineering techniques and is a prominent alternative for traditional meat production using live animals. In the last decade this notion gained increased attention in public opinion, popular media, investors and the scientific community, notably after the production of the first lab-grown cultured beef burger. Food produced using animals is considered to be inefficient, as the animals consume large amounts of food throughout their lives, which 80-90% of the calories are wasted on the animal's metabolism and the production of non-edible tissues. When comparing different industries, beef has the heaviest environmental impact, however in general, all animal-based products have a larger environmental footprint compared to plant-based products in terms of soil and water demand, and greenhouse gas (GHG) emission. According to the report of the Food and Agriculture Organization of the United Nations, the livestock sector is responsible for 18% of the GHG emissions, uses 30% of earth's terrain or 70% of the arable lands, and 8% of the global freshwater. In addition, the world's demand for meat is expected to double by 2050, meaning traditional meat production systems are not sustainable. Compared to several meat sources, cultured meat is estimated to decrease 7-45% of energy use, 78-96% of the GHG emissions, 99% of land use and 82-96% of water use.

[004] Intensive factory farming and poor animal welfare conditions are a cause for foodborne illnesses such as swine and avian influenza, and the spreading of E.coli, Salmonella and Campylobacter which can be found in meat. Producing meat in a sterile controlled environment could help improve food safety. In addition, 70% of all antibiotics used in the United States are given to farm animals as a food additive which promote the selection of antimicrobial resistant strains and increase the likelihood of multidrug resistant bacteria. Antibiotic overuse is the primary cause for the emergence of antibiotic resistant bacteria, which in the U.S alone is responsible for an economic burden of $55b, 2 million infections, 250,000 hospitalizations and at least 23,000 deaths per year. Bacteria with resistance to colistin, the last resort of antibiotics, has recently emerged in Chinese pig farms.

[005] Current cultured meat technology focuses on satellite cell culture. Cells are being grown in two-dimensional (2D) flasks or on micro carriers in suspension, isolated, differentiated and harvested. However, tissues do not consist solely of cells; the extracellular matrix (ECM), which is composed of macromolecules such as glycoproteins and oligosaccharides and gives the tissue its biochemical and biomechanical properties, is a large portion of the tissue. The ECM regulates cells behavior and affects their composition. Thus, ECM producing cells are essential for cultured meat, and processes such as cell harvest must be avoided. Additionally, three- dimensional (3D) cell culture mimics the natural cell environment and is crucial for correct cell behavior which affects cell biochemical content.

SUMMARY OF THE INVENTION

[006] In some embodiments, the present invention is directed to edible compositions comprising myotubes, and method for producing the edible compositions.

[007] The invention is based in part on the findings that a cell culture comprising a plurality of cell types (e.g., satellite cells, ECM-secreting cells and endothelial cells), showed better survival, proliferation and myotube formation, compared to controls, such as when grown on three- dimensional porous scaffold.

[008] The invention is further based in part on the surprising findings that satellite cells, such as non-human satellite cells, had better differentiation activity when co-cultured with low endothelial cells (ECs) concentration rather than with high ECs concentration.

[009] According to one aspect, there is provided a method for producing an edible composition, the method comprising the steps of: (a) incubating a three-dimensional porous scaffold and a plurality of cell types comprising: (i) myoblasts or progenitor cells thereof; and at least one of: (ii) at least one type of extracellular matrix (ECM)-secreting cell; or (iii) endothelial cells or progenitor cells thereof, wherein the myoblasts or progenitor cells thereof and the endothelial cells or progenitor cells thereof are incubated at a ratio ranging between 10: 1 to 1: 10; and (b) inducing differentiation of myoblasts or progenitor cells thereof into myotubes, thereby producing the edible composition.

[010] In some embodiments, the plurality of cell types comprises myoblasts or progenitor cells thereof, at least one type of extracellular matrix (ECM)-secreting cell, and endothelial cells or progenitor cells thereof. [Oi l] In some embodiments, the ECM-secreting cell is selected from the group consisting of: stromal cells, fibroblasts, pericytes, smooth muscle cells and progenitor cells thereof.

[012] In some embodiments, the plurality of cell types comprises myoblasts, ECM-secreting cells and endothelial cells.

[013] In some embodiments, the progenitor cell of a myoblast is a satellite cell.

[014] In some embodiments, the endothelial cells are selected from skeletal microvascular endothelial cells, aortic smooth muscle cells, or a combination thereof.

[015] In some embodiments, the plurality of cell types comprises satellite cells, ECM-secreting cells, and endothelial cells.

[016] In some embodiments, the myoblasts or progenitor cells thereof and ECM-secreting cells are incubated at a ratio ranging between 10:1 to 1: 1.

[017] In some embodiments, the ECM-secreting cells and endothelial cells are incubated at a ratio ranging between 1 : 10 to 1: 1.

[018] In some embodiments, the satellite cells, the ECM-secreting cells, and the endothelial cells are incubated at a ratio ranging between 10: 1 : 1 to 2: 1 : 10.

[019] In some embodiments, the ECM-secreting cell is a fibroblast, a progenitor cell thereof, or a combination thereof.

[020] In some embodiments, the three-dimensional porous scaffold is selected from the group consisting of: a textured protein, a non-textured protein, and a polysaccharide. In some embodiments, the textured protein is a textured soy protein. In some embodiments, the three dimensional porous scaffold comprises pores with an average diameter ranging from 20 to 1,000 micrometers.

[021] In some embodiments, the plurality of cell types are non-human cells. In some embodiments, the plurality of cells are derived from a livestock mammal.

[022] In some embodiments, the myoblasts or progenitor cells thereof and the three- dimensional porous scaffold are incubated at a ratio ranging from 10 3 to 10 7 of the myoblasts or progenitor cells thereof to 10 mg of the three-dimensional porous scaffold. In some embodiments, the three-dimensional porous scaffold further comprises an extracellular matrix.

[023] According to another aspect, there is provided a composition comprising: (a) a three- dimensional porous scaffold; (b) myotubes comprising 100,000-250,000 myotube nuclei per mm 3 of the three-dimensional porous scaffold; and (c) a plurality of cell types selected from the group consisting of: (i) myoblasts or progenitor cells thereof; and at least one of: (ii) at least one type of ECM-secreting cells; or (iii) endothelial cells or progenitor cells thereof, wherein the endothelial cells or progenitor cells thereof comprise less than 15% of the plurality of cells.

[024] In some embodiments, the composition is edible.

[025] Unless otherwise defined, all technical and/or scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which the invention pertains. Although methods and materials similar or equivalent to those described herein can be used in the practice or testing of embodiments of the invention, exemplary methods and/or materials are described below. In case of conflict, the patent specification, including definitions, will control. In addition, the materials, methods, and examples are illustrative only and are not intended to be necessarily limiting.

[026] Further embodiments and the full scope of applicability of the present invention will become apparent from the detailed description given hereinafter. However, it should be understood that the detailed description and specific examples, while indicating preferred embodiments of the invention, are given by way of illustration only, since various changes and modifications within the spirit and scope of the invention will become apparent to those skilled in the art from this detailed description.

BRIEF DESCRIPTION OF THE DRAWINGS

[027] Figs. 1A-1C are photographs showing commercial TSPs. (A) shows large, medium and small chunks products of TSP. (B) and (C) show 2 other commercial TSP flake products, annotated as TVP (left) and Arcon (right).

[028] Figs. 2A-2B are photographs showing TSP scaffold preparation (A), and cell containing TSP scaffolds (B).

[029] Figs. 3A-3B are lOOx magnification SEM images of big TSP (A) and medium TSP (B).

[030] Figs. 4A-4B are 2 x 10 5 magnification SEM images of big TSP (A) and medium TSP (B).

[031] Figs. 5A-5C are confocal microscopy images of TSP scaffolds populated with fibroblasts (red) and endothelial (green) cells on days 8 (A), 18 (B), and 21 (C) following seeding of fibroblasts.

[032] Fig. 6 is confocal microscopy images taken 14 days following seeding of 200,000 fibroblast (red) onto three different samples of scaffolds (samples 1-3) obtained from different sources denoted as Large, Medium, Small, and Medium from 90°, scaffolds denoted as Medium from 90° were cut from the pore side of the TSP in the scaffold preparation procedure TSP scaffolds. Scale bar = 1 mm.

[033] Fig. 7 is a confocal microscopy image of myoblast cultured on a TSP scaffold for 14 days (Blue - DAPI, Green - Desmin).

[034] Fig. 8 is confocal microscopy images of bovine skeletal muscle cells cultured on a TSP scaffold for 14 days (Blue - DAPI, Green - Phalloidin).

[035] Figs. 9A-9E are micrographs of bovine aorta smooth muscle cells (BAOSMC) (passage 8) seeded in different media. (A) Basal medium; (B) Commercial medium; (C) Fetal bovine serum (15%) supplementation; (D) non-essential amino acids supplementation; and (E) Pyruvate supplementation. Scale bar = 100 μιη.

[036] Figs. 10A-10B are vertical bar graphs of the comparison of different media on bovine aorta smooth muscle cells proliferation at passage 9 (A) and passage 10 (B) after 3 days of culturing.

[037] Figs. 11A-11B are graph demonstrating growth kinetics of Bovine dermal fibroblasts (BDF) (A) and Bovine aorta smooth muscle cells (BAOSMC; B).

[038] Figs. 12A-12L are images of fluorescently labeled bovine aortic endothelial cells (BAEC) cells seeded in co-culture with supporting cells on day 2 (A, D, G and J), 6 (B, E, H and K), and 8 (C, F, I and L). BAEC cells (green) were seeded with either bovine aortic smooth muscle cells (SMC; A-F) or bovine dermal fibroblasts (BDF; G-L), both which are in red. A-C and G-I are tile scans of 3 x3 in x5 magnification. D-F and J-L are x20 magnifications. Scale bar = 100 μ m .

