DOI QR코드

DOI QR Code

Current Research, Industrialization Status, and Future Perspective of Cultured Meat

  • Seung Hyeon Yun (Department of Animal Science and Technology, Chung-Ang University) ;
  • Da Young Lee (Department of Animal Science and Technology, Chung-Ang University) ;
  • Juhyun Lee (Department of Animal Science and Technology, Chung-Ang University) ;
  • Ermie Mariano Jr (Department of Animal Science and Technology, Chung-Ang University) ;
  • Yeongwoo Choi (Department of Animal Science and Technology, Chung-Ang University) ;
  • Jinmo Park (Department of Animal Science and Technology, Chung-Ang University) ;
  • Dahee Han (Department of Animal Science and Technology, Chung-Ang University) ;
  • Jin Soo Kim (Department of Animal Science and Technology, Chung-Ang University) ;
  • Sun Jin Hur (Department of Animal Science and Technology, Chung-Ang University)
  • Received : 2023.12.15
  • Accepted : 2024.01.30
  • Published : 2024.03.01

Abstract

Expectations for the industrialization of cultured meat are growing due to the increasing support from various sectors, such as the food industry, animal welfare organizations, and consumers, particularly vegetarians, but the progress of industrialization is slower than initially reported. This review analyzes the main issues concerning the industrialization of cultured meat, examines research and media reports on the development of cultured meat to date, and presents the current technology, industrialization level, and prospects for cultured meat. Currently, over 30 countries have companies industrializing cultured meat, and around 200 companies that are developing or industrializing cultured meat have been surveyed globally. By country, the United States has over 50 companies, accounting for more than 20% of the total. Acquiring animal cells, developing cell lines, improving cell proliferation, improving the efficiency of cell differentiation and muscle production, or developing cell culture media, including serum-free media, are the major research themes related to the development of cultured meat. In contrast, the development of devices, such as bioreactors, which are crucial in enabling large-scale production, is relatively understudied, and few of the many companies invested in the development of cultured meat have presented products for sale other than prototypes. In addition, because most information on key technologies is not publicly available, it is not possible to determine the level of technology in the companies, and it is surmised that the technology of cultured meat-related startups is not high. Therefore, further research and development are needed to promote the full-scale industrialization of cultured meat.

Keywords

Introduction

Cultured meat, also called in vitro meat or laboratory-cultured meat, is an edible tissue produced by the isolation, proliferation, and differentiation of muscle satellite cells (MSCs) obtained from a small amount of livestock tissue (Lee et al., 2021). The production of livestock products based on stem cell and tissue culture technologies is seen as a future technology and an emerging industry that is not only resource-efficient but can effectively address environmental degradation and the uncertainties associated with food security in the face of a growing global population and dwindling natural resources (Risner et al., 2023). Several countries around the world have implemented or taken steps to create policies to categorize cultured meat as cellular agriculture (Soice and Johnston, 2021).

A report by GlobeNewswire (2023) ascertained that the estimated value of the global cultured meat market was USD 182 million in 2022 and will continue to grow, with a projected CAGR of 23.2%. Currently, the only marketable cultured meats that have been officially certified as safe by the United States Food and Drug Administration (FDA) are cell-cultured chicken from Upside Foods and GOOD Meat.

Following this official approval, several companies worldwide are seeking permission to sell cultured meat. In July 2023, an Israel-based company, Aleph Farms, submitted a regulatory approval application to the Swiss Federal Office for Food Safety and Veterinary Medicine (Aleph Farms, 2023). Subsequently, in January 2024, Israel’s Ministry of Health (MoH) approved the sale of cultured beef from Aleph Farms, making it the third country to offer cultured meat for sale and the first approval for a bovine species (Aleph Farms, 2024). In October 2023, CellMEAT requested the Ministry of Food and Drug Safety (MFDS) of the Republic of Korea certification for Dokdo shrimp (Lebbeus groenlandicus) cell culture as a temporary food ingredient (CellMEAT, 2023). In December 2023, Food Standards Australia New Zealand (FSANZ) announced new amendments to an application received from Vow seeking approval of cultured quail (FSANZ, 2023). Likewise, research is underway around the world to produce cultured beef, pork, lamb, turkey, foie gras, and various types of seafood (oysters, lobster, shrimp, salmon, and tuna) using cell culture technology, and the development of various materials and equipment for cultured meat production, including adipocytes, supports, microcarriers, growth factors, and bioreactors, is gaining traction.

Despite expectations, the full-scale industrialization of cultured meat has not yet been achieved, and the timing of the industrialization of cultured meat remains unclear. In addition, the terminology for cultured meat has not yet been standardized. The Food and Agriculture Organization (FAO) and the World Health Organization (WHO) use the term ‘cell-based food,’ the United States Department of Agriculture-Food Safety and Inspection Service (USDA-FSIS) uses ‘cell-cultured meat,’ and the U.S. Food and Drug Administration (FDA) uses ‘cultured animal cell material’ (e.g., cultured Gallus gallus cell material; FAO and WHO, 2023; FDA, 2023a; USDA and FSIS, 2023).

Therefore, this review analyzes the main issues related to the industrialization of cultured meat, as well as research reports and media reports on the development of cultured meat to date, with the aim to present the current technology, industrialization level, and prospects of cultured meat.

Cultured Meat and Food Safety

Cultured meat production facilities are considered to be safer than conventional meat production facilities against foodborne pathogens, such as Salmonella, Campylobacter, Escherichia coli, yeasts, molds, and parasites because they are designed with enclosed structures that can control the entry of external substances (Chriki and Hocquette, 2020). However, potential threats from the cultured meat production process cannot be completely ruled out. Among the anticipated food safety concerns are contamination with microorganisms and prion proteins that may occur during the cell culture phase, residues of antibiotics and cell freezing agents, the safety of cell lines (genetic manipulation and excessive passage culture), exogenous recombinant growth factors, unknown allergens, and the safety of support materials (Broucke et al., 2023; Ong et al., 2021).

Furthermore, it is crucial to adhere to the guidelines of food authorities, such as the FDA, when using a scaffold for the production of cultured meat. This includes following regulations regarding the use of materials, solvents, cross-linking agents, inedible substances, toxic compounds, allergens, and other related factors (Levi et al., 2022). However, challenges remain in the commercialization of scaffolds due to the need to establish safety evaluation and approval standards for solvents or cross-linking agents used in scaffold polymerization, potential decomposition by-products of biodegradable scaffolds, physicochemical modifications of synthetic polymer scaffolds, and recombinant proteins that improve cell attachment efficiency (Bomkamp et al., 2022).

In response to these concerns, several countries, such as those in Australasia and the European Union (EU), Korea, Singapore, the United Kingdom, and the United States, have taken steps toward establishing regulations and classification guidelines for cell-based foods or temporarily allowing them as food ingredients (EU, 2021; FDA, 2019; FDA, 2023b; FSA, 2023; FSANZ, 2023; MFDS, 2023; SFA, 2023; USDA and FSIS, 2023). These regulations are overseen by national agencies in each country (Table 1).

Table 1. Countries with established regulations and classification guidelines for cell-based foods

CSSPBQ_2024_v44n2_326_t0001.png 이미지

As there are many threats to the safety of cultured meat, it is essential to establish a “standard safety assessment procedure for cultured meat” that includes not only cell-cultured chicken but also other major livestock species, such as beef and pork, or cell-cultured seafood, to ensure the safety certification and commercialization of cultured meat (Ong et al., 2021). Furthermore, potential threats in cultured meat that are not yet well understood need to be further investigated, and the safe production of cultured meat should be based on the use of validated food ingredients in the product production process and appropriate regulations (Bhat et al., 2015). Safe consumption of the product is a prerequisite for cultured meat to be licensed and marketed as a new food, which requires standardization of the manufacturing process or the development of manufacturing guidelines (Mariano et al., 2023). It may also be necessary to evaluate the safety of the final product manufactured according to the standardized process or manufacturing guidelines, which can be done using methods similar to those used to evaluate new foods for authorization for human consumption (Lee et al., 2023c). In general, the safe consumption of food is assessed by short-term and long-term toxicity tests in laboratory animals.

Toxicity tests used to assess food for human consumption analyze the genotoxicity, reproductive toxicity, hematotoxicity, hepatotoxicity, or allergenicity in comparison to existing products. In assessing the safety of cultured meat for consumption, it may be necessary to standardize or set guidelines for the following five processes:

- Cell acquisition

- Cell culture preparation

- Cell culture and muscle differentiation

- Cultured muscle acquisition

- Manufacturing meat products using cultured muscle

Sustainability and Animal Welfare

According to the United Nations’ World Population Prospects 2022 report (UN, 2022), the world’s population is expected to reach 9.7 billion by 2050 from 8 billion in 2022, and a joint report prepared by the Organization for Economic Co-operation and Development (OECD) and the FAO (OECD and FAO, 2022) predicts that global meat consumption will increase by 15% by 2031 to keep pace with projected population growth. As a result, more land for growing feed is needed to keep up with the trend of increasing meat consumption.

The global livestock industry has drawn increased attention in recent years because of the magnitude of its environmental impact. Greenhouse gases from livestock production are estimated to be 14.5% of global greenhouse gas emissions, and agricultural water use is reported to be 29% of global water use, 98% of which is used for the production of animal feed (Gerber et al., 2013; Mekonnen and Hoekstra, 2012). The environmental costs of livestock production also include land degradation, eutrophication of lakes and rivers, lower soil fertility, reduced biodiversity, increased exposure to zoonotic diseases, and accumulation of livestock manure, which could contaminate surface and groundwater, and has been shown to contribute to the transmission of zoonotic diseases and antibiotic-resistant bacteria (Godfray et al., 2018; Morand et al., 2019; Xie et al., 2018; Young et al., 2014).

Cultured meat has been reported to involve 78%–96% less greenhouse gas emissions, 99% less land use, 82%–96% less water, and 7%–45% less energy use than conventional meat production methods, depending on the specific meat product being compared and the species involved (Reis et al., 2020; Tuomisto and Teixeira de Mattos, 2011). These data suggest that cultured meat could be a key promotional tool to induce positive consumer perceptions of its environmental benefits and engagement in environmental protection. Pakseresht et al. (2022) reviewed a total of 43 articles and identified environmental and ethical concerns among eight major factors determining the consumer acceptance of cultured meat. However, data quantifying the climate and environmental impacts of cultured meat production is highly speculative, based on forward-looking projections, and actual cultured meat production systems are often hidden due to intense competition, leaving little detailed information available for analysis (Lynch and Pierrehumbert, 2019; Tuomisto, 2018). Therefore, a systematic approach with a larger sample size of cultured meat production technologies needs to be developed to assess the environmental impact of cultured meat.

