1
|
Guo G, Wang W, Tu M, Zhao B, Han J, Li J, Pan Y, Zhou J, Ma W, Liu Y, Sun T, Han X, An Y. Deciphering adipose development: Function, differentiation and regulation. Dev Dyn 2024; 253:956-997. [PMID: 38516819 DOI: 10.1002/dvdy.708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 03/02/2024] [Accepted: 03/10/2024] [Indexed: 03/23/2024] Open
Abstract
The overdevelopment of adipose tissues, accompanied by excess lipid accumulation and energy storage, leads to adipose deposition and obesity. With the increasing incidence of obesity in recent years, obesity is becoming a major risk factor for human health, causing various relevant diseases (including hypertension, diabetes, osteoarthritis and cancers). Therefore, it is of significance to antagonize obesity to reduce the risk of obesity-related diseases. Excess lipid accumulation in adipose tissues is mediated by adipocyte hypertrophy (expansion of pre-existing adipocytes) or hyperplasia (increase of newly-formed adipocytes). It is necessary to prevent excessive accumulation of adipose tissues by controlling adipose development. Adipogenesis is exquisitely regulated by many factors in vivo and in vitro, including hormones, cytokines, gender and dietary components. The present review has concluded a comprehensive understanding of adipose development including its origin, classification, distribution, function, differentiation and molecular mechanisms underlying adipogenesis, which may provide potential therapeutic strategies for harnessing obesity without impairing adipose tissue function.
Collapse
Affiliation(s)
- Ge Guo
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, China
| | - Wanli Wang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, China
| | - Mengjie Tu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, China
| | - Binbin Zhao
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, China
| | - Jiayang Han
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, China
| | - Jiali Li
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, China
| | - Yanbing Pan
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, China
| | - Jie Zhou
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, China
| | - Wen Ma
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, China
| | - Yi Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, China
| | - Tiantian Sun
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, China
| | - Xu Han
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, China
| | - Yang An
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, China
| |
Collapse
|
2
|
Boychenko S, Egorova VS, Brovin A, Egorov AD. White-to-Beige and Back: Adipocyte Conversion and Transcriptional Reprogramming. Pharmaceuticals (Basel) 2024; 17:790. [PMID: 38931457 PMCID: PMC11206576 DOI: 10.3390/ph17060790] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 06/11/2024] [Accepted: 06/13/2024] [Indexed: 06/28/2024] Open
Abstract
Obesity has become a pandemic, as currently more than half a billion people worldwide are obese. The etiology of obesity is multifactorial, and combines a contribution of hereditary and behavioral factors, such as nutritional inadequacy, along with the influences of environment and reduced physical activity. Two types of adipose tissue widely known are white and brown. While white adipose tissue functions predominantly as a key energy storage, brown adipose tissue has a greater mass of mitochondria and expresses the uncoupling protein 1 (UCP1) gene, which allows thermogenesis and rapid catabolism. Even though white and brown adipocytes are of different origin, activation of the brown adipocyte differentiation program in white adipose tissue cells forces them to transdifferentiate into "beige" adipocytes, characterized by thermogenesis and intensive lipolysis. Nowadays, researchers in the field of small molecule medicinal chemistry and gene therapy are making efforts to develop new drugs that effectively overcome insulin resistance and counteract obesity. Here, we discuss various aspects of white-to-beige conversion, adipose tissue catabolic re-activation, and non-shivering thermogenesis.
Collapse
Affiliation(s)
- Stanislav Boychenko
- Gene Therapy Department, Center for Translational Medicine, Sirius University of Science and Technology, 354340 Sirius, Russia; (S.B.); (A.B.)
| | - Vera S. Egorova
- Biotechnology Department, Center for Translational Medicine, Sirius University of Science and Technology, 354340 Sirius, Russia
| | - Andrew Brovin
- Gene Therapy Department, Center for Translational Medicine, Sirius University of Science and Technology, 354340 Sirius, Russia; (S.B.); (A.B.)
| | - Alexander D. Egorov
- Gene Therapy Department, Center for Translational Medicine, Sirius University of Science and Technology, 354340 Sirius, Russia; (S.B.); (A.B.)
| |
Collapse
|
3
|
Tan Z, Jiang H. Molecular and Cellular Mechanisms of Intramuscular Fat Development and Growth in Cattle. Int J Mol Sci 2024; 25:2520. [PMID: 38473768 DOI: 10.3390/ijms25052520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 02/15/2024] [Accepted: 02/19/2024] [Indexed: 03/14/2024] Open
Abstract
Intramuscular fat, also referred to as marbling fat, is the white fat deposited within skeletal muscle tissue. The content of intramuscular fat in the skeletal muscle, particularly the longissimus dorsi muscle, of cattle is a critical determinant of beef quality and value. In this review, we summarize the process of intramuscular fat development and growth, the factors that affect this process, and the molecular and epigenetic mechanisms that mediate this process in cattle. Compared to other species, cattle have a remarkable ability to accumulate intramuscular fat, partly attributed to the abundance of sources of fatty acids for synthesizing triglycerides. Compared to other adipose depots such as subcutaneous fat, intramuscular fat develops later and grows more slowly. The commitment and differentiation of adipose precursor cells into adipocytes as well as the maturation of adipocytes are crucial steps in intramuscular fat development and growth in cattle. Each of these steps is controlled by various factors, underscoring the complexity of the regulatory network governing adipogenesis in the skeletal muscle. These factors include genetics, epigenetics, nutrition (including maternal nutrition), rumen microbiome, vitamins, hormones, weaning age, slaughter age, slaughter weight, and stress. Many of these factors seem to affect intramuscular fat deposition through the transcriptional or epigenetic regulation of genes directly involved in the development and growth of intramuscular fat. A better understanding of the molecular and cellular mechanisms by which intramuscular fat develops and grows in cattle will help us develop more effective strategies to optimize intramuscular fat deposition in cattle, thereby maximizing the quality and value of beef meat.
Collapse
Affiliation(s)
- Zhendong Tan
- School of Animal Sciences, Virginia Tech, Blacksburg, VA 24061, USA
| | - Honglin Jiang
- School of Animal Sciences, Virginia Tech, Blacksburg, VA 24061, USA
| |
Collapse
|
4
|
Santos MM, Costa TC, Silva W, Pistillo LZ, Junior DTV, Verardo LL, Paulino PVR, Sampaio CB, Gionbelli MP, Du M, Duarte MS. Nutrient supplementation of beef female calves at pre-weaning enhances the commitment of fibro-adipogenic progenitor cells to preadipocytes. Meat Sci 2023; 204:109286. [PMID: 37494740 DOI: 10.1016/j.meatsci.2023.109286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 07/13/2023] [Accepted: 07/18/2023] [Indexed: 07/28/2023]
Abstract
We aimed to evaluate the impact of nutrient supplementation of beef female calves at pre-weaning on adipogenic determination. Thirty-four female calves were assigned to two experimental treatments: Control (CON, n = 17), where animals were supplemented only with mineral mixture; Supplemented (SUP, n = 17), where animals received energy-protein supplement containing minerals (5 g/kg of BW per day) of their body weight. Animals were supplemented from 100 to 250 days of age, and muscle samples were biopsied at the end of the supplementation period. Regarding the performance variables, there were no differences between treatments for initial body weight (P = 0.75). The final body weight (P = 0.07), average daily gain (P = 0.07), rib eye area (P = 0.03), and rib fat thickness (P = 0.08) were greater in SUP female calves compared with CON treatment. The number of fibro-adipogenic progenitor cells (P = 0.69) did not differ between treatments, while a greater number of intramuscular pre-adipocytes were observed in SUP than CON female calves (P = 0.01). The expression of miRNA-4429 (P = 0.20) did not differ between treatments, while the expression of miRNA-129-5p (P = 0.09) and miRNA-129-2-3p (P = 0.05) was greater in CON than SUP female calves. Our results suggest that nutrient supplementation at early postnatal stages of development enhances the commitment of fibro-adipogenic progenitor cells into the adipogenic lineages allowing to an increase in intramuscular fat deposition potential of the animals later in life.
Collapse
Affiliation(s)
- M M Santos
- Department of Animal Science, Universidade Federal de Viçosa, Viçosa, Brazil; Muscle Biology and Nutrigenomics Laboratory, Universidade Federal de Viçosa, Viçosa, Brazil
| | - T C Costa
- Muscle Biology and Nutrigenomics Laboratory, Universidade Federal de Viçosa, Viçosa, Brazil; Department of Animal Science, Universidade Federal de Lavras, Lavras, MG, Brazil
| | - W Silva
- Department of Animal Science, Universidade Federal de Viçosa, Viçosa, Brazil; Department of Animal Biosciences, University of Guelph, Guelph, ON, Canada
| | - L Z Pistillo
- Department of Animal Science, Universidade Federal de Viçosa, Viçosa, Brazil
| | - D T Valente Junior
- Department of Animal Science, Universidade Federal de Viçosa, Viçosa, Brazil; Muscle Biology and Nutrigenomics Laboratory, Universidade Federal de Viçosa, Viçosa, Brazil; Department of Animal Biosciences, University of Guelph, Guelph, ON, Canada
| | - L L Verardo
- Department of Animal Science, Universidade Federal dos Vales do Jequitinhonha e Mucuri, Diamantina, Brazil
| | | | - C B Sampaio
- Department of Animal Science, Universidade Federal de Viçosa, Viçosa, Brazil
| | - M P Gionbelli
- Department of Animal Science, Universidade Federal de Lavras, Lavras, MG, Brazil
| | - M Du
- Department of Animal Sciences, Washington State University, Pullman, WA, USA
| | - M S Duarte
- Department of Animal Biosciences, University of Guelph, Guelph, ON, Canada.
| |
Collapse
|
5
|
Jara TC, Park K, Vahmani P, Van Eenennaam AL, Smith LR, Denicol AC. Stem cell-based strategies and challenges for production of cultivated meat. NATURE FOOD 2023; 4:841-853. [PMID: 37845547 DOI: 10.1038/s43016-023-00857-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 09/05/2023] [Indexed: 10/18/2023]
Abstract
Cultivated meat scale-up and industrial production will require multiple stable cell lines from different species to recreate the organoleptic and nutritional properties of meat from livestock. In this Review, we explore the potential of stem cells to create the major cellular components of cultivated meat. By using developments in the fields of tissue engineering and biomedicine, we explore the advantages and disadvantages of strategies involving primary adult and pluripotent stem cells for generating cell sources that can be grown at scale. These myogenic, adipogenic or extracellular matrix-producing adult stem cells as well as embryonic or inducible pluripotent stem cells are discussed for their proliferative and differentiation capacity, necessary for cultivated meat. We examine the challenges for industrial scale-up, including differentiation and culture protocols, as well as genetic modification options for stem cell immortalization and controlled differentiation. Finally, we discuss stem cell-related safety and regulatory challenges for bringing cultivated meat to the marketplace.
Collapse
Affiliation(s)
- T C Jara
- Department of Animal Science, University of California Davis, Davis, CA, USA
| | - K Park
- Department of Animal Science, University of California Davis, Davis, CA, USA
| | - P Vahmani
- Department of Animal Science, University of California Davis, Davis, CA, USA
| | - A L Van Eenennaam
- Department of Animal Science, University of California Davis, Davis, CA, USA
| | - L R Smith
- Department of Neurobiology, Physiology and Behavior, University of California Davis, Davis, CA, USA.
| | - A C Denicol
- Department of Animal Science, University of California Davis, Davis, CA, USA
| |
Collapse
|
6
|
Bomkamp C, Musgrove L, Marques DMC, Fernando GF, Ferreira FC, Specht EA. Differentiation and Maturation of Muscle and Fat Cells in Cultivated Seafood: Lessons from Developmental Biology. MARINE BIOTECHNOLOGY (NEW YORK, N.Y.) 2023; 25:1-29. [PMID: 36374393 PMCID: PMC9931865 DOI: 10.1007/s10126-022-10174-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 10/10/2022] [Indexed: 06/16/2023]
Abstract
Cultivated meat, also known as cultured or cell-based meat, is meat produced directly from cultured animal cells rather than from a whole animal. Cultivated meat and seafood have been proposed as a means of mitigating the substantial harms associated with current production methods, including damage to the environment, antibiotic resistance, food security challenges, poor animal welfare, and-in the case of seafood-overfishing and ecological damage associated with fishing and aquaculture. Because biomedical tissue engineering research, from which cultivated meat draws a great deal of inspiration, has thus far been conducted almost exclusively in mammals, cultivated seafood suffers from a lack of established protocols for producing complex tissues in vitro. At the same time, fish such as the zebrafish Danio rerio have been widely used as model organisms in developmental biology. Therefore, many of the mechanisms and signaling pathways involved in the formation of muscle, fat, and other relevant tissue are relatively well understood for this species. The same processes are understood to a lesser degree in aquatic invertebrates. This review discusses the differentiation and maturation of meat-relevant cell types in aquatic species and makes recommendations for future research aimed at recapitulating these processes to produce cultivated fish and shellfish.
Collapse
Affiliation(s)
- Claire Bomkamp
- Department of Science & Technology, The Good Food Institute, Washington, DC USA
| | - Lisa Musgrove
- University of the Sunshine Coast, Sippy Downs, Queensland Australia
| | - Diana M. C. Marques
- Department of Bioengineering and Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal
- Associate Laboratory i4HB—Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal
| | - Gonçalo F. Fernando
- Department of Science & Technology, The Good Food Institute, Washington, DC USA
| | - Frederico C. Ferreira
- Department of Bioengineering and Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal
- Associate Laboratory i4HB—Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal
| | - Elizabeth A. Specht
- Department of Science & Technology, The Good Food Institute, Washington, DC USA
| |
Collapse
|
7
|
Jaborek JR, Fluharty FL, Lee K, Zerby HN, Relling AE. Lipid metabolism mRNA expression and cellularity of intramuscular adipocytes within the Longissimus muscle of Angus- and Wagyu-sired cattle fed for a similar days on feed or body weight endpoint. J Anim Sci 2023; 101:skac371. [PMID: 36753534 PMCID: PMC9907753 DOI: 10.1093/jas/skac371] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Accepted: 11/03/2022] [Indexed: 02/09/2023] Open
Abstract
This study investigates intramuscular (IM) adipocyte development in the Longissimus muscle (LM) between Wagyu- and Angus-sired steers compared at a similar age and days on feed (D) endpoint or similar body weight (B) endpoint by measuring IM adipocyte cell area and lipid metabolism mRNA expression. Angus-sired steers (AN, n = 6) were compared with steers from two different Wagyu sires (WA), selected for either growth (G) or marbling (M), to be compared at a similar days on feed (DOF; 258 ± 26.7 d; WA-GD, n = 5 and WA-MD, n = 5) in Exp. 1 or body weight (BW; 613 ± 18.0 kg; WA-GB, n = 4 and WA-MB, n = 5) in Exp. 2, respectively. In Exp. 1, WA-MD steers had a greater (P ≤ 0.01) percentage of IM fat in the LM compared with AN and WA-GD steers. In Exp. 2, WA-MB steers had a greater (P ≤ 0.01) percentage of IM fat in the LM compared with AN and WA-GB steers. The distribution of IM adipocyte area was unimodal at all biopsy collections, with IM adipocyte area becoming progressively larger as cattle age (P ≤ 0.01) and BW increased (P ≤ 0.01). Peroxisome proliferator activated receptor delta (PPARd) was upregulated earlier for WA-MD and WA-MB cattle compared with other steers at a similar DOF and BW (P ≤ 0.02; treatment × biopsy interaction). Peroxisome proliferator activated receptor gamma was upregulated (PPARg) at a lesser BW for WA-MB steers (P = 0.09; treatment × biopsy interaction), while WA-MD steers had a greater (P ≤ 0.04) overall mean PPARg mRNA expression compared with other steers. Glycerol-3-phosphate acyltransferase, lipin 1, and hormone sensitive lipase demonstrated mRNA expression patterns similar to PPARg and PPARd or CCAAT enhancer binding protein beta, which emphasizes their importance in marbling development and growth. Additionally, WA-MD and WA-MB steers often had a greater early mRNA expression of fatty acid transporters (fatty acid transport protein 1; P < 0.02; treatment × biopsy interaction) and binding proteins (fatty acid binding protein 4) compared with other steers. Cattle with a greater marbling propensity appear to upregulate adipogenesis at a younger chronological and physiological maturity through PPARd, PPARg, and possibly adipogenic regulating compounds, lysophosphatidic acid, and diacylglycerol. These genes and compounds could be used as potential markers for marbling propensity of cattle in the future.