[039] Figs. 13A-13L are images of fluorescently labeled bovine skeletal muscle microvascular endothelial cells (BSkMVEC) cells seeded in co-culture with supporting cells on day 2 (A, D, G and J), 6 (B, E, H and K), and 8 (C, F, I and L). BSkMVEC cells (green) were seeded with either bovine aortic smooth muscle cells (SMC; A-F) or bovine dermal fibroblasts (BDF; G-L), both which are in red. A-C and G-I are tile scans of 3 x3 in x5 magnification. D-F and J-L are x20 magnifications. Scale bar = 100 μιη.

[040] Figs. 14A-14F are images of fluorescently labeled cell co-cultures comprising BAEC and SMC in different ratios. (A and D) 5:1 BAEC to SMC; (B and E) 1: 1 BAEC to SMC. (C and F) are higher magnification of B and E, respectively. A-C are image taken on day 2 and D-F were taken on day 6. BAEC is in red; Scale bar = 100 μιη. [041] Figs. 15A-15E are images of fluorescently labeled cell co-cultures comprising BAEC and BDF in different ratios. (A and C) 5: 1 BAEC to SMC; (B and D) 1: 1 BAEC to SMC. (E) is a higher magnification of D. A and B are image taken on day 2 and C and D were taken on day 6. BAEC is in red; Scale bar = 100 μιη.

[042] Figs. 16A-16L are images of fluorescently labeled cell co-cultures comprising BAEC and SMC in different ratios. (A-D) 1:3 BAEC to SMC; (E-H) 1:1 BAEC to SMC; and (I-L and

F) 5:1 BAEC to SMC. (B, D, F, H, J, and L) are higher magnification of (A, C, E, G, I, and K), respectively. (A, B, E, F, I and J) are image taken on day 2 and (C, D, G, H, K and L) were taken on day 6. BAEC is in red; Scale bar = 100 μιη.

[043] Figs. 17A-17G demonstrate differentiation of bovine satellite cells (BSC) and formation of myotubes. (A-C) are images of BSC cells before differentiation; (D-F) are images of BSC cells after differentiation. (G) is a vertical bar graph showing the fusion index (FI) of the samples. (A and D) light microscopy; (B and E) Hoechst; and (C and F) Myogenin. The difference in the automatic FI before and after differentiation was found to be statistically significant (P-value =0.00003), while the difference between the automatic and manual FI was not.

[044] Figs. 18A-18G are bright field micrographs of BSC at day 0 of differentiation. (A) Control; (B) bovine fibroblast growth factor (bFGF); (C) epidermal growth factor (EGF); (D) IGF-1 (insulin-like growth factor 1); (E) Pro-LIF (Proliferation media without the growth factor LIF (Leukemia inhibitory factor)); (F) Weaning; and (G) Dil. Scale bar= 300 μιη.

[045] Figs. 19A-19G are bright field micrographs of BSC at day 4 of differentiation. (A) Control; (B) bovine fibroblast growth factor (bFGF); (C) epidermal growth factor (EGF); (D) IGF-1 (insulin-like growth factor 1); (E) Pro-LIF; (F) Weaning; and (G) Dil. Scale bar= 300 μιη.

[046] Figs. 20A-20H demonstrate myotube formation of BSC at 7 days of differentiation. (A-

G) are bright field micrographs of (A) Control; (B) bovine fibroblast growth factor (bFGF); (C) epidermal growth factor (EGF); (D) IGF-1 (insulin-like growth factor 1); (E) Pro-LIF ; (F) Weaning; and (G) Dil. Scale bar= 300 μιη. (H) is a vertical bar graph of % myotube area quantified for each of the different expansion conditions.

[047] Fig. 21 is a vertical bar graph demonstrating quantification values of cell coverage on porous scaffolds at day 7 with growth media or Pro-LIF media.

[048] Figs. 22A-22L are confocal microscopy images of fluorescently labeled BSC differentiated on a porous scaffold. Expansion phase included: (A-D) Cells cultured on growth media; (E-H) Cells cultured on Pro-LIF media; (I-L) Cells cultured on Pro-LIF-bFGF media. Differentiation phase included: (A, E and I) No growth factors (GFs) were added to the differentiation media; (B, F and J) IGF-1 was added to the differentiation media; (C, G and K) EGF was added to the differentiation media; and (D, H and L) IGF-1 and EGF were added to the differentiation media. Dil (red); Desmin (green); Scale bar = 100 μιη.

[049] Figs. 23A-230 are immunofluorescent micrographs of tri-cultures grown on PLLA/PLGA scaffolds taken 14 days post seeding. BSC, SMC and BAEC were seeded on PLLA/PLGA scaffolds in different cellular densities of (A-E) 2: 1: 1 and (F-J) 2: 1:5. (K-O) are magnifications of (A-E). Scaffolds were stained for (A, F and K) DAPI; (B, G and L) Myogenin; (C, H and M) Dil; (D, I and N) CD31. (E, J and O) are merged image. Scale bars = 100 μιη (A- J); 10 μιη (K-O).

[050] Figs. 24A-24F are immunofluorescent micrographs of PLLA/PLGA scaffolds 14 days post seeding. BSC, SMC and BEC were seeded on PLLA/PLGA scaffolds in different cellular densities of (A-C) 2: 1: 1 and (D-F) 2:1:5. (A and D) BSC and SMC (2: 1); (B and E) BSC and BEC (2: 1); and (C and F) monoculture of BSC alone. Scaffolds were stained for DAPI (blue); Myogenin (gray); Dil (red); and CD31 (green). Scale Bar of = 100 μιη.

[051] Figs. 25A-25H are immunofluorescent micrographs of BSC co-cultured with SMC on textured soy protein scaffolds, 14 days post seeding. Different cellular densities of (A-D) 2: 1: 1 and (E-H) 2:1:5, were tested. Scaffolds were stained for (A and E) DAPI; (B and F) Myogenin; (C and G) Dil; (D) and (H) are merged images of (A-C) and (E-G), respectively. Scale Bar of = 100 μηι.

[052] Figs. 26A-26E are immunofluorescent micrographs of BSC, SMC and BEC tri-cultured on textured soy protein scaffolds, 14 days post seeding. Scaffolds were stained for (A) DAPI; (B) Myogenin; (C) Dil; and (D) CD31. (E) is a merged image of (A-D).

[053] Figs. 27A-27B are micrographs of tri-chrome staining of tri-culture of BSC, SMC and BEC seeded on PLLA/PLGA scaffold at a ratio of 2: 1: 1, 14 days post seeding. (A) Tile scan of x5 magnification and (B) a xlO magnification. Scaffolds were stained to tri-chrome. Positive extracellular matrix (ECM) stains are encircled. Scale bar = 100 μιη.

[054] Figs. 28A-28C are vertical bar graphs showing the effect of the supporting cells on myogenic differentiation and the importance of the different cells' ratio within the culture. Myogenic differentiation is shown for tri-cultures (BSC:SMC:BEC) and controls (co-culture or monoculture) at cellular densities of (A) 2: 1: 1 and (B) 2:1:5 ON PLLA/PLGA scaffold. (C) 2:1: 1 on textured soy protein with a statistical analysis. [055] Figs. 29A-29N describe differentiation of tri-cultures on textured protein scaffolds. (A-L) are fluorescent images of immunostained textured soy protein scaffolds, 14 days post seeding. Cells were seeded on textured soy protein scaffolds (A, E and I) BSC, (B, F and J) BSC + BEC; (C, G and K) BSC + SMC; and (D, H and L) tri-culture of BSC + SMC + BEC. (A-D) x20 magnification, (E-H) tile scan of x20 and (I-L) myogenin staining, x63 magnification. Scaffolds were stained to DAPI (blue), Myogenin (grey), Dil (red) and CD31 (green). Scale bar = 100 μηι. (M) is a vertical bar graph of myogenin expression quantification on the TSP scaffolds in mono-culture (BSC only), co-cultures (BSC + BEC; or BSC + SMC) and tri-culture (BSC+ SMC + BEC). (N) is a vertical bar graph of BSC coverage quantification on the TSP scaffolds in a mono-culture and a co-culture with SMC.

[056] Fig. 30 is fluorescent images of immunostained BSC seeded without gel on Gelfoam scaffolds, 14 days post seeding. Scaffolds were stained to DAPI (blue), Myogenin (grey), Dil (red); and CD31 (green). Scale bar = 50 μιη.

DETAILED DESCRIPTION OF THE INVENTION

[057] The present invention is directed to compositions, kits and methods for producing cultured meat for food consumption. The present invention, in some embodiments, is directed to an edible composition comprising a porous textured protein and a three-dimensional multi-type cellular tissue comprising myoblasts and one or more cell types, attached thereto. The invention is further directed to methods of producing the composition by in-vitro culturing myoblasts and one or more cell types with a three-dimensional porous scaffold (e.g., a porous textured protein) under specific conditions.

[058] In some embodiments, the composition of the invention may be intended for consumption by human beings, non-human animals, or both. In some embodiments, the cultured meat products are food products for human consumption. In other embodiments, the cultured meat products are used for animal feed such as feed for livestock, feed for aquaculture, or feed for domestic pets.

[059] In some embodiments, the invention is directed to a method for producing an edible composition, the method comprising the steps of: a. incubating a three-dimensional porous scaffold and a plurality of cell types comprising myoblasts and at least one ECM-secreting cell type selected from the group consisting of: adipocytes, fibroblasts, and progenitor cells thereof; and an endothelial cell or progenitor cell thereof b. allowing the plurality of cell types to expand on the three-dimensional porous scaffold; and c. inducing myoblasts differentiation into myotubes; thereby allowing cells to form an edible composition comprising a three-dimensional multi-type cellular tissue comprising muscle cells and the porous textured protein.

[060] As used herein, the term "myotube" refers to a multi -nucleated fiber that is formed from the fusion of a plurality of myoblasts and/or myocytes. As used herein, the term "muscle cell" refers to any cell that contributes to muscle tissue and may encompass: myoblasts, satellite cells (SC), myotubes, myofibers, and myofibril tissues.

