NCCC210: Regulation of Adipose Tissue Accretion in Meat-Producing Animals

(Multistate Research Coordinating Committee and Information Exchange Group)

Status: Active

SAES-422 Reports

Annual/Termination Reports:

[09/14/2020] [07/09/2021] [11/15/2022]

Date of Annual Report: 09/14/2020

Report Information

Annual Meeting Dates: 07/14/2020 - 07/14/2020
Period the Report Covers: 07/14/2019 - 07/14/2020

Participants

1. Remote presentations (through Zoom): Eric Testroet/Ratan Choudhary, Eric Testroet/Michelle LaCasse, Yuan-Yu Lin, Steve Smith, Yan Huang, Kichoon Lee, Woo Kyun Kim,Stone Ding
2. Total in attendance - 43

Brief Summary of Minutes


  • Attendees arrived by 8 am EST

  • Donald Beitz gave a plenary talk on the history of the multistate group.

  • Invited talks started at 8:30 AM EST



  1. Remote presentations (through Zoom): Eric Testroet/Ratan Choudhary, Eric Testroet/Michelle LaCasse, Yuan-Yu Lin, Steve Smith, Yan Huang, Kichoon Lee, Woo Kyun Kim,Stone Ding


    1. Total in attendance - 43




  • Administrative business: Woo kyun Kim elected to be co-chair for 2020-2021.

  • Station reports from:

