US20050084958A1 - Tissue engineered meat for consumption and a method for producing tissue engineered meat for consumption - Google Patents

Tissue engineered meat for consumption and a method for producing tissue engineered meat for consumption Download PDF

Info

Publication number
US20050084958A1
US20050084958A1 US10/943,489 US94348904A US2005084958A1 US 20050084958 A1 US20050084958 A1 US 20050084958A1 US 94348904 A US94348904 A US 94348904A US 2005084958 A1 US2005084958 A1 US 2005084958A1
Authority
US
United States
Prior art keywords
human
cells
meat product
muscle
muscle cells
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US10/943,489
Inventor
Jon Vein
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Individual
Original Assignee
Individual
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Individual filed Critical Individual
Priority to US10/943,489 priority Critical patent/US20050084958A1/en
Publication of US20050084958A1 publication Critical patent/US20050084958A1/en
Abandoned legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/0062General methods for three-dimensional culture
    • AHUMAN NECESSITIES
    • A23FOODS OR FOODSTUFFS; TREATMENT THEREOF, NOT COVERED BY OTHER CLASSES
    • A23LFOODS, FOODSTUFFS, OR NON-ALCOHOLIC BEVERAGES, NOT COVERED BY SUBCLASSES A21D OR A23B-A23J; THEIR PREPARATION OR TREATMENT, e.g. COOKING, MODIFICATION OF NUTRITIVE QUALITIES, PHYSICAL TREATMENT; PRESERVATION OF FOODS OR FOODSTUFFS, IN GENERAL
    • A23L13/00Meat products; Meat meal; Preparation or treatment thereof
    • AHUMAN NECESSITIES
    • A23FOODS OR FOODSTUFFS; TREATMENT THEREOF, NOT COVERED BY OTHER CLASSES
    • A23LFOODS, FOODSTUFFS, OR NON-ALCOHOLIC BEVERAGES, NOT COVERED BY SUBCLASSES A21D OR A23B-A23J; THEIR PREPARATION OR TREATMENT, e.g. COOKING, MODIFICATION OF NUTRITIVE QUALITIES, PHYSICAL TREATMENT; PRESERVATION OF FOODS OR FOODSTUFFS, IN GENERAL
    • A23L13/00Meat products; Meat meal; Preparation or treatment thereof
    • A23L13/50Poultry products, e.g. poultry sausages
    • AHUMAN NECESSITIES
    • A23FOODS OR FOODSTUFFS; TREATMENT THEREOF, NOT COVERED BY OTHER CLASSES
    • A23LFOODS, FOODSTUFFS, OR NON-ALCOHOLIC BEVERAGES, NOT COVERED BY SUBCLASSES A21D OR A23B-A23J; THEIR PREPARATION OR TREATMENT, e.g. COOKING, MODIFICATION OF NUTRITIVE QUALITIES, PHYSICAL TREATMENT; PRESERVATION OF FOODS OR FOODSTUFFS, IN GENERAL
    • A23L17/00Food-from-the-sea products; Fish products; Fish meal; Fish-egg substitutes; Preparation or treatment thereof
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10TECHNICAL SUBJECTS COVERED BY FORMER USPC
    • Y10STECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10S426/00Food or edible material: processes, compositions, and products
    • Y10S426/802Simulated animal flesh