[061] In some embodiments, the method includes culturing myoblasts in vitro or ex vivo and allowing these cells to differentiate into specific types of muscle cells such as skeletal muscle cells or smooth muscle cells.

[062] In some embodiments, the invention is directed to an edible composition comprising: a porous textured protein; and a three-dimensional multi-type cellular tissue comprising muscle cells, wherein the three-dimensional multi-type cellular tissue is attached to the porous textured protein, and wherein the three-dimensional multi-type cellular tissue is derived from in-vitro culturing a plurality of cell types comprising: myoblasts and one or more cell types selected from: adipocytes, fibroblasts, smooth muscle cells, endothelial cells and progenitor cells thereof with the porous textured protein.

[063] As exemplified hereinbelow, the composition may comprise fibroblasts which secrete extracellular molecules such as to form the extracellular matrix (ECM) providing further structural and mechanical support to the cells.

[064] As further exemplified hereinbelow, the composition may comprise endothelial cells (EC) or endothelial progenitor cells (EPC) for providing tissue support, providing signaling and/or forming capillary endothelium.

[065] In some embodiments, the three-dimensional multi-type cellular tissue comprises muscle cells including skeletal muscle cells, smooth muscle cells and satellite cells. In some embodiments, the three-dimensional multi-type cellular tissue comprises fat cells (e.g., adipocytes). In some embodiments, the three-dimensional multi-type cellular tissue comprises an extra cellular matrix secreted by specialized cells (e.g., fibroblasts). In some embodiments, the three-dimensional multi-type cellular tissue comprises endothelial cells or capillary endothelium formed by endothelial cells, including, but not limited to aortic endothelial cells and skeletal microvascular endothelial cells. In some embodiments, the three-dimensional multi-type cellular tissue further comprises an extra cellular matrix. In some embodiments, the three-dimensional multi-type cellular tissue further comprises adipocytes. In some embodiments, the three- dimensional multi-type cellular tissue further comprises capillaries.

[066] In some embodiments, the invention is directed to a composition suitable for cell growth comprising a porous textured protein and a cell culture medium. In another embodiment, the composition suitable for cell growth further comprises growth factor, cytokines, bioactive agents, nutrients, amino acids, antibiotic compounds, anti-inflammatory compounds, or any combination thereof. Suitable medium and compounds suitable for viability and growth of the cells are known to one skilled in the art.

[067] In some embodiments, the invention is directed to a composition comprising: a plurality of cell types comprising: myoblasts and one or more cell types selected from: adipocytes, fibroblasts endothelial cells, smooth muscle cells, and progenitor cells thereof, attached to a porous textured protein.

[068] In some embodiments, the invention provides a kit comprising: a three-dimensional porous scaffold (e.g., a porous textured protein); and a plurality of cell types comprising: myoblasts and one or more cell types selected from: adipocytes, fibroblasts, smooth muscle cells, endothelial cells, and progenitor cells thereof. In some embodiments, the kit is for producing an edible composition. In some embodiments, the kit further comprises at least one component selected from: a cell culture medium, growth factor, differentiation medium, differentiation inducers. In some embodiments, the kit further comprises a cell culture medium. In another embodiment, the cell culture medium is selected from a dry powder media, a granulated preparation, an aqueous liquid or a media concentrate. In some embodiments, the plurality of cell types are frozen. In some embodiments, the kit further comprises instructions for use.

Plurality of cell types

[069] As will be appreciated by one skilled in the art, numerous cell types or population of cells may be cultured with the three-dimensional porous scaffold, so as to form a three-dimensional multi-type cellular tissue architecture. In some embodiments, the one or more cell types are selected from: myoblasts, ECM- secreting cells, endothelial cells.

[070] In some embodiments, the one or more cell types is a progenitor cell of a myoblast. In some embodiments, the one or more cell types is a progenitor cell of an ECM-secreting cell. In some embodiments, the one or more cell types is a progenitor cell of an endothelial cell.

[071] As used herein, a progenitor cell comprises a mesenchymal stem cell (MSc), an embryonic stem cell (ESc), an adult stem cell, a differentiated ESc, a differentiated adult Stem cell, and an induced pluripotent Stem cell (iPSc). As used herein, the term "progenitor cell" refers to a cell capable of giving rise to differentiated cells in multiple lineages, such as, myoblasts, fibroblasts, adipocytes, stromal cells, fibroblasts, pericytes, smooth muscle cells, and endothelial cells. "Progenitor cells" differ from stem cells in that they typically do not have the extensive self-renewal capacity.

[072] In some embodiments, a cell of the plurality of cell types of the present invention is a progenitor cell. In some embodiment, the progenitor cell is cultured in a monoculture. In some embodiment, the progenitor cell is differentiated in a monoculture. In some embodiment, the progenitor cell is differentiated in a monoculture and is then incubated on a three-dimensional porous scaffold with a plurality of cells according to the method of the present invention. A non- limiting example include, but is not limited to, culturing and differentiating a mesenchymal stem cell into a myoblast cell and thereafter seeding and subsequently incubating differentiated myoblast on a three-dimensional porous scaffold. Methods of culturing and inducing differentiation of progenitor cells to mature cells would be apparent to one of ordinary skill in the art.

[073] In some embodiments, the plurality of cell types comprises myoblasts and fibroblasts. In some embodiments, the plurality of cell types comprises myoblasts, fibroblasts and/or fibroblasts progenitor cells. In some embodiments, the plurality of cell types comprises myoblasts, fibroblasts and adipocytes. In some embodiments, the plurality of cell types comprises myoblasts, fibroblasts, adipocytes, and/or fibroblast progenitor cells and/or adipocyte progenitor cells. In some embodiments, the plurality of cell types comprises myoblasts, fibroblasts and endothelial cells. In some embodiments, the plurality of cell types comprises myoblasts, fibroblasts and endothelial cells, and/or fibroblast progenitor cells, and/or endothelial progenitor cells. In some embodiments, the plurality of cell types comprises myoblasts and smooth muscle cells. In some embodiments, the plurality of cell types comprises myoblasts, smooth muscle cells and endothelial cells. In some embodiments, the plurality of cell types comprises myoblasts, smooth muscle cells, endothelial cells, and adipocytes. In some embodiments, the plurality of cell types comprises myoblasts, fibroblasts, endothelial cells, and adipocytes. In some embodiments, the plurality of cell types comprises myoblasts, fibroblasts, endothelial cells, adipocytes and/or fibroblast progenitor cells, and/or adipocyte progenitor cells, and/or endothelial progenitor cells.

[074] In some embodiments, the plurality of cell types are obtained from a live animal and cultured as a primary cell line. For a non-limiting example, the cells may be obtained by biopsy and cultured ex vivo. For another non-limiting example, the cells may be obtained from commercial sources.

[075] In some embodiments, the plurality of cell types are derived from stem cells such as pluripotent embryonic stem cells. In another embodiment, mesenchymal stem cells (MSCs) are used. As known to one skilled in the art, MSCs may give rise to muscle cells, fat cells, bone cells, and cartilage cells. In another embodiment, the cells are induced pluripotent stem cells (iPS or iPSCs). In another embodiment, the cells are derived from totipotent embryonic stem cells such as cells from the blastocyst stage, fertilized eggs, placenta, or umbilical cords of these animals.

[076] In some embodiments, the plurality of cell types are derived from non-human cells. In some embodiments, the plurality of cell types are derived from non-human cells selected from the group consisting of: mammals, birds, fishes, invertebrates, reptiles, amphibians, and combinations thereof. In some embodiments, the plurality of cell types are derived from mammals. In some embodiments, the plurality of cell types are derived from non-human mammals. In some embodiments, the plurality of cell types are derived from livestock mammals. As used herein, "livestock" comprises any domestic mammal, semi-domestic mammal or captive wild mammal. Non-limiting examples of non-human mammals include: antelope, bear, beaver, bison, boar, camel, caribou, cattle, deer, elephant, elk, fox, giraffe, goat, hare, horse, ibex, kangaroo, lion, llama, moose, peccary, pig, rabbit, seal, sheep, squirrel, tiger, whale, yak, and zebra, or combinations thereof. In some embodiments, the plurality of cell types are derived from bird cells. Non-limiting examples of birds include: chicken, duck, emu, goose, grouse, ostrich, pheasant, pigeon, quail, and turkey, or combinations thereof. In some embodiments, the plurality of cell types are derived from fishes. Non-limiting examples of fishes include: bass, catfish, carp, cod, eel, flounder, fugu, grouper, haddock, halibut, herring, mackerel, mahi, marlin, orange roughy, perch, pike, pollock, salmon, sardine, shark, snapper, sole, swordfish, tilapia, trout, tuna, and walleye, or combinations thereof. In some embodiments, the plurality of cell types are derived from invertebrates. Non-limiting examples of invertebrates include: lobster, crab, shrimp, clams, oysters, mussels, and sea urchin. In some embodiments, the plurality of cell types are derived from reptiles. Non-limiting examples of reptiles include: snake, alligator, and turtle. In some embodiments, the plurality of cell types are derived from amphibians. Non- limiting example of amphibians includes frogs.

Seeding and culturing cells

[077] As will be appreciated by one skilled in the art, each type of cell used in the composition and method described herein may have a preferred or optimal range of cell density and prefers medium or growth factors suitable for the cell's viability. In some embodiments, each cell type is seeded in a specific cell density. In some embodiments, cells are seeded simultaneously or in a sequential manner.