As global meat consumption is on the rise, the scale of farming and the number of animals slaughtered are expected to increase, and the religious, ethical, and environmental controversies that arise from the slaughter process are likely to become more intense than before (Heidemann et al., 2020). Over the years, continued efforts have been made to improve the efficiency of the livestock industry for mass production, but equally important is prioritizing animal welfare and, accordingly, the movement to improve animal welfare, such as developing standards for animal welfare certification and labeling schemes, is reported to be increasing every year (Anomaly, 2015; Parker et al., 2017). Given that cultured meat would reduce the need for raising livestock for slaughter, it can improve animal welfare concerns (Hocquette, 2016), and studies of consumers suggest that the emotional benefits of cultured meat in terms of animal welfare contribute to positive perceptions of cultured meat (Bryant and Barnett, 2020; Lin-Hi et al., 2023; Rolland et al., 2020). Conversely, some consumers have expressed concerns that cultured meat will affect the demand for industrial animals, leading to a decrease in the number of live animals, which poses a potential threat to traditional livestock farming, ultimately leading to a disruption of the balance between animals and nature (Laestadius and Caldwell, 2015; Newton and Blaustein-Rejto, 2021). In response to these concerns, scenario analysis studies have been conducted on the possibility of cultured meat partially replacing traditional livestock farming, but cultured meat is still considered to be at a technological plateau, requiring extensive research and large capital investments to replace conventional meat production (Mateti et al., 2022; Moritz et al., 2023).

Consumer Perception

Cultured meat producers are emphasizing the benefits of environmental efficiency, sustainability, eco-branding, and environmental costs to win over consumers and are actually creating added value for cultured meat products by reducing the negative environmental impact of product production and providing differentiated products that make consumers feel like they are investing in environmental protection (Reis et al., 2020). In the review by Pakseresht et al. (2022) mentioned above, 43 (17.7%) of 243 screened articles on cultured meat development and technology concerned consumer attitudes, highlighting the scarcity of studies exploring consumers responses to this technology. In a choice experiment using a randomized group of 533 consumers, it was found that taste, health, price, animal welfare, environmental impact, and social impact were the most important factors in determining the purchase of a burger product and that only 11% of consumers would choose a burger made with cultured meat if all burgers had the same price (Slade, 2018). However, when presented with a positive framing of cultured meat, more than 66%–70% of consumers were willing to try or purchase cultured meat, and those who were willing to purchase had a favorable evaluation of cultured meat, citing improvements in environmental and animal welfare as benefits of cultured meat (Bryant and Barnett, 2020; Rolland et al., 2020; Wilks and Phillips, 2017; Zhang et al., 2020). Furthermore, in a system dynamics model study to estimate the demand for cultured meat, the price of the product had the greatest impact on the speed of promotion and purchase decision-making for cultured meat, with low prices showing high demand regardless of the promotion strategy, suggesting the importance of proper pricing in the launch of cultured meat products (Skinner and Blake, 2023).

However, cultured meat is categorized as a novel food that is only available for purchase or tasting in limited quantities in a handful of countries, and all the consumer research published as of November 2023 is based on hypothetical product settings. Additionally, consumer response has been found to be largely consistent, and it is expected that the production of affordable cultured meat to meet consumer satisfaction will be paramount. As alluded to above, another consumer concern regarding the development of the cultured meat industry is that it will negatively impact traditional livestock farmers (Wilks and Phillips, 2017). A survey of the acceptance of alternative meat products among farmers and non-farmers found that both farmers and non-farmers expressed concerns about the impact of cultured meat on traditional livestock farming, with farmers reporting a lower preference for alternative meat products than non-farmers (Crawshaw and Piazza, 2023).

Cultured meat also provokes ethical, cultural, and religious discussions. According to Islamic beliefs, halal means exception in Arabic, and whether cultured meat is halal is a determining factor in Muslims’ acceptance of cultured meat consumption (Hamdan et al., 2018). Muslims in the United Kingdom were less likely to try new foods than non-Muslims due to uncertainty about halal status, but Muslims were found to be more likely to purchase cultured meat than non-Muslims (Boereboom et al., 2022). Muslims in Singapore also considered the safety and halal status of cultured meat before accepting it, and there was a link between food safety and religious acceptance (Ho et al., 2023). To enter the kosher and halal markets, cultivated meat must comply with specific standards and requirements, including those related to its origin and method of production. In September 2023, Orthodox Union Kosher, the world’s largest and most influential kosher certification authority, certified poultry products from SuperMeat as kosher, marking a major advancement for the food technology’s acceptance under Jewish dietary law (Tress, 2023). At the time of writing, Aleph Farms (the first to receive approval for cultured meat for a bovine species) is awaiting a decision on kosher and halal certification of its beef steaks after seeking consultation from several religious authorities.

Therefore, the strategies necessary for consumer acceptance of cultured meat must consider the positions of various sectors, such as government policy, food safety, traditional livestock farming and cultured meat, and religious/cultural/ethical perspectives. Accurate data and research are needed to compare the sustainability of the conventional meat industry and cultured meat industry, not only to highlight the positive aspects of cultured meat but also to consider the coexistence of traditional livestock farming and cultured meat (Bryant and Barnett, 2018). However, market-based information on actual cultured meat technologies is inconsistent, making it difficult to evaluate and analyze, and environmental impact analysis is based on data with higher uncertainty compared to traditional livestock farming (Rodríguez Escobar et al., 2021). In addition, because most consumers’ positive perception and acceptance of cultured meat is based on trust in the government, it is necessary to establish strict standards for food safety (Ho et al., 2023).

In conclusion, to assess the ideal sustainability of cultured meat, bridging the knowledge and information gap is a must, and collaboration between relevant companies and researchers is needed to integrate the entire production process and scenarios so that the environmental impact of cultured meat can be reasonably predicted. Furthermore, the government should take into account the proposed scenarios and establish regulations to enable consumers to choose safe cultured meat.

Domestic and International Cultured Meat Companies

Information on domestic and international cultured meat companies as of 2023 is presented in Table 2, with a total of 195 companies producing food-grade cultured meat-related products in 35 countries. The largest number of cultured meat companies were identified in the United States (53), followed by the United Kingdom (17), Israel (14), Singapore and Canada (11), South Korea (10), Germany (9), the Netherlands and Japan (6), India, France and mainland China (5), South Africa, Argentina, and Australia (4), the Czech Republic (3), Belgium, Switzerland, Spain, Austria, and Chile (2), and other countries (New Zealand, Denmark, Russia, Malaysia, Mexico, Vietnam, Sweden, Iceland, Croatia, Turkey, and Portugal). Furthermore, of the 307 product categories mentioned as being researched by companies, the top 10 categories are meat, beef, fish, pork, chicken, seafood, scaffold, culture media, ingredients, and others, accounting for 79.5% of the total, indicating that current trends in company-level cultured meat research are centered on cultured meat (beef>fish>pork>chicken=seafood), supports, and media (Fig. 1). However, it is necessary to be cautious in identifying trends as there are many cases where the researchers do not clearly mention the animal species under research and refer to it as meat or seafood.

Table 2. Cultured meat-related companies

CSSPBQ_2024_v44n2_326_t0002.png 이미지

CSSPBQ_2024_v44n2_326_t0003.png 이미지

CSSPBQ_2024_v44n2_326_t0004.png 이미지

CSSPBQ_2024_v44n2_326_t0005.png 이미지

CSSPBQ_2024_v44n2_326_t0006.png 이미지

CSSPBQ_2024_v44n2_326_t0007.png 이미지

3D, three-dimensional.

CSSPBQ_2024_v44n2_326_f0001.png 이미지

Fig. 1. Major product trends for cultured meat companies.

Production of Cultured Meat

Muscle satellite cells

MSCs are muscle-derived adult stem cells that are responsible for the regenerative capacity of muscle following damage to myofibers. MSCs are characterized by rapid proliferation in a highly active state early in life, while the proportion entering a quiescent state increases with age (Mesires and Doumit, 2002). Myofibrils are composed of structures surrounded by an inner sarcolemma and an outer basement membrane, and the basal lamina, which is close to the myofibrils, has been identified as an extracellular matrix (ECM) that is in direct contact with MSCs and is involved in the maintenance of physiological functions and the development of skeletal muscle (Holmberg and Durbeej, 2013; Zhang et al., 2021). The basal lamina is composed mainly of type IV collagen, which plays a role in maintaining MSCs in a quiescent state by sequestering various growth factors and signaling molecules involved in their activation and proliferation (Kann et al., 2021; Sanes, 2003). Furthermore, quiescent MSCs located in the niche between the basal lamina and myofibrils have a fusiform morphology with little cytoplasm and organelles and have been shown to express MSC-specific genes, such as paired box protein 3 (Pax3) and Pax7, and myoblast determination protein 1 (MyoD) at the beginning of quiescence or proliferation entry (Fu et al., 2015; Kuang et al., 2006; Zhang et al., 2010).

Gene expression and signaling pathways

Understanding the regeneration process of MSCs is necessary for cultured meat production, and the genes and signal transduction pathways that regulate proliferation and differentiation that have been widely reported to date are shown in Fig. 2. Pax3 is considered one of the important genes responsible for MSC survival during embryogenesis. It is also purported to be involved in the formation and underlying development of early muscles by affecting the expression of MyoD and myogenic factor 5 (Myf5) to regulate the development of limb muscles (MyoD) and peri-spinal and intercostal muscles (Myf5) in early embryos (Kablar et al., 1997). Pax7 is an essential gene for MSC maintenance, and individuals with Pax7 knockout show a decreased rate of muscle regeneration in muscle injury treatments and difficulty in generating MSCs (Kuang et al., 2006). In addition, Pax7 has been found to act as an antagonist of MyoD, resulting in an increased number of Pax7-positive cells in the muscles of individuals with MyoD knockout (Kuang et al., 2006; Olguin and Olwin, 2004; Seale et al., 2000).

CSSPBQ_2024_v44n2_326_f0002.png 이미지

Fig. 2. Gene regulation and signaling pathways in myogenesis.

Activation of MSCs is an early step in myogenesis. When a muscle is damaged, the disruption of the basal plate and reorganization of the environment leads to interactions between signaling molecules that were previously sequestered by the basal plate and MSCs, leading to their activation (Li et al., 2018). Muscle formation is mainly regulated by myogenic regulatory factors (MRFs) expressed in activated MSCs. Some representative MRFs are MyoD, Myf5, myogenin, and muscle-specific regulatory factor 4 (MRF4, also known as Myf6; Kim et al., 2023a). Activated MSCs divide to produce satellite cell-derived myoblasts that continue to divide and proliferate before committing to differentiation and fusing to form myotubes, which then mature into myofibers. When satellite cells are activated, they initiate differential expression of MRFs depending on the asymmetry of cell orientation after division (Kuang et al., 2007). Accordingly, it has been shown that if the orientation of the cells formed after somatic cell division is on the myofibrillar side, they upregulate origin regulatory factors, such as MyoD and Myf5, whereas cells on the basal plate side do not express Myf5 and retain stemness (Kuang et al., 2007; Troy et al., 2012).