Collapse
Affiliation(s)
- J R Jaborek
- Department of Animal Sciences, The Ohio State University, Columbus, OH 43210, USA
- Michigan State University Extension - Sanilac County, Sandusky, MI 48471, USA
| | - F L Fluharty
- Department of Animal and Dairy Science, University of Georgia, Athens, GA 30602, USA
| | - Kichoon Lee
- Department of Animal Sciences, The Ohio State University, Columbus, OH 43210, USA
| | | | - A E Relling
- Department of Animal Sciences, The Ohio State University, Wooster, OH 44691, USA
| |
Collapse
|
8
|
Kumar A, Sood A, Han SS. Technological and structural aspects of scaffold manufacturing for cultured meat: recent advances, challenges, and opportunities. Crit Rev Food Sci Nutr 2022; 63:585-612. [PMID: 36239416 DOI: 10.1080/10408398.2022.2132206] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
In vitro cultured meat is an emerging area of research focus with an innovative approach through tissue engineering (i.e., cellular engineering) to meet the global food demand. The manufacturing of lab-cultivated meat is an innovative business that alleviates life-threatening environmental issues concerning public health and animal well-being on the global platform. There has been a noteworthy advancement in cultivating artificial meat, but still, there are numerous challenges that impede the swift headway of lab-grown meat production at a commercially large scale. In this review, we focus on the manufacturing of edible scaffolds for cultured meat production. In brief, first an introduction to cultivating artificial meat and its current scenario in the market is provided. Further, a discussion on the understanding of composition, cellular, and molecular communications in muscle tissue is presented, which are vital to scaling up the production of lab-grown meat. In continuation, the major components (e.g., cells, biomaterial scaffolds, and their manufacturing technologies, media, and potential bioreactors) for cultured meat production are conferred followed by a comprehensive discussion on the most recent advances in lab-cultured meat. Finally, existing challenges and opportunities including future research perspectives for scaling-up cultured meat production are discussed with conclusive interpretations.
Collapse
Affiliation(s)
- Anuj Kumar
- School of Chemical Engineering, Yeungnam University, Gyeongsan, South Korea.,Research Institute of Cell Culture, Yeungnam University, Gyeongsan, South Korea
| | - Ankur Sood
- School of Chemical Engineering, Yeungnam University, Gyeongsan, South Korea
| | - Sung Soo Han
- School of Chemical Engineering, Yeungnam University, Gyeongsan, South Korea.,Research Institute of Cell Culture, Yeungnam University, Gyeongsan, South Korea
| |
Collapse
|
9
|
Wang H, Wang X, Yan D, Sun H, Chen Q, Li M, Dong X, Pan Y, Lu S. Genome-wide association study identifying genetic variants associated with carcass backfat thickness, lean percentage and fat percentage in a four-way crossbred pig population using SLAF-seq technology. BMC Genomics 2022; 23:594. [PMID: 35971078 PMCID: PMC9380336 DOI: 10.1186/s12864-022-08827-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 08/05/2022] [Indexed: 12/12/2022] Open
Abstract
Background Carcass backfat thickness (BFT), carcass lean percentage (CLP) and carcass fat percentage (CFP) are important to the commercial pig industry. Nevertheless, the genetic architecture of BFT, CLP and CFP is still elusive. Here, we performed a genome-wide association study (GWAS) based on specific-locus amplified fragment sequencing (SLAF-seq) to analyze seven fatness-related traits, including five BFTs, CLP, and CFP on 223 four-way crossbred pigs. Results A total of 227, 921 highly consistent single nucleotide polymorphisms (SNPs) evenly distributed throughout the genome were used to perform GWAS. Using the mixed linear model (MLM), a total of 20 SNP loci significantly related to these traits were identified on ten Sus scrofa chromosomes (SSC), of which 10 SNPs were located in previously reported quantitative trait loci (QTL) regions. On SSC7, two SNPs (SSC7:29,503,670 and rs1112937671) for average backfat thickness (ABFT) exceeded 1% and 10% Bonferroni genome-wide significance levels, respectively. These two SNP loci were located within an intron region of the COL21A1 gene, which was a protein-coding gene that played an important role in the porcine backfat deposition by affecting extracellular matrix (ECM) remodeling. In addition, based on the other three significant SNPs on SSC7, five candidate genes, ZNF184, ZNF391, HMGA1, GRM4 and NUDT3 were proposed to influence BFT. On SSC9, two SNPs for backfat thickness at 6–7 ribs (67RBFT) and one SNP for CLP were in the same locus region (19 kb interval). These three SNPs were located in the PGM2L1 gene, which encoded a protein that played an indispensable role in glycogen metabolism, glycolysis and gluconeogenesis as a key enzyme. Finally, one significant SNP on SSC14 for CLP was located within the PLBD2 gene, which participated in the lipid catabolic process. Conclusions A total of two regions on SSC7 and SSC9 and eight potential candidate genes were found for fatness-related traits in pigs. The results of this GWAS based on SLAF-seq will greatly advance our understanding of the genetic architecture of BFT, CLP, and CFP traits. These identified SNP loci and candidate genes might serve as a biological basis for improving the important fatness-related traits of pigs. Supplementary Information The online version contains supplementary material available at 10.1186/s12864-022-08827-8.
Collapse
Affiliation(s)
- Huiyu Wang
- Faculty of Animal Science and Technology, Yunnan Agricultural University, No. 95 of Jinhei Road, Kunming, 650201, Yunnan, China.,Faculty of Animal Science, Xichang University, Xichang, 615000, Sichuan, China
| | - Xiaoyi Wang
- Faculty of Animal Science and Technology, Yunnan Agricultural University, No. 95 of Jinhei Road, Kunming, 650201, Yunnan, China
| | - Dawei Yan
- Faculty of Animal Science and Technology, Yunnan Agricultural University, No. 95 of Jinhei Road, Kunming, 650201, Yunnan, China
| | - Hao Sun
- Faculty of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Qiang Chen
- Faculty of Animal Science and Technology, Yunnan Agricultural University, No. 95 of Jinhei Road, Kunming, 650201, Yunnan, China
| | - Mingli Li
- Faculty of Animal Science and Technology, Yunnan Agricultural University, No. 95 of Jinhei Road, Kunming, 650201, Yunnan, China
| | - Xinxing Dong
- Faculty of Animal Science and Technology, Yunnan Agricultural University, No. 95 of Jinhei Road, Kunming, 650201, Yunnan, China
| | - Yuchun Pan
- Faculty of Animal Sciences, Zhejiang University, Hangzhou, 310058, Zhejiang, China.
| | - Shaoxiong Lu
- Faculty of Animal Science and Technology, Yunnan Agricultural University, No. 95 of Jinhei Road, Kunming, 650201, Yunnan, China.
| |
Collapse
|
10
|
Costa T, Lourenço P, Souza R, Lopes M, Araújo R, Santos M, Luciano L, Massensini J, Chalfun L, Rennó L, Sampaio C, Veroneze R, Paulino P, Gionbelli M, Duarte M. Ruminal undegradable protein enriched diet during late gestation of beef cows affects maternal metabolism and offspring’s skeletal muscle development. Anim Feed Sci Technol 2022. [DOI: 10.1016/j.anifeedsci.2022.115400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
11
|
Johnson CD, Zhou LY, Kopinke D. A Guide to Examining Intramuscular Fat Formation and its Cellular Origin in Skeletal Muscle. JOURNAL OF VISUALIZED EXPERIMENTS : JOVE 2022:10.3791/63996. [PMID: 35695517 PMCID: PMC9741761 DOI: 10.3791/63996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Fibro-adipogenic progenitors (FAPs) are mesenchymal stromal cells that play a crucial role during skeletal muscle homeostasis and regeneration. FAPs build and maintain the extracellular matrix that acts as a molecular myofiber scaffold. In addition, FAPs are indispensable for myofiber regeneration as they secrete a multitude of beneficial factors sensed by the muscle stem cells (MuSCs). In diseased states, however, FAPs are the cellular origin of intramuscular fat and fibrotic scar tissue. This fatty fibrosis is a hallmark of sarcopenia and neuromuscular diseases, such as Duchenne Muscular Dystrophy. One significant barrier in determining why and how FAPs differentiate into intramuscular fat is effective preservation and subsequent visualization of adipocytes, especially in frozen tissue sections. Conventional methods of skeletal muscle tissue processing, such as snap-freezing, do not properly preserve the morphology of individual adipocytes, thereby preventing accurate visualization and quantification. To overcome this hurdle, a rigorous protocol was developed that preserves adipocyte morphology in skeletal muscle sections allowing visualization, imaging, and quantification of intramuscular fat. The protocol also outlines how to process a portion of muscle tissue for RT-qPCR, enabling users to confirm observed changes in fat formation by viewing differences in the expression of adipogenic genes. Additionally, it can be adapted to visualize adipocytes by whole-mount immunofluorescence of muscle samples. Finally, this protocol outlines how to perform genetic lineage tracing of Pdgfrα-expressing FAPs to study the adipogenic conversion of FAPs. This protocol consistently yields high-resolution and morphologically accurate immunofluorescent images of adipocytes, along with confirmation by RT-qPCR, allowing for robust, rigorous, and reproducible visualization and quantification of intramuscular fat. Together, the analysis pipeline described here is the first step to improving our understanding of how FAPs differentiate into intramuscular fat, and provides a framework to validate novel interventions to prevent fat formation.
Collapse
Affiliation(s)
- Connor D. Johnson
- Department of Pharmacology and Therapeutics, University of Florida College of Medicine
| | - Lylybell Y. Zhou
- Department of Pharmacology and Therapeutics, University of Florida College of Medicine
| | - Daniel Kopinke
- Department of Pharmacology and Therapeutics, University of Florida College of Medicine,Myology Institute, University of Florida College of Medicine
| |
Collapse
|
12
|
Knežić T, Janjušević L, Djisalov M, Yodmuang S, Gadjanski I. Using Vertebrate Stem and Progenitor Cells for Cellular Agriculture, State-of-the-Art, Challenges, and Future Perspectives. Biomolecules 2022; 12:699. [PMID: 35625626 PMCID: PMC9138761 DOI: 10.3390/biom12050699] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 05/01/2022] [Accepted: 05/07/2022] [Indexed: 12/19/2022] Open
Abstract
Global food systems are under significant pressure to provide enough food, particularly protein-rich foods whose demand is on the rise in times of crisis and inflation, as presently existing due to post-COVID-19 pandemic effects and ongoing conflict in Ukraine and resulting in looming food insecurity, according to FAO. Cultivated meat (CM) and cultivated seafood (CS) are protein-rich alternatives for traditional meat and fish that are obtained via cellular agriculture (CA) i.e., tissue engineering for food applications. Stem and progenitor cells are the building blocks and starting point for any CA bioprocess. This review presents CA-relevant vertebrate cell types and procedures needed for their myogenic and adipogenic differentiation since muscle and fat tissue are the primary target tissues for CM/CS production. The review also describes existing challenges, such as a need for immortalized cell lines, or physical and biochemical parameters needed for enhanced meat/fat culture efficiency and ways to address them.
Collapse
Affiliation(s)
- Teodora Knežić
- Center for Biosystems, BioSense Institute, University of Novi Sad, Dr. Zorana Djindjica 1, 21000 Novi Sad, Serbia; (T.K.); (L.J.); (M.D.)
| | - Ljiljana Janjušević
- Center for Biosystems, BioSense Institute, University of Novi Sad, Dr. Zorana Djindjica 1, 21000 Novi Sad, Serbia; (T.K.); (L.J.); (M.D.)
| | - Mila Djisalov
- Center for Biosystems, BioSense Institute, University of Novi Sad, Dr. Zorana Djindjica 1, 21000 Novi Sad, Serbia; (T.K.); (L.J.); (M.D.)
| | - Supansa Yodmuang
- Research Affairs, Faculty of Medicine, Chulalongkorn University, 1873 Rama 4 Rd, Pathumwan, Bangkok 10330, Thailand;
| | - Ivana Gadjanski
- Center for Biosystems, BioSense Institute, University of Novi Sad, Dr. Zorana Djindjica 1, 21000 Novi Sad, Serbia; (T.K.); (L.J.); (M.D.)
| |
Collapse
|
13
|
The horizon of bone organoid: A perspective on construction and application. Bioact Mater 2022; 18:15-25. [PMID: 35387160 PMCID: PMC8961298 DOI: 10.1016/j.bioactmat.2022.01.048] [Citation(s) in RCA: 71] [Impact Index Per Article: 35.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Revised: 01/29/2022] [Accepted: 01/30/2022] [Indexed: 12/19/2022] Open
Abstract
Bone defects repair and regeneration by various causes such as tumor resection, trauma, degeneration, etc. have always been a key issue in the clinics. As one of the few organs that can regenerate after adulthood, bone itself has a strong regenerative ability. In recent decades, bone tissue engineering technology provides various types of functional scaffold materials and seed cells for bone regeneration and repair, which significantly accelerates the speed and quality of bone regeneration, and many clinical problems are gradually solved. However, the bone metabolism mechanism is complicated, the research duration is long and difficult, which significantly restricts the progress of bone regeneration and repair research. Organoids as a new concept, which is built in vitro with the help of tissue engineering technology based on biological theory, can simulate the complex biological functions of organs in vivo. Once proposed, it shows broad application prospects in the research of organ development, drug screening, mechanism study, and so on. As a complex and special organ, bone organoid construction itself is quite challenging. This review will introduce the characteristics of bone microenvironment, the concept of organoids, focus on the research progress of bone organoids, and propose the strategies for bone organoid construction, study direction, and application prospects. This review introduces the concept and recent progress of bone organoids. This review proposes the study focus and strategies for constructing bone organoids. This review summarizes the potential applications of bone organoids.