[078] In some embodiments, myoblasts are seeded in a cell density of 10 3 to 10 7 cells to 10 mg of the porous textured protein. In some embodiments, different cell types are seeded in a specific ratio. In some embodiments, a ratio of seeded myoblast to seeded fibroblasts ranges between 1:1,000 and 1,000: 1. In some embodiments, a ratio of seeded myoblast and seeded fibroblasts to seeded endothelial cells ranges between 1:20 and 20: 1. In some embodiments, a ratio of seeded myoblast and seeded fibroblasts to seeded adipocytes ranges between 1:5000 and 5000: 1. In some embodiments, a ratio of seeded satellite cells to seeded smooth muscle cells ranges between 5: 1 and 1:5. In some embodiments, a ratio of seeded satellite cells to seeded skeletal microvascular endothelial cells ranges between 10: 1 to 1: 10. In some embodiments, a ratio of seeded smooth muscle cells to seeded skeletal microvascular endothelial cells ranges between 10:1 to 1: 10. In some embodiments, a ratio of seeded satellite cells to seeded smooth muscle cells to seeded skeletal microvascular endothelial cells ranges between 10: 1: 1 to 2: 1: 10. In one embodiment, a ratio of seeded satellite cells to seeded smooth muscle cells to seeded skeletal microvascular endothelial cells ranges between is 2: 1: 1 to 2: 1:5, or any ratio there between. In one embodiment, a ratio of seeded satellite cells to seeded smooth muscle cells to seeded skeletal microvascular endothelial cells ranges between is 2: 1: 1 to 2: 1:2, or any ratio there between. In one embodiment, a ratio of seeded satellite cells to seeded smooth muscle cells to seeded skeletal microvascular endothelial cells ranges between is 2: 1: 1 to 2: 1:3, or any ratio there between. In one embodiment, a ratio of seeded satellite cells to seeded smooth muscle cells to seeded skeletal microvascular endothelial cells ranges between is 2: 1: 1 to 2:1:4, or any ratio there between. Each possibility represents a separate embodiment of the present invention.

[079] In some embodiments, the seeded cell density and incubated cell density are comparably the same.

[080] In one embodiment, "coverage %" refers to the area or volume of a porous scaffold that is in contact with cells or myotubes. In another embodiment, coverage % refers to the area or volume of a porous scaffold that is occupied by cells or myotubes. As used herein, cells in contact with the scaffold are on, within, or a combination thereof.

[081] In some embodiments, coverage % of the plurality of cells is at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90, or at least 99%. In some embodiments, coverage % of the plurality of cells is 5-20%, 15-30%, 25- 40%, 35-50%, 45-60%, 55-70%, 65-80%, 75-90%, 85-100%, or any range therebetween. Each possibility represents a separate embodiment of the present invention.

[082] In some embodiments, the coverage % of satellite cells are at least 35%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90, or at least 99%. In some embodiments, coverage % of satellite cells 25-40%, 35-50%, 45-60%, 55-70%, 65-80%, 75-90%, 85-100%, or any range therebetween. Each possibility represents a separate embodiment of the present invention.

[083] In some embodiments, the coverage % of endothelial cells is 2% at most, 5% at most, is 10% at most, is 15% at most, is 20% at most, is 25% at most, is 30% at most, is 35% at most, is 40% at most, is 45% at most, or is 50% at most. In some embodiments, the coverage % of endothelial cells is 5-15%, 10-25%, 20-35%, 30-50%, or any range therebetween. Each possibility represents a separate embodiment of the present invention.

[084] In some embodiments, endothelial cells are used to increase proliferation of myoblasts or progenitor cells thereof. In some embodiments, the endothelial cells inhibit differentiation of myoblasts or progenitor cells thereof to myotubes. In some embodiments, endothelial cells support growth and proliferation of myoblasts or progenitor cells thereof. In some embodiments, endothelial cells are not required for differentiation of myoblasts or progenitor cells thereof. In some embodiments, according to the method of the present invention, endothelial cells activity (e.g., secretion of myogenic agents, supporting myoblasts growth, survival, or both) is maintained for a defined period. In some embodiments, the defined period for endothelial activity is the period required for myoblasts or progenitor cells thereof to achieve a coverage of at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, or at least 99% of the three-dimensional porous scaffold. In some embodiments, the defined period for endothelial activity is the period required for myoblasts or progenitor cells thereof to achieve a coverage of at least 30%,

[085] In some embodiments, the coverage % of myotubes is at least 5%, at least 20%, at least 35%, at least 50%, at least 70%, at least 85%, at least 90%, at least 90%, or at least 99%. In some embodiments, the coverage % of myotubes is 1-10%, 5-20%, 15-35%, 30-50%, 40-65%, 60- 85%, 80-90%, 90-100%, or any range therebetween. Each possibility represents a separate embodiment of the present invention.

[086] In some embodiments, myotubes of the composition of the present invention comprise 10,000-100,000 nuclei per mm 3 of the three-dimensional porous scaffold, 10,000-100,000 nuclei per mm 3 of the three-dimensional porous scaffold 15,000-200,000 nuclei per mm 3 of the three- dimensional porous scaffold 50,000-500,000 nuclei per mm 3 of the three-dimensional porous scaffold 5,000-1,000,000 nuclei per mm 3 of the three-dimensional porous scaffold 100,000- 250,000 nuclei per mm 3 of the three-dimensional porous scaffold 10,000-1,500,000 nuclei per mm 3 of the three-dimensional porous scaffold, or any range therebetween. Each possibility represents a separate embodiment of the present invention.

[087] As would be apparent to a skilled artisan, ECM affects myoblasts differentiation to myotubes. In some embodiments, ECM-secreting cells utilized according to methods of the present invention improve myoblasts differentiation. In some embodiments, ECM-secreting cells improved myoblasts differentiation by simulating tissue physical properties.

[088] As defined herein, the terms "improved" and "increased" are interchangeable.

[089] In some embodiments, improved is by at least 5%, by at least 20%, by at least 35%, by at least 50%, by at least 75%, by at least 90%, by at least 100%, by at least 250%, by at least 500%, by at least 750%, by at least 1,000%, by at least 2,500%, or by at least 5,000%. In some embodiments, improved is by 5-15%, 10-35%, 25-45%, 40-70%, 65-90%, 85-150%, 100-500%, or 250-1,000%. Each possibility represents a separate embodiment of the present invention.

[090] Non-limiting examples of a tissue physical properties include, but are not limited to stiffness, porosity, flexibility, rigidity, etc.). A tissue can be physically defined according to its Young's modulus, viscosity modulus, or other parameters, all of which would be apparent to one of ordinary skill in the art.

[091] In some embodiment, the method of the present invention further comprises a step of sterilizing the porous textured protein. In some embodiments, the porous textured protein is sterilized prior to seeding or incubating the plurality of cell types. In some embodiments, sterilization is by gamma radiation. In another embodiment, sterilization is ethanol-based sterilization. Procedures of sterilization would be apparent to a person of ordinary skill in the art.

[092] A skilled artisan will appreciate that the seeding and/or the culturing of cells is performed in the presence of a cell culture medium. In another embodiment, the cell culture medium comprises growth factor, cytokines, bioactive agents, nutrients, amino acids, antibiotic compounds, anti-inflammatory compounds, or any combination thereof. Suitable medium and compounds suitable for viability and growth of the cells are known to one skilled in the art.

[093] Growth factors that can be used in the methods and compositions of the invention include but are not limited to platelet-derived growth factors (PDGF), insulin-like growth factor (IGF-1). PDGF and IGF-1 are known to stimulate mitogenic, chemotactic and proliferate (differentiate) cellular responses. The growth factor can be, but is not limited to, one or more of the following: PDGF, e.g., PDGF AA, PDGF BB; IGF, e.g., IGF-I, IGF-II; fibroblast growth factors (FGF), e.g., acidic FGF, basic FGF, β-endothelial cell growth factor, FGF 4, FGF 5, FGF 6, FGF 7, FGF 8, and FGF 9; transforming growth factors (TGF), e.g., TGF-P1, TGF βΐ .2, TGF-p2, TGF-p3, TGF-p5; bone morphogenic proteins (BMP), e.g., BMP 1, BMP 2, BMP 3, BMP 4; vascular endothelial growth factors (VEGF), e.g., VEGF, placenta growth factor; epidermal growth factors (EGF), e.g., EGF, amphiregulin, betacellulin, heparin binding EGF; interleukins, e.g., IL- 1, IL-2, IL-3, IL-4, IL-5, IL-6, IL-7, IL-8, IL-9, IL-10, IL-11, IL-12, IL-13, IL-14; colony stimulating factors (CSF), e.g., CSF-G, CSF-GM, CSF-M; nerve growth factor (NGF); stem cell factor; hepatocyte growth factor, and ciliary neurotrophic factor.

[094] As exemplified hereinbelow, in the case of bovine-derived cells, an optimized satellite cells (SC) expansion media (i.e., Pro-LIF media) comprises Bovine SC (BSC) growth media (99%), amphotericin B (AB/AM; 1% lx), ZnCl 2 (50 μΜ), EGF (62 ng/ml), IGF-1 (100 ng/ml), and bFGF (10 ng/ml).

[095] As exemplified hereinbelow, in the case of bovine-derived cells, an optimized SC growth media comprises DMEM/HEPES (43.5%), Ham's F-10 Nutrient Mix (43.5%), Fetal bovine serum (10%), MEM NEAA (1% lx), GlutaMAX (1%), and AB/AM (1% lx).

[096] As exemplified hereinbelow, in the case of bovine-derived cells, an optimized SC differentiation media comprises DMEM/HEPES (97%), Donor Horse Serum (2%), AB/AM (1% lx), IGF-1 (100 ng/ml), and EGF (62 ng/ml).

Porous scaffold

[002] In some embodiments, plurality of cells of the invention are incubated with a porous scaffold. As used herein, the term "scaffold" refers to a structure comprising a material that provides a surface suitable for adherence/attachment, maturation, differentiation, and proliferation of cells. A scaffold may further provide mechanical stability and support. A scaffold may be in a particular shape or form so as to influence or delimit a three-dimensional shape or form assumed by a population of proliferating cells. In some embodiments, a porous scaffold of the invention is three dimensional.