MyoD and Myf5 are genes that activate myogenin and MRF4 and participate in the late stages of muscle formation by influencing the fusion of myoblasts and the initiation of their final differentiation, leading to cell maturatiNagahisa on and ultimately the formation of multinucleated myotubes (Cornelison et al., 2000; Hawke and Garry, 2001; Punch et al., 2009; Smith et al., 1993). MyoD has somewhat overlapping roles with myogenin, but when myogenin is deleted, MyoD is unable to take over its role, and individuals with myogenin deletion have been shown to die at birth due to impaired skeletal muscle formation (Adhikari et al., 2021; Nabeshima et al., 1993). It was also found that in C2C12 cultures with myogenin deletion, myomaker and myomixer, two genes that regulate the fusion of skeletal muscle, were significantly downregulated, leading to the inhibition of differentiation (Adhikari et al., 2021). MRF4 is an origin regulator that is predominantly expressed in fully differentiated muscle fibers and plays a role in maintaining the MSC pool. It has been reported that deletion of MRF4 can significantly reduce the number of Pax7-positive MSCs in postnatal individuals (Lazure et al., 2020).

Signals that regulate the stemness of MSCs are known to include p38α/β mitogen-activated protein kinase (MAPK) or Notch. First, inhibition of p38 has been reported to induce self-renewal of MSCs by blocking the MyoD expression pathway and maintaining Pax7 expression along with inhibition of cell cycle entry to sustain an undifferentiated and proliferative state (Ding et al., 2018; Li et al., 2023; Troy et al., 2012). Among Notch signaling components, Notch1 is activated upon muscle injury in vivo by binding to myofilament ligands to induce cell cycle exit, Notch2 is activated in MSCs to maintain the stemness of the MSC population by inhibiting differentiation, and Notch3 has been shown to inhibit the p38α/β MAPK pathway to suppress myocyte enhancer factor 2 (MEF2) expression associated with differentiation (Conboy and Rando, 2002; Gagan et al., 2012; Jo et al., 2022).

It has been reported that activated MSCs are proliferation-induced and differentiation-inhibited by the phosphoinositide 3-kinase (PI3K)/Akt pathway or the extracellular signal-regulated kinase 1/2 (ERK1/2) pathway (Li et al., 2023; Mohammadabadi et al., 2021; Ohashi et al., 2015). Growth factors known to be involved in PI3K/Akt activation include fibroblast growth factor (FGF), insulin-like growth factor (IGF)-1/2, hepatocyte growth factor (HGF)/c-Met, epidermal growth factor (EGF), and interleukin-6/Janus kinase 2/signal transducer and activator of transcription 3 (IL-6/JAK2/STAT3). These factors have been shown to act as activators of mammalian target of rapamycin complex 1 (mTORC1), which can regulate the proliferation of muscle progenitor cells (Brandt et al., 2018; Holterman and Rudnicki, 2005; Lu et al., 2017; Messersmith et al., 2021; Ohashi et al., 2015; Ornitz and Itoh, 2015; Relaix et al., 2021; Rhoads et al., 2016; Wang et al., 2023a). Furthermore, it has been confirmed that EGF and FGF are involved in the ERK1/2 pathway, one of the MAPK family signaling pathways, which can activate myoblast proliferation and impair the initiation and maintenance of differentiation (Li et al., 2023; Mohammadabadi et al., 2021; Ohashi et al., 2015). Additionally, the Wnt pathway can activate both mTORC1/2, with mTORC1 regulating metabolism in response to environmental factors (growth factors, amino acids, energy, and stress) and mTORC2 involved in the maintenance of MSC populations through phosphatase family pathways (Oh and Jacinto, 2011; Rion et al., 2019; Wei et al., 2019).

The p38α/β MAPK pathway activates MEF2 and plays a major role in the differentiation of myoblasts. Myotube formation is inhibited when MEF2 is removed because of the involvement of MEF2 in the proliferation and differentiation of MSCs (Chen et al., 2017; Shao et al., 2022; Wang et al., 2018). Furthermore, when the mTOR pathway is inhibited by rapamycin in MSC cultures, the expression of myogenic genes (Pax7, Myf5, and MyoG) is inhibited, indicating that the mTOR pathway is essential for the proliferation and differentiation of MSCs (Zhang et al., 2015). In addition, previous studies on MSC differentiation have shown that Wnt1 and Wnt7a signaling, along with activation of the Wnt/β-catenin pathway, increases β-catenin to induce myogenic differentiation of mesenchymal stem cells and activate the myogenic regulators Myf5 and MyoD to influence skeletal muscle development (Eng et al., 2013; Zhu et al., 2022). Signals that inhibit differentiation include ERK, myostatin, and protein kinase A (PKA), with myostatin reported to inhibit muscle formation by co-inhibiting the Akt pathway and PKA reported to induce proteolytic cleavage to produce factors that inhibit MEF2 signaling (Backs et al., 2011; Mohammadabadi et al., 2021; Trendelenburg et al., 2009).

The nuclear factor of activated T-cells (NFAT) can activate signaling molecules that regulate the fusion of myoblasts and myotubes, such as MEF2 and IL-4, by the calcineurin and p38/MAPK pathways; however, PKA has been reported to prevent premature differentiation of myoblasts by rephosphorylating MEF2 and NFAT while inhibiting their differentiation (Horsley et al., 2003; Knight and Kothary, 2011; McKinsey et al., 2002; Stork and Schmitt, 2002; Wu et al., 2007; Yue et al., 2023). In addition, it has been shown that mTOR regulates the proliferation of MSCs but can also regulate myotube fusion by both kinase-dependent and -independent pathways (Park and Chen, 2005).

In conclusion, an understanding of the various gene expression and signaling processes within MSCs for cultured meat production is required, and further research is needed to control and regulate cell cycle arrest and activation, proliferation, differentiation, and even fusion.

Obtaining muscle satellite cells

MSCs can be obtained by biopsy of muscle tissue from living animals and by harvesting muscle tissue from animals immediately after slaughter. The most used processes for harvested muscle tissue are disinfection, removal of fat and connective tissue, fragmentation, digestive enzyme treatment, sequential filtration, centrifugation, pre-culture, and finally, cell recovery to obtain primary cells (Lee et al., 2021). The obtained primary cells are then subjected to immunofluorescence staining or polymerase chain reaction to determine the proportion of MSCs from the proportion of progenitor regulatory factors in the primary cells. Typically, Pax7 and MyoD are used to determine the purity of MSCs, and by comparing their expression levels, the activation of the MSCs used in the experiment can be determined (Ding et al., 2017; Kim et al., 2023a; Pasut et al., 2013). In addition, flow cytometry methods, such as fluorescence-activated cell sorting (FACS) and magnetic-activated cell sorting (MACS), can be used to obtain pure MSCs labeled with MSC-specific markers, which can then be proliferated to sufficient quantities for use in cultured meat experiments and production (Ding et al., 2018; Gromova et al., 2015; Kim et al., 2023a; Motohashi et al., 2014).

Culture of muscle satellite cells

The culture of MSCs has been performed since before the 1990s, and the methods can be broadly divided into two types: culture of isolated single strands of muscle fibers and culture of cells isolated from enzymatically treated muscle tissue (Anderson and Pilipowicz, 2002; Bischoff, 1986; Doumit and Merkel, 1992; McFarland et al., 1988). Fetal bovine serum (FBS) is a key ingredient added to the basal medium for culture, but the exact nature of FBS is still poorly understood, and commercialization of cultured meat is currently limited by the need to replace FBS completely (Lee et al., 2022; Lee et al., 2023a). It is difficult to avoid the ethical issues associated with the production of FBS, as more than 2 million bovine fetuses derived from slaughtered mothers are used for FBS production each year (Lee et al., 2022). In addition to ethical concerns, the high price of FBS has led numerous research teams to investigate serum-free media as an alternative to FBS, and along with research to refine the active ingredients of FBS, results support that serum can be effectively replaced with proteins required for cell growth or a combination of various growth factors (Messmer et al., 2022; Schenzle et al., 2022; Skrivergaard et al., 2023; Stout et al., 2022; Stout et al., 2023). Furthermore, to meet halal standards, the use of blood in cultured meat production is also limited (Hamdan et al., 2018). However, challenges remain, such as the use of recombinant growth factors in the preparation of serum-free media or chemically composed media and the cost of expensive additives (Stout et al., 2022; Stout et al., 2023).

Once the medium in which the cells are to be cultured is prepared, the method of culturing the cells must be chosen according to each cell type. Cell culture techniques for cultured meat production can be broadly divided into adherent culture and floating culture, and among the cells, MSCs and fibroblasts have been studied, as well as adipocytes (Bodiou et al., 2020; Ge et al., 2023; Humbird, 2021; Lee et al., 2021). Approximately 1014 cells and 10,000 L of culture medium are required to produce 1 t of cultured meat, assuming a cell density of 107 cells/mL in the bioreactor (Guan et al., 2021). However, the larger the bioreactor size, the higher the stirring intensity needed to maintain a homogeneous environment in the vessel, which can lead to shear stresses of a magnitude that can cause cell damage (Allan et al., 2019). In a modeling study of cultured meat production scenarios, it was emphasized that optimal cell selection to reduce the consumption rate of medium, completely replace or decrease the cost of growth factors, and increase the size of perfusable bioreactors are necessary for mass production environments (Risner et al., 2021).

As such, cultured meat is a tissue engineering technique under investigation based on the theory that the self-renewal ability of MSCs can be harnessed to produce dozens of times the amount of muscle tissue from a small piece of muscle. Cultured meat is one of the promising future technologies that can be used as an important source of meat for some countries because it is less sensitive to climatic conditions than conventional meat production, but there is a need to improve economic issues, such as cell acquisition, mass production and cost, and the amount of culture fluid and energy required for production compared to real meat. Additionally, research is being conducted worldwide to improve the qualitative limitations, such as flavor, texture and structure, meat color, and nutritional content, which are different from those of real meat.

Recent Trends in Muscle Satellite Cell Culture Technologies

Isolation

Bovine MSC isolation techniques for cultured meat production reported in 2023 are shown in Table 3. The goal of the isolation process is to obtain the raw material for cultured meat. The isolation techniques used can be broadly categorized into 1) enzymatic reactions and centrifugation to obtain MSCs and pre-culture and 2) flow cytometry to increase the purity of the MSCs.

Table 3. Isolation methods for bovine muscle satellite cells published in 2023

CSSPBQ_2024_v44n2_326_t0008.png 이미지

N/A, not applicable.

First, an enzymatic reaction is performed to obtain primary cells from muscle tissue. The cells are minced to increase the surface area, and connective tissue is removed to facilitate the reaction. Enzymes used for MSC isolation include collagenase, dispase, trypsin, and pronase in various concentrations. Centrifugation is a method that uses centrifugal force and density gradients to remove unwanted tissue and isolate desired cells. In the isolation process of MSCs, the centrifugal acceleration was 76–1,200×g, and the time was generally around 5–15 min.

