Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Elife ; 122023 04 04.
Article in English | MEDLINE | ID: mdl-37014056

ABSTRACT

We present a method of producing bulk cell-cultured fat tissue for food applications. Mass transport limitations (nutrients, oxygen, waste diffusion) of macroscale 3D tissue culture are circumvented by initially culturing murine or porcine adipocytes in 2D, after which bulk fat tissue is produced by mechanically harvesting and aggregating the lipid-filled adipocytes into 3D constructs using alginate or transglutaminase binders. The 3D fat tissues were visually similar to fat tissue harvested from animals, with matching textures based on uniaxial compression tests. The mechanical properties of cultured fat tissues were based on binder choice and concentration, and changes in the fatty acid compositions of cellular triacylglyceride and phospholipids were observed after lipid supplementation (soybean oil) during in vitro culture. This approach of aggregating individual adipocytes into a bulk 3D tissue provides a scalable and versatile strategy to produce cultured fat tissue for food-related applications, thereby addressing a key obstacle in cultivated meat production.


Subject(s)
Adipocytes , Adipose Tissue , Swine , Animals , Mice , Fatty Acids
2.
Sci Rep ; 13(1): 5098, 2023 03 29.
Article in English | MEDLINE | ID: mdl-36991012

ABSTRACT

Cell-cultivated fish offers the potential for a more ethical, sustainable, and safe seafood system. However, fish cell culture is relatively understudied in comparison to mammalian cells. Here, we established and characterized a continuous Atlantic mackerel (Scomber scombrus) skeletal muscle cell line ("Mack" cells). The cells were isolated from muscle biopsies of fresh-caught fish, with separate isolations performed from two distinct fish. Mack1 cells (cells from the first isolation) were cultured for over a year and subcultured over 130 times. The cells proliferated at initial doubling times of 63.9 h (± 19.1 SD). After a spontaneous immortalization crisis from passages 37-43, the cells proliferated at doubling times of 24.3 h (± 4.91 SD). A muscle phenotype was confirmed through characterization of muscle stemness and differentiation via paired-box protein 7 and myosin heavy chain immunostaining, respectively. An adipocyte-like phenotype was also demonstrated for the cells through lipid accumulation, confirmed via Oil Red O staining and quantification of neutral lipids. New qPCR primers (HPRT, PAX3B, MYOD1, MYOG, TNNT3A, and PPARG) were tailored to the mackerel genome and used to characterize mackerel cell genotypes. This work provides the first spontaneously immortalized fish muscle cell line for research, ideally serving as a reference for subsequent investigation.


Subject(s)
Muscles , Perciformes , Animals , Fishes , Perciformes/genetics , Muscle Cells , Cell Line , Phenotype , Mammals
3.
Biomaterials ; 296: 122092, 2023 05.
Article in English | MEDLINE | ID: mdl-36965281

ABSTRACT

The development of cost-effective serum-free media is essential for the economic viability of cultured meat. A key challenge facing this goal is the high-cost of recombinant albumin which is necessary in many serum-free media formulations, including a recently developed serum-free medium for bovine satellite cell (BSC) culture termed Beefy-9. Here we alter Beefy-9 by replacing recombinant albumin with rapeseed protein isolate (RPI), a bulk-protein solution obtained from agricultural waste through alkali extraction (pH 12.5), isoelectric protein precipitation (pH 4.5), dissolution of physiologically soluble proteins (pH 7.2), and concentration of proteins through 3 kDa ultrafiltration. This new medium, termed Beefy-R, was then used to culture BSCs over four passages, during which cells grew with an average doubling time of 26.6 h, showing improved growth compared with Beefy-9. In Beefy-R, BSCs maintained cell phenotype and myogenicity. Together, these results offer an effective, low-cost, and sustainable alternative to albumin for serum-free culture of muscle stem cells, thereby addressing a key hurdle facing cultured meat production.