[097] In some embodiments, an average pore diameter of the porous scaffold ranges from 20 micrometers (μιη) to 1000 μιη, 20 μιη to 900 μιη, 20 μιη to 800 μιη, 20 μιη to 700 μιη, 20 μιη to 600 μιη, 20 μιη to 500 μιη, 20 μιη to 400 μιη, 20 μιη to 300 μιη, 20 μιη to 200 μιη, 20 μιη to 100 μιη, 50 μιη to 1000 μιη, 50 μιη to 900 μιη, 50 μιη to 800 μιη, 50 μιη to 700 μιη, 50 μιη to 600 μιη, 50 μιη to 500 μιη, 50 μιη to 400 μιη, 50 μιη to 300 μιη, 50 μιη to 200 μιη, 50 μιη to 100 μιη, 100 μιη to 1000 μιη, 100 μιη to 900 μιη, 100 μιη to 800 μιη, 100 μιη to 700 μιη, 100 μιη to 600 μιη, 100 μιη to 500 μιη, 100 μιη to 400 μιη, 100 μιη to 300 μιη, 100 μιη to 200 μιη, 500 μιη to 1000 μηι, 500 μηι to 900 μηι, 500 μηι to 800 μηι, 500 μηι to 700 μηι, or 500 μηι to 600 μηι. Each possibility represents a separate embodiment of the present invention. In some embodiments, an average pore diameter of the porous scaffold ranges from 20 μιη to 1000 μιη.

[098] In some embodiments, the porous scaffold is edible. In some embodiments, the porous scaffold comprises a textured protein. In some embodiment, the porous scaffold comprises a polysaccharide. In some embodiments, the textured protein is a textured vegetable protein. In some embodiments, the textured protein is a textured soy protein (e.g., TSP).

[099] The term "texture" is used herein to refer to a rigid mass, or flexible mass, of individual cells which can be readily formed into various sizes, shapes and configurations and which is non-dispersible in water.

[0100] Suitable particulate textured protein materials for use herein can consist of from 30% to 100% protein, on a dry weight basis, and from 0% to 70% materials associated with the protein source material or added adjuvant materials. Examples of adjuvant materials are carbohydrates, vitamins, flavors, colorings or others. In some embodiments, the protein particles consist of 50% to 100% protein, or 50% to 80% protein by dry weight.

[0101] Suitable un-textured proteins which can be texturized to form textured particulate protein materials are available from a variety of sources. For a non- limiting example, a source of such proteins is a vegetable protein and certain fungal proteins; however, animal protein can be employed. Examples of suitable animal proteins are casein, collagen, and egg white. Examples of suitable vegetable protein sources are soybeans, safflower seed, corn, peanuts, wheat, wheat gluten, peas, sunflower seed, chickpea, cottonseed, coconut, rapeseed, sesame seed, leaf proteins, gluten, single cell proteins such as yeast, and the like.

[0102] Another example of a suitable protein source is mushroom. In some embodiment, a mushroom protein source comprises a protein amount of 15-20% (w/w), 20-30% (w/w), 28-45% (w/w), or 10-40% (w/w) by dry weight.

[0103] Generally, if the protein source is a vegetable protein, the protein prior to use is placed in a relatively pure form. Thus, for example if the protein source is soybeans, the soybeans can be solvent extracted, such as with hexane, to remove the oil therefrom. The resulting oil-free soybean meal contains about 50% protein.

[0104] The soybean meal can be processed in a known manner to remove carbohydrates and obtain products with higher levels of protein, for example, soy protein concentrates containing about 70% protein or soy protein isolates containing about 90% or more protein. In turn, a variety of suitable prior art processes can be employed to convert the soybean meal, concentrate, isolate and other edible protein bearing materials into suitable texturized particulate protein materials.

[0105] Suitable methods for converting un-textured animal and vegetable protein bearing materials into particulate textured proteins are disclosed, for example, in the following U.S. Pat. Nos. 2,682,466, granted June 29, 1954, to Boyer; 3,142,571, granted July 28, 1964, to Kitchel; 3,488,770, granted Jan. 6, 1970, to Atkinson; 3,498,794, granted Mar. 3, 1970, to Calvert et al; 3,759,715, granted Sept. 18, 1973, to Loepiktie et al.; 3,778,522, granted Dec. 11, 1973, to Strommer; 3,794,731, granted Feb. 26, 1974, to Dannert et al.; 3,814,823, granted June 4, 1974, to Yang et al.; and commonly assigned U.S. patent application Ser. No. 248,581, filed Apr. 28, 1972, now U.S. Pat. No. 3,840,679, granted Oct. 8, 1974 to Liepa et al.; all these patents being incorporated herein by reference.

[0106] In alternative embodiments, the porous textured protein may be substituted by other porous compositions which are ingestible edible as well as chewable. As used herein the terms "ingestible" and "edible" refer to compositions which can be safely taken into the body. These compositions include those which are adsorbed, and those which are not absorbed as well as those which are digestible and non-digestible. As used herein, the term "chewable" refers to a composition which can be broken/crushed into smaller pieces by chewing prior to swallowing. One skilled in the art will appreciate that a suitable edible composition may be selected according to physical properties (e.g., Young's modulus, viscosity modulus, stiffness, etc.) to a desired use (e.g., consumption by a human adult).

[0107] According to the method of the present invention, a plurality of cell types is seeded on a three dimensional porous scaffold per se. In some embodiments, a plurality of cell types seeded on a three dimensional porous scaffold requires no solidifying agents. In some embodiments, a plurality of cell types seeded on a three dimensional porous scaffold requires solidifying agents. In some embodiments, solidifying agents increase the adherence or attachment of the plurality of cell types to a three dimensional porous scaffold. Non-limiting examples for solidifying agents include but are not limited to thrombin or fibrin.

[0108] As used herein, the terms "gelation agent" and "solidifying agent" are interchangeable.

[0109] In the discussion unless otherwise stated, adjectives such as "substantially" and "about" modifying a condition or relationship characteristic of a feature or features of an embodiment of the invention, are understood to mean that the condition or characteristic is defined to within tolerances that are acceptable for operation of the embodiment for an application for which it is intended. Unless otherwise indicated, the word "or" in the specification and claims is considered to be the inclusive "or" rather than the exclusive or, and indicates at least one of, or any combination of items it conjoins.

[0110] It should be understood that the terms "a" and "an" as used above and elsewhere herein refer to "one or more" of the enumerated components. It will be clear to one of ordinary skill in the art that the use of the singular includes the plural unless specifically stated otherwise. Therefore, the terms "a," "an" and "at least one" are used interchangeably in this application.

[0111] For purposes of better understanding the present teachings and in no way limiting the scope of the teachings, unless otherwise indicated, all numbers expressing quantities, percentages or proportions, and other numerical values used in the specification and claims, are to be understood as being modified in all instances by the term "about". Accordingly, unless indicated to the contrary, the numerical parameters set forth in the following specification and attached claims are approximations that may vary depending upon the desired properties sought to be obtained. At the very least, each numerical parameter should at least be construed in light of the number of reported significant digits and by applying ordinary rounding techniques.

[0112] In the description and claims of the present application, each of the verbs, "comprise", "include" and "have" and conjugates thereof, are used to indicate that the object or objects of the verb are not necessarily a complete listing of components, elements or parts of the subject or subjects of the verb.

[0113] Other terms as used herein are meant to be defined by their well-known meanings in the art.

[0114] Additional objects, advantages, and novel features of the present invention will become apparent to one ordinarily skilled in the art upon examination of the following examples, which are not intended to be limiting. Additionally, each of the various embodiments and aspects of the present invention as delineated hereinabove and as claimed in the claims section below finds experimental support in the following examples.

[0115] It is appreciated that certain features of the invention, which are, for clarity, described in the context of separate embodiments, may also be provided in combination in a single embodiment. Conversely, various features of the invention, which are, for brevity, described in the context of a single embodiment, may also be provided separately or in any suitable subcombination or as suitable in any other described embodiment of the invention. Certain features described in the context of various embodiments are not to be considered essential features of those embodiments, unless the embodiment is inoperative without those elements.

EXAMPLES [0116] Generally, the nomenclature used herein, and the laboratory procedures utilized in the present invention include molecular, biochemical, microbiological and recombinant DNA techniques. Such techniques are thoroughly explained in the literature. See, for example, "Molecular Cloning: A laboratory Manual" Sambrook et al., (1989); "Current Protocols in Molecular Biology" Volumes I-III Ausubel, R. M., ed. (1994); Ausubel et al., "Current Protocols in Molecular Biology", John Wiley and Sons, Baltimore, Maryland (1989); Perbal, "A Practical Guide to Molecular Cloning", John Wiley & Sons, New York (1988); Watson et al., "Recombinant DNA", Scientific American Books, New York; Birren et al. (eds) "Genome Analysis: A Laboratory Manual Series", Vols. 1-4, Cold Spring Harbor Laboratory Press, New York (1998); methodologies as set forth in U.S. Pat. Nos. 4,666,828; 4,683,202; 4,801,531; 5,192,659 and 5,272,057; "Cell Biology: A Laboratory Handbook", Volumes I-III Cellis, J. E., ed. (1994); "Culture of Animal Cells - A Manual of Basic Technique" by Freshney, Wiley-Liss, N. Y. (1994), Third Edition; "Current Protocols in Immunology" Volumes I-III Coligan J. E., ed. (1994); Stites et al. (eds), "Basic and Clinical Immunology" (8th Edition), Appleton & Lange, Norwalk, CT (1994); Mishell and Shiigi (eds), "Strategies for Protein Purification and Characterization - A Laboratory Course Manual" CSHL Press (1996); "Bacteriophage Methods and Protocols", Volume 1: Isolation, Characterization, and Interactions, all of which are incorporated by reference. Other general references are provided throughout this document.