    • Purdue University – Kola Ajuwon

    • University of Arkansas – Sean Adams

    • Industry representation – Theo van Kempen

    • Washington State University – Min Du

    • National Taiwan University – Judy Lin, Yuan-Yu Lin, and Stone Ding

    • Auburn University – Werner Bergen

    • University of Tennessee – Brynn Voy



  • Adjourned at 12:00 PM EST

Accomplishments

<p><strong><span style="text-decoration: underline;">Station Reports 2020</span></strong></p><br /> <ol><br /> <li>The Ohio State University</li><br /> <li>Kichoon Lee</li><br /> </ol><br /> <p><strong>Participant:</strong> Kichoon Lee (<a href="mailto:lee.2626@osu.edu">lee.2626@osu.edu</a>) The Ohio State University</p><br /> <p>&nbsp;</p><br /> <p><strong>Two major Accomplishments:</strong></p><br /> <p><strong>&nbsp;</strong></p><br /> <p><strong>Establishment of genome-editing technology in avian by our unique CRISPR-Cas9</strong>.</p><br /> <p>CRISPR/Cas9 system has been successfully used to generate targeted mutation in livestock animals and rodents for academic and industrial purposes. However, there are some difficulties associated with using the CRISPR/Cas9 system in the generation of knockout birds. Therefore, CRISPR/Cas9-mediated gene knockout in the avian species is limitedly reported because of requiring a genetic modification of primordial germ cells (PGC) in culture and transfer to embryos by microinjection of the genetically modified PGCs into embryos. Yet, the PGCs culture and manipulation require laboriously high skilled-techniques and establishment of optimized condition. We have developed a poultry-specific CRISPR/Cas9 adenoviral delivery system to efficiently introduce targeted mutation in chromosomes of quail. Through this project, we generated several lines of genome-edited quail with mutations in melanophilin gene, resulting in change in feather color from brown to gray plumage. In addition, quail with genome editing in myostatin gene were generated and had increased muscle mass and decreased fat.&nbsp;&nbsp;&nbsp; &nbsp;</p><br /> <p>&nbsp;</p><br /> <p><strong>Short-term Outcomes: </strong>We have developed a poultry-specific CRISPR/Cas9 adenoviral delivery system to efficiently introduce targeted mutation in chromosomes of quail. &nbsp;<strong>Outputs:</strong> The results of this study led to three publications in Proceedings of the National Academy of Sciences of the USA and International Journal of Molecular Science.</p><br /> <p><strong>Activities:</strong> Use of the adenoviral vector system enables us to provide a more rapid way to generate a knockout bird by skipping steps for modification of the PGC genome prior to microinjection into embryos. The new CRISPR system developed from my laboratory resulted in recent awards of two research grants from USDA-NIFA.</p><br /> <p><strong>Milestones:</strong> Our goal is to further improve efficiency of genome editing in other avian species including chicken and turkey by 2022.</p><br /> <p><strong>Impacts: </strong>The method used in the present study allows avian knockout studies in broad research areas to be conducted for the advancement of developing more productive genomic lines of poultry.</p><br /> <p><strong>&nbsp;</strong></p><br /> <p><strong>Identification of novel imprinted genes by generating parthenogenetic pig fetus.</strong></p><br /> <p>While the majority of genes are expressed from both paternal and maternal alleles, a subset of genes often in clusters are subjected to genomic imprinting that leads to monoallelic gene expression.&nbsp; To date, about 150 imprinted genes are identified in the mouse and 100 imprinted genes in the human. Experimental evidence proved Haig&rsquo;s parental conflict theory that paternally derived genes favor increased growth, whereas maternally derived genes favor reduced growth. Therefore, genomic imprinting mainly contributes to growth traits of domestic animals and is important for the selection of superior animals to maximize their performance. However, a relatively few numbers of imprinted genes have been identified: 26, 17, and 10 genes in cattle, pigs, and sheep. With collaboration with the National Institute of Animal Biotechnology in Korea, we generated parthenogenetic porcine fetuses (two sets of maternal chromosomes and no paternal chromosomes) and normal controls (one set of maternal and one set of paternal chromosomes). Novel imprinted transcripts in porcine GNAS and SGCE/PEG10 complex loci were identified using whole genome methylome and transcriptome of parthenogenetic fetuses.</p><br /> <p>&nbsp;</p><br /> <p><strong>Short-term Outcomes:</strong> Using whole genome scale analyses including whole genome bisulfite sequencing to detect differential DNA methylation and RNAseq to identify differentially expressed genes between parthenotes and normal pigs, we were able to identify couple of hundreds imprinted genes including novel imprinted genes that have never been reported in human and mouse.</p><br /> <p><strong>Outputs:</strong> The results opened a new direction of my research program and the first two papers were published in Genes and Animals and others were being prepared for submission.</p><br /> <p><strong>Activities:</strong> We had high impact discoveries from this project yielding valuable preliminary data to garner federal grant funding and publications. We were also submitted a grant proposal to USDA-NIFA in March 2020.</p><br /> <p><strong>Milestones:</strong> Our goal is to document all imprinted genes in pigs and confirmation of their homologues in other animals and humans by 2022.</p><br /> <p><strong>Impacts: </strong>Imprinted genes are involved in regulation of adipose and fat growth in animals and humans. Successful completion of this project is expected to provide a scientific foundation that specific imprinted genes can serve as an excellent marker to assist in the selection of superior lines of swine for enhanced growth, as well as, of other food animal species. <strong>&nbsp;</strong></p><br /> <p><strong>&nbsp;</strong></p><br /> <p><strong>Publications in 2019-Current:</strong></p><br /> <ol><br /> <li>Ahn J, Wu H, <strong>Lee K</strong>. 2020. Integrative Analysis Revealing Human Heart-Specific Genes and Consolidating Heart-Related GWAS Loci. <strong>Frontiers in Genetics </strong>11:777.</li><br /> <li>Kim DH, Choi YM, Suh Y, Shin S, Lee J, Hwang S,<strong> Lee K. </strong>2020. Association of temporal expression of myostatin with hypertrophic muscle growth in different Japanese quail lines. <strong>Poultry Science</strong> 99(6): 2926-2930.</li><br /> <li>Lee J, Kim DH, <strong>Lee K.