Definitions

  • the field of the present invention relates to producing and harvesting meat products for consumption.
  • it relates to tissue engineered meat for consumption.
  • Meat products such as beef, pork, lamb, poultry, or fish are desirable products for food consumption. Meat products are currently produced from whole animals, which is a highly inefficient production method because a significant portion of all agriculturally produced grain is used for animal rather than human consumption.
  • livestock feed accounts for approximately 70% of all the wheat, corn, and other grain produced.
  • to produce one pound of beef thousands of pounds of water are required for the animal to drink and to grow the livestock feed. Meanwhile, throughout the world, by some account, over 800 million people are malnourished and 50,000 people die of starvation every day.
  • campylobacter In the poultry industry, for example, as much as 25% of broiler chickens and 45% of ground chickens are reportedly allowed to test positive for salmonella.
  • the Center for Disease Control estimates that campylobacter infects 70% to 90% of all chickens. Campylobacter infections cause cramps, bloody diarrhea, and fever. Every year in the United States, campylobacter infection results in about 800 deaths. Infections with campylobacter may also lead to Guillian-Barre syndrome, a disease that requires intensive care for several weeks. The incidence of serious illness and death from these bacteria may increase as more antibiotic-resistant strains develop. This has caused some scientists to question the continued use of antibiotics as a feed supplement for livestock.
  • the present invention is directed to tissue engineered meat products and methods for producing such meat products.
  • the meat product comprises muscle cells that are grown ex vivo. These muscle cells may be grown and attached to a support structure and may be derived from any non-human cells.
  • the meat product is substantially free from any harmful microbial or parasitic contamination.
  • Another embodiment of the invention is directed to a meat product comprising muscle cells and other cells such as fat cells or cartilage cells, or both, that are grown ex vivo together with the muscle cells.
  • the meat product comprises muscle cells that have been exposed to an electric or oscillating current.
  • meat products are taken from the muscles of animals. Butchers carve out corresponding cuts of beef, poultry, lamb, fish, or pork to be sold as steak, chicken breast, lamb chops, fish fillet, pork chops, etc. Meat products also include meat-product derivatives such as ground meat that may be processed into meatball, hamburger patty, fishball, sausage, salami, bologna, ham, etc. Meat products may also include muscle tissues or meat that has been seasoned or dried such as jerky.
  • One embodiment of the present invention involves a method for producing meat products that may be used for consumption.
  • the method may include culturing muscle stem cells in vitro and allowing these cells to differentiate into specific types of muscle cells such as skeletal muscle cells or smooth muscle cells ex vivo.
  • Muscle cells may be derived from any non-human animals consumed by humans such as mammals (e.g. cattle, buffalo, pigs, sheep, deer, etc.), birds (e.g. chicken, ducks, ostrich, turkey, pheasant, etc.), fish (e.g. swordfish, salmon, tuna, sea bass, trout, catfish, etc.), invertebrates (e.g.
  • muscle cells are derived from pluri-potent embryonic mesenchymal stem cells that give rise to muscle cells, fat cells, bone cells, and cartilage cells.
  • the muscle cells may also be derived from toti-potent embryonic stem cells such as cells from the blastocyst stage, fertilized eggs, placenta, or umbilical cords of these animals.
  • Muscle cells may be grown in culture into muscle tissues that are attached to a support structure such as a two or three-dimensional scaffold or support structure.
  • the muscle cells may be grown on the two dimensional support structure such as a petri-dish forming several layers of cells that may be peeled and processed for consumption.
  • Other examples of two dimensional support structures may include porous membranes that allow for diffusion of nutrients from culture media on one side of the membrane to the other side where the cells are attached. In this type of culture conditions, additional layers of cells may be achieved by exposing the cells to culture media from both sides of the membrane, i.e., cells received nutrients through diffusion from one side of the membrane and also from the culture media covering the cells growing on the membrane.
  • Muscle cells may also be grown on, around, or inside a three-dimensional support structure.
  • the support structure may be sculpted into different sizes, shapes, and forms, as desired, to provide the shape and form for the muscle cells to grow and resemble different types of muscle tissues such as steak, tenderloin, shank, chicken breast, drumstick, lamb chops, fish fillet, lobster tail, etc.
  • the support structure may be made from natural or synthetic biomaterials that are preferably non-toxic so that they may not be harmful if ingested. Natural biomaterials may include, for example, collagen, fibronectin, laminin, or other extracellular matrices. Synthetic biomaterials may include, for example, hydroxyapatite, alginate, polyglycolic acid, polylactic acid, or their copolymers.
  • the support structure may be formed as a solid or semisolid support.
  • the support structure preferably, has high porosity to provide maximal surface area for cell attachment.
  • a three-dimensional support structure may also be molded to include a branched vascular network providing for delivery of nutrients into and shuttling out of metabolites from the cells at the inner mass of the meat product.
  • the branch vascular network may be edible by using non-toxic natural or synthetic biomaterials as mentioned above.
  • the support structure may also include adhesion peptides, cell adhesion molecules, or other growth factors covalently or non-covalently associated with the support structure. Examples of the peptides include sequences such as Arg-Gly-Asp or Arg-Glu-Asp-Val. Niklason, L., et. al., Advances in Tissue Engineering of Blood Vessels and Other Tissues, Transplant Immunology, 5(4):303-306 (1997). This reference is hereby incorporated by reference as if fully set forth herein.
  • culture conditions for these muscle cells may include static, stirred, or dynamic flow conditions.
  • the preferred method is to use a bioreactor, which produces greater volume of cells and allows greater control over the flow of nutrients, gases, metabolites, and regulatory molecules.
  • bioreactors may provide physical and mechanical signals such as compression to stimulate cells to produce specific biomolecules.
  • meat products derived from muscle cells grown ex vivo may include fat cells derived also from any non-human animals.
  • Fattier meat is generally tastier, but with greater fat content comes greater risk of adverse health consequences such as heart disease.
  • the ratio of muscle cells to fat cells may be regulated in vitro to produce the meat products with optimal flavor and health effects. Regulation may be achieved by controlling the ratio of muscle and fat cells that are initially seeded in culture and/or by varying, as desired, the concentrations and ratio of growth factors or differentiation factors that act upon the muscle cells or fat cells.
  • cartilage derived from chondrocytes may first form an underlying support layer or structure together with the support structure. Afterwards, muscle cells or fat cells, or both, may be seeded onto the chondrocyte layer. The interaction of muscle cells and chondrocytes may further provide the necessary regulatory signals required for tissue formation. Examples of meat products that have muscle cells and cartilage cells include chicken breast or pork ribs.
  • aseptic techniques may be used to culture the muscle cells resulting in meat products that are substantially free from harmful microbes such as bacteria, fungi, viruses, prions, protozoa, or any combination of the above.
  • Harmful microbes may include pathogenic type microorganisms such as salmonella, campylobacter, E. coli — 0156:H7, etc.
  • muscle cells grown in culture may be substantially free from parasites such as tapeworms that infect muscles of whole animals and that are transferred to humans through consumption of insufficiently cooked meat.
  • Aseptic techniques may also be employed in packaging the meat products as they come off the biological production line. Such quality assurance may be monitored by standard assays for microorganisms or chemicals that are already known in the art. “Substantially free” means that the concentration of microbes or parasites is below a clinically significant level of contamination, i.e., below a level wherein ingestion would lead to disease or adverse health conditions.
  • the meat product derived from muscle cells grown ex vivo may be exposed to an electric or oscillating current.
  • muscle tissues grown ex vivo or in vitro may have never been exercised (e.g. never been used to move a leg).
  • exposing the muscle cells, muscle tissue, or the meat products in vitro to an electric or oscillating current may mimic exercise and increase the similarity in texture between meat grown ex vivo and meat derived from whole animals.
  • the electric or oscillating current may also increase the growth rate of muscle cells ex vivo.
  • the electric or oscillating current may be applied to the muscle stem cells or to the muscle cells after they have differentiated from the stem cells.
  • other nutrients such as vitamins that are normally lacking in meat products from whole animals may be added to increase the nutritional value of the meat. This may be achieved either through straight addition of the nutrients to the growth medium or through genetic engineering techniques.
  • the gene or genes for enzymes responsible for the biosynthesis of a particular vitamin, such as Vitamin D, A, or the different Vitamin B complexes may be transfected in the cultured muscle cells to produce the particular vitamin.
  • regulatory factors, growth factors, or other gene products may also be genetically introduced into the muscle cells.
  • These factors known as myogenic regulatory factors (“MRFs”), may stimulate and regulate the growth of muscles in vivo, but may not normally be produced by muscle cells in vivo or in vitro.
  • MRFs myogenic regulatory factors
  • expressing myogenic regulatory factors in cultured muscle cells may increase the production of muscle cells in vitro.