Collapse
|
14
|
Dohmen RGJ, Hubalek S, Melke J, Messmer T, Cantoni F, Mei A, Hueber R, Mitic R, Remmers D, Moutsatsou P, Post MJ, Jackisch L, Flack JE. Muscle-derived fibro-adipogenic progenitor cells for production of cultured bovine adipose tissue. NPJ Sci Food 2022; 6:6. [PMID: 35075125 PMCID: PMC8786866 DOI: 10.1038/s41538-021-00122-2] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Accepted: 12/20/2021] [Indexed: 12/30/2022] Open
Abstract
Cultured meat is an emergent technology with the potential for significant environmental and animal welfare benefits. Accurate mimicry of traditional meat requires fat tissue; a key contributor to both the flavour and texture of meat. Here, we show that fibro-adipogenic progenitor cells (FAPs) are present in bovine muscle, and are transcriptionally and immunophenotypically distinct from satellite cells. These two cell types can be purified from a single muscle sample using a simple fluorescence-activated cell sorting (FACS) strategy. FAPs demonstrate high levels of adipogenic potential, as measured by gene expression changes and lipid accumulation, and can be proliferated for a large number of population doublings, demonstrating their suitability for a scalable cultured meat production process. Crucially, FAPs reach a mature level of adipogenic differentiation in three-dimensional, edible hydrogels. The resultant tissue accurately mimics traditional beef fat in terms of lipid profile and taste, and FAPs thus represent a promising candidate cell type for the production of cultured fat.
Collapse
Affiliation(s)
- Richard G J Dohmen
- Mosa Meat B.V., Maastricht, The Netherlands
- Department of Physiology, Maastricht University, Maastricht, The Netherlands
| | - Sophie Hubalek
- Mosa Meat B.V., Maastricht, The Netherlands
- Department of Physiology, Maastricht University, Maastricht, The Netherlands
| | | | - Tobias Messmer
- Mosa Meat B.V., Maastricht, The Netherlands
- Department of Physiology, Maastricht University, Maastricht, The Netherlands
| | | | | | - Rui Hueber
- Mosa Meat B.V., Maastricht, The Netherlands
| | - Rada Mitic
- Mosa Meat B.V., Maastricht, The Netherlands
- Department of Physiology, Maastricht University, Maastricht, The Netherlands
| | | | | | - Mark J Post
- Mosa Meat B.V., Maastricht, The Netherlands
- Department of Physiology, Maastricht University, Maastricht, The Netherlands
| | | | | |
Collapse
|
15
|
Gao Q, Wang L, Wang S, Huang B, Jing Y, Su J. Bone Marrow Mesenchymal Stromal Cells: Identification, Classification, and Differentiation. Front Cell Dev Biol 2022; 9:787118. [PMID: 35047499 PMCID: PMC8762234 DOI: 10.3389/fcell.2021.787118] [Citation(s) in RCA: 53] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 11/25/2021] [Indexed: 12/20/2022] Open
Abstract
Bone marrow mesenchymal stromal cells (BMSCs), identified as pericytes comprising the hematopoietic niche, are a group of heterogeneous cells composed of multipotent stem cells, including osteochondral and adipocyte progenitors. Nevertheless, the identification and classification are still controversial, which limits their application. In recent years, by lineage tracing and single-cell sequencing, several new subgroups of BMSCs and their roles in normal physiological and pathological conditions have been clarified. Key regulators and mechanisms controlling the fate of BMSCs are being revealed. Cross-talk among subgroups of bone marrow mesenchymal cells has been demonstrated. In this review, we focus on recent advances in the identification and classification of BMSCs, which provides important implications for clinical applications.
Collapse
Affiliation(s)
- Qianmin Gao
- Institute of Translational Medicine, Shanghai University, Shanghai, China.,School of Medicine, Shanghai University, Shanghai, China.,School of Life Sciences, Shanghai University, Shanghai, China.,Shanghai University Institute of Advanced Interdisciplinary Materials Science, Shanghai, China
| | - Lipeng Wang
- Institute of Translational Medicine, Shanghai University, Shanghai, China
| | - Sicheng Wang
- Department of Orthopedics, Shanghai Zhongye Hospital, Shanghai, China
| | - Biaotong Huang
- Institute of Translational Medicine, Shanghai University, Shanghai, China.,Shanghai University Institute of Advanced Interdisciplinary Materials Science, Shanghai, China.,Wenzhou Institute of Shanghai University, Wenzhou, China
| | - Yingying Jing
- Institute of Translational Medicine, Shanghai University, Shanghai, China.,Shanghai University Institute of Advanced Interdisciplinary Materials Science, Shanghai, China
| | - Jiacan Su
- Department of Orthopedics Trauma, Shanghai Changhai Hospital, Naval Medical University, Shanghai, China
| |
Collapse
|
16
|
Ghnaimawi S, Rebello L, Baum J, Huang Y. DHA but not EPA induces the trans-differentiation of C2C12 cells into white-like adipocytes phenotype. PLoS One 2021; 16:e0249438. [PMID: 34473703 PMCID: PMC8412409 DOI: 10.1371/journal.pone.0249438] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Accepted: 08/12/2021] [Indexed: 01/21/2023] Open
Abstract
Muscle derived stem cells (MDSCs) and myoblast play an important role in myotube regeneration when muscle tissue is injured. However, these cells can be induced to differentiate into adipocytes once exposed to PPARγ activator like EPA and DHA that are highly suggested during pregnancy. The objective of this study aims at determining the identity of trans-differentiated cells by exploring the effect of EPA and DHA on C2C12 undergoing differentiation into brown and white adipocytes. DHA but not EPA committed C2C12 cells reprograming into white like adipocyte phenotype. Also, DHA promoted the expression of lipolysis regulating genes but had no effect on genes regulating β-oxidation referring to its implication in lipid re-esterification. Furthermore, DHA impaired C2C12 cells differentiation into brown adipocytes through reducing the thermogenic capacity and mitochondrial biogenesis of derived cells independent of UCP1. Accordingly, DHA treated groups showed an increased accumulation of lipid droplets and suppressed mitochondrial maximal respiration and spare respiratory capacity. EPA, on the other hand, reduced myogenesis regulating genes, but no significant differences were observed in the expression of adipogenesis key genes. Likewise, EPA suppressed the expression of WAT signature genes indicating that EPA and DHA have an independent role on white adipogensis. Unlike DHA treatment, EPA supplementation had no effect on the differential of C2C12 cells into brown adipocytes. In conclusion, DHA is a potent adipogenic and lipogenic factor that can change the metabolic profile of muscle cells by increasing myocellular fat.
Collapse
Affiliation(s)
- Saeed Ghnaimawi
- Department of Cell and Molecular Biology Program, University of Arkansas, Fayetteville, North Carolina, United States of America
| | - Lisa Rebello
- Department of Cell and Molecular Biology Program, University of Arkansas, Fayetteville, North Carolina, United States of America
| | - Jamie Baum
- Department of Food Science, Division of Agriculture, University of Arkansas, Fayetteville, AR, United States of America
| | - Yan Huang
- Department of Animal Science, Division of Agriculture, University of Arkansas, Fayetteville, North Carolina, United States of America
| |
Collapse
|
17
|
Reiss J, Robertson S, Suzuki M. Cell Sources for Cultivated Meat: Applications and Considerations throughout the Production Workflow. Int J Mol Sci 2021; 22:7513. [PMID: 34299132 PMCID: PMC8307620 DOI: 10.3390/ijms22147513] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 07/08/2021] [Accepted: 07/09/2021] [Indexed: 12/11/2022] Open
Abstract
Cellular agriculture is an emerging scientific discipline that leverages the existing principles behind stem cell biology, tissue engineering, and animal sciences to create agricultural products from cells in vitro. Cultivated meat, also known as clean meat or cultured meat, is a prominent subfield of cellular agriculture that possesses promising potential to alleviate the negative externalities associated with conventional meat production by producing meat in vitro instead of from slaughter. A core consideration when producing cultivated meat is cell sourcing. Specifically, developing livestock cell sources that possess the necessary proliferative capacity and differentiation potential for cultivated meat production is a key technical component that must be optimized to enable scale-up for commercial production of cultivated meat. There are several possible approaches to develop cell sources for cultivated meat production, each possessing certain advantages and disadvantages. This review will discuss the current cell sources used for cultivated meat production and remaining challenges that need to be overcome to achieve scale-up of cultivated meat for commercial production. We will also discuss cell-focused considerations in other components of the cultivated meat production workflow, namely, culture medium composition, bioreactor expansion, and biomaterial tissue scaffolding.
Collapse
Affiliation(s)
- Jacob Reiss
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI 53706, USA; (J.R.); (S.R.)
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Samantha Robertson
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI 53706, USA; (J.R.); (S.R.)
| | - Masatoshi Suzuki
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI 53706, USA; (J.R.); (S.R.)
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI 53706, USA
- Stem Cell and Regenerative Medicine Center, University of Wisconsin-Madison, Madison, WI 53706, USA
| |
Collapse
|
18
|
Peng DQ, Smith SB, Lee HG. Vitamin A regulates intramuscular adipose tissue and muscle development: promoting high-quality beef production. J Anim Sci Biotechnol 2021; 12:34. [PMID: 33663602 PMCID: PMC7934359 DOI: 10.1186/s40104-021-00558-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Accepted: 01/18/2021] [Indexed: 01/07/2023] Open
Abstract
During growth in cattle, the development of intramuscular adipose tissue and muscle is dependent upon cell hyperplasia (increased number of adipocytes) and hypertrophy (increased size of adipocytes). Based on the results of previous studies, other adipose tissue depots (e.g., perirenal and subcutaneous) develop from the fetal stage primarily as brown adipose tissue. The hyperplastic stage of intramuscular adipose is considered to develop from late pregnancy, but there is no evidence indicating that intramuscular adipose tissue develops initially as brown adipose tissue. Hyperplastic growth of intramuscular adipose continues well into postweaning and is dependent on the timing of the transition to grain-based diets; thereafter, the late-stage development of intramuscular adipose tissue is dominated by hypertrophy. For muscle development, hyperplasia of myoblasts lasts from early (following development of somites in the embryo) to middle pregnancy, after which growth of muscle is the result of hypertrophy of myofibers. Vitamin A is a fat-soluble compound that is required for the normal immunologic function, vision, cellular proliferation, and differentiation. Here we review the roles of vitamin A in intramuscular adipose tissue and muscle development in cattle. Vitamin A regulates both hyperplasia and hypertrophy in in vitro experiments. Vitamin A supplementation at the early stage and restriction at fattening stage generate opposite effects in the beef cattle. Appropriate vitamin A supplementation and restriction strategy increase intramuscular adipose tissue development (i.e., marbling or intramuscular fat) in some in vivo trials. Besides, hyperplasia and hypertrophy of myoblasts/myotubes were affected by vitamin A treatment in in vitro trials. Additionally, some studies reported an interaction between the alcohol dehydrogenase-1C (ADH1C) genotype and vitamin A feed restriction for the development of marbling and/or intramuscular adipose tissue, which was dependent on the timing and level of vitamin A restriction. Therefore, the feed strategy of vitamin A has the visible impact on the marbling and muscle development in the cattle, which will be helpful to promote the quality of the beef.
Collapse
Affiliation(s)
- Dong Qiao Peng
- Department of Animal Science and Technology, Sanghuh College of Life Sciences, Konkuk University, Seoul, 05029, South Korea
| | - Stephen B Smith
- Department of Animal Science, Texas A&M University, College Station, TX, 77843, USA
| | - Hong Gu Lee
- Department of Animal Science and Technology, Sanghuh College of Life Sciences, Konkuk University, Seoul, 05029, South Korea.
| |
Collapse
|
19
|
Pant R, Firmal P, Shah VK, Alam A, Chattopadhyay S. Epigenetic Regulation of Adipogenesis in Development of Metabolic Syndrome. Front Cell Dev Biol 2021; 8:619888. [PMID: 33511131 PMCID: PMC7835429 DOI: 10.3389/fcell.2020.619888] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Accepted: 12/14/2020] [Indexed: 12/12/2022] Open
Abstract
Obesity is one of the biggest public health concerns identified by an increase in adipose tissue mass as a result of adipocyte hypertrophy and hyperplasia. Pertaining to the importance of adipose tissue in various biological processes, any alteration in its function results in impaired metabolic health. In this review, we discuss how adipose tissue maintains the metabolic health through secretion of various adipokines and inflammatory mediators and how its dysfunction leads to the development of severe metabolic disorders and influences cancer progression. Impairment in the adipocyte function occurs due to individuals' genetics and/or environmental factor(s) that largely affect the epigenetic profile leading to altered gene expression and onset of obesity in adults. Moreover, several crucial aspects of adipose biology, including the regulation of different transcription factors, are controlled by epigenetic events. Therefore, understanding the intricacies of adipogenesis is crucial for recognizing its relevance in underlying disease conditions and identifying the therapeutic interventions for obesity and metabolic syndrome.
Collapse
Affiliation(s)
- Richa Pant
- National Centre for Cell Science, SP Pune University Campus, Pune, India
| | - Priyanka Firmal
- National Centre for Cell Science, SP Pune University Campus, Pune, India
| | - Vibhuti Kumar Shah
- National Centre for Cell Science, SP Pune University Campus, Pune, India
| | - Aftab Alam
- Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States
| | - Samit Chattopadhyay
- National Centre for Cell Science, SP Pune University Campus, Pune, India.,Department of Biological Sciences, BITS Pilani, Goa, India
| |
Collapse
|
20
|
Jakab J, Miškić B, Mikšić Š, Juranić B, Ćosić V, Schwarz D, Včev A. Adipogenesis as a Potential Anti-Obesity Target: A Review of Pharmacological Treatment and Natural Products. Diabetes Metab Syndr Obes 2021; 14:67-83. [PMID: 33447066 PMCID: PMC7802907 DOI: 10.2147/dmso.s281186] [Citation(s) in RCA: 120] [Impact Index Per Article: 40.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 12/09/2020] [Indexed: 12/14/2022] Open
Abstract
Obesity is recognized as a severe threat to overall human health and is associated with type 2 diabetes mellitus, dyslipidemia, hypertension, and cardiovascular diseases. Abnormal expansion of white adipose tissue involves increasing the existing adipocytes' cell size or increasing the number through the differentiation of new adipocytes. Adipogenesis is a process of proliferation and differentiation of adipocyte precursor cells in mature adipocytes. As a key process in determining the number of adipocytes, it is a possible therapeutic approach for obesity. Therefore, it is necessary to identify the molecular mechanisms involved in adipogenesis that could serve as suitable therapeutic targets. Reducing bodyweight is regarded as a major health benefit. Limited efficacy and possible side effects and drug interactions of available anti-obesity treatment highlight a constant need for finding novel efficient and safe anti-obesity ingredients. Numerous studies have recently investigated the inhibitory effects of natural products on adipocyte differentiation and lipid accumulation. Possible anti-obesity effects of natural products include the induction of apoptosis, cell-cycle arrest or delayed progression, and interference with transcription factor cascade or intracellular signaling pathways during the early phase of adipogenesis.