</strong> 2020. Current Approaches and Applications in Avian Genome Editing. <strong>International Journal of Molecular Science </strong>21:E3937.</li><br /> <li>Hwang IS, Park MR, Lee HS, Kwak TU, Son HY, Kang JK, Lee JW, <strong>Lee K</strong>, Park EW, Hwang S. 2020. Developmental and Degenerative Characterization of Porcine <br /> Parthenogenetic Fetuses during Early Pregnancy. <strong>Animals</strong> 10(4):622.</li><br /> <li>Kim DH, Lee J, Suh Y, Cressman M, Lee SS, <strong>Lee K</strong>. 2020. Adipogenic and Myogenic Potentials of Chicken Embryonic Fibroblasts in vitro: Combination of Fatty Acids and Insulin Induces Adipogenesis. <strong>Lipids</strong> 55(2):163-171.</li><br /> <li>Lee JB, Kim DH, <strong>Lee K</strong>. 2020. Muscle Hyperplasia in Japanese Quail by Single Amino Acid Deletion in MSTN Propeptide. <strong>International Journal of Molecular Science </strong>21:E1504.</li><br /> <li>Ahn J, Wu H, Lee J, Hwang IS, Yu D, Ahn J, Lee JW, Hwang S, <strong>Lee K</strong>. 2020. Identification of a novel imprinted transcript in the porcine GNAS complex locus using methylome and transcriptome of parthernogenetic fetuses. <strong>Genes</strong> 11(1). pii: E96.</li><br /> <li>Choi I, Park HB, Ahn JS, Han SH, Lee JB, Lim HT, Yoo CK, Jung EJ, Kim DH, Sun WS, Ramayo-Caldas Y, Kim SG, Kang YJ, Kim YK, Shin HS, Seong PN, Hwang IS, Park BY, Hwang SS, Lee SS, Ryu YC, Lee JH, Ko MS, <strong>Lee K</strong>, Andersson G, P&eacute;rez-Enciso M, Lee JW. 2019. A functional regulatory variant of MYH3 influences muscle fiber-type composition and intramuscular fat content in pigs. <strong>PloS Genetics </strong>15(10):e1008279.</li><br /> <li>Chen PR, Suh Y, Shin S, Woodfint RM, Hwang S, <strong>Lee K</strong>. 2019. Exogenous expression of an alternative splicing variant of myostatin prompts leg muscle fiber hyperplasia in Japanese quail. <strong>International Journal of Molecular Science</strong> 20:E4617.</li><br /> <li>Lovelia LM, Kim SH, Biswas A, Yu Z, Cho KK, Kim SB, <strong>Lee K</strong>, Lee SS. 2019. Rumen fermentation and microbial community composition influenced by live Enterococcus faecium supplementation. <strong>Applied Microbiology and Biotechnology EXPRESS </strong>9:123.</li><br /> <li>Lee JB, Ma J, <strong>Lee K</strong>. 2019. Direct delivery of adenoviral CRISPR/Cas9 vector into quail blastoderms for generation of knockout birds. <strong>Proceedings of the National Academy of Sciences of the USA </strong>116:13288-13292.</li><br /> <li>Ahn J, Suh Y, <strong>Lee K</strong>. 2019. Adipose-specific expression and developmental and nutritional regulation of the gene encoding retinol-binding protein 7 (RBP7) in pigs. <strong>Lipids</strong> 54:359-367.</li><br /> <li>Coleman DN<strong>, </strong>Carranza AC, Jin Y, <strong>Lee K</strong>, Relling AE. 2019. Prepartum fatty acid supplementation in sheep IV. Effect of calcium salts with eicosapentaenoic acid and docosahexaenoic acid in the maternal and finishing diet on lamb liver and adipose tissue during the lamb finishing period. <strong>Journal of Animal Science</strong> 97:3071-3088.</li><br /> <li>Ahn J, Woodfint R, Lee J, Wu H, Ma J, Suh Y, Hwang S, Cressman M, <strong>Lee K</strong>. 2019. Comparative identification and nutritional and physiological regulation of chicken liver-specific genes. <strong>Poultry Science</strong> 98:3007-3013.</li><br /> <li>Choi YJ, Garcia L, <strong>Lee K</strong>. 2019. Correlations of Sensory Quality Characteristics with Intramuscular Fat Content and Bundle Characteristics in Bovine Longissimus Thoracis Muscle. <strong>Food Science of Animal Resources </strong>39(2):197-208.</li><br /> <li>Lee B, Lee BJ, Lee YK, Hur SW, Kim KD, Kim KW, Han HS, Shin S, Choe JH, <strong>Lee K</strong>, Choi YM. 2019. Muscle fiber growth in olive flounder, Paralichthys olivaceus: Fiber hyperplasia at a specific body weight period and continuous hypertrophy<strong>. Journal of the World Aquaculture Society </strong>90:593-603.</li><br /> <li>Ahn J, Wu H, <strong>Lee K</strong>. 2019. Integrative Analysis Revealing Human Adipose-Specific Genes and Consolidating Obesity Loci. <strong>Scientific Reports</strong> 9(1):3087.</li><br /> <li>Kim DH, Lee JW, <strong>Lee K</strong>. 2019. Supplementation of all-trans retinoic acid below cytotoxic levels promotes adipogenesis in 3T3-L1 cells. <strong>Lipids </strong>54:99-107.</li><br /> </ol><br /> <p><strong>&nbsp;</strong></p><br /> <p>&nbsp;</p><br /> <p>&nbsp;</p><br /> <ol><br /> <li>University of Vermont</li><br /> <li>Eric D. Testroet</li><br /> <li>Accomplishments:</li><br /> </ol><br /> <p>Currently we are in the process of finalizing an <em>in vitro </em>model of bovine hepatic lipidosis.&nbsp; We have isolated and cultured primary bovine hepatocytes and are in final stages of functional validation (i.e., lipid accumulation, urea production, albumin production, LDH leakage, and cytotoxicity markers).&nbsp; Using this model, we will perform an on-farm experiment using multiparous dairy cattle in which we will either induce hepatic lipidosis or not using established protocols.&nbsp; We have performed liver and adipose biopsies on 12 cattle that are either experience hepatic lipidosis or are not and examine key signaling pathways related to protein kinase A (PKA), AMP-activated protein kinase (AMPK), and phosphodiesterase 4b (PDE4B).&nbsp; It is our hypothesis that dysregulation of PDE4b results in hepatic lipidosis and that ultimately modulation of PDE4b activity can lead to prevention of hepatic lipidosis in the periparturient cow.&nbsp; Our research was, unfortunately, interrupted by COVID-19, and we have just restarted with the final 12 cattle.&nbsp; In addition, we have received funding to characterize the bovine hepatic lipidosis model as a model for human NAFLD and NASH, and to examine use of a novel siRNA as a preventative for development of hepatic lipidosis in the transition period of dairy cattle.&nbsp;</p><br /> <p>&nbsp;</p><br /> <p><span style="text-decoration: underline;">Publications</span></p><br /> <p><strong>Submitted</strong></p><br /> <p><strong>&nbsp;</strong></p><br /> <ol><br /> <li>Shome, A. Testroet, J. Reecy, K. Amin, K. Conley, J. Reecy, R. Jernigan, D. Dobbs, M. Du, S. Clark, and D. C. Beitz, E. D.<strong> Testroet</strong>. 2020. Non-Coding RNA in Raw and Commercially Processed Milk and Putative Targets Related to Growth and Immune-Response. <span style="text-decoration: underline;">BMC Genomics.</span> Submitted.</li><br /> </ol><br /> <p>&nbsp;</p><br /> <ol start="2"><br /> <li>R. O&rsquo;Neil, E. D. <strong>Testroet</strong>, R. L Stuart, and D. C. Beitz. 