  • the meat products derived from muscle cells in vitro may include different derivatives of meat products. These derivatives may be prepared, for example, by grounding or shredding the muscle tissues grown in vitro and mixed with appropriate seasoning to make meatballs, fishballs, hamburger patties, etc. The derivatives may also be prepared from layers of muscle cells cut and spiced into, for example, beef jerky, ham, bologna, salami, etc. Thus, the meat products of the present invention may be used to generate any kind of food product originating from the meat of an animal.
  • This example illustrates the isolation of pluri-potent mesenchymal stem cells for use in producing meat products in vitro.
  • Mesenchymal stem cells give rise to muscle cells (myocytes), fat cells (adipocytes), bone cells (osteocytes), and cartilage cells (chrondocytes).
  • Mesenchymal stem cells may be dissected and isolated from embryonic tissues of any non-human animal embryos. In cattle, for example, embryonic mesenchymal tissues that are rich in pluri-potent muscle stem cells are preferably isolated from embryos at day 30 to 40 or earlier. Once dissected, the embryonic tissues may be minced into small pieces about one millimeter by one millimeter in size in phosphate buffered saline (“PBS”) pH 7.45.
  • PBS phosphate buffered saline
  • the reaction may be stopped by adding medium such as DMEM or Ham's F-12, or both in 1:1 ratio, (Life Technologies, Rockville, Md.) that is supplemented with 10 mM Hepes, 2 mM L-glutamine (Sigma-Aldrich), 10-20% heat-inactivated fetal calf or bovine serum (Hyclone Laboratories, Logan, Utah), penicillin at 100 units/ml and streptomycin at 100 ⁇ g/ml (“complete medium”). Cells may be completely dissociated by gently pipetting the tissues up and down followed by washing the cells in complete medium once or twice using a centrifuge. The cells may then be plated onto an appropriate-sized petri dish which may be coated with natural biomaterials (e.g.
  • collagen fibronectin, laminin, or other extracellular matrices
  • synthetic biomaterials e.g. hydroxyapatite, alginate, polyglycolic acid, polylactic acid, or their copolymers, or both, and may be grown at 37° C. and equilibrated with 5% CO 2 .
  • mesenchymal stem cells After mesenchymal stem cells have been isolated, they may be enriched for myoblasts or muscle stem cells in culture. Initially, the cells may be differentially plated on different petri dishes after dissociation and washing as described in Example I. Using a 60 mm petri dish, the cells may first be incubated in complete medium for two to four hours. During this time, epithelial cells will tend to attach quickly to the petri dish while the myoblasts remain in the supernatant. The supernatant may then be collected and the myoblasts may be plated on a different petri dish coated with natural or synthetic biomaterials such as those mentioned in Example I. Myoblasts may be enriched by supplementing the growth media with growth factors such as skeletal muscle growth factor, prostaglandin F 2 ⁇ (“PGF 2 ⁇ ”), and insulin-like growth factor I (“IGF-1”).
  • growth factors such as skeletal muscle growth factor, prostaglandin F 2 ⁇ (“PGF 2 ⁇ ”), and insulin-like growth factor I
  • myoblasts may be differentiated into specific myoctes or muscle cells by culturing the myoblasts in complete medium or in minimal media (e.g. complete medium less the fetal calf serum) supplemented with muscle specific growth or differentiation factors such as PGF 2 ⁇ at concentrations ranging from 24 pg/ml to 28 pg/ml, and insulin from 10 ⁇ 6 M to 10 ⁇ 5 M.
  • muscle specific growth or differentiation factors such as PGF 2 ⁇ at concentrations ranging from 24 pg/ml to 28 pg/ml, and insulin from 10 ⁇ 6 M to 10 ⁇ 5 M.
  • the culture medium may also be supplemented with appropriate neurotransmitters such as acetylcholine.
  • myoblasts may be enriched from toti-potent embryonic stem cells.
  • Toti-potent cells may be derived from in vitro fertilized eggs of an animal using in vitro fertilization techniques, from stem cells present in umbilical cords or placenta, or from Embryonic Stem (ES) cells isolated from cells at the blastocyst stage.
  • ES cells for example, may be collected, gently dissociated by trypsin, and cultured in vitro with recombinant leukemia inhibitory factor (Chemicon, San Diego, Calif.) and feeder cells such as growth arrested embryonic fibroblasts cells.
  • These toti-potent cells may be treated with growth factors such as PGF 2 ⁇ or IGF-1 to induce the cells to differentiate into myoblasts.
  • myoblasts or myocytes may be identified. Briefly, myoblasts or myocytes grown in culture may be transferred into glass slides coated with appropriate extracellular matrix as described above. These cells may be grown to the desired number and differentiation using the conditions described above. After a sufficient growth and differentiation period, the cells may be fixed with 4% formaldehyde. If intracellular antibody markers or nucleotide probes are to be used, the cell membranes may be permeabilized with 1% NP-40 or Triton-X.
  • Antibodies against markers specific for myoblasts or myocytes such as myosin, titin, alpha-actinin available from Sigma® may be used to identify the cells using standard fluorescent immunohistochemistry techniques. Alternatively, single stranded RNA or DNA probes for these markers may also be used for in-situ hybridization.
  • muscle cells when they have been attached to a three dimensional support structure as disclosed below, they may be cryo-frozen, sectioned and identified using antibody markers such as antibodies against myosin, titin, 12101, troponin T, alpha actinin available from Sigma®.
  • Two or three dimensional scaffolds or supports may be sculpted from natural biomaterials (e.g. collagen, fibronectin, laminin, or other extracellular matrix) or synthetic biomaterials (e.g. hydroxyapatite, alginate, polyglycolic acid, polylactic acid, and their copolymers), or both.
  • the three dimensional scaffolds are sculpted with branch pathways for nutrients and culture media to reach the internal mass of the forming muscle tissues. Examples of materials and construction methods for these scaffolds are provided by U.S. Pat. Nos.
  • the support structure is preferably sculpted to different sizes, shapes, and forms to allow for growth of muscle tissues resembling different types of meat products such as steak, tenderloin, shank, chicken breast, drumstick, lamb chops, fish fillet, lobster tail, etc.
  • Adipocytes, chondrocytes, and osteoblasts are all capable of differentiating from pluri-potent mesenchymal stem cells or toti-potent embryonic stem cells.
  • the stem cells may be isolated as described in Example I or III.
  • the stem cells may be cultured in DMEM, or Ham's F-12, or both in a 1:1 ratio.
  • the medium may be supplemented with thyroid hormone, transferrin, insulin, as well as other growth factors, such as insulin-like growth factor (IGF), basic fibroblast growth factor, and growth hormone.
  • IGF insulin-like growth factor
  • BMP bone morphogenetic proteins
  • BMP-4 and BMP-2 bone morphogenetic proteins
  • Ahrens et. al. Expression of human bone morphogenetic proteins -2 or -4 in murine mesenchymal progenitor C 3 H 10 T 1/2 cells induces differentiation into distinct mesenchymal cell lineages, DNA Cell Biol., 12:871-880 (1993); Wang et. al., Bone Morphogenetic protein -2 causes commitment and differentiation in C 3 H 10 T 1/2 and 3 T 3 cells, Growth Factors 9:57 (1993).
  • adipocyte differentiation of adipocytes may be enhanced with agonist of peroxisome proliferator-activated receptor gamma (“PPAR gamma”) such as BRL 49653 (rosiglitazone).
  • PPAR gamma peroxisome proliferator-activated receptor gamma
  • BRL 49653 rosiglitazone
  • myoblasts may even be induced to trans-differentiate into adipoblasts (adipocyte precursors) by treating myoblasts cells or muscle satellite cells with long-chain fatty acids (“LCFA”) or thiazolidinediones, or both.
  • LCFA long-chain fatty acids
  • thiazolidinediones or both.
  • meat products with the desired amount of fat content may be produced by seeding and co-culturing muscle cells and adipocyte cells at a certain ratio.
  • stem cells may be allowed to differentiate initially into myoblasts and then at a later time, LCFA or thiadolidinediones may be added at different concentrations and different exposure times to trans-differentiate the myoblasts into adipocytes as desired.
  • the growth of muscle cells and fat cells may be regulated by controlling the concentration of the growth and differentiation factors. For example, if less fat cells are desired in the final meat product, lesser concentrations of BMP factors may be added to the culture while a higher concentration of PGF 2 ⁇ and/or insulin may be added to promote muscle cell growth.
  • Chondrocytes or cartilage cells may also be isolated from an animal's knee or rib cages. Using similar techniques as described in Example I, dissected tissue from the knee or rib cages may be minced, digested with collagenase, and washed with complete medium. The cells may then be differentially plated to increase the purity of chondrocyte cells.
  • chondrocytes differentiate in response to mechanical stress.
  • the cells may be subjected to shear flow stress as described in U.S. Pat. No. 5,928,945, entitled “Application of Shear Flow Stress to Chondrocytes or Chondrocyte Stem Cells to Produce Cartilage,” which is hereby incorporated by reference as if fully set forth herein.
  • Chondrocytes may initially form a first layer of support cells in a three-dimensional scaffold. Myoblasts or adipocyte cells, or both, may then be seeded onto the chondrocyte layer and grown to the desired size. As such, the chondrocyte layer may provide additional adhesion or growth factors to the muscle cells.
  • Muscle cells grown in vitro differ from muscle cells grown in vivo in that in vivo cells are used during exercise or body movements. As muscles are used in vivo, muscle cells, in limbs for example, contract and relax in accordance with the movement of the limbs. Hence, to more closely mimic the growth of muscle cells in vivo, the cells grown in vitro may be exposed to an electric or oscillating current, or pulses of electric or oscillating current to contract the muscle cells. Electric probes may be immersed into the culture media to deliver mild current.
  • the support structure may be coated with electrically conducting materials. Examples of electrically conducting materials and a method for coating them onto the support structure are described in U.S. Pat. No. 5,843,741, entitled “Method for Altering the Differentiation of Anchorage Dependent Cells on an Electrically Conducting Polymer,” which is hereby incorporated by reference as if fully set forth herein.