The cells obtained by enzymatic reaction and centrifugation are primary cells. Cell pre-plating or purification techniques, such as FACS and MACS, are employed to increase the purity of MSCs. Pre-culture is a technique for isolating specific cells from a mixture of different cell types, effectively increasing the purity of MSCs by exploiting differences in the adhesion properties of primary intracellular fibroblasts and MSCs (Richler and Yaffe, 1970). The preincubation time used in the isolation of bovine MSCs reported in 2023 was 1–3 h. Fibroblasts begin to adhere 5 min after incubation and adhere to surfaces faster than MSCs, indicating that a relatively high purity of Pax7- or MyoD-positive cells can be obtained using the preincubation process (Table 3; Kim et al., 2022; Xu et al., 2018; Yoshioka et al., 2020). In other studies, preincubation conditions have been shown to vary from 5 min to 24 h after fibroblasts begin to adhere (Table 3). One of the effective methods for rat MSCs was preincubation for up to 10 min with shaking every 5 min (Yoshioka et al., 2020). For chicken MSCs, it was up to 2 h with shaking every 8 min after 2 h of rest, indicating that the preincubation conditions may also vary depending on species-specific cell characteristics (Kim et al., 2022).

Even without the pre-culture step, the purity of the MSCs can be increased by using cell sorting techniques, such as FACS and MACS. Some drawbacks of the flow cytometry-based isolation process are that it requires expensive equipment and reagents, trained professionals, and is cumbersome because of sorter-induced cellular stress, such as high-pressure jets, high voltage, and laser exposure during the isolation process, and cytotoxicity that can occur when using specific markers (Lopez and Hulspas, 2020). Although FACS has the advantage of being able to separate cells based on their size or three-dimensional features using fluorescent labeling, it has the disadvantage of expensive equipment and long analysis times. MACS uses magnetic particles to sort cells more than four times faster than FACS and is less expensive, but it is difficult to apply to cells that are susceptible to magnetism or cannot be labeled (Gerashchenko, 2011).

Both FACS and MACS label cells with clusters of differentiation (CD), which are specific markers of MSCs, and each uses a fluorescent agent for FACS and magnetic particles for MACS. Specific markers for MSCs used for cell labeling are species-specific, but some examples are integrin α7, vascular cell adhesion protein 1 (Vcam1), and differentiation clusters, such as CD29 (integrin β1), CD34 (hematopoietic stem cell marker), CD56 (neural cell adhesion molecule), and CD82 (4-transmembrane glycoprotein) (Castiglioni et al., 2014; Uezumi et al., 2016; Yoshioka et al., 2020). After sorting the MSCs, they can then be cultured to check the expression of Pax7 or MyoD to confirm the purity of the isolated MSCs, and the proliferation and differentiation capacity of the cells can be assessed.

In conclusion, the cell biological characteristics necessary for the isolation of MSCs from each animal species have not yet been fully identified, and comprehensive research is limited by the lack of standardization of separation methods, which is an obstacle to industrialization.

Proliferation

MSCs obtained during the isolation process will multiply in number in a properly conditioned growth medium. The proliferation process is directly related to the yield of cultured meat, and various studies have been conducted to improve the proliferation efficiency. First, the basal media commonly used for MSC culture are Ham’s F-10, Dulbecco’s modified Eagle’s medium (DMEM), and DMEM/F12, with bovine fetal serum added to the media at a concentration of 10%–20% (v/v) in most cases (Table 4). Basal media is a solution of basic nutritional components (e.g., amino acids, glucose, lipids, nucleic acid bases, inorganic salts, vitamins, buffers, pH indicators) formulated in a certain proportion according to the culture conditions of the desired cells. In the culture of MSCs, the basal medium and serum concentrations are known to be closely related to the cell proliferation rate and myotube formation (McFarland et al., 1988). In a broiler MSC culture experiment based on culture medium composition, DMEM was found to be more effective than McCoy’s 5A medium in terms of proliferation rate and MRF expression (Flees et al., 2022). In addition, the common view that a low glucose content is effective for chicken and bovine MSC proliferation when using DMEM as basal medium was confirmed (Flees et al., 2022; Zygmunt et al., 2023).

Table 4. Proliferation methods for bovine muscle satellite cells published in 2023

CSSPBQ_2024_v44n2_326_t0009.png 이미지

DMEM, Dulbecco’s modified Eagle’s medium; DMEM/F12, Dulbecco’s modified Eagle’s medium and Ham’s F-12 Nutrient Mixture; LG-DMEM, low glucose-DMEM; HG-DMEM, high glucose-DMEM; FBS, fetal bovine serum; bFGF, basic fibroblast growth factor; HGF, hepatocyte growth factor; IGF-1, insulin-like growth factor-1; IL-6, interleukin-6; ITS, insulin-transferrin-selenium; ITSE, insulin-transferrin-selenium-ethanolamine; PDGF, platelet-derived growth factor; VEGF, vascular endothelial growth factor; N/A, not applicable.

As adherent cells, MSCs require an ECM-based coating for proliferation and differentiation. Representative ECMs used for bovine MSC proliferation have been shown to be gelatin, collagen I, laminin, and Matrigel (Table 4). Integrin α7β1, which is present in the cell membrane of MSCs, binds to collagen and laminin, and laminin induces the proliferation and migration of satellite cells (Öcalan et al., 1988; Sanes, 2003). However, C2C12 cells cultured on plates coated with ECM proteins had a better proliferation rate compared to highly elastic coatings, such as collagen I/laminin/fibronectin hydrogels, which were not conducive to inducing proliferation of MSCs (Palade et al., 2019).

Growth factors are cell signaling proteins. For MSCs, basic fibroblast growth factor (bFGF), vascular endothelial growth factor (VEGF), and HGF are commonly used in culture (Table 4). Typically, bFGF is added to the proliferation medium at a concentration of 5–10 ng/mL. Accordingly, in bovine MSC cultures, a bFGF content of 10 ng/mL in the medium led to a faster proliferation rate than when the bFGF content was 5 ng/mL (Zygmunt et al., 2023). In addition, the expression of various endothelial cell-derived growth factors (IGF-1, HGF, bFGF, and VEGF) can stimulate MSCs to proliferate and regenerate muscle (Yamamoto et al., 2020; Zygmunt et al., 2023).

However, research into cultured meat using FBS remains prevalent. Even when serum-free media is used, various culture ingredients, such as basal media, coating agents, and recombinant proteins (growth factors, hormones), are employed. To address this issue, the development of natural product-derived media that meet food regulatory requirements is underway. However, industrialization is inevitably delayed because companies and research teams cannot disclose it due to competitiveness.

Differentiation

MSC differentiation is commonly achieved by removing the proliferation medium and replacing it with the differentiation medium once the cells have reached a sufficient cell density in the proliferation medium (Ding et al., 2017). The differentiation process typically uses media containing 2% FBS or horse serum to induce serum starvation (Table 5). Serum starvation is often chosen to induce differentiation of MSCs (Pirkmajer and Chibalin, 2011). Induction of differentiation in studies published in 2023 was mainly performed at cell densities above 70% confluence, and the duration of differentiation varied by 1–10 d depending on the experimental conditions, but only one study was identified that varied the serum concentration within the culture period (Table 5).

Table 5. Differentiation methods for bovine muscle satellite cells published in 2023

CSSPBQ_2024_v44n2_326_t0010.png 이미지

DMEM, Dulbecco’s modified Eagle’s medium; DMEM/F12, DMEM and Ham’s F-12 Nutrient Mixture; LG-DMEM, low glucose-DMEM; HG-DMEM, high glucose-DMEM; HS, horse serum; FBS, fetal bovine serum; N/A, not applicable.

When a low-serum environment is used to induce differentiation of MSCs, extensive changes occur at the transcript level, with upregulation of progenitor transcription factors and markers associated with differentiation identified during the differentiation process (Dmitriev et al., 2013; Messmer et al., 2022). Transient and mild levels of serum starvation (15% serum, v/v) induce autophagy, which can promote cell metabolism and differentiation, but 5% serum starvation induces excessive autophagy, leading to cell death (Wang et al., 2023b).

A hypoxic environment (1%–10% O2) in MSC culture can create conditions that mimic oxygen saturation in mature skeletal muscle. Moreover, a hypoxic environment (2% O2) upregulates the myogenic regulators Pax7, Myf5, and MyoD, and intermittent hypoxic exposure increases the expression of VEGF released from MSCs (Koning et al., 2011; Nagahisa and Miyata, 2018; Urbani et al., 2012). The hypoxia-induced factor-1 signaling pathway, which is expressed in response to hypoxic conditions, is thought to be involved in the regulation of myoblast proliferation and differentiation. Under a hypoxic environment (1% O2), broiler MSC cultures exhibited a decrease in the level of MyoD-positive cells along with changes in the transcriptome profile (Jung et al., 2024; Li et al., 2007).

However, serum starvation tends to be the preferred method for induction of differentiation compared to hypoxic environments, and the signaling pathways involved and their effects on differentiation remain poorly understood. Furthermore, the regulations regarding cultured meat are extensive and do not clearly differentiate between cultured meat with or without differentiated tissue, leading to confusion within the industry.

Conclusion

This study analyzed the current technology, industrialization level, and future prospects of cultured meat by analyzing research reports and media reports related to the industrialization of cultured meat. At present, major companies are not entering mass production except for prototype development, and the reason they do not disclose related technologies is that they do not have enough technological capabilities. Therefore, when investing in cultured meat development companies, it is necessary to accurately assess the level of technology that the company has or has acquired. Much of the focus is currently on cell acquisition technology, cell line acquisition technology, and cell culture and muscle differentiation technology. While the level of technology related to the industrialization of cultured meat has reached the stage where prototypes can be produced, it is believed that it has not yet reached the stage where production costs can be dramatically reduced and the product sold to the market. Nevertheless, given the steady increase in the number and depth of studies related to the industrialization of cultured meat and the increasing number of companies involved, it is expected that the industrialization of cultured meat could begin in the not-too-distant future.

Conflicts of Interest

The authors declare no potential conflicts of interest.

Acknowledgements

This work was conducted with the support of Chung-Ang University. This work has supported by the National Research Foundation of Korea (NRF) grant funded by the Korea government (MSIT) (No.2023R1A2C100608811).

Author Contributions

Conceptualization: Hur SJ. Data curation: Yun SH, Lee DY, Lee J, Mariano E Jr, Choi Y, Park J, Han D, Kim JS. Validation: Yun SH, Lee DY, Hur SJ. Investigation: Yun SH, Lee DY, Lee J, Mariano E Jr, Choi Y, Park J, Han D, Kim JS. Writing - original draft: Yun SH. Writing - review & editing: Yun SH, Lee DY, Lee J, Mariano E Jr, Choi Y, Park J, Han D, Kim JS, Hur SJ.