Subject(s)
Brassica napus , Animals , Cattle , Culture Media, Serum-Free , Cell Culture Techniques/methods , Albumins , Culture Media , Cells, Cultured
4.
Chem Rev ; 123(5): 2242-2275, 2023 Mar 08.
Article in English | MEDLINE | ID: mdl-36603542

ABSTRACT

Engineered materials are ubiquitous throughout society and are critical to the development of modern technology, yet many current material systems are inexorably tied to widespread deterioration of ecological processes. Next-generation material systems can address goals of environmental sustainability by providing alternatives to fossil fuel-based materials and by reducing destructive extraction processes, energy costs, and accumulation of solid waste. However, development of sustainable materials faces several key challenges including investigation, processing, and architecting of new feedstocks that are often relatively mechanically weak, complex, and difficult to characterize or standardize. In this review paper, we outline a framework for examining sustainability in material systems and discuss how recent developments in modeling, machine learning, and other computational tools can aid the discovery of novel sustainable materials. We consider these through the lens of materiomics, an approach that considers material systems holistically by incorporating perspectives of all relevant scales, beginning with first-principles approaches and extending through the macroscale to consider sustainable material design from the bottom-up. We follow with an examination of how computational methods are currently applied to select examples of sustainable material development, with particular emphasis on bioinspired and biobased materials, and conclude with perspectives on opportunities and open challenges.

5.
ACS Biomater Sci Eng ; 8(9): 3785-3796, 2022 Sep 12.
Article in English | MEDLINE | ID: mdl-35977409

ABSTRACT

Cell-cultured fat could provide important elements of flavor, nutrition, and texture to enhance the quality and therefore expand consumer adoption of alternative meat products. In contrast to cells from livestock animals, insect cells have been proposed as a relatively low-cost and scalable platform for tissue engineering and muscle cell-derived cultured meat production. Furthermore, insect fat cells have long been cultured and characterized for basic biology and recombinant protein production but not for food production. To develop a food-relevant approach to insect fat cell cultivation and tissue engineering, Manduca sexta cells were cultured and induced to accumulate lipids in 2D and 3D formats within decellularized mycelium scaffolding. The resultant in vitro fat tissues were characterized and compared to in vivo fat tissue data by imaging, lipidomics, and texture analyses. The cells exhibited robust lipid accumulation when treated with a 0.1 mM soybean oil emulsion and had "healthier" fat profiles, as measured by the ratio of unsaturated to saturated fatty acids. Mycelium scaffolding provided a simple, food-grade approach to support the 3D cell cultures and lipid accumulation. This approach provides a low-cost, scalable, and nutritious method for cultured fat production.


Subject(s)
Fatty Acids , Manduca , Agriculture , Animals , Fatty Acids/analysis , Fatty Acids/metabolism , Manduca/metabolism
6.
Biomaterials ; 280: 121273, 2022 01.
Article in English | MEDLINE | ID: mdl-34933254

ABSTRACT

With rising global demand for food proteins and significant environmental impact associated with conventional animal agriculture, it is important to develop sustainable alternatives to supplement existing meat production. Since fat is an important contributor to meat flavor, recapitulating this component in meat alternatives such as plant based and cell cultured meats is important. Here, we discuss the topic of cell cultured or tissue engineered fat, growing adipocytes in vitro that could imbue meat alternatives with the complex flavor and aromas of animal meat. We outline potential paths for the large scale production of in vitro cultured fat, including adipogenic precursors during cell proliferation, methods to adipogenically differentiate cells at scale, as well as strategies for converting differentiated adipocytes into 3D cultured fat tissues. We showcase the maturation of knowledge and technology behind cell sourcing and scaled proliferation, while also highlighting that adipogenic differentiation and 3D adipose tissue formation at scale need further research. We also provide some potential solutions for achieving adipose cell differentiation and tissue formation at scale based on contemporary research and the state of the field.


Subject(s)
Adipocytes , Adipose Tissue , Adipogenesis , Animals , Cell Differentiation , Meat/analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...