Abstract

A non-human tissue engineered meat product and a method for producing such meat product are disclosed. The meat product comprises muscle cells that are grown ex vivo and is used for food consumption. The muscle cells may be grown and attached to a support structure and may be derived from any non-human cells. The meat product may also comprise other cells such as fat cells or cartilage cells, or both, that are grown ex vivo together with the muscle cells.

Description

    RELATED APPLICATIONS
  • The present application is a continuation of U.S. application Ser. No. 09/991,544, filed Nov. 16, 2001, which claims priority to U.S. Provisional Patent Application No. 60/60/249,993, filed Nov. 17, 2000, both of which are incorporated herein in their entirety by reference.
  • FIELD OF THE INVENTION
  • The field of the present invention relates to producing and harvesting meat products for consumption. In particular, it relates to tissue engineered meat for consumption.
  • BACKGROUND OF THE INVENTION
  • Meat products such as beef, pork, lamb, poultry, or fish are desirable products for food consumption. Meat products are currently produced from whole animals, which is a highly inefficient production method because a significant portion of all agriculturally produced grain is used for animal rather than human consumption. In the United States, for example, livestock feed accounts for approximately 70% of all the wheat, corn, and other grain produced. In addition, to produce one pound of beef, thousands of pounds of water are required for the animal to drink and to grow the livestock feed. Meanwhile, throughout the world, by some account, over 800 million people are malnourished and 50,000 people die of starvation every day.
  • Current meat production methods are also harmful to the environment. Rain forests are depleted at a rate of approximately 500 square feet of rain forest for every pound of beef to be grown. Likewise, modern techniques for fishing marine life have become so efficient that the oceans and lakes are over-fished. Species that were once common are now endangered or extinct.
  • Current scientific efforts to address these problems have focused on increasing the effectiveness of breeding or growing livestock. For example, growth hormones have been used to make livestock grow faster and thus, consume less grain and water. Growth hormones are typically injected into the livestock, but new methods of introducing the growth hormone have also been developed using genetic engineering technologies such as transgenics or cloning of the whole animal. Current meat production methods, nonetheless, require water, grain, and land to raise livestock.
  • Another problem with current meat production methods involves food contamination. Every year, on average, each American becomes sick and 9,000 people die from something they have injested. To control food contamination, the government's present strategy is to inspect meat during processing. The USDA and the FDA, however, rarely regulate the farms where pathogens originate because they lack the regulatory powers over the farms. Nonetheless, except for E. coli 0156:H7, dangerous bacteria are legally considered “inherent” to raw meat. Two of the “inherent bacteria,” however,—campylobacter and salmonella—account for 80% of all illnesses and 75% of all deaths from meat and poultry consumption.
  • In the poultry industry, for example, as much as 25% of broiler chickens and 45% of ground chickens are reportedly allowed to test positive for salmonella. The Center for Disease Control estimates that campylobacter infects 70% to 90% of all chickens. Campylobacter infections cause cramps, bloody diarrhea, and fever. Every year in the United States, campylobacter infection results in about 800 deaths. Infections with campylobacter may also lead to Guillian-Barre syndrome, a disease that requires intensive care for several weeks. The incidence of serious illness and death from these bacteria may increase as more antibiotic-resistant strains develop. This has caused some scientists to question the continued use of antibiotics as a feed supplement for livestock.
  • Thus, there exists a need to produce meat products for consumption that is more efficient, safer, and healthier than the current methods of production.
  • SUMMARY OF THE INVENTION
  • The present invention is directed to tissue engineered meat products and methods for producing such meat products. In one embodiment of the invention, the meat product comprises muscle cells that are grown ex vivo. These muscle cells may be grown and attached to a support structure and may be derived from any non-human cells. In a preferred embodiment of the invention, the meat product is substantially free from any harmful microbial or parasitic contamination. Another embodiment of the invention is directed to a meat product comprising muscle cells and other cells such as fat cells or cartilage cells, or both, that are grown ex vivo together with the muscle cells. In another embodiment of the invention, the meat product comprises muscle cells that have been exposed to an electric or oscillating current.
  • GENERAL DESCRIPTION OF THE PREFERRED EMBODIMENT
  • Generally, meat products are taken from the muscles of animals. Butchers carve out corresponding cuts of beef, poultry, lamb, fish, or pork to be sold as steak, chicken breast, lamb chops, fish fillet, pork chops, etc. Meat products also include meat-product derivatives such as ground meat that may be processed into meatball, hamburger patty, fishball, sausage, salami, bologna, ham, etc. Meat products may also include muscle tissues or meat that has been seasoned or dried such as jerky.
  • One embodiment of the present invention involves a method for producing meat products that may be used for consumption. The method may include culturing muscle stem cells in vitro and allowing these cells to differentiate into specific types of muscle cells such as skeletal muscle cells or smooth muscle cells ex vivo. Muscle cells may be derived from any non-human animals consumed by humans such as mammals (e.g. cattle, buffalo, pigs, sheep, deer, etc.), birds (e.g. chicken, ducks, ostrich, turkey, pheasant, etc.), fish (e.g. swordfish, salmon, tuna, sea bass, trout, catfish, etc.), invertebrates (e.g. lobster, crab, shrimp, clams, oysters, mussels, sea urchin, etc.), reptiles (e.g. snake, alligator, turtle, etc.), and amphibians (e.g. frog legs). Preferably, muscle cells are derived from pluri-potent embryonic mesenchymal stem cells that give rise to muscle cells, fat cells, bone cells, and cartilage cells. The muscle cells may also be derived from toti-potent embryonic stem cells such as cells from the blastocyst stage, fertilized eggs, placenta, or umbilical cords of these animals.
  • Muscle cells may be grown in culture into muscle tissues that are attached to a support structure such as a two or three-dimensional scaffold or support structure. The muscle cells may be grown on the two dimensional support structure such as a petri-dish forming several layers of cells that may be peeled and processed for consumption. Other examples of two dimensional support structures may include porous membranes that allow for diffusion of nutrients from culture media on one side of the membrane to the other side where the cells are attached. In this type of culture conditions, additional layers of cells may be achieved by exposing the cells to culture media from both sides of the membrane, i.e., cells received nutrients through diffusion from one side of the membrane and also from the culture media covering the cells growing on the membrane.
  • Muscle cells may also be grown on, around, or inside a three-dimensional support structure. The support structure may be sculpted into different sizes, shapes, and forms, as desired, to provide the shape and form for the muscle cells to grow and resemble different types of muscle tissues such as steak, tenderloin, shank, chicken breast, drumstick, lamb chops, fish fillet, lobster tail, etc. The support structure may be made from natural or synthetic biomaterials that are preferably non-toxic so that they may not be harmful if ingested. Natural biomaterials may include, for example, collagen, fibronectin, laminin, or other extracellular matrices. Synthetic biomaterials may include, for example, hydroxyapatite, alginate, polyglycolic acid, polylactic acid, or their copolymers. The support structure may be formed as a solid or semisolid support.
  • To provide for optimal cell and tissue growth, the support structure, preferably, has high porosity to provide maximal surface area for cell attachment. A three-dimensional support structure may also be molded to include a branched vascular network providing for delivery of nutrients into and shuttling out of metabolites from the cells at the inner mass of the meat product. In this particular embodiment, the branch vascular network may be edible by using non-toxic natural or synthetic biomaterials as mentioned above. Furthermore, the support structure may also include adhesion peptides, cell adhesion molecules, or other growth factors covalently or non-covalently associated with the support structure. Examples of the peptides include sequences such as Arg-Gly-Asp or Arg-Glu-Asp-Val. Niklason, L., et. al., Advances in Tissue Engineering of Blood Vessels and Other Tissues, Transplant Immunology, 5(4):303-306 (1997). This reference is hereby incorporated by reference as if fully set forth herein.
  • On the other hand, culture conditions for these muscle cells may include static, stirred, or dynamic flow conditions. For scaled up production, the preferred method is to use a bioreactor, which produces greater volume of cells and allows greater control over the flow of nutrients, gases, metabolites, and regulatory molecules. Furthermore, bioreactors may provide physical and mechanical signals such as compression to stimulate cells to produce specific biomolecules. Vacanti, J., et. al., Tissue Engineering: The Design and Fabrication of Living Replacement Devices for Surgical Reconstruction and Transplantation, Lancet, 354 Suppl. 1, pSI32-34 (1999). This reference is hereby incorporated by reference as if fully set forth herein.
  • In another embodiment of the invention, meat products derived from muscle cells grown ex vivo may include fat cells derived also from any non-human animals. Fattier meat is generally tastier, but with greater fat content comes greater risk of adverse health consequences such as heart disease. Thus, the ratio of muscle cells to fat cells may be regulated in vitro to produce the meat products with optimal flavor and health effects. Regulation may be achieved by controlling the ratio of muscle and fat cells that are initially seeded in culture and/or by varying, as desired, the concentrations and ratio of growth factors or differentiation factors that act upon the muscle cells or fat cells.
  • In another embodiment of the invention, cartilage derived from chondrocytes may first form an underlying support layer or structure together with the support structure. Afterwards, muscle cells or fat cells, or both, may be seeded onto the chondrocyte layer. The interaction of muscle cells and chondrocytes may further provide the necessary regulatory signals required for tissue formation. Examples of meat products that have muscle cells and cartilage cells include chicken breast or pork ribs.
  • In a preferred embodiment of the invention, aseptic techniques may be used to culture the muscle cells resulting in meat products that are substantially free from harmful microbes such as bacteria, fungi, viruses, prions, protozoa, or any combination of the above. Harmful microbes may include pathogenic type microorganisms such as salmonella, campylobacter, E. coli 0156:H7, etc. In addition, muscle cells grown in culture may be substantially free from parasites such as tapeworms that infect muscles of whole animals and that are transferred to humans through consumption of insufficiently cooked meat. Aseptic techniques may also be employed in packaging the meat products as they come off the biological production line. Such quality assurance may be monitored by standard assays for microorganisms or chemicals that are already known in the art. “Substantially free” means that the concentration of microbes or parasites is below a clinically significant level of contamination, i.e., below a level wherein ingestion would lead to disease or adverse health conditions.
  • In another preferred embodiment of the invention, the meat product derived from muscle cells grown ex vivo may be exposed to an electric or oscillating current. Unlike muscle tissues derived from whole animals, muscle tissues grown ex vivo or in vitro may have never been exercised (e.g. never been used to move a leg). Thus, exposing the muscle cells, muscle tissue, or the meat products in vitro to an electric or oscillating current may mimic exercise and increase the similarity in texture between meat grown ex vivo and meat derived from whole animals. The electric or oscillating current may also increase the growth rate of muscle cells ex vivo. The electric or oscillating current may be applied to the muscle stem cells or to the muscle cells after they have differentiated from the stem cells.
  • In another embodiment of the invention, other nutrients such as vitamins that are normally lacking in meat products from whole animals may be added to increase the nutritional value of the meat. This may be achieved either through straight addition of the nutrients to the growth medium or through genetic engineering techniques. For example, the gene or genes for enzymes responsible for the biosynthesis of a particular vitamin, such as Vitamin D, A, or the different Vitamin B complexes, may be transfected in the cultured muscle cells to produce the particular vitamin.