Materials and methods

Textured soy protein

[0117] Textured soy protein (TSP) is commercially available as several different products. TSP scaffolds were prepared out of 5 different sources of commercial TSP - 3 TSP chunks (Fig. 1A) bought at a local health food store and 2 TSP flakes (Figs. 1B-1C) bought from ADM. TSP was sterilized in gamma radiation of 25-40 kGy for 3.5 hours and kept in sterile environment or using 70% (v/v) ethanol for 15 minutes. Followed by 3 washes with PBS.

TSP scaffold preparation

[0118] TSP flakes (Arcon or TVP) were chosen using Graefe Forceps so as to have similar thickness of about 200-400 μιη (per 1 layer). Flakes were incubated in 5-10 ml of sterile DDW at 37 °C overnight (in a 50 ml falcon). The next day the flakes were cut into 6 mm scaffolds using 6 mm biopsy punch inside the hood on a non-TC 10 cm plate. Using a Pasteur pipette the scaffold was remove from the biopsy punch and transferred into a falcon with 5 ml growth media. TSP chunks were incubated in DDW at 37°C overnight. Then chunks were cut into cylinders (inset of Fig. 2A) using a biopsy punch and cut into 1 mm thick disks using a knife (Fig. 2A). The scaffolds were then placed in 5 ml growth media. Scaffold seeding

[0119] Per a single scaffold, 2 x 10 6 BSC were transferred to an Eppendorf and centrifuged for 4 minutes at 1,500 rcf. Meanwhile each scaffold was put in a well of a Non-TC 6-well plate with 50 μΐ with growth media. After centrifugation the media was aspirated from the Eppendorf tube leaving a pellet of cells. Then, per each Eppendorf the following steps were performed: (1) the media was thoroughly aspirated from the scaffold using forceps to gently squeeze the scaffold and media residues were vacuumed; (2) 7μ1 of thrombin (20MHU/ml sigma in PBS) and 7μ1 of fibrinogen (15 mg/ml sigma in PBS) were added; (3) the cells were then seeded on the scaffold; (4) scaffolds were incubated at 37 °C for 30 min till drying and then were added with 2ml of expansion media into the well; (5) the day after, scaffold was transferred to a new container (e.g., 24 non-TC well with coverslip bottom) using sterile Graefe forceps into and was added with 2 ml of expansion media; (6) the scaffold was incubated at 37 °C and 5% CO2; (7) the scaffold maintained in the expansion media for 7 days; (8) media was replaced every 2 days (2 ml); (9) after 7 days the expansion media was replaced by BSC differentiation media; and (10) differentiation media was replaced every 2 days for a period of 5 days.

Cell culture

[0120] Bovine satellite cells (BSC) were cultured in BSC proliferation media 43.5% DMEM/HEPES (Gibco), 43.5% F-10 Nut Mix (Gibco), 10% FBS (HyClone), 1% NEAA (Gibco), 1% GlutaMAX (Gibco) and 1% Penicillin-Streptomycin- Amphotericin B Solution (Biological Industries (Ab/Am, BI) supplemented with 50 μΜ ZnCh (Millipore), 62 ng/ml EGF (R&D Systems), 100 ng/ml IGF-1 (R&D Systems), 10 ng/ml LIF (R&D Systems) and 10 ng/ml bFGF (R&D Systems). BSC differentiation medium was comprised of DMEM/HEPES supplemented with 2% FBS and 1% Ab/Am. Red Fluorescent Protein (RFP) expressing Dermal Fibroblasts (Lonza, USA) are cultured in Dulbecco's minimal essential medium (DMEM; Gibco Life Technologies) supplemented with 10% FBS (HyClone; Thermo Fisher Scientific), 1% nonessential amino acids (Biological Industries), 0.2% β-mercaptoethanol (Biological Industries), and 100 units/ml penicillin and 0.1 mg/ml streptomycin (Pen-Strep Solution, Biological Industries, Israel). Myoblasts (American Type Culture Collection) are cultured in DMEM, supplemented with 10% FBS, 2.5% HEPES buffer (Biological Industries) and 1% Pen- strep solution. Myoblast differentiation medium is comprised of DMEM supplemented with 2% FBS, 2.5% HEPES buffer and 1% Pen-strep solution. All incubations are performed in a 5% (v/v) CO2 humidified atmosphere at 37 °C. Bovine smooth muscle cell (SMC; CellAplications) were cultured as described in table 3. Bovine aortic endothelial cells (BAEC; CellApplications) were cultured in the commercial media (CellApplications). Bovine skeletal muscle microvascular ECs (BSkMVEC; AngioProteomie) were cultured in ECM commercial medium (ScienCell) with 10% total FBS. Bovine dermal fibroblasts (BDF; Sciencell) were cultured either in the commercial medium (FM-2; ScienCell) or in DMEM high glucose medium supplemented with 15% FBS.

Immunohistochemistry

[0121] Scaffolds are rinsed twice in PBS, then fixed in 4% paraformaldehyde (PFA) for 20 minutes on a shaker. The scaffolds are washed thrice with PBS for 5 minutes on a shaker, and then permeabilized with 0.3% Triton X-100 (Bio Lab Ltd) for 10 min. Next, the scaffolds are washed thrice with PBS for 5 minutes and then immersed in 5% Bovine Serum Albumin (BSA; Millipore) in PBS overnight at 4°C. The scaffolds are incubated with 250μ1 Primary antibody (in 5% BSA in PBS) for 3 hours at room temperature. Following incubation, the scaffolds are washed 4 times with PBS for 5 minutes. Then, the cells are incubated in room temperature for 3 hours with Secondary antibody (in PBS) and 4',6-diamidino-2-phenylindole (DAPI, 1: 1000 in PBS; Vector Laboratories) in a plate covered with tin foil on shaker. The scaffolds are washed with PBS for 5 minutes on a shaker and imaged with a confocal microscope (LSM 700, Zeiss).

Bovine aortic smooth muscle cells (BAMSC)

[0122] The effect on cell proliferation of four different media formulations were compared with a commercial medium (BSM) (Table 3). At the beginning of the experiment, cells were at passage 8 and were seeded at 4,000 cells/cm 2 in a 6-well plate. At day 3, cells were trypsinized, counted with a hemocytometer, and reseeded at 4,000 cells/cm 2 . The experiment was performed twice, using the same cells for each condition. One-way ANOVA was used to test statistical significance.

Cell growth kinetics

[0123] Approximately 4 x 10 3 cells/cm 2 were seeded and counted every day with a hemocytometer, over a period of six days. Counted values were plotted.

Cell staining

[0124] Supporting cells (BDF or BASMC) were stained with 3 ml fresh media with 15 μΐ of lipophilic -tracers DiD (1 mg/ml diluted in absolute ethanol; #D7757, Molecular Probes®, USA). Bovine endothelial cells (BECs; BAEC or BSkMVEC) were stained with 3 ml fresh media with 5 μΐ of lipophilic-tracers Dil (3 mg/ml diluted in absolute ethanol; #D, Molecular Probes®, USA) and incubated for 30 min in 37 °C. Following one wash with medium and then two more washes with PBS, the cells were trypsinized and ready for seeding as described hereinbelow.

Seeding cells on a porous scaffold [0125] Bovine ECs (1.25 x 10 5 ) and supporting cells (0.25 x 10 5 ) were resuspended in 5 μΐ of a 1:1 mixture of 1 5mg/ml fibrinogen (Sigma Aldrich) and 20 NIHu/ml thrombin (Sigma Aldrich). The fibrinogen solution was prepared by diluting lyophilized human fibrinogen (Sigma Chemical) in 40 mM glycine-tris buffer. The thrombin solution was prepared by diluting thrombin (Sigma Aldrich) in 40 mM calcium chloride. The co-culture suspension was then seeded onto 4 mm diameter round PLLA-PLGA porous scaffolds and allowed to solidify in 12- well non-tissue plates for 30 min, in an incubator (37 °C, 5% C0 2 ). After solidification, 2 ml of endothelial culture media (ECM, ScienCell) supplemented with additional 5% FBS and 2 nM VEGF was added to each well. Medium was replaced every other day.

[0126] For further tuning of the ratio of cells seeded on porous scaffolds, the cells were seeded as described above with the following modifications: the cells were first seeded with 1: 1 medium (bEBM, AngioProteomie: commercial medium of the supporting cells) and the cells were resuspended in 5 μΐ of a 1: 1 mixture of 15 mg/ml fibrinogen (Johnson and Johnson) and 2 U/ml thrombin (Johnson and Johnson).

Bovine satellite cells (BSC) differentiation in 2D

[0127] 5 x 10 4 BSC (Passage 4) were seeded on a TC 24-well plate with coverslip bottom in triplicates. Cells were grown according to a myotube formation protocol, as follows: 4 days in growth media and 7 days in differentiation media (t=0 refers to the point when using differentiation media started). The cells were then stained for Hoechst and Myogenin at days -1 and 7.

Optimization of BSC differentiation in 2D

[0128] BSC (Passage 4) were grown for 4 days in expansion media with different GFs (as described below) and were then grown for 7 days in differentiation media (DMEM-HEPES + 2% HS). Day 0 was defined as the day the media was replaced to differentiation media.

The expansion media formulations included: (1) Control - Growth media (No GF added); (2) bovine fibroblast growth factor (bFGF; 10 ng/ml); (3) Epidermal growth factor (EGF; 62 ng/ml); (3) Insulin-like growth factor 1 (IGF-1; 100 ng/ml); (4) Pro-LIF; bFGF 10 ng/ml, EGF 62 ng/ml, IGF-1 100 ng/ml, and ZnCl 2 50 μΜ); (6) GF weaning: Pro-LIF media for 3 days, then 3 washes with growth media (every 5 minutes), then 1 day growth media; (7) cells were stained with Dil grown in growth media - to check whether Dil, a fluorescent color added for tracking the cells, affects cell differentiation.