Collapse
Affiliation(s)
- Jelena Jakab
- Faculty of Medicine, Josip Juraj Strossmayer University of Osijek, Osijek, Croatia
- Faculty of Dental Medicine and Health Osijek, Josip Juraj Strossmayer University of Osijek, Osijek, Croatia
- Correspondence: Jelena Jakab Faculty of Medicine, Josip Juraj Strossmayer University of Osijek, Crkvena 21, Osijek31 000, CroatiaTel +385 91 224 1502 Email
| | - Blaženka Miškić
- Faculty of Dental Medicine and Health Osijek, Josip Juraj Strossmayer University of Osijek, Osijek, Croatia
- Department of Internal Medicine, General Hospital “Dr. Josip Benčević”, Slavonski Brod, Croatia
| | - Štefica Mikšić
- Faculty of Dental Medicine and Health Osijek, Josip Juraj Strossmayer University of Osijek, Osijek, Croatia
| | - Brankica Juranić
- Faculty of Dental Medicine and Health Osijek, Josip Juraj Strossmayer University of Osijek, Osijek, Croatia
- Department of Cardiology, University Hospital Osijek, Osijek, Croatia
| | - Vesna Ćosić
- Faculty of Dental Medicine and Health Osijek, Josip Juraj Strossmayer University of Osijek, Osijek, Croatia
| | - Dragan Schwarz
- Faculty of Dental Medicine and Health Osijek, Josip Juraj Strossmayer University of Osijek, Osijek, Croatia
- Special Hospital Radiochirurgia Zagreb, Zagreb, Croatia
| | - Aleksandar Včev
- Faculty of Dental Medicine and Health Osijek, Josip Juraj Strossmayer University of Osijek, Osijek, Croatia
| |
Collapse
|
21
|
Liu H, Li B, Qiao L, Liu J, Ren D, Liu W. miR-340-5p inhibits sheep adipocyte differentiation by targeting ATF7. Anim Sci J 2020; 91:e13462. [PMID: 33190272 DOI: 10.1111/asj.13462] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 08/02/2020] [Accepted: 08/13/2020] [Indexed: 01/01/2023]
Abstract
Several microRNAs (miRNAs) have been identified to play roles in adipocyte differentiation. However, little is known about their involvement in the differentiation of ovine intramuscular adipocytes. Here, the role of one such miRNA, miR-340-5p, in ovine adipocyte differentiation was investigated. Stromal vascular (SV) cells were isolated from skeletal muscle tissues of 1-month-old lambs and induced to differentiate into mature adipocytes. miRNA mimics and inhibitors were used for miR-340-5p overexpression and knockdown assays. For overexpression and knockdown of activating transcription factor 7 (ATF7), lentivirus infection was performed. Luciferase reporter assay was performed to determine the relationship between miR-340-5p and ATF7. The expression of adipogenesis marker genes, PPARγ, C/EBPα, FABP4, ADIPOQ, and ACC, and formation of lipid droplets were detected after the overexpression and inhibition of miR-340-5p, or upon overexpression or knockdown of ATF7. miR-340-5p inhibited the expression of the marker genes and the formation of lipid droplets. ATF7 positively regulated the expression of the marker genes and the formation of lipids. Thus, ATF7 is the target of miR-340-5p in sheep. Overall, these findings indicate that miR-340-5p acts as an inhibitor of the differentiation of intramuscular adipocytes by targeting ATF7. Our study provides a new theoretical basis for improving sheep meat quality.
Collapse
Affiliation(s)
- Haodong Liu
- College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Taigu, China
| | - Baojun Li
- College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Taigu, China
| | - Liying Qiao
- College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Taigu, China
| | - Jianhua Liu
- College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Taigu, China
| | - Duanyang Ren
- College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Taigu, China
| | - Wenzhong Liu
- College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Taigu, China
| |
Collapse
|
22
|
Effects of Maternal Supplementation with an Injectable Trace Mineral Containing Copper, Manganese, Zinc, and Selenium on Subsequent Steer Finishing Phase Performance and Carcass Characteristics. Animals (Basel) 2020; 10:ani10122226. [PMID: 33261026 PMCID: PMC7760028 DOI: 10.3390/ani10122226] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 11/13/2020] [Accepted: 11/23/2020] [Indexed: 12/29/2022] Open
Abstract
Simple Summary The persistent effects of maternal nutrition on subsequent offspring health and performance have drawn great attention in both the livestock industry and human health field in recent years. Trace minerals play very important roles in nutrition and regulate many critical biological processes. Therefore, trace mineral status of the dam has the potential to influence early growth and development of the fetus, which leads to long-lasting effects on animal health and growth performance. This study demonstrated that maternal supplementation of trace minerals increased the percentage of carcasses graded as USDA Choice or greater, but maternal trace mineral injections had limited effects on finishing phase growth performance and other carcass characteristics of the offspring. Stakeholders of the cow/calf and feedlot operations should consider these results as they make decisions on maternal trace mineral administrations. Abstract The objective of this study was to investigate effects of maternal supplementation with an injectable trace mineral (Cu, Mn, Zn, and Se) on subsequent steer performance during the finishing phase. Seventy-six Angus cross steers (initial body weight 249 ± 41.5 kg) from dams administered either an injectable trace mineral (TM; Multimin 90) or sterilized physiological saline (CON) during prepartum stage were used. Individual feed intake during the finishing phase were recorded with GrowSafe feed bunks. Blood and liver biopsy samples were collected to evaluate trace mineral status. Steers were slaughtered at 413 ± 26 days of age and carcass data were obtained at a commercial abattoir. Growth performance or mineral status of the steers during the finishing phase was not affected (p ≥ 0.14) by maternal treatments. Carcass characteristics were not different (p ≥ 0.18), except steers from TM dams had greater (p = 0.05) percentage of carcasses graded as Choice or greater. In conclusion, maternal supplementation of an injectable trace mineral increased the percentage of carcasses graded as Choice or greater, other than that, maternal supplementation had limited influence on finishing phase growth performance, trace mineral status, or carcass characteristics of the subsequent steer progeny.
Collapse
|
23
|
IDH2 Deficiency Is Critical in Myogenesis and Fatty Acid Metabolism in Mice Skeletal Muscle. Int J Mol Sci 2020; 21:ijms21165596. [PMID: 32764267 PMCID: PMC7460611 DOI: 10.3390/ijms21165596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 07/31/2020] [Accepted: 08/01/2020] [Indexed: 11/22/2022] Open
Abstract
Mitochondrial NADP+-dependent isocitrate dehydrogenase (IDH2) catalyzes the oxidative decarboxylation of isocitrate into α-ketoglutarate with concurrent reduction of NADP+ to NADPH. However, it is not fully understood how IDH2 is intertwined with muscle development and fatty acid metabolism. Here, we examined the effects of IDH2 knockout (KO) on skeletal muscle energy homeostasis. Calf skeletal muscle samples from 10-week-old male IDH2 KO and wild-type (WT; C57BL/6N) mice were harvested, and the ratio of skeletal muscle weight to body and the ratio of mitochondrial to nucleic DNA were measured. In addition, genes involved in myogenesis, mitochondria biogenesis, adipogenesis, and thermogenesis were compared. Results showed that the ratio of skeletal muscle weight to body weight was lower in IDH2 KO mice than those in WT mice. Of note, a noticeable shift in fiber size distribution was found in IDH2 KO mice. Additionally, there was a trend of a decrease in mitochondrial content in IDH2 KO mice than in WT mice (p = 0.09). Further, mRNA expressions for myogenesis and mitochondrial biogenesis were either decreased or showed a trend of decrease in IDH2 KO mice. Moreover, genes for adipogenesis pathway (Pparg, Znf423, and Fat1) were downregulated in IDH2 KO mice. Interestingly, mRNA and protein expression of uncoupling protein 1 (UCP1), a hallmark of thermogenesis, were remarkably increased in IDH2 KO mice. In line with the UCP1 expression, IDH2 KO mice showed higher rectal temperature than WT mice under cold stress. Taken together, IDH2 deficiency may affect myogenesis, possibly due to impairments of muscle generation and abnormal fatty acid oxidation as well as thermogenesis in muscle via upregulation of UCP1.
Collapse
|
24
|
Adipogenesis, fibrogenesis and myogenesis related gene expression in longissimus muscle of high and low marbling beef cattle. Livest Sci 2019. [DOI: 10.1016/j.livsci.2019.09.032] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
25
|
Review: Enhancing intramuscular fat development via targeting fibro-adipogenic progenitor cells in meat animals. Animal 2019; 14:312-321. [PMID: 31581971 DOI: 10.1017/s175173111900209x] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
In the livestock industry, subcutaneous and visceral fat pads are considered as wastes, while intramuscular fat or marbling fat is essential for improving flavor and palatability of meat. Thus, strategies for optimizing fat deposition are needed. Intramuscular adipocytes provide sites for lipid deposition and marbling formation. In the present article, we addressed the origin and markers of intramuscular adipocyte progenitors - fibro-adipogenic progenitors (FAPs), as well as the latest progresses in mechanisms regulating the proliferation and differentiation of intramuscular FAPs. Finally, by targeting intramuscular FAPs, possible nutritional manipulations to improve marbling fat deposition are discussed. Despite recent progresses, the properties and regulation of intramuscular FAPs in livestock remain poorly understood and deserve further investigation.
Collapse
|
26
|
Raja Gopal Reddy M, Mahesh M, Manne M, Putcha UK, Jeyakumar SM. Vitamin A and its metabolic pathway play a determinant role in high-fructose-induced triglyceride accumulation of the visceral adipose depot of male Wistar rats. Cell Biochem Funct 2019; 37:578-590. [PMID: 31495961 DOI: 10.1002/cbf.3434] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 08/07/2019] [Accepted: 08/22/2019] [Indexed: 12/17/2022]
Abstract
Here, we tested a hypothesis that vitamin A and/or its metabolic pathways are involved in the high-fructose-mediated alteration in adipose tissue biology. For this purpose, weanling male Wistar rats were provided with one of the following diets: control (C), control with vitamin A deficiency (C-VAD), high fructose (HFr), and HFr with VAD (HFr-VAD) for 16 weeks, except that half of the C-VAD diet-fed rats were shifted to HFr diet (C-VAD(s)HFr), after 8-week period. Compared with control, feeding of HFr diet significantly increased the triglyceride content (P ≤ .01) and thus adipocyte size (hypertrophy) (P ≤ .001) in visceral adipose depot; retroperitoneal white adipose tissue (RPWAT) and these changes were corroborated with de novo lipogenesis, as evidenced by the increased glycerol-3-phosphate dehydrogenase activity (P ≤ .01) and up-regulation of lipogenic pathway transcripts, fructose transporter, and aldehyde dehydrogenase 1 A1. On the contrary, the absence of vitamin A in the HFr diet (HFr-VAD) failed to exert these changes; however, it induced adipocyte hyperplasia. Further, vitamin A deficiency-mediated changes were reversed by replenishment, as evident from the group that was shifted from C-VAD to HFr diet. In conclusion, vitamin A and its metabolic pathway play a key determinant role in the high-fructose-induced triglyceride accumulation and adipocyte hypertrophy of visceral white adipose depot. SIGNIFICANCE OF THE STUDY: Here, we report the metabolic impact of high-fructose feeding under vitamin A-sufficient and vitamin A-deficient conditions. Feeding of high-fructose diet induced triglyceride accumulation and adipocyte hypertrophy of the visceral white adipose depots. These changes corroborated with augmented expression of vitamin A and lipid metabolic pathway genes. Contrarily, absence of vitamin A in the high-fructose diet did not elicit such responses, while vitamin A replenishment reversed the changes exerted by vitamin A deficiency. To our knowledge, this is the first study to report the role of vitamin A and its metabolic pathway in the high-fructose-induced triglyceride synthesis and its accumulation in visceral adipose depot and thus provide a new insight and scope to understand these nutrients interaction in clinical conditions.
Collapse
Affiliation(s)
| | - Malleswarapu Mahesh
- Lipid Biochemistry Division, ICMR-National Institute of Nutrition, Hyderabad, India
| | - Munikumar Manne
- Biomedical Informatics Centre, ICMR-National Institute of Nutrition, Hyderabad, India
| | - Uday Kumar Putcha
- Pathology Division, ICMR-National Institute of Nutrition, Hyderabad, India
| | | |
Collapse
|
27
|
Ben-Arye T, Levenberg S. Tissue Engineering for Clean Meat Production. FRONTIERS IN SUSTAINABLE FOOD SYSTEMS 2019. [DOI: 10.3389/fsufs.2019.00046] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
|
28
|
Fu X, Li C, Liu Q, McMillin KW. GROWTH AND DEVELOPMENT SYMPOSIUM: STEM AND PROGENITOR CELLS IN ANIMAL GROWTH: The regulation of beef quality by resident progenitor cells1. J Anim Sci 2019; 97:2658-2673. [PMID: 30982893 PMCID: PMC6541817 DOI: 10.1093/jas/skz111] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Accepted: 04/01/2019] [Indexed: 12/11/2022] Open
Abstract
The intramuscular adipose tissue deposition in the skeletal muscle of beef cattle is a highly desired trait essential for high-quality beef. In contrast, the excessive accumulation of crosslinked collagen in intramuscular connective tissue contributes to beef toughness. Recent studies revealed that adipose tissue and connective tissue share an embryonic origin in mice and may be derived from a common immediate bipotent precursor in mice and humans. Having the same linkages in the development of adipose tissue and connective tissue in beef, the lineage commitment and differentiation of progenitor cells giving rise to these tissues may directly affect beef quality. It has been shown that these processes are regulated by some key transcription regulators and are subjective to epigenetic modifications such as DNA methylation, histone modifications, and microRNAs. Continued exploration of relevant regulatory pathways is very important for the identification of mechanisms influencing meat quality and the development of proper management strategies for beef quality improvement.
Collapse
Affiliation(s)
- Xing Fu
- School of Animal Sciences, Louisiana State University Agricultural Center, Baton Rouge, LA
| | - Chaoyang Li
- School of Animal Sciences, Louisiana State University Agricultural Center, Baton Rouge, LA
| | - Qianglin Liu
- School of Animal Sciences, Louisiana State University Agricultural Center, Baton Rouge, LA
| | - Kenneth W McMillin
- School of Animal Sciences, Louisiana State University Agricultural Center, Baton Rouge, LA
| |
Collapse
|
29
|
Fu YY, Hu BH, Chen KL, Li HX. Chemerin induces lipolysis through ERK1/2 pathway in intramuscular mature adipocytes of dairy bull calves. J Cell Biochem 2019; 120:1122-1132. [PMID: 30256444 DOI: 10.1002/jcb.27506] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2018] [Accepted: 07/18/2018] [Indexed: 01/24/2023]
Abstract
The adipokine Chemerin has been reported to regulate differentiation and metabolism of adipocytes, but the mechanism underlying lipolysis is still largely unknown. The purpose of this study was to explore whether ERK1/2 pathway is involved in regulating Chemerin during bovine intramuscular mature adipocyte lipolysis. Intramuscular mature adipocytes of dairy bull calves were cultured in vitro and were treated with Chemerin or U0126, which is an inhibitor of ERK1/2 pathway. The results showed that TG content in cells was significantly decreased, glycerol and free fatty acid were significantly increased in cell culture media, and the expression of phosphorylated ERK1/2 in cells was increased in Chemerin-treated group, suggested that ERK1/2 pathway was involved in regulation of lipolysis by Chemerin. In addition, the expression of lipolytic-related critical factors ATGL, HSL, LPL, PPARα, UCP3, and CPT1 were upregulated, but the expression of adipogenic key factors, including PPARγ and C/EBPα were downregulated by Chemerin. Interestingly, all the effects of Chemerin on genes expression in intramuscular mature adipocytes or fat tissue were inhibited by U0126, showed that the function of Chemerin to promote adipose decomposition will be significantly weakened if the ERK1/2 pathway is suppressed, and confirmed that ERK1/2 pathway is involved in mediate Chemerin-enhanced lipolysis. In conclusion, the study demonstrated that Chemerin induce intramuscular mature adipocytes lipolysis through activation of the ERK1/2 pathway. Our research at least provide partial mechanisms of Chemerin on lipolysis and deposition of intramuscular fat tissue of dairy bull calves.