Ethics Approval

This article does not require IRB/IACUC approval because there are no human and animal participants.

References

  1. Adhikari A, Kim W, Davie J. 2021. Myogenin is required for assembly of the transcription machinery on muscle genes during skeletal muscle differentiation. PLOS ONE 16:e0245618.
  2. Aleph Farms. 2023. Aleph Farms submits application to Swiss regulators, marking the first-ever submission for cultivated meat in Europe. Available from: https://aleph-farms.com/journals/aleph-farms-submits-application-to-swiss-regulators-marking-the-first-ever-submission-for-cultivated-meat-in-europe/. Accessed at Jan 18, 2024.
  3. Aleph Farms. 2024. Aleph Farms granted world's first regulatory approval for cultivated beef. Available from: https://aleph-farms.com/journals/aleph-farms-granted-worlds-first-regulatory-approval-for-cultivated-beef/. Accessed at Jan 18, 2024.
  4. Allan SJ, De Bank PA, Ellis MJ. 2019. Bioprocess design considerations for cultured meat production with a focus on the expansion bioreactor. Front Sustain Food Syst 3:44.
  5. Anderson J, Pilipowicz O. 2002. Activation of muscle satellite cells in single-fiber cultures. Nitric Oxide 7:36-41. https://doi.org/10.1016/S1089-8603(02)00011-3
  6. Anomaly J. 2015. What's wrong with factory farming? Public Health Ethics 8:246-254. https://doi.org/10.1093/phe/phu001
  7. Backs J, Worst BC, Lehmann LH, Patrick DM, Jebessa Z, Kreusser MM, Sun Q, Chen L, Heft C, Katus HA, Olson EN. 2011. Selective repression of MEF2 activity by PKA-dependent proteolysis of HDAC4. J Cell Biol 195:403-415. https://doi.org/10.1083/jcb.201105063
  8. Bhat ZF, Kumar S, Fayaz H. 2015. In vitro meat production: Challenges and benefits over conventional meat production. J Integr Agric 14:241-248. https://doi.org/10.1016/S2095-3119(14)60887-X
  9. Bischoff R. 1986. Proliferation of muscle satellite cells on intact myofibers in culture. Dev Biol 115:129-139. https://doi.org/10.1016/0012-1606(86)90234-4
  10. Bodiou V, Moutsatsou P, Post MJ. 2020. Microcarriers for upscaling cultured meat production. Front Nutr 7:10.
  11. Boereboom A, Sheikh M, Islam T, Achirimbi E, Vriesekoop F. 2022. Brits and British Muslims and their perceptions of cultured meat: How big is their willingness to purchase? Food Front 3:529-540. https://doi.org/10.1002/fft2.165
  12. Bomkamp C, Skaalure SC, Fernando GF, Ben-Arye T, Swartz EW, Specht EA. 2022. Scaffolding biomaterials for 3D cultivated meat: Prospects and challenges. Adv Sci 9:2102908.
  13. Brandt AM, Kania JM, Reinholt BM, Johnson SE. 2018. Human IL6 stimulates bovine satellite cell proliferation through a signal transducer and activator of transcription 3 (STAT3)-dependent mechanism. Domest Anim Endocrinol 62:32-38. https://doi.org/10.1016/j.domaniend.2017.08.004
  14. Broucke K, Van Pamel E, Van Coillie E, Herman L, Van Royen G. 2023. Cultured meat and challenges ahead: A review on nutritional, technofunctional and sensorial properties, safety and legislation. Meat Sci 195:109006.
  15. Bryant C, Barnett J. 2018. Consumer acceptance of cultured meat: A systematic review. Meat Sci 143:8-17. https://doi.org/10.1016/j.meatsci.2018.04.008
  16. Bryant C, Barnett J. 2020. Consumer acceptance of cultured meat: An updated review (2018-2020). Appl Sci 10:5201.
  17. Castiglioni A, Hettmer S, Lynes MD, Rao TN, Tchessalova D, Sinha I, Lee BT, Tseng YH, Wagers AJ. 2014. Isolation of progenitors that exhibit myogenic/osteogenic bipotency in vitro by fluorescence-activated cell sorting from human fetal muscle. Stem Cell Rep 2:92-106. https://doi.org/10.1016/j.stemcr.2013.12.006
  18. CellMEAT. 2023. Cell MEAT begins approval process for cell-cultured meat from the Ministry of Food and Drug Safety of Korea. Available from: https://www.thecellmeat.com/bbs/board.php?bo_table=gallery&wr_id=44. Accessed at Jan 18, 2024.
  19. Chen X, Gao B, Ponnusamy M, Lin Z, Liu J. 2017. MEF2 signaling and human diseases. Oncotarget 8:112152-112165. https://doi.org/10.18632/oncotarget.22899
  20. Chriki S, Hocquette JF. 2020. The myth of cultured meat: A review. Front Nutr 7:7.
  21. Conboy IM, Rando TA. 2002. The regulation of Notch signaling controls satellite cell activation and cell fate determination in postnatal myogenesis. Dev Cell 3:397-409. https://doi.org/10.1016/S1534-5807(02)00254-X
  22. Cornelison DDW, Olwin BB, Rudnicki MA, Wold BJ. 2000. MyoD-/- satellite cells in single-fiber culture are differentiation defective and MRF4 deficient. Dev Biol 224:122-137. https://doi.org/10.1006/dbio.2000.9682
  23. Crawshaw C, Piazza J. 2023. Livestock farmers' attitudes towards alternative proteins. Sustainability 15:9253.
  24. Ding S, Swennen GNM, Messmer T, Gagliardi M, Molin DGM, Li C, Zhou G, Post MJ. 2018. Maintaining bovine satellite cells stemness through p38 pathway. Sci Rep 8:10808.
  25. Ding S, Wang F, Liu Y, Li S, Zhou G, Hu P. 2017. Characterization and isolation of highly purified porcine satellite cells. Cell Death Discov 3:17003.
  26. Dmitriev P, Barat A, Polesskaya A, O'Connell MJ, Robert T, Dessen P, Walsh TA, Lazar V, Turki A, Carnac G, Laoudj-Chenivesse D, Lipinski M, Vassetzky YS. 2013. Simultaneous miRNA and mRNA transcriptome profiling of human myoblasts reveals a novel set of myogenic differentiation-associated miRNAs and their target genes. BMC Genom 14:265.
  27. Doumit ME, Merkel RA. 1992. Conditions for isolation and culture of porcine myogenic satellite cells. Tissue Cell 24:253-262. https://doi.org/10.1016/0040-8166(92)90098-R
  28. Eng D, Ma HY, Gross MK, Kioussi C. 2013. Gene networks during skeletal myogenesis. Int Sch Res Notices 2013:348704.
  29. European Union [EU]. 2021. Document 02015R2283-20210327: Regulation (EU) 2015/2283 of the European Parliament and of the council of 25 November 2015 on novel foods, amending Regulation (EU) No 1169/2011 of the European Parliament and of the Council and repealing Regulation (EC) No 258/97 of the European Parliament and of the Council and Commission Regulation (EC) No 1852/2001 (text with EEA relevance). Available from: https://eurlex.europa.eu/legal-content/EN/TXT/?uri=CELEX%3A02015R2283-20210327. Accessed at Jan 18, 2024.
  30. Flees JJ, Starkey CW, Starkey JD. 2022. Effect of different basal culture media and sera type combinations on primary broiler chicken muscle satellite cell heterogeneity during proliferation and differentiation. Animals 12:1425.
  31. Food and Agriculture Organization [FAO], World Health Organization [WHO]. 2023. Food safety aspects of cell-based food. Available from: http://www.fao.org/3/cc4855en/cc4855en.pdf. Accessed at Jan 18, 2024.
  32. Food and Drug Administration [FDA]. 2019. Formal agreement between FDA and USDA regarding oversight of human food produced using animal cell technology derived from cell lines of USDA-amenable species. Available from: https://www.fda.gov/food/domestic-interagency-agreements-food-expired/formal-agreement-between-fda-and-usda-regarding-oversight-human-food-produced-using-animal-cell. Accessed at Jan 18, 2024.
  33. Food and Drug Administration [FDA]. 2023a. Inventory of completed pre-market consultations for human food made with cultured animal cells. Available from: https://www.fda.gov/food/human-food-made-cultured-animal-cells/inventory-completed-pre-market-consultations-human-food-made-cultured-animal-cells. Accessed at Jan 18, 2024.
  34. Food and Drug Administration [FDA]. 2023b. Human food made with cultured animal cells. Available from: https://www.fda.gov/food/food-ingredients-packaging/human-food-made-cultured-animal-cells. Accessed at Jan 18, 2024. 
  35. Food Standards Agency [FSA]. 2023. Cell-cultivated products. Available from: https://www.food.gov.uk/business-guidance/cell-cultivated-products. Accessed at Jan 18, 2024.
  36. Food Standards Australia New Zealand [FSANZ]. 2023. A1269: Cultured quail as a novel food. Available from: https://www.foodstandards.gov.au/food-standards-code/applications/A1269-Cultured-Quail-as-a-Novel-Food. Accessed at Jan 18, 2024.
  37. Fu X, Wang H, Hu P. 2015. Stem cell activation in skeletal muscle regeneration. Cell Mol Life Sci 72:1663-1677. https://doi.org/10.1007/s00018-014-1819-5
  38. Gagan J, Dey BK, Layer R, Yan Z, Dutta A. 2012. Notch3 and Mef2c proteins are mutually antagonistic via Mkp1 protein and miR-1/206 microRNAs in differentiating myoblasts. J Biol Chem 287:40360-40370. https://doi.org/10.1074/jbc.M112.378414
  39. Ge C, Selvaganapathy PR, Geng F. 2023. Advancing our understanding of bioreactors for industrial-sized cell culture: Health care and cellular agriculture implications. Am J Physiol Cell Physiol 325:C580-C591. https://doi.org/10.1152/ajpcell.00408.2022
  40. Gerashchenko BI. 2011. Choosing a cell sorting option to study the fate of bystander cells: FACS or MACS? Cytom A 79A:179-180. https://doi.org/10.1002/cyto.a.21021
  41. Gerber PJ, Steinfeld H, Henderson B, Mottet A, Opio C, Dijkman J, Falcucci A, Tempio G. 2013. Tackling climate change through livestock: A global assessment of emissions and mitigation opportunities. Available from: https://www.fao.org/3/i3437e/i3437e.pdf. Accessed at Jan 18, 2024.