2020. Effect of breed and stage of maturity on concentration of serum &beta;-carotene, vitamin E, 25-hydroxyvitamin D3, and vitamin A of Angus and Holstein cows and heifer calves. <span style="text-decoration: underline;">Livestock Science. </span>Submitted.</li><br /> </ol><br /> <p>&nbsp;</p><br /> <ol start="3"><br /> <li>S. Dankwa, U. Humagain, C. Yeoman, S. Clark, D. C. Beitz, S. Ishaq, and E. D. <strong>Testroet</strong>. 2020. Reduced-fat dried distillers grains with solubles does not negatively impact gut bacteria. <span style="text-decoration: underline;">J. Dairy Sci.</span> Submitted.</li><br /> </ol><br /> <p>&nbsp;</p><br /> <p><strong>&nbsp;</strong></p><br /> <p><strong>In Revision</strong><br /> </p><br /> <ol><br /> <li><strong>Testroet</strong>, E. D., J. M. de Avila, S. Clark D. C. Beitz, and M. Du. 2020.The effect of palmitate and TNF&alpha; on abattoir-derived Holstein cow liver primary cell culture. In preparation to be re-submitted to <span style="text-decoration: underline;"> Dairy Sci.</span></li><br /> </ol><br /> <p>&nbsp;</p><br /> <p><strong>In Preparation</strong></p><br /> <p>&nbsp;</p><br /> <ol><br /> <li>Choudhary, M. LaCasse, R. K. Choudhary, M. Rincon, D. C. Beitz, and E. D. <strong>Testroet.</strong> 2020. <em>In vivo</em> and <em>in vitro </em>expression of mitochondrial complex 1 inhibitor in bovine liver. In preparation to be submitted to <span style="text-decoration: underline;">J. Dairy Sci.</span></li><br /> </ol><br /> <p><strong>Published Abstracts </strong></p><br /> <p><strong>&nbsp;</strong></p><br /> <ol><br /> <li>LaCasse, M., S. Choudhary, R. Choudhary. J. de Avila, D. C. Beitz, M. Du, and <strong> D. Testroet.</strong> A nonperfusion-based method of hepatic cell isolation and development of fatty liver disease model for dairy cattle. Poster. To be presented at <span style="text-decoration: underline;">the 2020 Experimental Biology Annual Meeting,</span> San Diego, CA.</li><br /> </ol><br /> <p>&nbsp;</p><br /> <ol start="2"><br /> <li>Choudhary, R. Choudhary, LaCasse, M., J. de Avila, D. C. Beitz, M. Du, M. Rincon and <strong>E. D. Testroet.</strong> Expression of mitochondrial complex 1 inhibitor in bovine tissue, primary hepatic cells, and detection of its&rsquo; transcript in conditioned media mimicking fatty liver disease. Poster.&nbsp; To be presented at <span style="text-decoration: underline;">the 2020 Experimental Biology Annual Meeting,</span> San Diego, CA.</li><br /> </ol><br /> <p>&nbsp;</p><br /> <p>&nbsp;</p><br /> <p>&nbsp;</p><br /> <p>&nbsp;</p><br /> <p>&nbsp;</p><br /> <ol><br /> <li>Auburn University</li><br /> <li>Werner G. Bergen</li><br /> </ol><br /> <p>Graduate students: Kamille Piacquadio, Robert Mihelic</p><br /> <p>Collaborators in NCC210: Woo Kim (UGA)</p><br /> <p>Other collaborators: Shawn Campagna (UTK), Jeanna Wilson (UGA)</p><br /> <ol start="3"><br /> <li>Accomplishments:</li><br /> </ol><br /> <p>Over the last year, I have concentrated on defining the role of the&nbsp;&nbsp;&nbsp;&nbsp;&nbsp;&nbsp;&nbsp;&nbsp;&nbsp;&nbsp;&nbsp;&nbsp;&nbsp;&nbsp;&nbsp;&nbsp;&nbsp;&nbsp;&nbsp;&nbsp; mTOR &gt;AKT&gt; signal pathway in enhancing protein translation in skeletal muscle of finishing Yorkshire barrows. We will assay for the phosphorylated and not-phosphorylated signal proteins to describe the pathway&rsquo;s involvement in ractopamine (beta-agonist) treated late finishing barrows. . For many years, the hypothesis has floated that beta-agonist likely affect both translation (protein synthesis) and protein turnover. Based on available data this hypothesis can be extended to the notion while beta-agonist enhancement of muscle protein synthesis is a short term initial response, while depressing protein turnover may be occurring secondarily via transcriptional control of the proteasomal-protein degradation mechanisms. Plans are to investigate this possibility that beta-agonist enhancement of protein accretion is due to early translation enhancement and later (or more sustained) diminution of protein degradation.</p><br /> <p>&nbsp;</p><br /> <p>&nbsp;&nbsp;&nbsp;&nbsp;&nbsp;&nbsp;&nbsp;&nbsp;&nbsp;&nbsp;&nbsp; &nbsp;&nbsp;&nbsp;&nbsp;&nbsp;&nbsp;&nbsp;&nbsp;&nbsp;&nbsp;&nbsp;&nbsp;&nbsp;&nbsp;&nbsp;&nbsp;&nbsp;&nbsp;&nbsp;&nbsp;&nbsp;&nbsp;&nbsp;&nbsp;&nbsp;&nbsp;&nbsp;&nbsp;&nbsp;&nbsp;&nbsp;&nbsp;&nbsp;&nbsp;&nbsp;&nbsp;&nbsp;&nbsp;&nbsp;&nbsp;&nbsp;&nbsp;&nbsp;&nbsp;&nbsp;&nbsp;&nbsp; Citations</p><br /> <p>Huang C., L. Chiba, WE Magee, Y Wang, DA Griffing, LM Toressa, SP Rodning, , CL Bratcher, WG Bergen, EA Spangler. 2019. Effect of flaxseed oil, animal fat, and vitamin E supplementation on growth performance, serum metabolites, and carcass characteristics of finisher pigs, and physical characteristics of pork. Livestock Science 220:143-151.</p><br /> <p>&nbsp;</p><br /> <p>Huang C., L. Chiba, WE Magee, Y Wang, SP Rodning, , CL Bratcher, WG Bergen, EA Spangler. 2019. Effect of flaxseed oil, poultry fat, and vitamin E on physical and organoleptic characteristics and fatty acid profiles of pork, and expression of genes associated with lipid metabolism. /https:// doi org 10, 1016, Livestock Sci 2019. 103849.</p><br /> <p>&nbsp;</p><br /> <p>Bergen WG, 2020. Amino Acids in Beef Cattle Nutrition and Production. Advances in Experimental Medicine and Biology&nbsp;(A Springer Nature publication-Guo Wu, editor; accepted and in press).</p><br /> <p>&nbsp;</p><br /> <p>&nbsp;</p><br /> <ol><br /> <li>University of Georgia</li><br /> <li>Woo Kyun Kim</li><br /> <li>Accomplishments:</li><br /> </ol><br /> <p><strong>The effects of maternal fish oil supplementation on offspring-broiler growth performance, body composition (fat, muscle and bone) at market age. </strong>S</p><br /> <p>Maternal supplement of fish oil which contains high levels of long chain omega-3 polyunsaturated fatty acids has been shown to benefit the growth and development of offspring in humans and mice. It positively influences bone formation in animal models as well as osteogenesis in cell culture, however, the effect in poultry still remains unknown. In order to evaluate the effect of maternal dietary fish oil on growth performance, body composition, and bone quality in market-age broilers, breeder hens were fed the experimental diets containing 2.3 % food-grade soybean oil (SO) or fish oil (FO) for 4 wk. Fertilized eggs were collected over a period of 7 days and incubated under standard conditions (99.5 <sup>o</sup>F and 60% humidity). A total of 240 one-day-old chicks from different maternal diet groups (SO group and FO group) were randomly selected and allocated in12 floor pens (6 replicates /20 birds). All chicks were raised for 42 d. Growth performance were recorded at d 1, 14, 28 and 42, and feed conversion ratio was calculated in each feeding phase and overall. On d 42, 3 birds per pen (18 birds/ treatment) were randomly selected for body composition measurement by a Dual Energy X-ray Densitometry (DEXA), and the femurs were collected for bone microarchitectural analyses by micro-CT. Bone marrow from femurs was collected to measure expression of key osteogenic and adipogenic genes by qPCR. One-way ANOVA was performed, and means were compared by student&rsquo;s t-test (P&lt;0.05).