[0129] The inventors further checked the effect of GFs: IGF-1, EGF and their combination during the differentiation phase. BSC (Passage 4) were grown for 4 days in growth media and were then grown for 7 days in differentiation media with different GFs (as described below). The differentiation media formulations included: (1) Control - the original differentiation media (DMEM-HEPES + 2% HS); (2) IGF-1 (100 ng/ml); (3) EGF (62 ng/ml); and (4) IGF-1 (lOOng/ml) + EGF (62 ng/ml). Cells were visualized using brightfield microscopy for all cases described above.

Optimization of BSC expansion on 3D scaffolds

[0130] For this purpose, a double factorial experiment (seeding volume and expansion media formulation) was set with duplicates, as follows: (1) Seeding volume and cell number: (a) 10 μΐ seeding volume and 1 x 10 6 BSC; (b) 10 μΐ seeding volume and 2 x 10 6 BSC; and (c) 20 μΐ seeding volume and 2 x 10 6 BSC. (2) Expansion media: (a) Growth media; and (b) Pro-LIF media (growth media + bFGF + EGF + IGF-1 + ZnCl 2 ). BSC (Passage 4) were stained using Dil and were then seeded on porous scaffolds (Arcon) according to the Arcon seeding protocol.

Optimization of BSC differentiation on 3D scaffolds

[0131] For this purpose, a double factorial experiment (expansion media in the first week and differentiation media in the second week) was set, as follows: (1) Expansion media: (a) Control (Growth media); (b) Pro-LIF; and (c) Pro-LIF - bFGF. (2) Differentiation media either alone or supplemented as follows: (a) Control (differentiation media with 2% Horse serum); (b) + IGF-1; (c) + EGF; and (d) + IGF-1 + EGF. BSC (Passage 4) were stained using Dil. Four (4) mm porous scaffolds were seeded with 0.5 x 10 6 BSC (Passage 4) in 5 μΐ of fibrin. The cells were imaged at days 2, 7 and 14 post seeding. The scaffolds were then stained for desmin. Staining was done according a whole mount staining protocol with the primary polyclonal antibody 1: 100 goat a desmin (Santa Cruz Cat. No. sc-7559) and the secondary antibody Alexa 1:200 488 Donkey a goat (Invitrogen Cat. No. A11055).

Staining bovine satellite cells (BSC) prior seeding

[0132] BSC were stained with 1 ml fresh media with 5 μΐ of lipophilic-tracers Dil (3 mg/ml diluted in absolute ethanol, Molecular Probes®, USA) and incubated for 30 min in 37 °C. Following 1 wash with medium and then 2 washes with PBS, after which the cells were trypsinized and ready for seeding as described.

Seeding cells on PLLA-PLGA

[0133] Bovine satellite cells (BSC) with or without bovine ECs (BEC) and with or without supporting cells were resuspended in 6 μΐ of a 1: 1 mixture of 15 mg/ml fibrinogen (Sigma Aldrich) and 20 NIHU/ml thrombin (Sigma Aldrich). The fibrinogen solution was prepared by diluting lyophilized human fibrinogen (Sigma Chemical) in 40 mM glycine-tris buffer. The thrombin solution was prepared by diluting thrombin (Sigma Aldrich) in 40 mM calcium chloride. The cells' suspension was then seeded onto 4 mm diameter round PLLA-PLGA scaffolds and allowed to solidify in 12-well non-tissue plates for 30 min, inside an incubator (37 °C, 5% C0 2 ). After solidification, 1 ml of 1: 1 mixture of endothelial culture media with BSC growth medium (Pro-LIF) was added to each well. Medium was replaced every other day. After a week, the medium was changed to BSC differentiation medium (supplemented with IGF-1 and EGF) for additional week.

Seeding cells on textured soy protein scaffolds

[0134] Bovine satellite cells (BSC) with or without bovine ECs and with or without supporting cells were resuspended in 15 μΐ of a 1: 1 mixture of 15mg/ml fibrinogen (Sigma Aldrich) and 20 NIHU/ml thrombin (Sigma Aldrich). The fibrinogen solution was prepared by diluting lyophilized human fibrinogen (Sigma Chemical) in 40 mM glycine-tris buffer. The thrombin solution was prepared by diluting thrombin (Sigma Aldrich) in 40 mM calcium chloride. The cells' suspension was then seeded on both sides of 6 mm diameter round textured soy protein scaffolds and allowed to solidify in 12-well non-tissue plates for 45 min, inside an incubator (37 °C, 5% CO2). After solidification, 1 ml of 1: 1 mixture of endothelial culture media with BSC growth medium (Pro-LIF) was added to each well. Medium was replaced every other day. After a week, the medium was changed to BSC differentiation medium (supplemented with IGF-1 and EGF) for additional week.

[0135] Two seeding ratios and 2 types of endothelial cells (and their appropriate medium) were tested as described in the tables below:

Table 1 - Seeding on PLLAXPLGA scaffolds

Table 2 - Seeding on textured soy protein scaffolds Culture BSC SMC BEC TOTAL

Tri-culture 2: 1: 1 750,000 375,000 375,000 1,500,000

Tri-culture 2: 1:5 375,000 187,500 937,500 1,500,000

Monoculture control

750,000 750,000 for 2: 1: 1

Co-culture 2:1 750,000 375,000 1,125,00

Co-culture 2:1 750,000 375,000 1,125,00

Monoculture control

375,000 375,000 for 2: 1:5

Co-culture 2:1 375,000 187,500 562,500

Co-culture 2:5 375,000 937,500 1,312,500

Seeding cells on Gelfoam

[0136] Bovine satellite cells (BSC) were resuspended in 15 μΐ of BSC growth media. The cells' suspension was then seeded on both sides of 6 mm diameter celinder Gelfoam © scaffolds and allowed to solidify in 12-well non-tissue plates for 30 min, inside an incubator (37 °C, 5% C02). After solidification, 1 ml of BSC growth medium (Pro - LIF) was added to each well. Medium was replaced every other day. After a week, the medium was changed to BSC differentiation medium (supplemented with IGF-1 and EGF) for additional week.

Cell growth on scaffolds

[0137] One and two weeks post seeding, Dil labeled-BSC seeded on PLLA/PLGA and Arcon scaffolds were imaged using confocal microscopy for assessing cell growth.

Whole -mount immunofluorescence staining

[0138] Two weeks post seeding, scaffolds were fixed with 4% paraformaldehyde (PFA; Electron Microscopy Sciences) for 20 min, followed by three washes with PBS (Gibco® Life Technologies). Permeabilization was achieved by incubating the scaffolds for 15 min at room temperature (RT), in 0.3% Triton X-100 (Bio Lab Ltd). After three washes with PBS, scaffolds were incubated overnight in blocking buffer (10% FBS, 1% (w/v) glycine and 0.01% triton, in PBS), at 4 °C. Scaffolds were then incubated overnight at 4 °C with primary antibodies: 1:50 goat-anti-CD31 (Santa Cruz), with or without 1:50 mouse anti-MYH (Santa Cruz), diluted in blocking buffer. Following several washes with PBS, samples were incubated with 1:400 donkey anti-goat Alexa Fluor®488 (Jackson ImmunoResearch), 1:50 Alexa Flour 647 -conjugated mouse anti-myogenin (Santa Cruz) and 1:1,000 DAPI (4',6-diamidino-2-phenylindole, Sigma- Aldrich) with or without 1:300 Alexa Flour 647 anti-mouse, for 3 h at RT. Following several washes with PBS, scaffolds were immediately imaged with a Zeiss LSM700 confocal microscope (Carl Zeiss), using the Zen software. Image processing and further analyses were performed using FIJI software. PLLA/PLGA scaffolds were also whole-mount immunofluorescence-stained one week post seeding.

Cryosectioning and trichrome staining

[0139] At the end of each experiment, scaffolds were incubated overnight in a 30% (w/v) sucrose solution at 4 °C. Scaffolds were then embedded in optimal cutting temperature (OCT) compound (Tissue-Tec, USA), and frozen for subsequent cryosectioning. The OCT embedded scaffolds were cryosectioned into 5 and 10 μιη-thick sections. 5 μιη-thick sections were then stained according to trichrome standard staining protocol.

EXAMPLE 1

Three dimensional structure analysis of a TSP porous scaffold

[0140] TSP scaffolds were examined using scanning electron microscopy (SEM) to analyze their three-dimensional (3D) structure both at the micron and the nano scale. The lOOx images showed the TSP was porous with 100-1,000 μηι pore size, which can be used for cell culture. It was shown that the wall in the big-TSP were thicker compared to the medium sourced TSP (Figs. 3A and 3B). SEM images (2xl0 5 magnification) of the large TSP showed that the TSP comprised 30 nm ball shaped protein clusters. Analysis of the medium TSP showed more amorphous glue-like material between those clusters (Figs. 4A and 4B).

EXAMPLE 2

Growth and proliferation of fibroblasts cultured on a porous scaffold

[0141] Attachment and proliferation of fibroblasts on a TSP scaffold were examined. First, 50,000 dermal fibroblasts which were labeled with RFP, were seeded on a TSP scaffold, and cultured. Following 14 days of incubation, the TSP scaffold, which was populated with dermal fibroblasts, was additionally seeded with 200,000 endothelial cells, and incubated for additional 7 days. Samples of TSP populated with cells were immune-stained and imaged using a confocal microscopy, at 8, 18 and 21 days following seeding of the fibroblasts. Results demonstrated that the fibroblasts proliferated and covered the TSP scaffold even when seeded at the low cell number of 50,000 cells (Fig. 5A). Following the seeding of the endothelial cells (Fig. 5B), the endothelial cells gathered into clusters (Fig. 5C). Next, scaffolds from each scaffold source and scaffolds cut from 90° to the surface direction of the TSP were examined. Each of the scaffolds was seeded with 200,000 fibroblast cells and incubated for 14 days. Results demonstrated that the fibroblasts cells grew and proliferated on the big and medium TSP scaffold (Fig 6). These results indicate that cells can fill the scaffold after 14 days of culture.

EXAMPLE 3

Growth of myoblasts cultured on a porous scaffold

[0142] In order to assess the applicability of generating a muscle tissue, experiments were conducted to examine whether muscle cells can grow on the TSP scaffold.