  42. GlobeNewswire. 2023. Cultured meat market to witness 23.2% CAGR by 2032 driven by consumer shift towards sustainable and ethical food choices: Exclusive report by Market.us. Available from: https://www.globenewswire.com/en/news-release/2023/04/10/2643902/0/en/Cultured-Meat-Market-to-Witness-23-2-CAGR-by-2032-Driven-by-Consumer-Shift-Towards-Sustainable-and-Ethical-Food-Choices-Exclusive-Report-by-Market-us.html. Accessed at Jan 18, 2024.
  43. Godfray HCJ, Aveyard P, Garnett T, Hall JW, Key TJ, Lorimer J, Pierrehumbert RT, Scarborough P, Springmann M, Jebb SA. 2018. Meat consumption, health, and the environment. Science 361:eaam5324.
  44. Gromova A, Tierney MT, Sacco A. 2015. FACS-based satellite cell isolation from mouse hind limb muscles. Bio-protocol 5:e1558.
  45. Guan X, Lei Q, Yan Q, Li X, Zhou J, Du G, Chen J. 2021. Trends and ideas in technology, regulation and public acceptance of cultured meat. Future Foods 3:100032.
  46. Hamdan MN, Post MJ, Ramli MA, Mustafa AR. 2018. Cultured meat in Islamic perspective. J Relig Health 57:2193-2206. https://doi.org/10.1007/s10943-017-0403-3
  47. Hawke TJ, Garry DJ. 2001. Myogenic satellite cells: Physiology to molecular biology. J Appl Physiol 91:534-551. https://doi.org/10.1152/jappl.2001.91.2.534
  48. Heidemann MS, Molento CFM, Reis GG, Phillips CJC. 2020. Uncoupling meat from animal slaughter and its impacts on human-animal relationships. Front Psychol 11:1824.
  49. Ho SS, Ou M, Vijayan AV. 2023. Halal or not? Exploring Muslim perceptions of cultured meat in Singapore. Front Sustain Food Syst 7:1127164.
  50. Hocquette JF. 2016. Is in vitro meat the solution for the future? Meat Sci 120:167-176. https://doi.org/10.1016/j.meatsci.2016.04.036
  51. Holmberg J, Durbeej M. 2013. Laminin-211 in skeletal muscle function. Cell Adhes Migr 7:111-121. https://doi.org/10.4161/cam.22618
  52. Holterman CE, Rudnicki MA. 2005. Molecular regulation of satellite cell function. Semin Cell Dev Biol 16:575-584. https://doi.org/10.1016/j.semcdb.2005.07.004
  53. Horsley V, Jansen KM, Mills ST, Pavlath GK. 2003. IL-4 acts as a myoblast recruitment factor during mammalian muscle growth. Cell 113:483-494. https://doi.org/10.1016/S0092-8674(03)00319-2
  54. Humbird D. 2021. Scale-up economics for cultured meat. Biotechnol Bioeng 118:3239-3250. https://doi.org/10.1002/bit.27848
  55. Jo YW, Park I, Yoo K, Woo HY, Kim YL, Kim YE, Kim JH, Kong YY. 2022. Notch1 and Notch2 signaling exclusively but cooperatively maintain fetal myogenic progenitors. Stem Cells 40:1031-1042.  https://doi.org/10.1093/stmcls/sxac056
  56. Jung U, Kim M, Dowker-Key P, Noe S, Bettaieb A, Shepherd E, Voy B. 2024. Hypoxia promotes proliferation and inhibits myogenesis in broiler satellite cells. Poult Sci 103:103203.
  57. Kablar B, Krastel K, Ying C, Asakura A, Tapscott SJ, Rudnicki MA. 1997. MyoD and Myf-5 differentially regulate the development of limb versus trunk skeletal muscle. Development 124:4729-4738. https://doi.org/10.1242/dev.124.23.4729
  58. Kann AP, Hung M, Krauss RS. 2021. Cell-cell contact and signaling in the muscle stem cell niche. Curr Opin Cell Biol 73:78-83. https://doi.org/10.1016/j.ceb.2021.06.003
  59. Kim B, Ko D, Choi SH, Park S. 2023a. Bovine muscle satellite cells in calves and cattle: A comparative study of cellular and genetic characteristics for cultivated meat production. Curr Res Food Sci 7:100545.
  60. Kim J, Kim W. 2023. Arginine and lysine promote skeletal muscle hypertrophy by regulating the mTOR signaling pathway in bovine myocytes. Meat Muscle Biol 7:16876.
  61. Kim SH, Kim CJ, Lee EY, Son YM, Hwang YH, Joo ST. 2022. Optimal pre-plating method of chicken satellite cells for cultured meat production. Food Sci Anim Resour 42:942-952. https://doi.org/10.5851/kosfa.2022.e61
  62. Kim WS, Daddam JR, Keng BH, Kim J, Kim J. 2023b. Heat shock protein 27 regulates myogenic and self-renewal potential of bovine satellite cells under heat stress. J Anim Sci 101:skad303.
  63. Kim Y, Oh S, Park G, Park S, Park Y, Choi H, Kim M, Choi J. 2023c. Characteristics of bovine muscle satellite cell from different breeds for efficient production of cultured meat. J Anim Sci Technol (in press). doi: 10.5187/jast.2023.e115.
  64. Knight JDR, Kothary R. 2011. The myogenic kinome: Protein kinases critical to mammalian skeletal myogenesis. Skelet Muscle 1:29.
  65. Koning M, Werker PMN, van Luyn MJA, Harmsen MC. 2011. Hypoxia promotes proliferation of human myogenic satellite cells: A potential benefactor in tissue engineering of skeletal muscle. Tissue Eng A 17:1747-1758. https://doi.org/10.1089/ten.tea.2010.0624
  66. Kuang S, Charge SB, Seale P, Huh M, Rudnicki MA. 2006. Distinct roles for Pax7 and Pax3 in adult regenerative myogenesis. J Cell Biol 172:103-113. https://doi.org/10.1083/jcb.200508001
  67. Kuang S, Kuroda K, Le Grand F, Rudnicki MA. 2007. Asymmetric self-renewal and commitment of satellite stem cells in muscle. Cell 129:999-1010. https://doi.org/10.1016/j.cell.2007.03.044
  68. Laestadius LI, Caldwell MA. 2015. Is the future of meat palatable? Perceptions of in vitro meat as evidenced by online news comments. Public Health Nutr 18:2457-2467. https://doi.org/10.1017/S1368980015000622
  69. Lazure F, Blackburn DM, Corchado AH, Sahinyan K, Karam N, Sharanek A, Nguyen D, Lepper C, Najafabadi HS, Perkins TJ, Jahani-Asl A, Soleimani VD. 2020. Myf6/MRF4 is a myogenic niche regulator required for the maintenance of the muscle stem cell pool. EMBO Rep 21:e49499.
  70. Lee DY, Lee SY, Yun SH, Jeong JW, Kim JH, Kim HW, Choi JS, Kim GD, Joo ST, Choi I, Hur SJ. 2022. Review of the current research on fetal bovine serum and the development of cultured meat. Food Sci Anim Resour 42:775-799. https://doi.org/10.5851/kosfa.2022.e46
  71. Lee DY, Yun SH, Lee SY, Lee J, Mariano E JR, Joo ST, Choi JS, Kim GD, Lee J, Choi SH, Hur SJ. 2023a. Analysis of commercial fetal bovine serum (FBS) and its substitutes in the development of cultured meat. Food Res Int 174:113617.
  72. Lee JH, Peng DQ, Jin XC, Smith SB, Lee HG. 2023b. Vitamin D3 decreases myoblast fusion during the growth and increases myogenic gene expression during the differentiation phase in muscle satellite cells from Korean native beef cattle. J Anim Sci 101:skad192.
  73. Lee SY, Kang HJ, Lee DY, Kang JH, Ramani S, Park S, Hur SJ. 2021. Principal protocols for the processing of cultured meat. J Anim Sci Technol 63:673-680. https://doi.org/10.5187/jast.2021.e40
  74. Lee SY, Lee DY, Jeong JW, Kim JH, Yun SH, Mariano E Jr, Lee J, Park S, Jo C, Hur SJ. 2023c. Current technologies, regulation, and future perspective of animal product analogs: A review. Anim Biosci 36:1465-1487.
  75. Levi S, Yen FC, Baruch L, Machluf M. 2022. Scaffolding technologies for the engineering of cultured meat: Towards a safe, sustainable, and scalable production. Trends Food Sci Technol 126:13-25. https://doi.org/10.1016/j.tifs.2022.05.011
  76. Li B, Wang J, Raza SHA, Wang S, Liang C, Zhang W, Yu S, Shah MA, Al Abdulmonem W, Alharbi YM, Aljohani ASM, Pant SD, Zan L. 2023. MAPK family genes' influences on myogenesis in cattle: Genome-wide analysis and identification. Res Vet Sci 159:198-212. https://doi.org/10.1016/j.rvsc.2023.04.024
  77. Li EW, McKee-Muir OC, Gilbert PM. 2018. Cellular biomechanics in skeletal muscle regeneration. Curr Top Dev Biol 126:125-176. https://doi.org/10.1016/bs.ctdb.2017.08.007
  78. Li X, Zhu L, Chen X, Fan M. 2007. Effects of hypoxia on proliferation and differentiation of myoblasts. Med Hypotheses 69:629-636. https://doi.org/10.1016/j.mehy.2006.12.050
  79. Lin-Hi N, Reimer M, Schafer K, Bottcher J. 2023. Consumer acceptance of cultured meat: An empirical analysis of the role of organizational factors. J Bus Econ 93:707-746.
  80. Lopez PA, Hulspas R. 2020. Special issue on enhancement of reproducibility and rigor. Cytom A 97:105-106. https://doi.org/10.1002/cyto.a.23972
  81. Lu Y, Bradley JS, McCoski SR, Gonzalez JM, Ealy AD, Johnson SE. 2017. Reduced skeletal muscle fiber size following caloric restriction is associated with calpain-mediated proteolysis and attenuation of IGF-1 signaling. Am J Physiol Regul Integr Comp Physiol 312:R806-R815. https://doi.org/10.1152/ajpregu.00400.2016
  82. Lynch J, Pierrehumbert R. 2019. Climate impacts of cultured meat and beef cattle. Front Sustain Food Syst 3:5.
  83. Mariano E Jr, Lee DY, Yun SH, Lee J, Lee SY, Hur SJ. 2023. Checkmeat: A review on the applicability of conventional meat authentication techniques to cultured meat. Food Sci Anim Resour 43:1055-1066. https://doi.org/10.5851/kosfa.2023.e48
  84. Mateti T, Laha A, Shenoy P. 2022. Artificial meat industry: Production methodology, challenges, and future. JOM 74:3428-3444. https://doi.org/10.1007/s11837-022-05316-x
  85. McFarland DC, Doumit ME, Minshall RD. 1988. The turkey myogenic satellite cell: Optimization of in vitro proliferation and differentiation. Tissue Cell 20:899-908. https://doi.org/10.1016/0040-8166(88)90031-6