Collapse
Affiliation(s)
- Yuan-Yuan Fu
- Department of Animal Genetics and Breeding, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Bian-Hong Hu
- Department of Animal Genetics and Breeding, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Kun-Lin Chen
- Department of Animal Genetics and Breeding, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Hui-Xia Li
- Department of Animal Genetics and Breeding, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| |
Collapse
|
30
|
Van Bibber-Krueger CL, Vahl CI, Narayanan SK, Amachawadi RG, Taylor EA, Scott HM, Drouillard JS. Effects of supplemental zinc sulfate on growth performance, carcass characteristics, and antimicrobial resistance in feedlot heifers. J Anim Sci 2019; 97:424-436. [PMID: 30388223 PMCID: PMC6313150 DOI: 10.1093/jas/sky411] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Accepted: 10/16/2018] [Indexed: 11/13/2022] Open
Abstract
Effects of supplemental Zn as Zn sulfate on feedlot performance, carcass traits, and antimicrobial resistance were evaluated using 480 crossbred heifers (BW = 385 kg ± 13.08) in a randomized complete block design. Heifers were blocked by BW and randomly assigned within block to diets with 0, 30, 60, or 90 mg supplemental Zn/kg DM. Heifers were housed in dirt-surfaced pens (20 animals per pen; 6 pens per treatment) equipped with fence-line feed bunks and automatic water fountains. Heifers were fed once daily to ensure ad libitum intake. Plasma was collected on day 0 from five randomly selected heifers per pen and repeated on days 63 and 115 to determine plasma Zn concentrations. Random samples of freshly voided feces were collected from the surface of each pen the day of harvest to determine antibiotic resistance. Heifers were transported on day 144 to a commercial abattoir where hot carcass weight (HCW) and incidence of liver abscesses were recorded at harvest and HCW, dressed yield, ribeye area, 12th rib fat, quality and yield grades were recorded after 36 h of refrigeration. Plasma Zn concentration increased (P = 0.02) linearly in response to increasing concentrations of dietary Zn. Final BW and ADG were unaffected by supplementation (P ≥ 0.29). Quantified levels of resistance to ceftriaxone and tetracycline among fecal Escherichia coli were not impacted (P > 0.05) by dietary zinc concentrations. Increasing Zn concentrations tended to decrease (linear effect, P = 0.07) DMI, resulting in a linear (P = 0.03) and tendency for quadratic (P = 0.12) improvement in feed efficiency with increasing Zn concentration. No differences were detected for HCW, dressed yield, ribeye area, 12th rib fat, percentages of carcasses grading Select or Choice, or yield grade (P > 0.53), but added Zn tended to affect percentage of carcasses that graded Prime, peaking at 60 mg/kg added Zn (quadratic effect, P = 0.07). In vitro fermentations were performed using ruminal fluid cultures containing 0, 30, 60, 90, 120, or 150 mg Zn/kg substrate DM to determine impact of Zn on gas production, VFA concentrations, and in vitro DM disappearance (IVDMD). There were no effects of Zn on in vitro gas production, IVDMD, or most VFA (P > 0.15), but isovalerate decreased linearly in response to added Zn (P = 0.05). Supplementing finishing heifers up to 60 mg Zn/kg diet DM improved feed efficiency compared to other treatments.
Collapse
Affiliation(s)
| | - Chris I Vahl
- Department of Statistics, Kansas State University, Manhattan, KS
| | - Sanjeev K Narayanan
- Department of Comparative Pathobiology, Purdue University, West Lafayette, IN
| | | | - Ethan A Taylor
- Department of Veterinary Pathobiology, Texas A & M University, College Station, TX
| | - Harvey Morgan Scott
- Department of Veterinary Pathobiology, Texas A & M University, College Station, TX
| | - James S Drouillard
- Department of Animal Sciences and Industry, Kansas State University, Manhattan, KS
| |
Collapse
|
31
|
Harris CL, Wang B, Deavila JM, Busboom JR, Maquivar M, Parish SM, McCann B, Nelson ML, Du M. Vitamin A administration at birth promotes calf growth and intramuscular fat development in Angus beef cattle. J Anim Sci Biotechnol 2018; 9:55. [PMID: 30062009 PMCID: PMC6055337 DOI: 10.1186/s40104-018-0268-7] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2017] [Accepted: 05/25/2018] [Indexed: 01/05/2023] Open
Abstract
Background Marbling, or intramuscular fat, is an important factor contributing to the palatability of beef. Vitamin A, through its active metabolite, retinoic acid, promotes the formation of new fat cells (adipogenesis). As intramuscular adipogenesis is active during the neonatal stage, we hypothesized that vitamin A administration during the neonatal stage would enhance intramuscular adipogenesis and marbling. Methods Angus steer calves (n = 30), in a completely randomized design, were randomly allotted to three treatment groups at birth, receiving 0, 150,000, or 300,000 IU of vitamin A at both birth and one month of age. A biopsy of the biceps femoris muscle was collected at two months of age. After weaning at 210 d of age, steers were fed a backgrounding diet in a feedlot until 308 d of age, when they were transitioned to a high concentrate finishing diet and implanted with trenbolone/estradiol/tylosin mixture. Steers were harvested at an average of 438 d of age. All diets were formulated to meet nutrient requirements. Results Weaning weight and weight during the backgrounding phase were linearly increased (P < 0.05) by vitamin A level, though no difference in body weight was observed at harvest. Intramuscular fat of steers at 308 d of age, measured by ultrasound, quadratically increased (P < 0.05) with vitamin A level from 4.0±0.26 % to 4.9±0.26 %. Similarly, carcass marbling score in the ribeye quadratically increased (P < 0.05). Conclusion Administration of vitamin A at birth increased weaning weight and enhanced marbling fat development. Thus, vitamin A administration provides a practical method for increasing marbling and early growth of beef cattle.
Collapse
Affiliation(s)
- Corrine L Harris
- 1Department of Animal Sciences, Washington State University, Pullman, WA 99164 USA
| | - Bo Wang
- 1Department of Animal Sciences, Washington State University, Pullman, WA 99164 USA.,2State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193 People's Republic of China
| | - Jeneane M Deavila
- 1Department of Animal Sciences, Washington State University, Pullman, WA 99164 USA
| | - Jan R Busboom
- 1Department of Animal Sciences, Washington State University, Pullman, WA 99164 USA
| | - Martin Maquivar
- 1Department of Animal Sciences, Washington State University, Pullman, WA 99164 USA
| | - Steven M Parish
- 3College of Veterinary Medicine, Washington State University, Pullman, WA 99164 USA
| | - Brent McCann
- 1Department of Animal Sciences, Washington State University, Pullman, WA 99164 USA
| | - Mark L Nelson
- 1Department of Animal Sciences, Washington State University, Pullman, WA 99164 USA
| | - Min Du
- 1Department of Animal Sciences, Washington State University, Pullman, WA 99164 USA
| |
Collapse
|
32
|
Park SJ, Beak SH, Jung DJS, Kim SY, Jeong IH, Piao MY, Kang HJ, Fassah DM, Na SW, Yoo SP, Baik M. Genetic, management, and nutritional factors affecting intramuscular fat deposition in beef cattle - A review. ASIAN-AUSTRALASIAN JOURNAL OF ANIMAL SCIENCES 2018; 31:1043-1061. [PMID: 29879830 PMCID: PMC6039335 DOI: 10.5713/ajas.18.0310] [Citation(s) in RCA: 147] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Accepted: 05/10/2018] [Indexed: 02/07/2023]
Abstract
Intramuscular fat (IMF) content in skeletal muscle including the longissimus dorsi muscle (LM), also known as marbling fat, is one of the most important factors determining beef quality in several countries including Korea, Japan, Australia, and the United States. Genetics and breed, management, and nutrition affect IMF deposition. Japanese Black cattle breed has the highest IMF content in the world, and Korean cattle (also called Hanwoo) the second highest. Here, we review results of research on genetic factors (breed and sex differences and heritability) that affect IMF deposition. Cattle management factors are also important for IMF deposition. Castration of bulls increases IMF deposition in most cattle breeds. The effects of several management factors, including weaning age, castration, slaughter weight and age, and environmental conditions on IMF deposition are also reviewed. Nutritional factors, including fat metabolism, digestion and absorption of feed, glucose/starch availability, and vitamin A, D, and C levels are important for IMF deposition. Manipulating IMF deposition through developmental programming via metabolic imprinting is a recently proposed nutritional method to change potential IMF deposition during the fetal and neonatal periods in rodents and domestic animals. Application of fetal nutritional programming to increase IMF deposition of progeny in later life is reviewed. The coordination of several factors affects IMF deposition. Thus, a combination of several strategies may be needed to manipulate IMF deposition, depending on the consumer’s beef preference. In particular, stage-specific feeding programs with concentrate-based diets developed by Japan and Korea are described in this article.
Collapse
Affiliation(s)
- Seung Ju Park
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Sciences, College of Agriculture and Life Sciences, Seoul National University, Seoul 08826, Korea
| | - Seok-Hyeon Beak
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Sciences, College of Agriculture and Life Sciences, Seoul National University, Seoul 08826, Korea
| | - Da Jin Sol Jung
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Sciences, College of Agriculture and Life Sciences, Seoul National University, Seoul 08826, Korea
| | - Sang Yeob Kim
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Sciences, College of Agriculture and Life Sciences, Seoul National University, Seoul 08826, Korea
| | - In Hyuk Jeong
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Sciences, College of Agriculture and Life Sciences, Seoul National University, Seoul 08826, Korea
| | - Min Yu Piao
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Sciences, College of Agriculture and Life Sciences, Seoul National University, Seoul 08826, Korea
| | - Hyeok Joong Kang
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Sciences, College of Agriculture and Life Sciences, Seoul National University, Seoul 08826, Korea
| | - Dilla Mareistia Fassah
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Sciences, College of Agriculture and Life Sciences, Seoul National University, Seoul 08826, Korea
| | - Sang Weon Na
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Sciences, College of Agriculture and Life Sciences, Seoul National University, Seoul 08826, Korea
| | - Seon Pil Yoo
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Sciences, College of Agriculture and Life Sciences, Seoul National University, Seoul 08826, Korea
| | - Myunggi Baik
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Sciences, College of Agriculture and Life Sciences, Seoul National University, Seoul 08826, Korea.,Institutes of Green Bio Science Technology, Pyeongchang 25354, Korea
| |
Collapse
|
33
|
Van Bibber-Krueger CL, Amachawadi RG, Scott HM, Gonzalez JM, Drouillard JS. Interactive effects of supplemental Zn sulfate and ractopamine hydrochloride on growth performance, carcass traits, and plasma urea nitrogen in feedlot heifers. J Anim Sci 2018; 95:4638-4645. [PMID: 29108074 DOI: 10.2527/jas2017.1764] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Interactive effects of supplemental Zn and ractopamine hydrochloride (RH) were evaluated using 156 crossbred heifers (initial BW = 527 kg ± 6.61; gross BW × 0.96) to determine the impact on feedlot performance, plasma urea nitrogen (PUN), and carcass characteristics. The study was conducted as a randomized complete block design with a 2 × 2 factorial arrangement of treatments. Factors consisted of 1) 30 or 100 mg supplemental Zn/kg diet DM (30Zn or 100Zn) as Zn sulfate and 2) 0 or 200 mg RH/heifer daily. Heifers were blocked by BW and assigned randomly within block to treatments for a 43-d trial. Heifers were housed in partially covered feeding pens (3 heifers/pen; 13 pens/treatment) and provided ad libitum access to feed. Ractopamine hydrochloride was fed for 42 d and removed from the diet until cattle were harvested on d 43. Zinc treatments were fed until harvest. Plasma samples were collected on d 0 and 36 to assess changes in plasma Zn and PUN. On d 43, heifers were weighed, then transported to a commercial abattoir where HCW and incidence of liver abscesses were recorded. Carcass data were collected after 32 h of refrigeration. No Zn × RH interactions were observed for plasma Zn or PUN ( ≥ 0.58); however, there was a tendency for a RH × day interaction for PUN ( = 0.08). Supplementing 100Zn resulted in increased plasma Zn ( = 0.02) compared to 30Zn. No RH × Zn interactions were observed for feedlot performance ( ≥ 0.24). Final BW and ADG increased with RH supplementation ( ≤ 0.02), but DMI was not affected ( = 0.63); thus, feed efficiency improved ( < 0.01) when heifers were fed RH. Supplementing 100Zn tended to reduce ADG ( = 0.07) but did not affect other measures of feedlot performance ( ≥ 0.12). Zinc × RH interactions were observed for LM area and yield grade ( ≤ 0.01); LM area decreased and yield grade increased when heifers were supplemented 100Zn with no RH compared to other treatments. A tendency for a Zn × RH interaction was observed for dressed yield ( = 0.08), but no other interactions or effects of Zn were detected for carcass traits ( ≥ 0.11). Supplementing RH increased HCW ( = 0.03) but did not affect other carcass traits ( ≥ 0.13). In conclusion, supplemental Zn had little impact on feedlot performance or PUN concentration but may alter muscle and fat deposition when fed in conjunction with RH.
Collapse
|
34
|
Bond HM, Scicchitano S, Chiarella E, Amodio N, Lucchino V, Aloisio A, Montalcini Y, Mesuraca M, Morrone G. ZNF423: A New Player in Estrogen Receptor-Positive Breast Cancer. Front Endocrinol (Lausanne) 2018; 9:255. [PMID: 29867779 PMCID: PMC5968090 DOI: 10.3389/fendo.2018.00255] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Accepted: 05/03/2018] [Indexed: 01/13/2023] Open
Abstract
Preventive therapy can target hormone-responsive breast cancer (BC) by treatment with selective estrogen receptor modulators (SERMs) and reduce the incidence of BC. Genome-wide association studies have identified single nucleotide polymorphisms (SNPs) with relevant predictive values, SNPs in the ZNF423 gene were associated with decreased risk of BC during SERM therapy, and SNPs in the Cathepsin O gene with an increased risk. ZNF423, which was not previously associated with BC is a multifunctional transcription factor known to have a role in development, neurogenesis, and adipogenesis and is implicated in other types of cancer. ZNF423 is transcriptionally controlled by the homolog ZNF521, early B cell factor transcription factor, epigenetic silencing of the promoter by CpG island hyper-methylation, and also by ZNF423 itself in an auto-regulatory loop. In BC cells, ZNF423 expression is found to be induced by estrogen, dependent on the binding of the estrogen receptor and calmodulin-like 3 to SNPs in ZNP423 intronic sites in proximity to consensus estrogen response elements. ZNF423 has also been shown to play a mechanistic role by trans-activating the tumor suppressor BRCA1 and thus modulating the DNA damage response. Even though recent extensive trial studies did not classify these SNPs with the highest predictive values, for inclusion in polygenic SNP analysis, the mechanism unveiled in these studies has introduced ZNF423 as a factor important in the control of the estrogen response. Here, we aim at providing an overview of ZNF423 expression and functional role in human malignancies, with a specific focus on its implication in hormone-responsive BC.