</p><br /> <p>Maternal FO diet significantly improved the body weight and body weight gain in offspring broilers at the finisher stage (p&lt;0.05) and overall period (p&lt;0.05), whereas there was no difference in the feed conversion ratio during the experiment. At d 1, a higher percentage of body fat (p&lt;0.05) was observed in the FO group by DEXA, however, a higher percentage of lean mass was detected in the FO group at d 42 (p&lt;0.05). Microarchitectural analysis indicated that there were no significant differences in total bone mineral content and bone mineral density, or trabecular bone microarchitecture of the distal end of the femurs. However, in the middle shaft of femurs, the FO group had significantly lower cortical porosity, pore space volume, total porosity percentage, open porosity percentage, and open pore space volume (p&lt;0.05), and a higher number of closed-pore (P&lt;0.05). In addition, cortical bone mineral density (BMDCt) in was relatively higher (P= 0.054). Moreover, maternal FO supplementation suppressed the expression of adipogenesis-related gene, such as PPAR&gamma;, FABP4 and C/EBP&beta;, but did not significantly affect osteogenesis gene expression in the bone marrow. Cortical porosity and bone marrow adiposity are determinants of the bone strength and mechanical competence that has adverse effects on bone quality, the changes in porosity may be seen as a result of an alternation in bone growth and turnover. Based on those findings, the maternal fish oil intake not only enhanced body composition and lean mass in offspring, but also decreased adipogenesis in the bone marrow that coupled with lower porosity on cortical bone which indicated an improvement in bone quality.&nbsp;&nbsp;&nbsp;&nbsp;&nbsp;&nbsp;&nbsp;&nbsp;</p><br /> <p>&nbsp;</p><br /> <ol><br /> <li><strong>Published Papers</strong></li><br /> <li>Su*, Y. Wang, C. Chen*, M. Suh, M. Azain and <strong>W.K. Kim</strong> (2020) Fatty acid composition and regulatory gene expression in late-term embryos of ACRB and COBB broilers. Frontiers in Veterinary Science 7:317. <a href="https://doi.org/10.3389/fvets.2020.00317">https://doi.org/10.3389/fvets.2020.00317</a></li><br /> </ol><br /> <p><strong>&nbsp;</strong></p><br /> <p><strong>&nbsp;</strong></p><br /> <p><strong>&nbsp;</strong></p><br /> <ol start="2"><br /> <li><strong>Impacts</strong></li><br /> </ol><br /> <p>&nbsp;</p><br /> <p>We found that maternal supplementation of fish oil containing high omega-3 fatty acids affects body composition of offspring, increasing muscle and bone mass and reducing fat mass. This could be significant impact on human and animal health and wellbeing.</p><br /> <p>&nbsp;</p><br /> <p>&nbsp;</p><br /> <ol><br /> <li>Purdue University</li><br /> <li>Kola Ajuwon</li><br /> <li>Accomplishments:</li><br /> </ol><br /> <p>Publications:</p><br /> <p>Our focus in 2019 was understanding the mechanism involved in the regulation of allergic response to soy proteins using the pig model. We also investigated effects of fiber in the form of wheat bran and soy extract on glucose metabolism in the rat model. A major finding in 2019 was on the role of live yeast supplementation to sows in regulating growth performance and nutrient digestibility the offspring, demonstrating a carryover effect from mother to offspring. Additionally, our work on phytase elucidated the mechanism of the growth enhancing effect of phytase, the gene expression responses and metabolites that might mediate these responses. This work showed that phytase led to increased expression of muscle GLUT4 expression such that phytase could be increasing glucose uptake in the muscle through this glucose transporter. However, this observed effect of phytase might not be mediated by <em>myo</em>-inositol.</p><br /> <p>&nbsp;</p><br /> <p><strong>Publications:</strong></p><br /> <ol><br /> <li>Hashimoto-Hill S., M. Kim, L. Friesen, <strong>M. Ajuwon</strong>, E. Herman, A. Schinckel, and C.H. Kim. 2019. Differential food protein-induced inflammatory responses in swine lines 1 selected for reactivity to soy antigens. <em>Allergy. </em>&nbsp;74:1566-1569. doi: 10.1111/all.13757.</li><br /> <li>Alexis D. Stamatikos, A.D., J. E Davis, N.F. Shay, <strong>M. Ajuwon</strong>, F. Deyhim, and W. J. Banz. 2019. Consuming Diet Supplemented with Either Red Wheat Bran or Soy Extract Changes Glucose and Insulin Levels in Female Obese Zucker Rats. Int J Vitam Nutr Res. 90:23-32. doi: 10.1024/0300-9831/a000547.</li><br /> <li>Lu, H., P. Wilcock, O. Adeola, <strong> M. Ajuwon</strong>*.<sup>. </sup>2019. Effect of live yeast supplementation to gestating sows and nursery piglets on postweaning growth performance and nutrient digestibility. <em>J Anim Sci. 97:2534-2540. doi: 10.1093/jas/skz150.</em></li><br /> <li>Lu, H., I. K&uuml;hn, M.R. Bedford, H. Whitfield, Brearley, O. Adeola, and <strong>K. M. Ajuwon</strong>*. 2019. Effect of phytase on intestinal phytate breakdown, plasma inositol concentrations and glucose transporter type 4 abundance in muscle membranes of weanling pigs. <em>J Anim Sci. 2019 97:3907-3919. doi: 10.1093/jas/skz234.</em></li><br /> <li>Lu, H., A. J. Cowieson, J. W. Wilson, <strong>M. Ajuwon</strong>*, and O. Adeola<sup>. </sup>2019. Extra-phosphoric effects of super dosing phytase on growth performance of pigs is not solely due to release of <em>myo</em>-inositol. <em>J Anim Sci. 97:3898-3906. doi: 10.1093/jas/skz232.