  86. McKinsey TA, Zhang CL, Olson EN. 2002. MEF2: A calcium-dependent regulator of cell division, differentiation and death. Trends Biochem Sci 27:40-47. https://doi.org/10.1016/S0968-0004(01)02031-X
  87. Mekonnen MM, Hoekstra AY. 2012. A global assessment of the water footprint of farm animal products. Ecosystems 15:401-415. https://doi.org/10.1007/s10021-011-9517-8
  88. Mesires NT, Doumit ME. 2002. Satellite cell proliferation and differentiation during postnatal growth of porcine skeletal muscle. Am J Physiol Cell Physiol 282:C899-C906. https://doi.org/10.1152/ajpcell.00341.2001
  89. Messersmith EM, Smerchek DT, Hansen SL. 2021. The crossroads between zinc and steroidal implant-induced growth of beef cattle. Animals 11:1914.
  90. Messmer T, Dohmen RGJ, Schaeken L, Melzener L, Hueber R, Godec M, Didoss C, Post MJ, Flack JE. 2023. Single-cell analysis of bovine muscle-derived cell types for cultured meat production. Front Nutr 10:1212196.
  91. Messmer T, Klevernic I, Furquim C, Ovchinnikova E, Dogan A, Cruz H, Post MJ, Flack JE. 2022. A serum-free media formulation for cultured meat production supports bovine satellite cell differentiation in the absence of serum starvation. Nat Food 3:74-85. https://doi.org/10.1038/s43016-021-00419-1
  92. Ministry of Food and Drug Safety of Korea [MFDS]. 2023. Administrative notice of partial amendment notice (public notice No. 2023-507) for the "Temporary standards and recognition standards of specification for food, etc.". Available from: https://www.mfds.go.kr/brd/m_209/view.do?seq=43852&srchFr=&srchTo=&srchWord=&srchTp=&itm_seq_1=0&itm_seq_2=0&multi_itm_seq=0&company_cd=&company_nm=&page=1. Accessed at Jan 18, 2024.
  93. Mohammadabadi M, Bordbar F, Jensen J, Du M, Guo W. 2021. Key genes regulating skeletal muscle development and growth in farm animals. Animals 11:835.
  94. Morand S, Blasdell K, Bordes F, Buchy P, Carcy B, Chaisiri K, Chaval Y, Claude J, Cosson JF, Desquesnes M, Jittapalapong S, Jiyipong T, Karnchanabanthoen A, Pornpan P, Rolain JM, Tran A. 2019. Changing landscapes of Southeast Asia and rodent-borne diseases: Decreased diversity but increased transmission risks. Ecol Appl 29:e01886.
  95. Moritz J, McPartlin M, Tuomisto HL, Ryynanen T. 2023. A multi-level perspective of potential transition pathways towards cultured meat: Finnish and German political stakeholder perceptions. Res Policy 52:104866.
  96. Motohashi N, Asakura Y, Asakura A. 2014. Isolation, culture, and transplantation of muscle satellite cells. J Vis Exp 86:e50846.
  97. Nabeshima Y, Hanaoka K, Hayasaka M, Esuml E, Li S, Nonaka I, Nabeshima Y. 1993. Myogenin gene disruption results in perinatal lethality because of severe muscle defect. Nature 364:532-535. https://doi.org/10.1038/364532a0
  98. Nagahisa H, Miyata H. 2018. Influence of hypoxic stimulation on angiogenesis and satellite cells in mouse skeletal muscle. PLOS ONE 13:e0207040.
  99. Newton P, Blaustein-Rejto D. 2021. Social and economic opportunities and challenges of plant-based and cultured meat for rural producers in the US. Front Sustain Food Syst 5:624270.
  100. Ocalan M, Goodman SL, Kuhl U, Hauschka SD, von der Mark K. 1988. Laminin alters cell shape and stimulates motility and proliferation of murine skeletal myoblasts. Dev Biol 125:158-167. https://doi.org/10.1016/0012-1606(88)90068-1
  101. Oh S, Park S, Park Y, Kim Y, Park G, Cui X, Kim K, Joo S, Hur S, Kim G, Choi J. 2023. Culturing characteristics of Hanwoo myosatellite cells and C2C12 cells incubated at 37℃ and 39℃ for cultured meat. J Anim Sci Technol 65:664-678. https://doi.org/10.5187/jast.2023.e10
  102. Oh WJ, Jacinto E. 2011. mTOR complex 2 signaling and functions. Cell Cycle 10:2305-2316. https://doi.org/10.4161/cc.10.14.16586
  103. Ohashi K, Nagata Y, Wada E, Zammit PS, Shiozuka M, Matsuda R. 2015. Zinc promotes proliferation and activation of myogenic cells via the PI3K/Akt and ERK signaling cascade. Exp Cell Res 333:228-237. https://doi.org/10.1016/j.yexcr.2015.03.003
  104. Olguin HC, Olwin BB. 2004. Pax-7 up-regulation inhibits myogenesis and cell cycle progression in satellite cells: A potential mechanism for self-renewal. Dev Biol 275:375-388. https://doi.org/10.1016/j.ydbio.2004.08.015
  105. Ong KJ, Johnston J, Datar I, Sewalt V, Holmes D, Shatkin JA. 2021. Food safety considerations and research priorities for the cultured meat and seafood industry. Compr Rev Food Sci Food Saf 20:5421-5448. https://doi.org/10.1111/1541-4337.12853
  106. Organisation for Economic Co‑operation and Development [OECD], Food and Agricultural Organization [FAO]. 2022. OECD-FAO agricultural outlook 2022-2031. Available from: https://www.oecd-ilibrary.org/agriculture-and-food/oecd-fao-agricultural-outlook-2022-2031_f1b0b29c-en. Accessed at Jan 18, 2024.
  107. Ornitz DM, Itoh N. 2015. The fibroblast growth factor signaling pathway. Wiley Interdiscip Rev Dev Biol 4:215-266. https://doi.org/10.1002/wdev.176
  108. Pakseresht A, Kaliji SA, Canavari M. 2022. Review of factors affecting consumer acceptance of cultured meat. Appetite 170:105829.
  109. Palade J, Pal A, Rawls A, Stabenfeldt S, Wilson-Rawls J. 2019. Molecular analysis of muscle progenitor cells on extracellular matrix coatings and hydrogels. Acta Biomater 97:296-309. https://doi.org/10.1016/j.actbio.2019.08.019
  110. Park IH, Chen J. 2005. Mammalian target of rapamycin (mTOR) signaling is required for a late-stage fusion process during skeletal myotube maturation*[boxs]. J Biol Chem 280:32009-32017. https://doi.org/10.1074/jbc.M506120200
  111. Park S, Park G, Oh S, Park Y, Kim Y, Kim J, Choi J. 2023. Investigating proliferation and differentiation capacities of Hanwoo steer myosatellite cells at different passages for developing cell-cultured meat. Sci Rep 13:15614. 
  112. Parker C, Carey R, De Costa J, Scrinis G. 2017. Can the hidden hand of the market be an effective and legitimate regulator? The case of animal welfare under a labeling for consumer choice policy approach. Regul Gov 11:368-387. https://doi.org/10.1111/rego.12147
  113. Pasut A, Jones AE, Rudnicki MA. 2013. Isolation and culture of individual myofibers and their satellite cells from adult skeletal muscle. J Vis Exp 73:e50074.
  114. Pirkmajer S, Chibalin AV. 2011. Serum starvation: Caveat emptor. Am J Physiol Cell Physiol 301:C272-C279. https://doi.org/10.1152/ajpcell.00091.2011
  115. Punch VG, Jones AE, Rudnicki MA. 2009. Transcriptional networks that regulate muscle stem cell function. Wiley Interdiscip Rev Syst Biol Med 1:128-140. https://doi.org/10.1002/wsbm.11
  116. Reis GG, Heidemann MS, de Matos KHO, Molento CFM. 2020. Cell-based meat and firms' environmental strategies: New rationales as per available literature. Sustainability 12:9418.
  117. Relaix F, Bencze M, Borok MJ, Der Vartanian A, Gattazzo F, Mademtzoglou D, Perez-Diaz S, Prola A, Reyes-Fernandez PC, Rotini A, Taglietti V. 2021. Perspectives on skeletal muscle stem cells. Nat Commun 12:692.
  118. Rhoads RP, Baumgard LH, El-Kadi SW, Zhao LD. 2016. Physiology and endocrinology symposium: Roles for insulin-supported skeletal muscle growth. J Anim Sci 94:1791-1802. https://doi.org/10.2527/jas.2015-0110
  119. Richler C, Yaffe D. 1970. The in vitro cultivation and differentiation capacities of myogenic cell lines. Dev Biol 23:1-22. https://doi.org/10.1016/S0012-1606(70)80004-5
  120. Rion N, Castets P, Lin S, Enderle L, Reinhard JR, Ruegg MA. 2019. mTORC2 affects the maintenance of the muscle stem cell pool. Skelet Muscle 9:30.
  121. Risner D, Kim Y, Nguyen C, Siegel JB, Spang ES. 2023. Environmental impacts of cultured meat: A cradle-to-gate life cycle assessment. Available from: https://www.biorxiv.org/content/10.1101/2023.04.21.537778v1. Accessed at Jan 18, 2024.
  122. Risner D, Li F, Fell JS, Pace SA, Siegel JB, Tagkopoulos I, Spang ES. 2021. Preliminary techno-economic assessment of animal cell-based meat. Foods 10:3.
  123. Rodriguez Escobar MI, Cadena E, Nhu TT, Cooreman-Algoed M, De Smet S, Dewulf J. 2021. Analysis of the cultured meat production system in function of its environmental footprint: Current status, gaps and recommendations. Foods 10:2941.
  124. Rolland NCM, Markus CR, Post MJ. 2020. The effect of information content on acceptance of cultured meat in a tasting context. PLOS ONE 15:e0231176.
  125. Sanes JR. 2003. The basement membrane/basal lamina of skeletal muscle. J Biol Chem 278:12601-12604. https://doi.org/10.1074/jbc.R200027200
  126. Schenzle L, Egger K, Fuchs A, Pichler H. 2022. Never let me down: Optimizing performance of serum free culture medium for bovine satellite cells. Available from: https://www.biorxiv.org/content/10.1101/2022.11.13.516330v1. Accessed at Jan 18, 2024.
  127. Seale P, Sabourin LA, Girgis-Gabardo A, Mansouri A, Gruss P, Rudnicki MA. 2000. Pax7 is required for the specification of myogenic satellite cells. Cell 102:777-786. https://doi.org/10.1016/S0092-8674(00)00066-0