Collapse
Affiliation(s)
- Heather M. Bond
- Laboratory of Molecular Haematopoiesis and Stem Cell Biology, Department of Experimental and Clinical Medicine, Magna Græcia University of Catanzaro, Catanzaro, Italy
- *Correspondence: Heather M. Bond, ; Maria Mesuraca, ; Giovanni Morrone,
| | - Stefania Scicchitano
- Laboratory of Molecular Haematopoiesis and Stem Cell Biology, Department of Experimental and Clinical Medicine, Magna Græcia University of Catanzaro, Catanzaro, Italy
| | - Emanuela Chiarella
- Laboratory of Molecular Haematopoiesis and Stem Cell Biology, Department of Experimental and Clinical Medicine, Magna Græcia University of Catanzaro, Catanzaro, Italy
| | - Nicola Amodio
- Laboratory of Medical Oncology, Department of Experimental and Clinical Medicine, Magna Græcia University of Catanzaro, Catanzaro, Italy
| | - Valeria Lucchino
- Laboratory of Molecular Haematopoiesis and Stem Cell Biology, Department of Experimental and Clinical Medicine, Magna Græcia University of Catanzaro, Catanzaro, Italy
| | - Annamaria Aloisio
- Laboratory of Molecular Haematopoiesis and Stem Cell Biology, Department of Experimental and Clinical Medicine, Magna Græcia University of Catanzaro, Catanzaro, Italy
| | - Ylenia Montalcini
- Laboratory of Molecular Haematopoiesis and Stem Cell Biology, Department of Experimental and Clinical Medicine, Magna Græcia University of Catanzaro, Catanzaro, Italy
| | - Maria Mesuraca
- Laboratory of Molecular Haematopoiesis and Stem Cell Biology, Department of Experimental and Clinical Medicine, Magna Græcia University of Catanzaro, Catanzaro, Italy
- *Correspondence: Heather M. Bond, ; Maria Mesuraca, ; Giovanni Morrone,
| | - Giovanni Morrone
- Laboratory of Molecular Haematopoiesis and Stem Cell Biology, Department of Experimental and Clinical Medicine, Magna Græcia University of Catanzaro, Catanzaro, Italy
- *Correspondence: Heather M. Bond, ; Maria Mesuraca, ; Giovanni Morrone,
| |
Collapse
|
35
|
Yanting C, Yang QY, Ma GL, Du M, Harrison JH, Block E. Dose- and type-dependent effects of long-chain fatty acids on adipogenesis and lipogenesis of bovine adipocytes. J Dairy Sci 2017; 101:1601-1615. [PMID: 29153512 DOI: 10.3168/jds.2017-13312] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Accepted: 10/05/2017] [Indexed: 12/17/2022]
Abstract
Differentiation and lipid metabolism of adipocytes have a great influence on milk performance, health, and feed efficiency of dairy cows. The effects of dietary long-chain fatty acids (FA) on adipogenesis and lipogenesis of dairy cows are often confounded by other nutritional and physiological factors in vivo. Therefore, this study used an in vitro approach to study the effect of dose and type of long-chain FA on adipogenesis and lipogenesis of bovine adipocytes. Stromal vascular cells were isolated from adipose tissue of dairy cows and induced into mature adipocytes in the presence of various long-chain FA including myristic, palmitic, stearic, oleic, or linoleic acid. When concentrations of myristic, palmitic, and oleic acids in adipogenic mediums were 150 and 200 μM, the induced mature adipocytes had greater lipid content compared with other concentrations of FA. In addition, mature adipocytes induced at 100 μM stearic acid and 300 μM linoleic acid had the greatest content of lipid than at other concentrations. High concentrations of saturated FA were more toxic for cells than the same concentration of unsaturated FA during the induction. When commitment stage was solely treated with FA, the number of differentiated mature adipocytes was greater for oleic and linoleic acids than other FA. When the maturation stage was treated with FA, the number of mature adipocytes was not affected, but the lipid content in adipocytes was affected and ranked oleic > linoleic > myristic > stearic > palmitic. In summary, this study showed that adipogenesis and lipogenesis of bovine adipocytes were differentially affected by long-chain FA, with unsaturated FA more effective than saturated FA.
Collapse
Affiliation(s)
- Chen Yanting
- Department of Animal Science, Washington State University, Pullman 99164
| | - Q Y Yang
- Department of Animal Science, Washington State University, Pullman 99164
| | - G L Ma
- Department of Animal Science, Washington State University, Pullman 99164
| | - M Du
- Department of Animal Science, Washington State University, Pullman 99164
| | - J H Harrison
- Department of Animal Science, Washington State University, Puyallup 98731.
| | - E Block
- Church and Dwight Animal Nutrition, Princeton, NJ 08543
| |
Collapse
|
36
|
Dalrymple BP, Guo B. TRIENNIAL GROWTH AND DEVELOPMENT SYMPOSIUM: Intramuscular fat deposition in ruminants and pigs: A transcriptomics perspective. J Anim Sci 2017; 95:2272-2283. [PMID: 28727003 DOI: 10.2527/jas.2016.1112] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The genomics era has led to an explosion in the study of gene expression in production animals. Intramuscular fat (IMF) content (both high and low) and composition are major quality attributes of meat, and more than 90 transcriptomic studies of IMF deposition have been undertaken in the ruminants and pigs since 2001, with the majority since 2008. The studies have implicated many genes involved in the control of adipogenesis, lipogenesis, and deposition of IMF, but there is relatively little consistency between the different studies. However, the genes encoding the synthesis enzymes acetyl-CoA carboxylase α, fatty acid synthase, and stearoyl-CoA desaturase; the fatty acid binding protein 4; the potential signaling protein thyroid hormone responsive; and the regulators C/EBPα, PPARγ, and sterol regulatory element binding transcription factor 1 are supported by 5 or more of the 90 studies. By combining the results of all the studies, complete pathways for long-chain fatty acid (LCFA) and triacylglyceride (TAG) synthesis are identified, as are a number of genes encoding proteins probably associated with the storage of TAG and genes encoding a number of known and potential adipokines. In contrast, support for the association of lipolytic pathways with IMF percentage is less strong. Differences in experimental design-in particular, the age of the animals, the rate of IMF deposition at sampling, the past nutritional history of the animals used, and the complexities of using a tissue with mixed cell types-have contributed to the differences in results and interpretation. Biomarkers predictive of future IMF percentage, facilitating reaching optimal IMF content at slaughter, may have industry utility, but to be useful in animal biopsy and postslaughter samples, where multiple cell types are present, genes must be carefully chosen to ensure that they are informative about the expected processes. Despite these problems, candidate biomarkers for estimation of de novo intramuscular adipocyte LCFA synthesis, LCFA uptake rate by intramuscular adipocytes, and IMF deposition rate have been identified and examples of their utility have been published. However, further work is required to demonstrate how best to apply the assays for the benefit of the relevant livestock production industries.
Collapse
|
37
|
Meng X, Cui J, Wang Y, Zhang X, Li D, Hai Y, Du H. Heterogeneous nuclear ribonucleoprotein A1 interacts with microRNA-34a to promote chondrogenic differentiation of mesenchymal stem cells. Am J Transl Res 2017; 9:1774-1782. [PMID: 28469782 PMCID: PMC5411925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Accepted: 02/01/2017] [Indexed: 09/28/2022]
Abstract
The mesenchymal stem cell (MSC) shows potential in degenerative disc disease (DDD) treatment. However, little is known about the function of heterogeneous nuclear ribonucleoprotein A1 (hnRNPA1) in modulating the chondrogenic differentiation of MSCs. This study aimed to investigate the role of hnRNPA1 in the chondrogenic differentiation of MSCs and potential mechanisms. Mouse MSCs C3H10 and chondrogenic ATDC5 cells were used to quantify hnRNPA1 expression. The hnRNPA1 overexpression vectors were transfected into C3H10 cells, cell viability and chondrogenic factors expressions were assessed by MTT assay, qPCR and Western blot, respectively. After microRNA-34a (miR-34a) inhibitor transfection, expressions of chondrogenic factors and the Wnt signaling were detected. RNA-binding protein immunoprecipitation (RIP) was performed to reveal the interaction between hnRNPA1 and miR-34a. Results showed that hnRNPA1 was significantly down-regulated in C3H10 compared to ATDC5.Overexpression of hnRNPA1 markedly promoted C3H10 cell viability and expressions of chondrogenic factors SRY-box 9 (SOX9), collagen II, hyaluronan synthase 2 (HAS2) and aggrecan, without significant influence on adipogenic factors. miR-34a inhibitor suppressed chondrogenic factors expressions. RIP results showed the interaction between miR-34a and hnRNPA1. Besides, hnRNPA1 promoted expressions of Wnt family member 3A (WNT3A), WNT5A and β-catenin, and these effects were abrogated by miR-34a inhibitor. We fund the promotive effect of hnRNPA1 on chondrogenic factors, which might require the interaction with miR-34a and the regulation of the Wnt signaling. Thus hnRNPA1 might induce the chondrogenic differentiation of MSCs that facilitate the MSC therapy for DDD.
Collapse
Affiliation(s)
- Xianglong Meng
- Department of Orthopedic, Beijing Chao-Yang Hospital Affiliated to Capital Medical UniversityBeijing 100020, China
| | - Jiawen Cui
- 112 Lab, School of Chemistry and Biological Engineering, University of Science and Technology BeijingBeijing 100083, China
| | - Yunsheng Wang
- Department of Orthopedic, Beijing Chao-Yang Hospital Affiliated to Capital Medical UniversityBeijing 100020, China
| | - Xinuo Zhang
- Department of Orthopedic, Beijing Chao-Yang Hospital Affiliated to Capital Medical UniversityBeijing 100020, China
| | - Dongyue Li
- Department of Orthopedic, Beijing Chao-Yang Hospital Affiliated to Capital Medical UniversityBeijing 100020, China
| | - Yong Hai
- Department of Orthopedic, Beijing Chao-Yang Hospital Affiliated to Capital Medical UniversityBeijing 100020, China
| | - Hongwu Du
- 112 Lab, School of Chemistry and Biological Engineering, University of Science and Technology BeijingBeijing 100083, China
| |
Collapse
|
38
|
Guan L, Hu X, Liu L, Xing Y, Zhou Z, Liang X, Yang Q, Jin S, Bao J, Gao H, Du M, Li J, Zhang L. bta-miR-23a involves in adipogenesis of progenitor cells derived from fetal bovine skeletal muscle. Sci Rep 2017; 7:43716. [PMID: 28255176 PMCID: PMC5334644 DOI: 10.1038/srep43716] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2016] [Accepted: 01/27/2017] [Indexed: 12/30/2022] Open
Abstract
Intramuscular fat deposition or marbling is essential for high quality beef. The molecular mechanism of adipogenesis in skeletal muscle remains largely unknown. In this study, we isolated Platelet-derived growth factor receptor α (PDGFRα) positive progenitor cells from fetal bovine skeletal muscle and induced into adipocytes. Using miRNAome sequencing, we revealed that bta-miR-23a was an adipogenic miRNA mediating bovine adipogenesis in skeletal muscle. The expression of bta-miR-23a was down-regulated during differentiation of PDGFRα+ progenitor cells. Forced expression of bta-miR-23a mimics reduced lipid accumulation and inhibited the key adipogenic transcription factor peroxisome proliferative activated receptor gamma (PPARγ) and CCAAT/enhancer binding protein alpha (C/EBPα). Whereas down-regulation of bta-miR-23a by its inhibitors increased lipid accumulation and expression of C/EBPα, PPARγ and fatty acid-binding protein 4 (FABP4). Target prediction analysis revealed that ZNF423 was a potential target of bta-miR-23a. Dual-luciferase reporter assay revealed that bta-miR-23a directly targeted the 3′-UTR of ZNF423. Together, our data showed that bta-miR-23a orchestrates early intramuscular adipogeneic commitment as an anti-adipogenic regulator which acts by targeting ZNF423.
Collapse
Affiliation(s)
- Long Guan
- Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Xin Hu
- Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Li Liu
- Institute of Animal Husbandry, Heilongjiang Academy of Agricultural Sciences, Harbin 150086, China
| | - Yishen Xing
- Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Zhengkui Zhou
- Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Xingwei Liang
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, Guangxi High Education Laboratory for Animal Reproduction and Biotechnology, Guangxi University, Guangxi 530004, China
| | - Qiyuan Yang
- Department of Animal Sciences, Washington State University, Pullman, WA 99164, USA
| | - Shengyun Jin
- Animal Husbandry Station of Wulagai, Wulagai 026321, China
| | - Jinshan Bao
- Animal Husbandry Station of Wulagai, Wulagai 026321, China
| | - Huijiang Gao
- Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Min Du
- Department of Animal Sciences, Washington State University, Pullman, WA 99164, USA
| | - Junya Li
- Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Lupei Zhang
- Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| |
Collapse
|
39
|
Yun UJ, Song NJ, Yang DK, Kwon SM, Kim K, Kim S, Jo DG, Park WJ, Park KW, Kang H. miR-195a inhibits adipocyte differentiation by targeting the preadipogenic determinator Zfp423. J Cell Biochem 2016; 116:2589-97. [PMID: 25903991 DOI: 10.1002/jcb.25204] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2015] [Accepted: 04/15/2015] [Indexed: 01/27/2023]
Abstract
MicroRNAs (miRNAs) play essential roles in various cellular processes including proliferation and differentiation. In this study, we identified miRNA-195a (miR-195a) as a regulator of adipocyte differentiation. Differential expression of miR-195a in preadipocytes and adipocytes suggests its role in lipid accumulation and adipocyte differentiation. Forced expression of miR-195a mimics suppressed lipid accumulation and inhibited expression of adipocyte markers such as PPARγ and aP2 in 3T3-L1 and C3H10T1/2 cells. Conversely, downregulation of miR-195a by anti-miR-195a increased lipid accumulation and expression of adipocyte markers. Target prediction analysis suggested zinc finger protein 423 (Zfp423), a preadipogenic determinator, as a potential gene recognized by miR-195a. In line with this, mimicked expression of miR-195a reduced the expression of Zfp423, whereas anti-miR-195a increased its expression. Predicted targeting sequences in Zfp423 3'UTR, but not mutated sequences fused to luciferase, were regulated by miR-195a. Ectopic Zfp423 expression in 3T3-L1 cells increased lipid accumulation and expression of adipocyte markers, consistent with the observation that miR-195a targets Zfp423, resulting in suppressed adipocyte differentiation. In addition, miR-195a and Zfp423 were inversely correlated in obese fat tissues, raising the possibility of miRNA's role in obesity. Together, our data show that miR-195a is an anti-adipogenic regulator, which acts by targeting Zfp423, and further suggest the roles of miR-195a in obesity and metabolic diseases.