</em></li><br /> </ol><br /> <p>&nbsp;</p><br /> <p>&nbsp;</p><br /> <ol><br /> <li>Texas A&amp;M</li><br /> <li>Stephen Smith</li><br /> <li>Accomplishments:</li><br /> </ol><br /> <p><strong>Accomplishments:</strong></p><br /> <p><strong>&nbsp;</strong></p><br /> <p><span style="text-decoration: underline;">Impact of beef&rsquo;s unique food matrix on human health &ndash; Cholesterol metabolism and voluntary nutrient intake in men consuming low-fat and high-fat ground beef (funded by NCBA)</span></p><br /> <p>We have documented the effects of low-fat (6.4% fat) and high-fat (26.9%) on risk factors for cardiovascular disease in men. We hypothesized that consumption of high-fat ground beef would increase high-density lipoprotein cholesterol (HDL-C) concentrations in men, whereas consumption of low-fat ground beef would have no effect or may even decrease HDL-C concentrations. Ground beef consumption had no effect on total energy intake (<em>P</em> &ge; 0.05). The high-fat ground beef intervention decreased percent energy (%EN) from carbohydrates (<em>P</em> &lt;0.05) and increased %EN from fat (<em>P</em> &lt; 0.05), relative to entry. Consumption of the high-fat ground beef decreased plasma total cholesterol (<em>P</em> &lt; 0.05, LDL-C (<em>P</em> &lt; 0.05), and HDL-C (<em>P</em> &lt; 0.05), but did not change the HDL:LDL (<em>P</em> &gt; 0.05), relative to entry and washout values (Table 1). The low-fat ground beef also decreased HDL-C (<em>P</em> &lt; 0.05) but had no effect of LDL-C (<em>P</em> &gt; 0.05). Contrary to our initial hypothesis, the high-fat ground beef intervention decreased rather than increased HDL-C concentrations.</p><br /> <p><strong>Short-term Outcomes:</strong> This research has demonstrated that the consumption of ground does not increase LDL-C, thereby providing U.S. consumers with a healthful protein source.</p><br /> <p><strong>Outputs:</strong> The results of this study are being prepared for publication in the Journal of Nutrition.</p><br /> <p><strong>Activities:</strong> This study represents one of several studies by the laboratory of S. B. Smith documenting the effects of high-fat beef on risk factors for cardiovascular disease. The data generated by this study will be used to obtain additional funding from commodity and national funding agencies.</p><br /> <p><strong>Milestones:</strong> Our goal is to improve the perception of high-fat beef as a healthful component of the U.S. diet by 2025.</p><br /> <p><strong>Impacts: </strong>The targeted impact of the research is to improve the economic, social, and health benefits for the U.S. consumer. Ground beef constitutes approximately 50% of beef consumption in the U.S., and high-fat ground beef (&gt; 25% fat) represents the most often purchase type of ground. This is especially true of lower economic groups.</p><br /> <p><strong>&nbsp;</strong></p><br /> <p>&nbsp;</p><br /> <p>&nbsp;</p><br /> <ol><br /> <li>Washington State University</li><br /> <li>Min Du</li><br /> <li>Accomplishments:</li><br /> </ol><br /> <p><strong>Objective 1 and 2: </strong></p><br /> <p>During the past year, we are continuing to define mechanisms regulating the differentiation of adipogenic and fibrogenic differentiation of progenitor cells. In mice, we found that maternal nutrition and stress during pregnancy alter fetal brown adipose tissue development which has long-term negative effects on metabolic health of offspring. In addition, we found that vitamin A supplementation during the pregnancy and lactation enhances fetal intramuscular fat development and overall animal growth, which is consistent with enhanced marbling fat deposition in beef cattle through neonatal vitamin A administration. In addition, we are exploring the mechanisms responsible for the difference in marbling and tenderness between Wagyu and Angus cattle, focusing on the differentiation of adipogenic/fibrogenic progenitor cells. In the following year, we will further define mechanisms regulating intramuscular fat development and its long-term effects on growth performance and meat quality of offspring. &nbsp;</p><br /> <p><strong>Impacts</strong>:</p><br /> <ol><br /> <li>Maternal nutrition has a major impact on fetal and neonatal adipose development, exerting long-term impacts on offspring growth performance.</li><br /> <li>Proper nutritional management of beef cattle during early development improves intramuscular adipose development, beef production efficiency and quality.</li><br /> <li>Differentiation of adipogenic/fibrogenic progenitors inside muscle has profound impacts on meat quality.</li><br /> </ol><br /> <p><strong>Peer-reviewed Journals: </strong></p><br /> <ol><br /> <li>Ren, L., Q. Li, X. Hu, Q. Yang, <strong> Du</strong>, Y. Xing, Y. Wang, J. Li, and L. Zhang. (2020). A novel mechanism of bta-miR-210 in bovine early intramuscular adipogenesis. <em>Genes</em>, In press.</li><br /> <li>Chen, Y.T., Y. Hu, Q.Y. Wang, J.S. Son, X.D. Liu, J.M. de Avila, M. J. Zhu, and <strong> Du</strong>. (2020). Excessive glucocorticoids during pregnancy impair fetal brown fat development and predisposes offspring to metabolic dysfunctions. <em>Diabetes</em>, In press.</li><br /> <li>Tian Q.Y., X.D. Liu, and <strong> Du</strong>. (2020). Alpha-ketoglutarate for adipose tissue rejuvenation. <em>Aging</em>, In press.</li><br /> <li>Bordbar F., J. Jensen, <strong> Du</strong>, A. Abied, W. Guo, L. Xu, H. Gao, L. Zhang, and J. Li. (2020). Identification and validation of a novel candidate gene regulating net meat weight in Simmental beef cattle based on imputed next-generation sequencing. <em>Cell Proliferation</em>, In press.</li><br /> <li>Hu, Y., Y. Feng, L. Zhang, Y. Jia, D. Cai, S.B. Qian, <strong> Du</strong>, and R. Zhao. (2020). GR-mediated FTO transactivation induces lipid accumulation in hepatocytes via demethylation of m6A on lipogenic mRNAs. <em>RNA Biology</em>, In press.</li><br /> <li>Li, T., <strong> Du</strong>, H. Wang, and X. Mao. (2020). Milk fat globule membrane and its component phosphatidylcholine induce adipose browning both in vivo and in vitro. <em>Journal of Nutritional Biochemistry</em>, 81: 108372.</li><br /> <li>Sun, J., L. Zhao, Y. Chen, K. Chen, S. A. Chae, J. M. de Avila, H. Wang, M.J. Zhu, Z. Jiang, and <strong> Du</strong>. (2020). Maternal exercise via exerkine apelin enhances brown adipogenesis and prevents metabolic dysfunction in offspring mice. <em>Science Advances</em>, 6: eaaz0359.</li><br /> <li>Li, T., H. Gong, Q. Yuan, <strong> Du</strong>, F. Ren, and X. Mao. (2020). Supplementation of polar lipids-enriched milk fat globule membrane in high fat diet fed rats during pregnancy and lactation promotes brown/beige adipocyte development and prevents obesity in male offspring. <em>FASEB Journal</em>, 34: 4619-4634.</li><br /> <li>Yuan, Q., B. Zhan, R. Chang, <strong> Du</strong>, and X. Mao. (2020). Antidiabetic effect of casein glycomacropeptide hydrolysates on high-fat diet and STZ-induced diabetic mice via regulating insulin signaling in skeletal muscle and modulating gut microbiota. <em>Nutrients</em>, 12: E220.</li><br /> <li>Sun, Y.N., J.Q. Huang, Z.Z. Chen, <strong> Du</strong>, F.Z. Ren, J. Luo, and B. Fang. (2020). Amyotrophy induced by a high-fat diet is closely related to inflammation and protein degradation determined by quantitative phosphoproteomic analysis in skeletal muscle of C57BL/6 J mice. <em>Journal of Nutrition</em>, 150: 294-302.