  128. Shao X, Gong W, Wang Q, Wang P, Shi T, Mahmut A, Qin J, Yao Y, Yan W, Chen D, Chen X, Jiang Q, Guo B. 2022. Atrophic skeletal muscle fibre-derived small extracellular vesicle miR-690 inhibits satellite cell differentiation during ageing. J Cachexia Sarcopenia Muscle 13:3163-3180. https://doi.org/10.1002/jcsm.13106
  129. Singapore Food Agency [SFA]. 2023. Requirements for the safety assessment of novel foods and novel food ingredients. Available from: https://www.sfa.gov.sg/docs/default-source/food-information/requirements-for-the-safety-assessment-of-novel-foods-and-novel-food-ingredients.pdf. Accessed at Jan 18, 2024.
  130. Skinner D, Blake J. 2023. Modelling consumers' choice of novel food. PLOS ONE 18:e0290169.
  131. Skrivergaard S, Rasmussen MK, Sahebekhtiari N, Young JF, Therkildsen M. 2023. Satellite cells sourced from bull calves and dairy cows differs in proliferative and myogenic capacity: Implications for cultivated meat. Food Res Int 173:113217. 
  132. Slade P. 2018. If you build it, will they eat it? Consumer preferences for plant-based and cultured meat burgers. Appetite 125:428-437. https://doi.org/10.1016/j.appet.2018.02.030
  133. Smith TH, Block NE, Rhodes SJ, Miller JB. 1993. A unique pattern of expression of the four muscle regulatory factor proteins distinguishes somitic from embryonic, fetal and newborn mouse myogenic cells. Development 117:1125-1133. https://doi.org/10.1242/dev.117.3.1125
  134. Soice E, Johnston J. 2021. How cellular agriculture systems can promote food security. Front Sustain Food Syst 5:753996.
  135. Stork PJS, Schmitt JM. 2002. Crosstalk between cAMP and MAP kinase signaling in the regulation of cell proliferation. Trends Cell Biol 12:258-266. https://doi.org/10.1016/S0962-8924(02)02294-8
  136. Stout AJ, Mirliani AB, Rittenberg ML, Shub M, White EC, Yuen Jr. JSK, Kaplan DL. 2022. Simple and effective serum-free medium for sustained expansion of bovine satellite cells for cell cultured meat. Commun Biol 5:466.
  137. Stout AJ, Rittenberg ML, Shub M, Saad MK, Mirliani AB, Dolgin J, Kaplan DL. 2023. A Beefy-R culture medium: Replacing albumin with rapeseed protein isolates. Biomaterials 296:122092.
  138. Trendelenburg AU, Meyer A, Rohner D, Boyle J, Hatakeyama S, Glass DJ. 2009. Myostatin reduces Akt/TORC1/p70S6K signaling, inhibiting myoblast differentiation and myotube size. Am J Physiol Cell Physiol 296:C1258-C1270. https://doi.org/10.1152/ajpcell.00105.2009
  139. Tress L. 2023. In first, leading kosher authority Orthodox Union certifies lab-grown meat. Available from: https://www.timesofisrael.com/in-first-leading-kosher-authority-orthodox-union-certifies-lab-grown-meat/. Accessed at Jan 18, 2024.
  140. Troy A, Cadwallader AB, Fedorov Y, Tyner K, Tanaka KK, Olwin BB. 2012. Coordination of satellite cell activation and self-renewal by par-complex-dependent asymmetric activation of p38α/β MAPK. Cell Stem Cell 11:541-553. https://doi.org/10.1016/j.stem.2012.05.025
  141. Tuomisto HL. 2018. The eco-friendly burger: Could cultured meat improve the environmental sustainability of meat products? EMBO Rep 20:e47395.
  142. Tuomisto HL, Teixeira de Mattos MJ. 2011. Environmental impacts of cultured meat production. Environ Sci Technol 45:6117-6123. https://doi.org/10.1021/es200130u
  143. Tzimorotas D, Solberg NT, Andreassen RC, Moutsatsou P, Bodiou V, Pedersen ME, Ronning SB. 2023. Expansion of bovine skeletal muscle stem cells from spinner flasks to benchtop stirred-tank bioreactors for up to 38 days. Front Nutr 10:1192365.
  144. Uezumi A, Nakatani M, Ikemoto-Uezumi M, Yamamoto N, Morita M, Yamaguchi A, Yamada H, Kasai T, Masuda S, Narita A, Miyagoe-Suzuki Y, Takeda S, Fukada S, Nishino I, Tsuchida K. 2016. Cell-surface protein profiling identifies distinctive markers of progenitor cells in human skeletal muscle. Stem Cell Rep 7:263-278. https://doi.org/10.1016/j.stemcr.2016.07.004
  145. United Nations [UN]. 2022. World population prospects 2022. Available from: https://desapublications.un.org/file/989/download. Accessed at Dec 14, 2023.
  146. United States Department of Agriculture [USDA], Food Safety and Inspection Service [FSIS]. 2023. FSIS responsibilities in establishments producing cell-cultured meat and poultry food products. Available from: https://www.fsis.usda.gov/policy/fsis-directives/7800.1. Accessed at Jan 18, 2024.
  147. Urbani L, Piccoli M, Franzin C, Pozzobon M, De Coppi P. 2012. Hypoxia increases mouse satellite cell clone proliferation maintaining both in vitro and in vivo heterogeneity and myogenic potential. PLOS ONE 7:e49860.
  148. Uyen NT, Van Cuong D, Thuy PD, Son LH, Ngan NT, Quang NH, Tuan ND, Hwang I. 2023. A comparative study on the adipogenic and myogenic capacity of muscle satellite cells, and meat quality characteristics between Hanwoo and Vietnamese yellow steers. Food Sci Anim Resour 43:563-579. https://doi.org/10.5851/kosfa.2023.e19
  149. Wang K, Liufu S, Yu Z, Xu X, Ai N, Li X, Liu X, Chen B, Zhang Y, Ma H, Yin Y. 2023a. miR-100-5p regulates skeletal muscle myogenesis through the Trib2/mTOR/S6K signaling pathway. Int J Mol Sci 24:8906. 
  150. Wang Y, Gao J, Fan B, Hu Y, Yang Y, Wu Y, Li F, Ju H. 2023b. Different levels of autophagy induced by transient serum starvation regulate metabolism and differentiation of porcine skeletal muscle satellite cells. Sci Rep 13:13153.
  151. Wang YN, Yang WC, Li PW, Wang HB, Zhang YY, Zan LS. 2018. Myocyte enhancer factor 2A promotes proliferation and its inhibition attenuates myogenic differentiation via myozenin 2 in bovine skeletal muscle myoblast. PLOS ONE 13:e0196255.
  152. Wei X, Luo L, Chen J. 2019. Roles of mTOR signaling in tissue regeneration. Cells 8:1075.
  153. Wilks M, Phillips CJC. 2017. Attitudes to in vitro meat: A survey of potential consumers in the United States. PLOS ONE 12:e0171904.
  154. Wu H, Peisley A, Graef IA, Crabtree GR. 2007. NFAT signaling and the invention of vertebrates. Trends Cell Biol 17:251-260. https://doi.org/10.1016/j.tcb.2007.04.006
  155. Xie WY, Shen Q, Zhao FJ. 2018. Antibiotics and antibiotic resistance from animal manures to soil: A review. Eur J Soil Sci 69:181-195. https://doi.org/10.1111/ejss.12494
  156. Xu Z, Yu L, Lu H, Feng W, Chen L, Zhou J, Yang X, Qi Z. 2018. A modified preplate technique for efficient isolation and proliferation of mice muscle-derived stem cells. Cytotechnology 70:1671-1683. https://doi.org/10.1007/s10616-018-0262-0
  157. Yamamoto N, Oyaizu T, Enomoto M, Horie M, Yuasa M, Okawa A, Yagishita K. 2020. VEGF and bFGF induction by nitric oxide is associated with hyperbaric oxygen-induced angiogenesis and muscle regeneration. Sci Rep 10:2744.
  158. Yoshioka K, Kitajima Y, Okazaki N, Chiba K, Yonekura A, Ono Y. 2020. A modified pre-plating method for high-yield and high-purity muscle stem cell isolation from human/mouse skeletal muscle tissues. Front Cell Dev Biol 8:793.
  159. Young HS, Dirzo R, Helgen KM, McCauley DJ, Billeter SA, Kosoy MY, Osikowicz LM, Salkeld DJ, Young TP, Dittmar K. 2014. Declines in large wildlife increase landscape-level prevalence of rodent-borne disease in Africa. Proc Natl Acad Sci USA 111:7036-7041. https://doi.org/10.1073/pnas.1404958111
  160. Yue J, Xu W, Xiang L, Chen S, Li X, Yang Q, Zhang R, Bao X, Wang Y, Mbadhi M, Liu Y, Yao L, Chen L, Zhao X, Hu C, Zhang J, Zheng H, Wu Y, Chen SY, Li S, Lv J, Shi L, Tang J. 2023. Continuous exposure to isoprenaline reduced myotube size by delaying myoblast differentiation and fusion through the NFAT-MEF2C signaling pathway. Sci Rep 13:436.
  161. Yun SH, Lee DY, Lee SY, Lee J, Mariano E Jr, Joo ST, Choi I, Choi JS, Kim GD, Hur SJ. 2023. Improved culture procedure for bovine muscle satellite cells for cultured meat. Food Res Int 174:113660.
  162. Zhang J, Sheng H, Pan C, Wang S, Yang M, Hu C, Wei D, Wang Y, Ma Y. 2023. Identification of key genes in bovine muscle development by co-expression analysis. PeerJ 11:e15093.
  163. Zhang K, Sha J, Harter ML. 2010. Activation of Cdc6 by MyoD is associated with the expansion of quiescent myogenic satellite cells. J Cell Biol 188:39-48. https://doi.org/10.1083/jcb.200904144
  164. Zhang M, Li L, Bai J. 2020. Consumer acceptance of cultured meat in urban areas of three cities in China. Food Control 118:107390.
  165. Zhang P, Liang X, Shan T, Jiang Q, Deng C, Zheng R, Kuang S. 2015. mTOR is necessary for proper satellite cell activity and skeletal muscle regeneration. Biochem Biophys Res Commun 463:102-108. https://doi.org/10.1016/j.bbrc.2015.05.032
  166. Zhang W, Liu Y, Zhang H. 2021. Extracellular matrix: An important regulator of cell functions and skeletal muscle development. Cell Biosci 11:65.
  167. Zhu Y, Li P, Dan X, Kang X, Ma Y, Shi Y. 2022. FHL2 regulates proliferation differentiation and autophagy of bovine skeletal muscle satellite cells through Wnt/β-catenin signaling pathway. Available from: https://assets.researchsquare.com/files/rs-1180493/v1/e2a3a74e-b5a8-428b-9003-292c62c6a085.pdf?c=1649420068. Accessed at Jan 18, 2024.
  168. Zygmunt K, Otwinowska-Mindur A, Piorkowska K, Witarski W. 2023. Influence of media composition on the level of bovine satellite cell proliferation. Animals 13:1855.