Collapse
Affiliation(s)
- Ui Jeong Yun
- School of Pharmacy, Sungkyunkwan University, Suwon 440-746, Republic of Korea
| | - No-Joon Song
- Department of Food Science and Biotechnology, Sungkyunkwan University, Suwon 440-746, Republic of Korea
| | - Dong Kwon Yang
- Icahn School of Medicine at Mount Sinai, New York, USA.,College of Life Science, Gwangju Institute of Science and Technology, Gwangju 500-712, Republic of Korea
| | - So-Mi Kwon
- Department of Food Science and Biotechnology, Sungkyunkwan University, Suwon 440-746, Republic of Korea
| | - Kwangho Kim
- Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon 406-772, Republic of Korea
| | - Sunghwan Kim
- New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu 701-310, Republic of Korea
| | - Dong-Gyu Jo
- School of Pharmacy, Sungkyunkwan University, Suwon 440-746, Republic of Korea
| | - Woo Jin Park
- College of Life Science, Gwangju Institute of Science and Technology, Gwangju 500-712, Republic of Korea
| | - Kye Won Park
- Department of Food Science and Biotechnology, Sungkyunkwan University, Suwon 440-746, Republic of Korea
| | - Hara Kang
- Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon 406-772, Republic of Korea
| |
Collapse
|
40
|
Liang X, Yang Q, Fu X, Rogers CJ, Wang B, Pan H, Zhu MJ, Nathanielsz PW, Du M. Maternal obesity epigenetically alters visceral fat progenitor cell properties in male offspring mice. J Physiol 2016; 594:4453-66. [PMID: 27060371 DOI: 10.1113/jp272123] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Accepted: 03/29/2016] [Indexed: 02/01/2023] Open
Abstract
KEY POINTS Maternal obesity reduces adipogenic progenitor density in offspring adipose tissue. The ability of adipose tissue expansion in the offspring of obese mothers is limited and is associated with metabolic dysfunction of adipose tissue when challenged with a high-fat diet. Maternal obesity induces DNA demethylation in the promoter of zinc finger protein 423, which renders progenitor cells with a high adipogenic capacity. Maternal obesity demonstrates long-term effects on the adipogenic capacity of progenitor cells in offspring adipose tissue, demonstrating a developmental programming effect. ABSTRACT Maternal obesity (MO) programs offspring obesity and metabolic disorders, although the underlying mechanisms remain poorly defined. Progenitor cells are the source of new adipocytes. The present study aimed to test whether MO epigenetically predisposes adipocyte progenitors in the fat of offspring to adipogenic differentiation and subsequent depletion, which leads to a failure of adipose tissue plasticity under positive energy balance, contributing to adipose tissue metabolic dysfunction. C57BL/6 female mice were fed either a control diet (10% energy from fat) or a high-fat diet (45% energy from fat) for 8 weeks before mating. Male offspring of control (Con) and obese (OB) dams were weaned onto a regular (Reg) or obesogenic (Obe) diet until 3 months of age. At weaning, male OB offspring had a higher expression of Zinc finger protein 423 (zfp423), a key transcription factor in adipogenesis, as well as lower DNA methylation of its promoter in progenitors of epididymal fat compared to Con offspring, which was correlated with enhanced adipogenic differentiation. At 3 months of age, progenitor density was 30.9 ± 9.7% lower in OB/Obe compared to Con/Obe mice, accompanied by a limited expansion of the adipocyte number when challenged with a high-energy diet. This difference was associated with lower DNA methylation in the zfp423 promoter in the epididymal fat of OB/Obe offspring, which was correlated with greater macrophage chemotactic protein-1 and hypoxia-inducible factor 1α expression. In summary, MO epigenetically limits the expansion capacity of offspring adipose tissue, providing an explanation for the adipose metabolic dysfunction of male offspring in the setting of MO.
Collapse
Affiliation(s)
- Xingwei Liang
- Washington Centre for Muscle Biology and Department of Animal Sciences, Washington State University, Pullman, WA, USA
| | - Qiyuan Yang
- Washington Centre for Muscle Biology and Department of Animal Sciences, Washington State University, Pullman, WA, USA
| | - Xing Fu
- Washington Centre for Muscle Biology and Department of Animal Sciences, Washington State University, Pullman, WA, USA
| | - Carl J Rogers
- Washington Centre for Muscle Biology and Department of Animal Sciences, Washington State University, Pullman, WA, USA
| | - Bo Wang
- Washington Centre for Muscle Biology and Department of Animal Sciences, Washington State University, Pullman, WA, USA
| | - Hong Pan
- Washington Centre for Muscle Biology and Department of Animal Sciences, Washington State University, Pullman, WA, USA
| | - Mei-Jun Zhu
- School of Food Sciences, Washington State University, Pullman, WA, USA
| | - Peter W Nathanielsz
- Wyoming Pregnancy and Life Course Health Centre, Department of Animal Science, University of Wyoming, Laramie, Wyoming, USA
| | - Min Du
- Washington Centre for Muscle Biology and Department of Animal Sciences, Washington State University, Pullman, WA, USA.,Beijing Advanced Innovation Centre for Food Nutrition and Human Health, College of Food Science & Nutritional Engineering, China Agricultural University, Beijing, China
| |
Collapse
|
41
|
Wang B, Yang Q, Harris CL, Nelson ML, Busboom JR, Zhu MJ, Du M. Nutrigenomic regulation of adipose tissue development - role of retinoic acid: A review. Meat Sci 2016; 120:100-106. [PMID: 27086067 DOI: 10.1016/j.meatsci.2016.04.003] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Revised: 04/02/2016] [Accepted: 04/05/2016] [Indexed: 12/17/2022]
Abstract
To improve the efficiency of animal production, livestock have been extensively selected or managed to reduce fat accumulation and increase lean growth, which reduces intramuscular or marbling fat content. To enhance marbling, a better understanding of the mechanisms regulating adipogenesis is needed. Vitamin A has recently been shown to have a profound impact on all stages of adipogenesis. Retinoic acid, an active metabolite of vitamin A, activates both retinoic acid receptors (RAR) and retinoid X receptors (RXR), inducing epigenetic changes in key regulatory genes governing adipogenesis. Additionally, Vitamin D and folates interact with the retinoic acid receptors to regulate adipogenesis. In this review, we discuss nutritional regulation of adipogenesis, focusing on retinoic acid and its impact on epigenetic modifications of key adipogenic genes.
Collapse
Affiliation(s)
- Bo Wang
- Department of Animal Sciences, Washington State University, Pullman, WA 99164, United States
| | - Qiyuan Yang
- Department of Animal Sciences, Washington State University, Pullman, WA 99164, United States
| | - Corrine L Harris
- Department of Animal Sciences, Washington State University, Pullman, WA 99164, United States
| | - Mark L Nelson
- Department of Animal Sciences, Washington State University, Pullman, WA 99164, United States
| | - Jan R Busboom
- Department of Animal Sciences, Washington State University, Pullman, WA 99164, United States
| | - Mei-Jun Zhu
- School of Food Science, Washington State University, Pullman, WA 99164, United States
| | - Min Du
- Department of Animal Sciences, Washington State University, Pullman, WA 99164, United States.
| |
Collapse
|
42
|
Signaroldi E, Laise P, Cristofanon S, Brancaccio A, Reisoli E, Atashpaz S, Terreni MR, Doglioni C, Pruneri G, Malatesta P, Testa G. Polycomb dysregulation in gliomagenesis targets a Zfp423-dependent differentiation network. Nat Commun 2016; 7:10753. [PMID: 26923714 PMCID: PMC4773478 DOI: 10.1038/ncomms10753] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Accepted: 01/19/2016] [Indexed: 02/07/2023] Open
Abstract
Malignant gliomas constitute one of the most significant areas of unmet medical need, owing to the invariable failure of surgical eradication and their marked molecular heterogeneity. Accumulating evidence has revealed a critical contribution by the Polycomb axis of epigenetic repression. However, a coherent understanding of the regulatory networks affected by Polycomb during gliomagenesis is still lacking. Here we integrate transcriptomic and epigenomic analyses to define Polycomb-dependent networks that promote gliomagenesis, validating them both in two independent mouse models and in a large cohort of human samples. We find that Polycomb dysregulation in gliomagenesis affects transcriptional networks associated with invasiveness and de-differentiation. The dissection of these networks uncovers Zfp423 as a critical Polycomb-dependent transcription factor whose silencing negatively impacts survival. The anti-gliomagenic activity of Zfp423 requires interaction with the SMAD proteins within the BMP signalling pathway, pointing to a novel synergic circuit through which Polycomb inhibits BMP signalling.
Collapse
Affiliation(s)
- Elena Signaroldi
- Department of Experimental Oncology, European Institute of Oncology, via Adamello 16, Milan 20139, Italy
| | - Pasquale Laise
- Department of Experimental Oncology, European Institute of Oncology, via Adamello 16, Milan 20139, Italy
| | - Silvia Cristofanon
- Department of Experimental Oncology, European Institute of Oncology, via Adamello 16, Milan 20139, Italy
| | - Arianna Brancaccio
- Department of Experimental Oncology, European Institute of Oncology, via Adamello 16, Milan 20139, Italy
| | - Elisa Reisoli
- Trasferimento Genico, IRCCS-AOU San Martino-IST, Largo Rosanna Benzi 10, Genoa 16132, Italy
| | - Sina Atashpaz
- Department of Experimental Oncology, European Institute of Oncology, via Adamello 16, Milan 20139, Italy
| | - Maria Rosa Terreni
- Pathology Department, IRCCS San Raffaele Scientific Institute, via Olgettina 60, Milan 20132, Italy
| | - Claudio Doglioni
- Pathology Department, IRCCS San Raffaele Scientific Institute, via Olgettina 60, Milan 20132, Italy
| | - Giancarlo Pruneri
- Division of Pathology and Laboratory Medicine, European Institute of Oncology, via Ripamonti 435, Milan 20141, Italy
| | - Paolo Malatesta
- Trasferimento Genico, IRCCS-AOU San Martino-IST, Largo Rosanna Benzi 10, Genoa 16132, Italy
- Department of Experimental Medicine (DiMES), University of Genoa, Via Leon Battista Alberti 2, Genoa 16132, Italy
| | - Giuseppe Testa
- Department of Experimental Oncology, European Institute of Oncology, via Adamello 16, Milan 20139, Italy
- Department of Oncology and Hemato-oncology, University of Milan, Via Festa del Perdono 7, Milan 20122, Italy
| |
Collapse
|
43
|
Moseti D, Regassa A, Kim WK. Molecular Regulation of Adipogenesis and Potential Anti-Adipogenic Bioactive Molecules. Int J Mol Sci 2016; 17:ijms17010124. [PMID: 26797605 PMCID: PMC4730365 DOI: 10.3390/ijms17010124] [Citation(s) in RCA: 481] [Impact Index Per Article: 60.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Revised: 12/27/2015] [Accepted: 01/07/2016] [Indexed: 11/24/2022] Open
Abstract
Adipogenesis is the process by which precursor stem cells differentiate into lipid laden adipocytes. Adipogenesis is regulated by a complex and highly orchestrated gene expression program. In mammalian cells, the peroxisome proliferator-activated receptor γ (PPARγ), and the CCAAT/enhancer binding proteins (C/EBPs) such as C/EBPα, β and δ are considered the key early regulators of adipogenesis, while fatty acid binding protein 4 (FABP4), adiponectin, and fatty acid synthase (FAS) are responsible for the formation of mature adipocytes. Excess accumulation of lipids in the adipose tissue leads to obesity, which is associated with cardiovascular diseases, type II diabetes and other pathologies. Thus, investigating adipose tissue development and the underlying molecular mechanisms is vital to develop therapeutic agents capable of curbing the increasing incidence of obesity and related pathologies. In this review, we address the process of adipogenic differentiation, key transcription factors and proteins involved, adipogenic regulators and potential anti-adipogenic bioactive molecules.
Collapse
Affiliation(s)
- Dorothy Moseti
- Department of Animal Science, University of Manitoba, 201 Animal Science building, Winnipeg, MB R3T 2N2, Canada.
| | - Alemu Regassa
- Department of Animal Science, University of Manitoba, 201 Animal Science building, Winnipeg, MB R3T 2N2, Canada.
| | - Woo-Kyun Kim
- Department of Poultry Science, University of Georgia, 303 Poultry Science Building, Athens, GA 30602-2772, USA.
| |
Collapse
|
44
|
Bell AW, Greenwood PL. Prenatal origins of postnatal variation in growth, development and productivity of ruminants. ANIMAL PRODUCTION SCIENCE 2016. [DOI: 10.1071/an15408] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
This review provides an update on recent research into the effects of maternal nutrition on fetal biology and the growth, development and productivity of progeny in postnatal life of ruminant livestock. Evidence is summarised for effects on postnatal growth and body composition, feed intake and efficiency, carcass characteristics and meat quality, wool production, reproduction and lactation performance. In general, these demonstrated effects are not large in relation to the effects of postnatal nutrition and other environmental influences. The mechanisms underpinning the above production outcomes are briefly discussed in terms of systemic endocrine and metabolic responses, and cellular and molecular effects in skeletal muscle, bone, adipose tissue, wool follicles and brain of fetal, neonatal and adult progeny. Treatments observed to elicit tissue responses include maternal under- and overnutrition at various stages of pregnancy and placental insufficiency caused by increased litter size, chronic maternal heat stress and premating carunclectomy in sheep. The as yet meagre evidence for epigenetic mediation of intergenerational effects in ruminants is considered, as is the likelihood that other, more conventional explanations may suffice in some cases. Finally, evidence is summarised for the proposition that the placenta is not merely a passive conduit for nutrient transfer from dam to fetus, but plays an active role in buffering the effects of variations in maternal nutrition on fetal growth and development, and thence, postnatal outcomes.