[0143] For this purpose, 0.5xl0 6 myoblasts were seeded on TSP scaffolds and cultured in the presence of a differentiation medium which was low on serum and thus fitted better for cultured meat production. The images of the myoblasts at day 14 (Fig. 7) showed that the myoblasts proliferated on the TSP scaffold, indicating that cells can fill the scaffold after 14 days of culture.

EXAMPLE 4

Proliferation of bovine skeletal muscle cells cultured on TSP

[0144] In order to assess the applicability for human consumption, bovine skeletal muscle cells (bSkMC) were seeded on the TSP scaffold. Specifically, 0.5xl0 6 bSkMC cells were seeded on each scaffold and cultured in the presence of a differentiation medium. On day 14, cells were imaged using a confocal microscopy. The images (Fig. 8) showed that the bSkMC proliferated on the TSP scaffold, indicating that the TSP is applicable for cultured meat production.

EXAMPLE 5

Proliferation of bovine aortic smooth muscle cells (SMC)

[0145] The inventors examined the effect of four different media formulations compared to a commercial medium (BSM; table 3) on the proliferation of SMC seeded (Figs. 9A-9E) in media described hereinbelow.

Table 3. Formulations of different media tested on SMC

[0146] At day 3, no significant difference in cell number could be detected across the five different media (Fig. 10A). On day 3 of a following experiment (Fig. lOError! Reference source not found. B), a significant difference between the four different treatments was observed (P<0.01) and a post-hoc analysis revealed that the number of cells exposed to pyruvate- supplemented medium was significantly higher compared to NEAA supplemented medium and 15% FBS supplemented medium (with P-values <0.05 and <0.01, respectively). The difference between the commercial medium and the sodium pyruvate- supplemented medium was marginally significant (P=0.052). Therefore, the inventors proceeded with sodium pyruvate supplemented medium (1 mM) for BAOSMC culturing.

EXAMPLE 6

Growth characterization of BDF and SMC under defined conditions

[0147] After having defined custom media formulations, the inventors have examined their growth kinetics under these conditions (Example 5). The growth rates of BDF (Fig. 11A) and BAOSMC were found to be highly similar (Fig. 11B).

EXAMPLE 7

Effect of EC and supporting cells on vascularization

[0148] In order to test if bovine endothelial cells (EC; BAEC - Bovine aortic endothelial cells or BSkMVEC - Bovine skeletal muscle microvascular endothelial cells) seeded in co-culture with supporting cells enhance vascularization and scaffold coverage, BAEC were co-seeded with either BDF or BASMC. Indeed, co-culturing fibroblasts together with myoblasts and endothelial cells has been shown to increase vascularization of engineered muscle constructs and promoted stabilization of the vessel structure over time. Cells were found to cover the entire scaffold (Fig. 12). Furthermore, when BAEC were seeded in co-culture with BASMC (Fig. 12Error! Reference source not found.A) the inventors noticed that the staining for ECs is reduced over time. Also, a vessel like structure was evident on day 2 in this combination. When BAEC were seeded in co- culture with BDF (Fig. 12B) the inventors noticed no vessel-like structures. When BSkMVEC were seeded in co-culture with BASMC (Fig. 13A) the inventors noticed much more BASMC than BSkMVEC. Also, vessel-like structures were evident with BASMC (Fig. 13A) and with BDF (Fig. 13B), throughout the entire experiment period.

EXAMPLE 8

Specific cell ratio improves blood vessel network formation

[0149] To further improve vessel networks the inventors seeded bovine BAEC with supporting cells (BDF or BASMC) in different ratios. The inventors examined 3 different ratios that were: 1:3, 1: 1 and 5: 1 ECs to supporting cells. The inventors clearly observed self-assembly of the BAEC cells (Figs. 14-16) which created vessels, as early as on day 2 in the presence of BASMC (Figs. 14 and 16). The inventors also noticed that by day 6 there were less ECs then on day 2 and that the BAEC created a region of vessel network in a 1: 1 BAEC to BASMC ratio. BAEC in the presence of BDF seemed not to create vessels (Figs. 15).

EXAMPLE 9

Bovine satellite cell (BSC) differentiation in a cell culture

[0150] The inventors then examined myogenin expression before and after myotube formation from BSC, and further assessed the ability to use DAPI-Myogenin for fusion index (FI) measurement. The inventors found little evidence of myogenin expression in undifferentiated BSC (Fig. 17). Myogenin expression occurred mainly in the nuclei of myotubes and in some nuclei around the myotubes. It was shown that the fusion Index (FI) can be calculated automatically by the ratio of myogenin/Hoechst nuclei count, which was found to be comparable to the manual FI calculation. This protocol was further used in quantification of myotube formations.

EXAMPLE 10

Optimization of BSC differentiation in a cell culture

[0151] The inventors then wanted to optimize BSC differentiation in 2D. The inventors checked which growth factors (GFs) can be added during (1) the expansion phase which will not inhibit myotube formation during the differentiation stage and (2) at the end of the differentiation phase which will increase myotube size and FI.

[0152] After the expansion phase and before the differentiation phase, spontaneous myotubes appeared in samples that did not contain bFGF (Fig. 18) suggesting bFGF inhibits myotube formation. EGF, IGF-1 and Dil did not seem to affect myotube formation. Increased cell density occurred in IGF-1, Pro -LIF and Weaning. On day 4 of differentiation, the inventors observe an increased in total myotubes under bFGF and Pro-LIF culture conditions. The bFGF maintained the sternness of the BSC and once removed the cells burst into differentiation (Fig. 19). On day 7 of differentiation, the inventors observed a high area of myotube in the control, weaning and Dil, as compared to the other tested conditions (Fig. 20). The inventors concluded that there is a fine line between myotube formation and cell death. Accordingly, cells should be grown in expansion media containing bFGF (alone or with Pro-LIF) until reaching confluency and then be transferred to differentiation media for a short period of time of 2-5 days. Further shown was high abundancy of myotubes when IGF-1 or IGF-1 and EGF were added to the cell culture media. EXAMPLE 11

Optimization of BSC expansion on a porous scaffold

[0153] The inventors then attempted to increase BSC coverage on the TSP scaffold of above 60% as previous experiments showed low cell coverage on the porous scaffold. Therefore, GFs were added to the expansion phase and initial cell number at seeding was increased. Quantification of the cell coverage area showed that cells cultured with growth media supplemented with GFs (i.e., Pro-LIF) covered 72 + 15% of the scaffold, while cells cultured solely on growth media covered approximately 18% of the scaffold (Fig. 21). A difference which was found to be statistically significant between the two treatments (P value = 0.0009).

EXAMPLE 12

Optimization of BSC differentiation on a porous scaffold

[0154] In order to find the optimal conditions for myotube formation on 3D scaffolds, the inventors scanned multiple combinations of expansion media and subsequently differentiation media. The myotubes formed without the GFs in the differentiation media (Figs. 22A, E and I) were small and rounded. Adding IGF-1 to the differentiation media resulted in myotubes - which were rarely observed in the growth media (Fig. 22B), common in the Pro-LIF- bFGF (Fig. 22J) and abundant in the Pro-LIF media (Figs. 22F). Adding EGF to the differentiation media resulted in myotubes in all expansion media (Figs. 22C, G and K). Adding IGF-1 + EGF resulted in myotubes in the growth media (Fig. 22D) and Pro-LIF- bFGF (Fig. 22L) and abundant myotubes in the Pro-LIF (Fig. 22H).

EXAMPLE 13

Tri-culture increases BSC proliferation, elongation, and coverage on a porous scaffold

[0155] The inventors then wanted to examine the effect of tri-culturing BSC with constant amount of smooth muscle cells (SMC) and different amounts of skeletal microvascular endothelial (SkMVEC) (2: 1: 1 and 2: 1:5, respectively) on BSC survival, proliferation and myotube differentiation (positive for the differentiation marker - Myogenin). The inventors observed that in the low SkMVEC concentration (2: 1: 1) BSC appeared to be more elongated compared to their appearance when incubated with higher concentration of SkMVEC (2: 1:5; Fig. 28). The inventors also observed that BSC had increased their coverage over the porous scaffold and appeared to be more differentiated compared to either monoculture or co-cultures comprising BSC (controls). The inventors concluded that tri-cultures better support BSC proliferation and differentiation compared to the other tested groups in both incubation ratios, and summarized BSC differentiation potential as follows: BSC alone < BSC + EC < BSC + SMC < BSC + EC + SMC (Figs. 23-26 and 28). The invnetors also observed extracellular matrix (ECM) secretion in tri-cultures of BSC + SMC + SkMVEC (2: 1:1) (Figs. 27A-27B).

EXAMPLE 14

Tri-culture increases BSC differentiation on a textures soy protein scaffold

[0156] The inventors then wanted to examine the effect of tri-culturing BSC with constant amount of smooth muscle cells (SMC) and skeletal microvascular endothelial (SkMVEC) (2:1: 1 respectively) on BSC survival, proliferation and myotube differentiation (positive for the differentiation marker - Myogenin). The inventors observed that not only BSC had increased their coverage over the TSP scaffold when seeded either in co-culture with SMC or in tri-culture with SMC and BEC but also appeared to be more differentiated compared to either monoculture (BSC only) or co-culture (BSC with BEC; Fig. 29).

EXAMPLE 15

Seeding BSC without the addition of solidifying gel

[0157] The inventors then wanted to examine the effect of seeding BSC on FDA-approved collagen based scaffold (Gelfoam ©) without the addition of solidifying agents (e.g., fibrin. The inventors observed successful BSC differentiation, as demonstrated by myotube formation (Fig. 30).

[0158] While certain features of the invention have been illustrated and described herein, many modifications, substitutions, changes, and equivalents will now occur to those of ordinary skill in the art. It is, therefore, to be understood that the appended claims are intended to cover all such modifications and changes as fall within the true spirit of the invention.