</li><br /> <li>Tian, Q., J. Zhao, Q. Yang, B. Wang, J. Deavila, M.J. Zhu, and <strong> Du</strong>. (2020). Dietary alpha-ketoglutarate promotes beige adipogenesis and prevents obesity in middle-aged mice. <em>Aging Cell</em>, 19: e13059.</li><br /> <li>Zhao, L., B. Wang, N. Gomez, J. Deavila, M. J. Zhu, and <strong> Du</strong>. (2020). Even a Low Dose of Tamoxifen Profoundly Induces Adipose Tissue Browning in Female Mice. <em>International Journal of Obesity</em>, 44,226-234.</li><br /> <li>Bordbar, F., J. Jensen, B. Zhu, Z. Wang, L. Xu, T. Chang, L. Xu, <strong> Du</strong>, L. Zhang, H. Gao, L. Xu, and J. Li. (2019). Identification of muscle-specific candidate genes in Simmental beef cattle using imputed next generation sequencing. <em>PLOS One</em>, 14: e0223671.</li><br /> <li>Li, M., X. Guo, B. Qin, X. Yang, J. Jia, J. Niu, M. Li, C. Cai, Y. Zhao, P. Gao, <strong> Du</strong>, B. Li, and G. Cao. (2019). Comparison of carcass traits, meat quality and expression of MyHCs in muscles between Mashan and Large White pigs.<em> Italian Journal of Animal Science</em>. 18: 1410-1418.</li><br /> <li>Li, X., X. Fu, G. Yang, and <strong> Du</strong>. (2019). Review: Enhancing intramuscular fat development via targeting fibro-adipogenic progenitor cells in meat animals. <em>Animal</em>, 14: 312-321.</li><br /> <li>Xie, S., Y. Li, W. Teng, <strong> Du</strong>, Y. Li, and B. Sun. (2019). Liensinine inhibits beige adipocytes recovering to white adipocytes through blocking mitophagy flux <em>in vitro</em> and <em>in vivo</em>. <em>Nutrients</em>, 11: 1640.</li><br /> <li>Teng, W., Y. Li, <strong> Du</strong>, X. Lei, S. Xie, and F. Ren. (2019). Sulforaphane prevents hepatic insulin resistance by blocking serine palmitoyltransferase 3-mediated ceramide biosynthesis. <em>Nutrients</em>, 11: 1185.</li><br /> <li>Costa, T. C., F. H. Moura, R. O. Souza, M. M. Lopes, M.S. Fontes, N. V. Serao, L. P. Sanglard, <strong> Du</strong>, M. P. Gionbelli, and M. S. Duarte. (2019). Effect of maternal feed restriction in dairy goats at different stages of gestation on skeletal muscle development and energy metabolism of kids at the time of births. <em>Animal Reproduction Science</em>, 206: 46-59.</li><br /> <li>Zhao, L., Y. Huang, and <strong> Du</strong>. (2019). Farm Animals for Studying Muscle Development and Metabolism: dual purposes for animal production and human health. <em>Animal Frontiers</em>, 9:3.</li><br /> <li>Wei, S., A. Li, L. Zhang, and <strong> Du</strong>. (2019). Long noncoding RNAs in adipogenesis and adipose development of meat animals. <em>Journal of Animal Science</em>, 97: 2644-2657.</li><br /> <li>Wang, H., Y. Chen, X. Mao, and <strong> Du</strong>. (2019). Maternal obesity impairs fetal mitochondriogenesis and brown adipose tissue development partially via upregulation of miR-204-5p. <em>BBA-Molecular Basis of Disease</em>, 1865: 2706-2715.</li><br /> </ol><br /> <p>&nbsp;</p><br /> <p>&nbsp;</p><br /> <p>&nbsp;</p><br /> <ol><br /> <li>The Ohio State University</li><br /> <li>Kichoon Lee</li><br /> <li>Accomplishments:</li><br /> </ol><br /> <p>Discovery of novel adipose-specific genes and consolidation of obesity loci.</p><br /> <p>Discovery of each adipose-specific gene (Leptin, Adiponectin, PPAR, FABP4, ATGL, G0S2 etc.) has profoundly contributed to our understanding of adipocyte biology and their etiologic implications for obesity and obesity-related diseases. Although an average of about 200 tissue-specific genes are estimated in each tissue, more adipose-specific genes in humans and animals need to be identified to understand their roles in adipose development and the development of obesity. To identify additional adipose-specific genes in humans, the most recent Genotype-Tissue Expression (GTEx) data v7 obtained from more than 400 individuals and 46 tissues were used for analysis. In addition, data from Gene Expression Omnibus and genome-wide association studies (GWASs) were also analyzed for this project. According to our analysis, 38 adipose-specific genes were identified. These genes were further analyzed for expression patterns at different developmental stages of adipogenesis and obesity conditions. In addition, 414 differentially expressed genes between subcutaneous and omental adipose depots were identified. Furthermore, some of the depot-specific genes (subcutaneous versus omental depots) were perfectly matched with particular GWAS loci for hip circumferences, waist circumferences and waist-to-hip ratio, consolidating association of these genes with the regional fat distribution. These results were published in Scientific Reports. Because genetic networks regulating adipose development and fat accretion are conserved in mammalian and avian species, the novel adipose-specific genes will be further studied to understand their roles in development of obesity and fat accretion in food animals.&nbsp;</p><br /> <p>Papers published:</p><br /> <ol><br /> <li>Ahn J, Woodfint R, Lee J, Wu H, Ma J, Suh Y, Hwang S, Cressman M, Lee K. 2019. Comparative identification and nutritional and physiological regulation of chicken liver-specific genes. Poultry Science DOI:10.3382/ps/pez057.</li><br /> <li>Lee B, Lee BJ, Lee YK, Hur SW, Kim KD, Kim KW, Han HS, Shin S, Choe JH, Lee K, Choi YM. 2019. Muscle fiber growth in olive flounder, Paralichthys olivaceus: Fiber hyperplasia at a specific body weight period and continuous hypertrophy. Journal of the World Aquaculture Society DOI: 10.1111/jwas.12580.</li><br /> <li>Ahn J, Wu H, Lee K. 2019. Integrative Analysis Revealing Human Adipose-Specific Genes and Consolidating Obesity Loci. Scientific Reports 9(1):3087.</li><br /> <li>Kim DH, Lee JW, Lee K. 2019. Supplementation of all-trans retinoic acid below cytotoxic levels promotes adipogenesis in 3T3-L1 cells. Lipids 54:99-107.</li><br /> <li>Woodfint R, Hanmlin E, Lee K. 2018. Avian Bioreactor Systems: A Review. Molecular Biotechnology 60(12):975-983.</li><br /> <li>Ahn J, Kim DH, Suh Y, Lee JW, Lee K. 2018. Adipose-specific expression of mouse Rbp7 gene and its developmental and metabolic changes. Gene 670:38-45.</li><br /> <li>Biswas A, Lee SS, Mamuad L, Kim SH, Choi YJ, Lee C, Lee K, Bae GS, Lee S. 2018. Effects of illite supplementation on in vitro and in vivo rumen fermentation, microbial population and methane emission of Hanwoo steers fed high concentrate diets. Animal Science Journal 89(1):114-121.</li><br /> </ol><br /> <p>&nbsp;</p><br /> <p>&nbsp;</p><br /> <ol><br /> <li>Purdue University</li><br /> <li>Kola Ajuwon</li><br /> <li>Accomplishments:</li><br /> </ol><br /> <p>Our focus in 2018 was understanding the mechanism involved in the regulation of adipose and intestinal tissue development and function. We continue to conduct research in the area of effects of fiber and fiber products on metabolism and function of intestinal tissue. A major finding in 2018 was on the role of 18C fatty acids with different degrees of saturation in the regulation of metabolism.