Collapse
|
45
|
Martins TS, Sanglard LMP, Silva W, Chizzotti ML, Rennó LN, Serão NVL, Silva FF, Guimarães SEF, Ladeira MM, Dodson MV, Du M, Duarte MS. Molecular Factors Underlying the Deposition of Intramuscular Fat and Collagen in Skeletal Muscle of Nellore and Angus Cattle. PLoS One 2015; 10:e0139943. [PMID: 26436893 PMCID: PMC4593631 DOI: 10.1371/journal.pone.0139943] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Accepted: 09/18/2015] [Indexed: 01/24/2023] Open
Abstract
Studies have shown that intramuscular adipogenesis and fibrogenesis may concomitantly occur in skeletal muscle of beef cattle. Thus, we hypothesized that the discrepancy of intramuscular fat content in beef from Nellore and Angus was associated with differences in intramuscular adipogenesis and fibrogenesis during the finishing phase. To test our hypothesis, longissimus muscle samples of Nellore (n = 6; BW = 372.5 ± 37.3 kg) and Angus (n = 6; BW = 382.8 ± 23.9 kg) cattle were collected for analysis of gene and protein expression, and quantification of intramuscular fat and collagen. Least-squares means were estimated for the effect of Breed and differences were considered at P ≤ 0.05. A greater intramuscular fat content was observed in skeletal muscle of Angus compared to Nellore cattle (P≤0.05). No differences were observed for mRNA expression of lipogenic and lipolytic markers ACC, FAS, FABP4, SERBP–1, CPT–2, LPL, and ACOX (P > 0.05) in skeletal muscle of Nellore and Angus cattle. Similarly, no differences were observed in mRNA expression of adipogenic markers Zfp423, PPARγ, and C/EBPα (P>0.05) However, a greater PPARγ protein content was observed in skeletal muscle of Angus compared to Nellore cattle (P≤0.05). A greater abundance of adipo/fibrogenic cells, evaluated by the PDGFRα content, was observed in skeletal muscle of Angus than Nellore cattle (P≤0.05). No differences in fibrogenesis were observed in skeletal muscle of Angus and Nellore cattle, which is in accordance with the lack of differences in intramuscular collagen content in beef from both breeds (P>0.05). These findings demonstrate that difference in intramuscular fat content is associated with a slightly enhanced adipogenesis in skeletal muscle of Angus compared to Nellore cattle, while no difference in fibrogenesis.
Collapse
Affiliation(s)
- Taiane S. Martins
- Department of Animal Science, Universidade Federal de Viçosa, Viçosa, Brazil
| | - Letícia M. P. Sanglard
- Department of Animal Science, Universidade Federal de Viçosa, Viçosa, Brazil
- Animal Biotechnology Laboratory—LABTEC, Universidade Federal de Viçosa, Viçosa, Minas Gerais, Brazil
| | - Walmir Silva
- Department of Animal Science, Universidade Federal de Viçosa, Viçosa, Brazil
- Animal Biotechnology Laboratory—LABTEC, Universidade Federal de Viçosa, Viçosa, Minas Gerais, Brazil
| | - Mário L. Chizzotti
- Department of Animal Science, Universidade Federal de Viçosa, Viçosa, Brazil
| | - Luciana N. Rennó
- Department of Animal Science, Universidade Federal de Viçosa, Viçosa, Brazil
| | - Nick V. L. Serão
- Department of Animal Science, North Carolina State University, Raleigh, North Carolina, United States of America
| | - Fabyano F. Silva
- Department of Animal Science, Universidade Federal de Viçosa, Viçosa, Brazil
- Animal Biotechnology Laboratory—LABTEC, Universidade Federal de Viçosa, Viçosa, Minas Gerais, Brazil
| | - Simone E. F. Guimarães
- Department of Animal Science, Universidade Federal de Viçosa, Viçosa, Brazil
- Animal Biotechnology Laboratory—LABTEC, Universidade Federal de Viçosa, Viçosa, Minas Gerais, Brazil
| | - Márcio M. Ladeira
- Department of Animal Science, Universidade Federal de Lavras, Lavras, Brazil
| | - Michael V. Dodson
- Department of Animal Science, Washington State University, Pullman, Washington, United States of America
| | - Min Du
- Department of Animal Science, Washington State University, Pullman, Washington, United States of America
| | - Marcio S. Duarte
- Department of Animal Science, Universidade Federal de Viçosa, Viçosa, Brazil
- Animal Biotechnology Laboratory—LABTEC, Universidade Federal de Viçosa, Viçosa, Minas Gerais, Brazil
- * E-mail:
| |
Collapse
|
46
|
Abstract
The abundance and cross-linking of intramuscular connective tissue contributes to the background toughness of meat, and is thus undesirable. Connective tissue is mainly synthesized by intramuscular fibroblasts. Myocytes, adipocytes and fibroblasts are derived from a common pool of progenitor cells during the early embryonic development. It appears that multipotent mesenchymal stem cells first diverge into either myogenic or non-myogenic lineages; non-myogenic mesenchymal progenitors then develop into the stromal-vascular fraction of skeletal muscle wherein adipocytes, fibroblasts and derived mesenchymal progenitors reside. Because non-myogenic mesenchymal progenitors mainly undergo adipogenic or fibrogenic differentiation during muscle development, strengthening progenitor proliferation enhances the potential for both intramuscular adipogenesis and fibrogenesis, leading to the elevation of both marbling and connective tissue content in the resulting meat product. Furthermore, given the bipotent developmental potential of progenitor cells, enhancing their conversion to adipogenesis reduces fibrogenesis, which likely results in the overall improvement of marbling (more intramuscular adipocytes) and tenderness (less connective tissue) of meat. Fibrogenesis is mainly regulated by the transforming growth factor (TGF) β signaling pathway and its regulatory cascade. In addition, extracellular matrix, a part of the intramuscular connective tissue, provides a niche environment for regulating myogenic differentiation of satellite cells and muscle growth. Despite rapid progress, many questions remain in the role of extracellular matrix on muscle development, and factors determining the early differentiation of myogenic, adipogenic and fibrogenic cells, which warrant further studies.
Collapse
|
47
|
da Silva JM, Giachetto PF, da Silva LOC, Cintra LC, Paiva SR, Caetano AR, Yamagishi MEB. Genomic Variants Revealed by Invariably Missing Genotypes in Nelore Cattle. PLoS One 2015; 10:e0136035. [PMID: 26305794 PMCID: PMC4549312 DOI: 10.1371/journal.pone.0136035] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2015] [Accepted: 07/29/2015] [Indexed: 12/18/2022] Open
Abstract
High density genotyping panels have been used in a wide range of applications. From population genetics to genome-wide association studies, this technology still offers the lowest cost and the most consistent solution for generating SNP data. However, in spite of the application, part of the generated data is always discarded from final datasets based on quality control criteria used to remove unreliable markers. Some discarded data consists of markers that failed to generate genotypes, labeled as missing genotypes. A subset of missing genotypes that occur in the whole population under study may be caused by technical issues but can also be explained by the presence of genomic variations that are in the vicinity of the assayed SNP and that prevent genotyping probes from annealing. The latter case may contain relevant information because these missing genotypes might be used to identify population-specific genomic variants. In order to assess which case is more prevalent, we used Illumina HD Bovine chip genotypes from 1,709 Nelore (Bos indicus) samples. We found 3,200 missing genotypes among the whole population. NGS re-sequencing data from 8 sires were used to verify the presence of genomic variations within their flanking regions in 81.56% of these missing genotypes. Furthermore, we discovered 3,300 novel SNPs/Indels, 31% of which are located in genes that may affect traits of importance for the genetic improvement of cattle production.
Collapse
Affiliation(s)
- Joaquim Manoel da Silva
- Faculdade de Ciências Agrárias, Biológicas e Sociais Aplicadas, Universidade do Estado de Mato Grosso (UNEMAT), Nova Xavantina, Mato Grosso, Brazil
- Programa de Pós-Graduação em Genética e Biologia Molecular–Instituto de Biologia, Universidade Estadual de Campinas (UNICAMP), Campinas, São Paulo, Brazil
- * E-mail:
| | - Poliana Fernanda Giachetto
- Laboratório Multiusuário de Bioinformática (LMB)—Embrapa Informática Agropecuária, Campinas, São Paulo, Brazil
| | | | - Leandro Carrijo Cintra
- Laboratório Multiusuário de Bioinformática (LMB)—Embrapa Informática Agropecuária, Campinas, São Paulo, Brazil
| | - Samuel Rezende Paiva
- Embrapa–Secretaria de Relações Internacionais, Brasília, Distrito Federal, Brazil
| | | | | |
Collapse
|
48
|
Du M, Wang B, Fu X, Yang Q, Zhu MJ. Fetal programming in meat production. Meat Sci 2015; 109:40-7. [PMID: 25953215 DOI: 10.1016/j.meatsci.2015.04.010] [Citation(s) in RCA: 95] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2015] [Revised: 04/14/2015] [Accepted: 04/16/2015] [Indexed: 12/23/2022]
Abstract
Nutrient fluctuations during the fetal stage affects fetal development, which has long-term impacts on the production efficiency and quality of meat. During the early development, a pool of mesenchymal progenitor cells proliferate and then diverge into either myogenic or adipogenic/fibrogenic lineages. Myogenic progenitor cells further develop into muscle fibers and satellite cells, while adipogenic/fibrogenic lineage cells develop into adipocytes, fibroblasts and resident fibro-adipogenic progenitor cells. Enhancing the proliferation and myogenic commitment of progenitor cells during fetal development enhances muscle growth and lean production in offspring. On the other hand, promoting the adipogenic differentiation of adipogenic/fibrogenic progenitor cells inside the muscle increases intramuscular adipocytes and reduces connective tissue, which improves meat marbling and tenderness. Available studies in mammalian livestock, including cattle, sheep and pigs, clearly show the link between maternal nutrition and the quantity and quality of meat production. Similarly, chicken muscle fibers develop before hatching and, thus, egg and yolk sizes and hatching temperature affect long-term growth performance and meat production of chicken. On the contrary, because fishes are able to generate new muscle fibers lifelong, the impact of early nutrition on fish growth performance is expected to be minor, which requires further studies.
Collapse
Affiliation(s)
- Min Du
- Department of Animal Sciences, Washington State University, Pullman, WA 99164, United States.
| | - Bo Wang
- Department of Animal Sciences, Washington State University, Pullman, WA 99164, United States
| | - Xing Fu
- Department of Animal Sciences, Washington State University, Pullman, WA 99164, United States
| | - Qiyuan Yang
- Department of Animal Sciences, Washington State University, Pullman, WA 99164, United States
| | - Mei-Jun Zhu
- School of Food Science, Washington State University, Pullman, WA 99164, United States
| |
Collapse
|
49
|
Carrió E, Díez-Villanueva A, Lois S, Mallona I, Cases I, Forn M, Peinado MA, Suelves M. Deconstruction of DNA methylation patterns during myogenesis reveals specific epigenetic events in the establishment of the skeletal muscle lineage. Stem Cells 2015; 33:2025-36. [PMID: 25801824 DOI: 10.1002/stem.1998] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Accepted: 02/06/2015] [Indexed: 12/17/2022]
Abstract
The progressive restriction of differentiation potential from pluripotent embryonic stem cells (ESCs) to tissue-specific stem cells involves widespread epigenetic reprogramming, including modulation of DNA methylation patterns. Skeletal muscle stem cells are required for the growth, maintenance, and regeneration of skeletal muscle. To investigate the contribution of DNA methylation to the establishment of the myogenic program, we analyzed ESCs, skeletal muscle stem cells in proliferating (myoblasts) and differentiating conditions (myotubes), and mature myofibers. About 1.000 differentially methylated regions were identified during muscle-lineage determination and terminal differentiation, mainly located in gene bodies and intergenic regions. As a whole, myogenic stem cells showed a gain of DNA methylation, while muscle differentiation was accompanied by loss of DNA methylation in CpG-poor regions. Notably, the hypomethylated regions in myogenic stem cells were neighbored by enhancer-type chromatin, suggesting the involvement of DNA methylation in the regulation of cell-type specific enhancers. Interestingly, we demonstrated the hypomethylation of the muscle cell-identity Myf5 super-enhancer only in muscle cells. Furthermore, we observed that upstream stimulatory factor 1 binding to Myf5 super-enhancer occurs upon DNA demethylation in myogenic stem cells. Taken altogether, we characterized the unique DNA methylation signature of skeletal muscle stem cells and highlighted the importance of DNA methylation-mediated regulation of cell identity Myf5 super-enhancer during cellular differentiation.
Collapse
Affiliation(s)
- Elvira Carrió
- Institut de Medicina Predictiva i Personalitzada del Càncer (IMPPC).,Institut Germans Trias i Pujol (IGTP), Campus Can Ruti, 08916, Badalona, Spain
| | - Anna Díez-Villanueva
- Institut de Medicina Predictiva i Personalitzada del Càncer (IMPPC).,Institut Germans Trias i Pujol (IGTP), Campus Can Ruti, 08916, Badalona, Spain
| | - Sergi Lois
- Institut de Medicina Predictiva i Personalitzada del Càncer (IMPPC)
| | - Izaskun Mallona
- Institut de Medicina Predictiva i Personalitzada del Càncer (IMPPC).,Institut Germans Trias i Pujol (IGTP), Campus Can Ruti, 08916, Badalona, Spain
| | - Ildefonso Cases
- Institut de Medicina Predictiva i Personalitzada del Càncer (IMPPC)
| | - Marta Forn
- Institut de Medicina Predictiva i Personalitzada del Càncer (IMPPC)
| | - Miguel A Peinado
- Institut de Medicina Predictiva i Personalitzada del Càncer (IMPPC).,Institut Germans Trias i Pujol (IGTP), Campus Can Ruti, 08916, Badalona, Spain
| | - Mònica Suelves
- Institut de Medicina Predictiva i Personalitzada del Càncer (IMPPC).,Institut Germans Trias i Pujol (IGTP), Campus Can Ruti, 08916, Badalona, Spain
| |
Collapse
|
50
|
Hausman GJ, Basu U, Du M, Fernyhough-Culver M, Dodson MV. Intermuscular and intramuscular adipose tissues: Bad vs. good adipose tissues. Adipocyte 2014; 3:242-55. [PMID: 26317048 DOI: 10.4161/adip.28546] [Citation(s) in RCA: 133] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2013] [Revised: 03/11/2014] [Accepted: 03/14/2014] [Indexed: 12/23/2022] Open
Abstract
Human studies of the influence of aging and other factors on intermuscular fat (INTMF) were reviewed. Intermuscular fat increased with weight loss, weight gain, or with no weight change with age in humans. An increase in INTMF represents a similar threat to type 2 diabetes and insulin resistance as does visceral adipose tissue (VAT). Studies of INTMF in animals covered topics such as quantitative deposition and genetic relationships with other fat depots. The relationship between leanness and higher proportions of INTMF fat in pigs was not observed in human studies and was not corroborated by other pig studies. In humans, changes in muscle mass, strength and quality are associated with INTMF accretion with aging. Gene expression profiling and intrinsic methylation differences in pigs demonstrated that INTMF and VAT are primarily associated with inflammatory and immune processes. It seems that in the pig and humans, INTMF and VAT share a similar pattern of distribution and a similar association of components dictating insulin sensitivity. Studies on intramuscular (IM) adipocyte development in meat animals were reviewed. Gene expression analysis and genetic analysis have identified candidate genes involved in IM adipocyte development. Intramuscular (IM) adipocyte development in human muscle is only seen during aging and some pathological circumstance. Several genetic links between human and meat animal adipogenesis have been identified. In pigs, the Lipin1 and Lipin 2 gene have strong genetic effects on IM accumulation. Lipin1 deficiency results in immature adipocyte development in human lipodystrophy. In humans, overexpression of Perilipin 2 (PLIN2) facilitates intramyocellular lipid accretion whereas in pigs PLIN2 gene expression is associated with IM deposition. Lipins and perilipins may influence intramuscular lipid regardless of species.
Collapse
|