  • In another embodiment of the invention, regulatory factors, growth factors, or other gene products may also be genetically introduced into the muscle cells. These factors, known as myogenic regulatory factors (“MRFs”), may stimulate and regulate the growth of muscles in vivo, but may not normally be produced by muscle cells in vivo or in vitro. Thus, expressing myogenic regulatory factors in cultured muscle cells may increase the production of muscle cells in vitro.
  • In another embodiment of the invention, the meat products derived from muscle cells in vitro may include different derivatives of meat products. These derivatives may be prepared, for example, by grounding or shredding the muscle tissues grown in vitro and mixed with appropriate seasoning to make meatballs, fishballs, hamburger patties, etc. The derivatives may also be prepared from layers of muscle cells cut and spiced into, for example, beef jerky, ham, bologna, salami, etc. Thus, the meat products of the present invention may be used to generate any kind of food product originating from the meat of an animal.
  • The following examples illustrate how one skilled in the art may make use of the current invention to produce meat products in vitro. Methods in cell biology, cell culture, and immunohistochemistry that are not explicitly described in this disclosure have already been amply reported in the scientific literature.
  • EXAMPLE I
  • This example illustrates the isolation of pluri-potent mesenchymal stem cells for use in producing meat products in vitro. Mesenchymal stem cells give rise to muscle cells (myocytes), fat cells (adipocytes), bone cells (osteocytes), and cartilage cells (chrondocytes). Mesenchymal stem cells may be dissected and isolated from embryonic tissues of any non-human animal embryos. In cattle, for example, embryonic mesenchymal tissues that are rich in pluri-potent muscle stem cells are preferably isolated from embryos at day 30 to 40 or earlier. Once dissected, the embryonic tissues may be minced into small pieces about one millimeter by one millimeter in size in phosphate buffered saline (“PBS”) pH 7.45. Five to ten pieces of the minced tissue may be incubated in 300 μl of 0.25% trypsin and 0.1% EDTA in PBS for thirty minutes at 37° C. with gentle agitation. Afterwards, the tissues may be allowed to settle on the bottom of the tube by gravity or gentle centrifugation. The supernatant containing the trypsin/EDTA solution may then be aspirated and replaced with 300 μl of 0.1% collagenase in PBS for ten to thirty minutes at 37° C. Colleganese digestion may be repeated for several cycles as desired. Depending of the viscosity of the solution because of DNA released from damaged cells, 40 μl of DNase I at 1 mg/ml in PBS may be added to the collagenase solution in between cycles.
  • The reaction may be stopped by adding medium such as DMEM or Ham's F-12, or both in 1:1 ratio, (Life Technologies, Rockville, Md.) that is supplemented with 10 mM Hepes, 2 mM L-glutamine (Sigma-Aldrich), 10-20% heat-inactivated fetal calf or bovine serum (Hyclone Laboratories, Logan, Utah), penicillin at 100 units/ml and streptomycin at 100 μg/ml (“complete medium”). Cells may be completely dissociated by gently pipetting the tissues up and down followed by washing the cells in complete medium once or twice using a centrifuge. The cells may then be plated onto an appropriate-sized petri dish which may be coated with natural biomaterials (e.g. collagen, fibronectin, laminin, or other extracellular matrices) or synthetic biomaterials (e.g. hydroxyapatite, alginate, polyglycolic acid, polylactic acid, or their copolymers), or both, and may be grown at 37° C. and equilibrated with 5% CO2.
  • EXAMPLE II
  • After mesenchymal stem cells have been isolated, they may be enriched for myoblasts or muscle stem cells in culture. Initially, the cells may be differentially plated on different petri dishes after dissociation and washing as described in Example I. Using a 60 mm petri dish, the cells may first be incubated in complete medium for two to four hours. During this time, epithelial cells will tend to attach quickly to the petri dish while the myoblasts remain in the supernatant. The supernatant may then be collected and the myoblasts may be plated on a different petri dish coated with natural or synthetic biomaterials such as those mentioned in Example I. Myoblasts may be enriched by supplementing the growth media with growth factors such as skeletal muscle growth factor, prostaglandin F(“PGF”), and insulin-like growth factor I (“IGF-1”).
  • Further, myoblasts may be differentiated into specific myoctes or muscle cells by culturing the myoblasts in complete medium or in minimal media (e.g. complete medium less the fetal calf serum) supplemented with muscle specific growth or differentiation factors such as PGFat concentrations ranging from 24 pg/ml to 28 pg/ml, and insulin from 10−6 M to 10−5 M. To more closely mimic in vivo muscle cells, which are normally innervated by neuronal cells, the culture medium may also be supplemented with appropriate neurotransmitters such as acetylcholine.
  • EXAMPLE III
  • Alternatively, myoblasts may be enriched from toti-potent embryonic stem cells. Toti-potent cells may be derived from in vitro fertilized eggs of an animal using in vitro fertilization techniques, from stem cells present in umbilical cords or placenta, or from Embryonic Stem (ES) cells isolated from cells at the blastocyst stage. ES cells, for example, may be collected, gently dissociated by trypsin, and cultured in vitro with recombinant leukemia inhibitory factor (Chemicon, San Diego, Calif.) and feeder cells such as growth arrested embryonic fibroblasts cells. These toti-potent cells may be treated with growth factors such as PGF or IGF-1 to induce the cells to differentiate into myoblasts.
  • EXAMPLE IV
  • Using standard immunohistochemistry or in-situ hybridization techniques, myoblasts or myocytes (differentiated muscle cells) may be identified. Briefly, myoblasts or myocytes grown in culture may be transferred into glass slides coated with appropriate extracellular matrix as described above. These cells may be grown to the desired number and differentiation using the conditions described above. After a sufficient growth and differentiation period, the cells may be fixed with 4% formaldehyde. If intracellular antibody markers or nucleotide probes are to be used, the cell membranes may be permeabilized with 1% NP-40 or Triton-X. Antibodies against markers specific for myoblasts or myocytes such as myosin, titin, alpha-actinin available from Sigma® may be used to identify the cells using standard fluorescent immunohistochemistry techniques. Alternatively, single stranded RNA or DNA probes for these markers may also be used for in-situ hybridization.
  • In addition, when the muscle cells have been attached to a three dimensional support structure as disclosed below, they may be cryo-frozen, sectioned and identified using antibody markers such as antibodies against myosin, titin, 12101, troponin T, alpha actinin available from Sigma®.
  • EXAMPLE V
  • Two or three dimensional scaffolds or supports may be sculpted from natural biomaterials (e.g. collagen, fibronectin, laminin, or other extracellular matrix) or synthetic biomaterials (e.g. hydroxyapatite, alginate, polyglycolic acid, polylactic acid, and their copolymers), or both. Preferably, the three dimensional scaffolds are sculpted with branch pathways for nutrients and culture media to reach the internal mass of the forming muscle tissues. Examples of materials and construction methods for these scaffolds are provided by U.S. Pat. Nos. 5,686,091, entitled “Biodegradable Foams For Cell Transplantation”; 5,863,984, entitled “Biostable Porous Material Comprising Composite Biopolymers”; 5,770,417, entitled “Three-Dimensional Fibrous Scaffold Containing Attached Cells for Producing Vascularized Tissue in vivo;” and 5,916,265, entitled “Method of Producing a Biological Extracellular Matrix for Use as a Cell Seeding Scaffold and Implant.” These patents are hereby incorporated by reference as if fully set forth herein.
  • The support structure is preferably sculpted to different sizes, shapes, and forms to allow for growth of muscle tissues resembling different types of meat products such as steak, tenderloin, shank, chicken breast, drumstick, lamb chops, fish fillet, lobster tail, etc.
  • EXAMPLE VI
  • Adipocytes, chondrocytes, and osteoblasts are all capable of differentiating from pluri-potent mesenchymal stem cells or toti-potent embryonic stem cells. The stem cells may be isolated as described in Example I or III. The stem cells may be cultured in DMEM, or Ham's F-12, or both in a 1:1 ratio. The medium may be supplemented with thyroid hormone, transferrin, insulin, as well as other growth factors, such as insulin-like growth factor (IGF), basic fibroblast growth factor, and growth hormone.
  • For adipocytes, differentiation may be achieved by treating the stem cells with bone morphogenetic proteins (“BMP”) such as BMP-4 and BMP-2, which are known to induce commitment to the adipocyte lineage. Ahrens et. al., Expression of human bone morphogenetic proteins-2 or -4 in murine mesenchymal progenitor C3H10T1/2 cells induces differentiation into distinct mesenchymal cell lineages, DNA Cell Biol., 12:871-880 (1993); Wang et. al., Bone Morphogenetic protein-2 causes commitment and differentiation in C3H10T1/2 and 3T3 cells, Growth Factors 9:57 (1993). These references are hereby incorporated by reference as if fully set forth herein.
  • In addition to BMPs, the differentiation of adipocytes may be enhanced with agonist of peroxisome proliferator-activated receptor gamma (“PPAR gamma”) such as BRL 49653 (rosiglitazone). Sottile and Seuwen, Bone morphogenetic protein-2 stimulates adipogenic differentiation of mesenchymal precursor cells in synergy with BRL 49653 9 (rosiglitzaone), FEBS Lett, 475(3):201-204 (2000). This reference is hereby incorporated by reference as if fully set forth herein.
  • In certain situations, myoblasts may even be induced to trans-differentiate into adipoblasts (adipocyte precursors) by treating myoblasts cells or muscle satellite cells with long-chain fatty acids (“LCFA”) or thiazolidinediones, or both. Grimaldi et. al., Trans-differentiation of myoblasts to adipoblasts: triggering effects of fatty acids and thiazolidinediones, Prostaglandins Leukot Essent Fatty Acids, 57(1):71-75 (1997); Teboul et. al., Thiazolidinediones and fatty acids convert myogenic cells into adipose-like cells, J. Biol. Chem. 270(47):28183-28187 (1995). These references are hereby incorporated by reference as if fully set forth herein.
  • Thus, meat products with the desired amount of fat content may be produced by seeding and co-culturing muscle cells and adipocyte cells at a certain ratio. Alternatively, stem cells may be allowed to differentiate initially into myoblasts and then at a later time, LCFA or thiadolidinediones may be added at different concentrations and different exposure times to trans-differentiate the myoblasts into adipocytes as desired. Furthermore, the growth of muscle cells and fat cells may be regulated by controlling the concentration of the growth and differentiation factors. For example, if less fat cells are desired in the final meat product, lesser concentrations of BMP factors may be added to the culture while a higher concentration of PGFand/or insulin may be added to promote muscle cell growth.
  • EXAMPLE VII
  • Chondrocytes or cartilage cells may also be isolated from an animal's knee or rib cages. Using similar techniques as described in Example I, dissected tissue from the knee or rib cages may be minced, digested with collagenase, and washed with complete medium. The cells may then be differentially plated to increase the purity of chondrocyte cells.
  • It is known that chondrocytes differentiate in response to mechanical stress. Thus, preferably, the cells may be subjected to shear flow stress as described in U.S. Pat. No. 5,928,945, entitled “Application of Shear Flow Stress to Chondrocytes or Chondrocyte Stem Cells to Produce Cartilage,” which is hereby incorporated by reference as if fully set forth herein.
  • Chondrocytes may initially form a first layer of support cells in a three-dimensional scaffold. Myoblasts or adipocyte cells, or both, may then be seeded onto the chondrocyte layer and grown to the desired size. As such, the chondrocyte layer may provide additional adhesion or growth factors to the muscle cells.
  • EXAMPLE VIII
  • Muscle cells grown in vitro differ from muscle cells grown in vivo in that in vivo cells are used during exercise or body movements. As muscles are used in vivo, muscle cells, in limbs for example, contract and relax in accordance with the movement of the limbs. Hence, to more closely mimic the growth of muscle cells in vivo, the cells grown in vitro may be exposed to an electric or oscillating current, or pulses of electric or oscillating current to contract the muscle cells. Electric probes may be immersed into the culture media to deliver mild current. Alternatively, the support structure may be coated with electrically conducting materials. Examples of electrically conducting materials and a method for coating them onto the support structure are described in U.S. Pat. No. 5,843,741, entitled “Method for Altering the Differentiation of Anchorage Dependent Cells on an Electrically Conducting Polymer,” which is hereby incorporated by reference as if fully set forth herein.
  • The preceding examples illustrate the procedures for producing meat products ex vivo. They are intended only as examples and are not intended to limit the invention to these examples. It is understood that modifying and combining the examples above do not depart from the spirit of the invention.

Claims (23)

1. A non-human meat product for consumption comprising non-human muscle cells grown ex vivo.
2. The non-human meat product in claim 1 further comprising:
a support structure; and
wherein the non-human muscle cells are attached to the support structure.
3. The non-human meat product in claim 1 wherein the non-human muscle cells are skeletal muscle cells.
4. The non-human meat product in claim 1 wherein the non-human muscle cells are derived from animals selected from the group consisting of mammals, birds, fishes, invertebrates, reptiles, and amphibians.
5. The non-human meat product in claim 1 wherein the non-human meat product is substantially free from harmful microbial contamination.
6. The non-human meat product in claim 1 wherein the non-human muscle cells are derived from pluri-potent or toti-potent cells.
7. The non-human meat product in claim 1 wherein the non-human muscle cells have been exposed to an electric current.
8. The non-human meat product in claim 1 further comprising non-human adipocyte cells grown ex vivo.
9. The non-human meat product in claim 8 wherein the non-human adipocyte cells are trans-differentiated from non-human myoblasts.
10. The non-human meat product in claim 8 wherein the non-human adipocyte cells are derived from pluri-potent or toti-potent non-human stem cells.
11. The non-human meat product in claim 1 further comprising non-human cartilage cells grown ex vivo.
12. The non-human meat product in claim 10 wherein the non-human cartilage cells are positioned between a support structure and the non-human muscle cells.
13. The non-human meat product in claim 10 wherein the non-human cartilage cells have been exposed to mechanical stress.
14. A method of producing non-human meat products for consumption comprising the steps:
culturing non-human muscle stem cells ex vivo;
seeding the non-human muscle stem cells onto a support structure; and
growing the non-human muscle stem cells to produce a non-human meat product.
15. The method in claim 13 wherein the step of growing the non-human muscle stem cells comprises:
differentiating the non-human muscle stem cells into different types of non-human muscle cells.
16. The method in claim 14 further comprising the step:
exposing the non-human muscle cells to an electric or oscillating current.
17. The method in claim 13 further comprising the step:
adding nutrients to be incorporated into the non-human meat products.
18. The method in claim 13 wherein the non-human muscle cells are derived from animals selected from the group consisting of mammmals, birds fishes, invertebrates, reptiles, and amphibians.
19. The method in claim 13 wherein the non-human meat product is substantially free from harmful microbial contamination.
20. A method of producing non-human meat for consumption comprising the steps:
co-culturing non-human muscle cells and non-human fat cells ex vivo;
seeding the non-human muscle cells and the non-human fat cells to a support structure; and growing the non-human muscle cells and the non-human fat cells to produce a non-human meat product.
21. A method of producing non-human meat for consumption comprising the steps of:
culturing non-human muscle stem cells ex vivo;
seeding the non-human muscle stem cells to a support structure;
treating the non-human muscle stem cells with fatty acids to trans-differentiate the non-human muscle stem cells into adipocytes; and
growing the adipocytes to produce a non-human meat product.
22. A method of producing non-human meat products for consumption comprising the steps:
culturing non-human cartilage cells ex vivo;
seeding the non-human cartilage cells to a support structure;
culturing non-human muscle cells together with the non-human cartilage cells on or around the support structure; and
growing the non-human muscle cells to produce a non-human meat product.
23. The method in claim 20 wherein the non-human cartilage cells have been exposed to mechanical stress.
US10/943,489 2000-11-17 2004-09-17 Tissue engineered meat for consumption and a method for producing tissue engineered meat for consumption Abandoned US20050084958A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US10/943,489 US20050084958A1 (en) 2000-11-17 2004-09-17 Tissue engineered meat for consumption and a method for producing tissue engineered meat for consumption

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US24999300P 2000-11-17 2000-11-17
US09/991,544 US6835390B1 (en) 2000-11-17 2001-11-16 Method for producing tissue engineered meat for consumption
US10/943,489 US20050084958A1 (en) 2000-11-17 2004-09-17 Tissue engineered meat for consumption and a method for producing tissue engineered meat for consumption

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US09/991,544 Continuation US6835390B1 (en) 2000-11-17 2001-11-16 Method for producing tissue engineered meat for consumption

Publications (1)

Publication Number Publication Date
US20050084958A1 true US20050084958A1 (en) 2005-04-21

Family

ID=33518749

Family Applications (2)

Application Number Title Priority Date Filing Date
US09/991,544 Expired - Lifetime US6835390B1 (en) 2000-11-17 2001-11-16 Method for producing tissue engineered meat for consumption
US10/943,489 Abandoned US20050084958A1 (en) 2000-11-17 2004-09-17 Tissue engineered meat for consumption and a method for producing tissue engineered meat for consumption

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US09/991,544 Expired - Lifetime US6835390B1 (en) 2000-11-17 2001-11-16 Method for producing tissue engineered meat for consumption

Country Status (1)

Country Link
US (2) US6835390B1 (en)

Cited By (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20060029922A1 (en) * 1997-12-18 2006-02-09 Van Eelen Willem F Industrial production of meat
US20130029008A1 (en) * 2011-07-26 2013-01-31 The Curators Of The University Of Missouri Engineered comestible meat
WO2015038988A1 (en) * 2013-09-13 2015-03-19 Modern Meadow, Inc. Edible and animal-product-free microcarriers for engineered meat
WO2020100143A1 (en) * 2018-11-15 2020-05-22 Aleph Farms Ltd. High quality cultured meat, compositions and methods for producing same
US11147300B2 (en) 2019-11-20 2021-10-19 Upside Foods, Inc. Apparatuses and systems for preparing a meat product
CN114072494A (en) * 2019-05-28 2022-02-18 阿普赛德食品公司 Apparatus and method for preparing edible meat products
EP3918117A4 (en) * 2019-02-01 2022-11-16 Nanofiber Solutions, LLC Electrospun polymer fibers for cultured meat production
US11576927B2 (en) 2018-12-11 2023-02-14 Nanofiber Solutions, Llc Methods of treating chronic wounds using electrospun fibers
US11737990B2 (en) 2012-01-12 2023-08-29 Nfs Ip Holdings, Llc Nanofiber scaffolds for biological structures
WO2023164746A1 (en) * 2022-03-03 2023-09-07 Vow Group Pty Ltd Cell enrichment

Families Citing this family (25)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6835390B1 (en) * 2000-11-17 2004-12-28 Jon Vein Method for producing tissue engineered meat for consumption
US20060121006A1 (en) * 2004-09-10 2006-06-08 Chancellor Michael B Production of nutritional and therapeutic products from cultured animal cells
JP4279233B2 (en) * 2004-10-25 2009-06-17 国立大学法人広島大学 Sheet for inducing mesenchymal tissue regeneration and method for producing the same
US20110091604A1 (en) * 2009-10-21 2011-04-21 Seth Adrian Miller Synthetic meat
EP3459558B1 (en) 2010-06-25 2020-07-29 Aston University Glycoproteins having lipid mobilizing properties and therapeutic uses thereof
WO2012024072A1 (en) * 2010-08-19 2012-02-23 Allergan, Inc. Compositions comprising adipose tissue and a pge2 analogue and their use in the treatment of a soft tissue condition
JP6728049B2 (en) 2013-10-30 2020-07-22 ザ キュレイターズ オブ ザ ユニバーシティ オブ ミズーリ Expandable skeletal muscle lineage formation and culture methods
CA2938156C (en) * 2014-02-05 2022-05-10 Modern Meadow, Inc. Dried food products formed from cultured muscle cells
ES2842501T5 (en) 2015-09-21 2023-04-13 Modern Meadow Inc Fiber Reinforced Fabric Composite Materials
KR20170096093A (en) 2016-02-15 2017-08-23 브렌던 패트릭 퍼셀 Composite biofabricated material
CN106282093A (en) * 2016-10-08 2017-01-04 上海生乐康生物技术发展有限公司 A kind of production method of edible cell
EP3609344A1 (en) 2017-04-09 2020-02-19 Supermeat the Essence of Meat Ltd. Cultured meat-containing hybrid food
CA3066060A1 (en) * 2017-06-07 2018-12-13 Wild Type, Inc. Ex vivo meat production
WO2019014652A1 (en) 2017-07-13 2019-01-17 Memphis Meats, Inc. Compositions and methods for increasing the efficiency of cell cultures used for food production
AU2018253595A1 (en) 2017-11-13 2019-05-30 Modern Meadow, Inc. Biofabricated leather articles having zonal properties
US20220025334A1 (en) * 2018-12-12 2022-01-27 Wild Type, Inc. Synthetic food compositions
EP3704202A4 (en) 2019-01-17 2020-12-16 Modern Meadow, Inc. Layered collagen materials and methods of making the same
GB2603289A (en) * 2019-05-21 2022-08-03 Univ Kent State Ohio Animal cell lines for foods containing cultured animal cells
CN115867149A (en) 2020-07-02 2023-03-28 莱托生物股份有限公司 Microbial control of novel food substances
WO2022164858A1 (en) * 2021-01-26 2022-08-04 The Regents Of The University Of California Method for producing food products using cells grown in culture as an alternative to animal husbandry
WO2022189505A1 (en) 2021-03-09 2022-09-15 Ants Innovate Pte. Ltd. Scalable methods for manufacturing alternative meat cuts
WO2022192441A1 (en) * 2021-03-10 2022-09-15 Terasaki Institute For Biomedical Innovation Systems and methods of producing fat tissue for cell-based meat products
WO2023067595A1 (en) 2021-10-18 2023-04-27 Supermeat The Essence Of Meat Ltd. Methods for preparing a food ingredient and compositions produced thereby
WO2023069991A1 (en) 2021-10-19 2023-04-27 Eat Scifi Inc. Plant base/animal cell hybrid meat substitute
WO2023232677A1 (en) 2022-05-31 2023-12-07 Lactobio A/S Strains, compositions and methods of use

Citations (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5686091A (en) * 1994-03-28 1997-11-11 The Johns Hopkins University School Of Medicine Biodegradable foams for cell transplantation
US5746649A (en) * 1996-07-03 1998-05-05 Kraft Foods, Inc. Desinewing of boneless trim
US5770417A (en) * 1986-11-20 1998-06-23 Massachusetts Institute Of Technology Children's Medical Center Corporation Three-dimensional fibrous scaffold containing attached cells for producing vascularized tissue in vivo
US5843741A (en) * 1994-08-01 1998-12-01 Massachusetts Insitute Of Technology Method for altering the differentiation of anchorage dependent cells on an electrically conducting polymer
US5863531A (en) * 1986-04-18 1999-01-26 Advanced Tissue Sciences, Inc. In vitro preparation of tubular tissue structures by stromal cell culture on a three-dimensional framework
US5863984A (en) * 1995-12-01 1999-01-26 Universite Laval, Cite Universitaire Biostable porous material comprising composite biopolymers
US5916265A (en) * 1994-03-30 1999-06-29 Hu; Jie Method of producing a biological extracellular matrix for use as a cell seeding scaffold and implant
US5928945A (en) * 1996-11-20 1999-07-27 Advanced Tissue Sciences, Inc. Application of shear flow stress to chondrocytes or chondrocyte stem cells to produce cartilage
US6348069B1 (en) * 1995-05-19 2002-02-19 Children's Medical Center Corporation Engineering of strong, pliable tissues
US6379962B1 (en) * 1997-11-14 2002-04-30 Bonetec Corporation Polymer scaffold having microporous polymer struts defining interconnected macropores
US6592623B1 (en) * 1999-08-31 2003-07-15 Virginia Commonwealth University Intellectual Property Foundation Engineered muscle
US6709860B1 (en) * 1997-05-26 2004-03-23 Biovitrum Ab Animal model
US6835390B1 (en) * 2000-11-17 2004-12-28 Jon Vein Method for producing tissue engineered meat for consumption

Patent Citations (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5863531A (en) * 1986-04-18 1999-01-26 Advanced Tissue Sciences, Inc. In vitro preparation of tubular tissue structures by stromal cell culture on a three-dimensional framework
US5770417A (en) * 1986-11-20 1998-06-23 Massachusetts Institute Of Technology Children's Medical Center Corporation Three-dimensional fibrous scaffold containing attached cells for producing vascularized tissue in vivo
US5686091A (en) * 1994-03-28 1997-11-11 The Johns Hopkins University School Of Medicine Biodegradable foams for cell transplantation
US5916265A (en) * 1994-03-30 1999-06-29 Hu; Jie Method of producing a biological extracellular matrix for use as a cell seeding scaffold and implant
US5843741A (en) * 1994-08-01 1998-12-01 Massachusetts Insitute Of Technology Method for altering the differentiation of anchorage dependent cells on an electrically conducting polymer
US6348069B1 (en) * 1995-05-19 2002-02-19 Children's Medical Center Corporation Engineering of strong, pliable tissues
US5863984A (en) * 1995-12-01 1999-01-26 Universite Laval, Cite Universitaire Biostable porous material comprising composite biopolymers
US5746649A (en) * 1996-07-03 1998-05-05 Kraft Foods, Inc. Desinewing of boneless trim
US5928945A (en) * 1996-11-20 1999-07-27 Advanced Tissue Sciences, Inc. Application of shear flow stress to chondrocytes or chondrocyte stem cells to produce cartilage
US6709860B1 (en) * 1997-05-26 2004-03-23 Biovitrum Ab Animal model
US6379962B1 (en) * 1997-11-14 2002-04-30 Bonetec Corporation Polymer scaffold having microporous polymer struts defining interconnected macropores
US6592623B1 (en) * 1999-08-31 2003-07-15 Virginia Commonwealth University Intellectual Property Foundation Engineered muscle
US6835390B1 (en) * 2000-11-17 2004-12-28 Jon Vein Method for producing tissue engineered meat for consumption

Cited By (24)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20060029922A1 (en) * 1997-12-18 2006-02-09 Van Eelen Willem F Industrial production of meat
US7270829B2 (en) 1997-12-18 2007-09-18 Willem Frederik Van Eelen Industrial production of meat using cell culture methods
CN103747693A (en) * 2011-07-26 2014-04-23 密苏里大学董事会 Engineered comestible meat
WO2013016547A2 (en) 2011-07-26 2013-01-31 The Curators Of The University Of Missouri Engineered comestible meat
WO2013016547A3 (en) * 2011-07-26 2013-05-10 The Curators Of The University Of Missouri Engineered comestible meat
US8703216B2 (en) * 2011-07-26 2014-04-22 The Curators Of The University Of Missouri Engineered comestible meat
US20130029008A1 (en) * 2011-07-26 2013-01-31 The Curators Of The University Of Missouri Engineered comestible meat
EP2736357A4 (en) * 2011-07-26 2015-05-06 Univ Missouri Engineered comestible meat
US11707077B2 (en) 2011-07-26 2023-07-25 The Curators Of The University Of Missouri Engineered comestible meat
US11737990B2 (en) 2012-01-12 2023-08-29 Nfs Ip Holdings, Llc Nanofiber scaffolds for biological structures
WO2015038988A1 (en) * 2013-09-13 2015-03-19 Modern Meadow, Inc. Edible and animal-product-free microcarriers for engineered meat
WO2020100143A1 (en) * 2018-11-15 2020-05-22 Aleph Farms Ltd. High quality cultured meat, compositions and methods for producing same
CN113038841A (en) * 2018-11-15 2021-06-25 阿利夫农场公司 High-quality cultured meat, composition, and method for producing high-quality cultured meat, composition
US11576927B2 (en) 2018-12-11 2023-02-14 Nanofiber Solutions, Llc Methods of treating chronic wounds using electrospun fibers
EP3918117A4 (en) * 2019-02-01 2022-11-16 Nanofiber Solutions, LLC Electrospun polymer fibers for cultured meat production
US11357244B2 (en) * 2019-05-28 2022-06-14 Upside Foods, Inc. Apparatuses and methods for preparing a comestible meat product
US11559073B2 (en) 2019-05-28 2023-01-24 Upside Foods, Inc. Substrates for preparing a comestible meat product
US11559072B2 (en) 2019-05-28 2023-01-24 Upside Foods, Inc. Methods for preparing a comestible meat product
CN114072494A (en) * 2019-05-28 2022-02-18 阿普赛德食品公司 Apparatus and method for preparing edible meat products
US11344050B2 (en) 2019-11-20 2022-05-31 Upside Foods, Inc. Apparatuses and systems for preparing a meat product
US11576411B2 (en) 2019-11-20 2023-02-14 Upside Foods, Inc. Apparatuses and systems for preparing a meat product
US11627751B2 (en) 2019-11-20 2023-04-18 Upside Foods, Inc. Apparatuses and systems for preparing a meat product
US11147300B2 (en) 2019-11-20 2021-10-19 Upside Foods, Inc. Apparatuses and systems for preparing a meat product
WO2023164746A1 (en) * 2022-03-03 2023-09-07 Vow Group Pty Ltd Cell enrichment

Also Published As

Publication number Publication date
US6835390B1 (en) 2004-12-28
US20050010965A1 (en) 2005-01-13

Similar Documents

Publication Publication Date Title
US6835390B1 (en) Method for producing tissue engineered meat for consumption
IL181859A (en) Non-human meat product for tissue consumption comprising non-human cells grown ex-vivo and method of production
US7270829B2 (en) Industrial production of meat using cell culture methods
Bhat et al. In vitro meat production: Challenges and benefits over conventional meat production
AU2015214092B2 (en) Dried food products formed from cultured muscle cells
US20200140821A1 (en) Ex vivo meat production
US20230115567A1 (en) Cultured meat-containing hybrid food
Bhat et al. Tissue engineered meat-future meat
AU2018214029A1 (en) Engineered Comestible Meat
Bhat et al. Prospects for in vitro cultured meat–a future harvest
KR20200071061A (en) Cultured meat composition
US20110301249A1 (en) Stem cell enhanced protein products and uses therof
US20230151330A1 (en) Systems and methods for cell conversion
US20210348129A1 (en) Non-decellularized plant leaf cultures for meat
CN101132803A (en) Tissue engineering edible meat and method of manufacturing the same
NZ554226A (en) Tissue engineered meat for consumption and a method for producing tissue engineered meat for consumption
KR102636615B1 (en) A method for preparing non-human cell line, the cell line prepared accordingly, and a method for culturing the cell line
RU2778255C2 (en) Compositions out of cultured meat
CN117004555A (en) Preparation method of cell culture meat
CN117795058A (en) Production of cell-based products for human consumption

Legal Events

Date Code Title Description
STCB Information on status: application discontinuation

Free format text: ABANDONED -- AFTER EXAMINER'S ANSWER OR BOARD OF APPEALS DECISION