&nbsp; We also investigated the response of adipose tissue to heat stress in pigs. This response was in favor of increased adipose tissue triglyceride storage. We determined the phosphoenolpyruvate carboxy kinase (PEPCK) played a major role in triglyceride storage in adipose tissue during heat stress. We also determined the identity of metabolites that might be involved in the response to heat stress in adipose tissue using metabolomics.</p><br /> <p>Publications:</p><br /> <ol><br /> <li>Qu. H., and K.M. 2018. Adipose tissue specific responses reveal an important role of lipogenesis during heat stress adaptation in pigs. J. Anim. Sci. 96:975-989. doi: 10.1093/jas/sky022.</li><br /> <li>Shin, S. and K.M. Ajuwon. 2018. Divergent response of murine and porcine adipocytes to stimulation of browning genes by 18-carbon polyunsaturated fatty acids and beta-adrenergic receptor agonists. Lipids. 53:65-75. doi: 10.1002/lipd.12010.</li><br /> <li>Shin, S. and K.M. Ajuwon. 2018. Effects of diets differing in composition of 18-C fatty acids on adipose tissue thermogenic gene expression in mice fed high-fat diets. Nutrients. 10(2). pii: E256. doi: 10.3390/nu10020256.</li><br /> <li>Almeida, V.V., H. Yan, C. H. Nakatsu and K. M. Ajuwon. 2018. Investigation of carry-over effect of prior fiber consumption on diet-induced obesity susceptibility and metabolic health indicators in Ossabaw pigs. J. Anim. Physiol. Anim. Nutr. (Berl). 102:1053-1061. doi: 10.1111/jpn.12900. Contribution: Almeida was Ajuwon&rsquo;s postdoctoral scholar.</li><br /> <li>Qu. H., and K.M. 2018. Cytosolic phosphoenolpyruvate is a response gene involved in porcine adipocyte adaptation to heat stress. J. Anim. Sci. 96:1724-1735. doi: 10.1093/jas/sky126.</li><br /> <li>Qu. H., and K.M. Ajuwon. 2018. Metabolomics of heat stress response in pig adipose tissue reveals alteration of phospholipid and fatty acid composition during heat stress. J. Anim. Sci. 96:3184-3195. doi: 10.1093/jas/sky127.</li><br /> <li>Shin, S. and K.M. Ajuwon. 2018. Lipopolysaccharide Alters Thermogenic and Inflammatory Genes in White Adipose Tissue in Mice Fed Diets with Distinct 18-Carbon Fatty-Acid Composition. Lipids. 53:885-896. doi: 10.1002/lipd.12101.</li><br /> </ol><br /> <p>&nbsp;</p><br /> <p>&nbsp;</p><br /> <ol><br /> <li>University of Arkansas</li><br /> <li>Yan Huang</li><br /> <li>Accomplishments</li><br /> </ol><br /> <p><span style="text-decoration: underline;">Effect of EPA and DHA on C2C12 myoblasts adipogenesis</span></p><br /> <p>We have demonstrated that n-3 polyunsaturated fatty (PUFAs) acids affect myoblast differentiation during myogenesis. The hypothesis is that the PUFAs such as EPA and DHA promotes adipogenesis during C2C12 myoblast differentiation. Data showed up-regulation of white adipogenic marker genes in EPA and DHA treated cells, while no effect was observed among the brown adipogenic marker genes expression. Mitochondrial biosynthesis and activity were inhibited by EPA and DHA treatment. During brown adipogenic transdifferentiating, EPA and DHA inhibited Krebs cycle and electron transport chain.</p><br /> <p><strong>Short-term Outcomes: </strong>Data suggest that excessive levels of EPA and DHA could promote the adipogenic potential of C2C12 myoblasts, and are associated with mitochondrial dysfunction in trans-differentiation into brown adipocytes.</p><br /> <p><strong>Outputs: </strong>The results of this study have been published in Animal Cells and Systems, and Frontiers in Genetics.</p><br /> <p><strong>Activities: </strong>This is one of several studies conducted by the laboratory of Yan Huang to study the effect of maternal nutrition on offspring muscle and fat growth. The data generated by this study will be used to apply for additional funding from the USDA-NIFA program.</p><br /> <p><strong>Milestone:</strong> To demonstrate the regulation of intramuscular fat deposition in livestock animals and childhood obesity in medical science through maternal nutrition by 2025.</p><br /> <p><strong>Impacts:</strong> The targeted impacts of the research are to improve the efficiency of producing high-quality meat from livestock animals and to discover the treatment of childhood obesity by monitoring maternal nutrition.</p><br /> <p>&nbsp;</p><br /> <p><strong>Publications: </strong>(July 2019 - July 2020)</p><br /> <p>1)&nbsp;&nbsp;&nbsp;&nbsp;&nbsp;&nbsp;&nbsp;&nbsp; S. Ghnaimawi, J.I. Baum, R. Liyanage, and <strong>Y. Huang</strong>. Concurrent EPA and DHA Supplementation Impairs Brown Adipogenesis of C2C12 Cells. Front. Genet. 11:531. DOI: 10.3389/fgene.2020.00531</p><br /> <p>2)&nbsp;&nbsp;&nbsp;&nbsp;&nbsp;&nbsp;&nbsp;&nbsp; H. Wu, S. Dridi, <strong>Y. Huang</strong>, and J.I. Baum. Leucine Decreases Intramyocellular Lipid Deposition in an mTORC1-independent Manner in Palmitate-treated C2C12 Myotubes. 2019. Am J Physiol Endocrinol Metab. 2019 Nov 26. DOI: 10.1152/ajpendo.00241.2019.</p><br /> <p>3)&nbsp;&nbsp;&nbsp;&nbsp;&nbsp;&nbsp;&nbsp;&nbsp; S. Ghnaimawi, S.Shelby, J.I. Baum, and <strong>Y. Huang</strong>. Effects of Eicosapentaenoic Acid and Docosahexaenoic Acid on C2C12 Cell Adipogenesis and Inhibition of Myotube Formation. 2019. Animal Cells and Systems. 23 (5), 355-364. DOI: 10.1080/19768354.2019.1661282</p><br /> <p>4)&nbsp;&nbsp;&nbsp;&nbsp;&nbsp;&nbsp;&nbsp;&nbsp; G. Liu, <strong>Y. Huang</strong>, F. S. Reis, D. Song, and H. Ni. Impact of Nutritional and Environmental Factors on Inflammation, Oxidative Stress, and the Microbiome. BioMed Research International. Volume 2019. DOI: 10.1155/2019/ 5716241</p><br /> <p>5)&nbsp;&nbsp;&nbsp;&nbsp;&nbsp;&nbsp;&nbsp;&nbsp; L. Zhao, <strong>Y.</strong> <strong>Huang</strong>, and M. Du. Farm Animals are Important Biomedical Models. Animal Frontiers. 2019. Volume 9, Issue 3, July 2019, Pages 21&ndash;27. DOI: 10.1093/af/vfz015.</p><br /> <p>6)&nbsp;&nbsp;&nbsp;&nbsp;&nbsp;&nbsp;&nbsp;&nbsp; B. Alrubaye, M. Abraha, A. Almansour, M. Bansal, H. Wang, Y.M. Kwon, <strong>Y. Huang</strong>, B. Hargis, and X. Sun. Microbial metabolite deoxycholic acid shapes microbiota against Campylobacter jejuni chicken colonization. PLoS ONE. 2019. 14(7): e0214705. DOI: 10.1371/journal.pone.0214705</p><br /> <p>&nbsp;</p>

Publications

Impact Statements

Back to top

Date of Annual Report: 07/09/2021

Report Information

Annual Meeting Dates: 07/09/2021 - 07/09/2021
Period the Report Covers: 07/08/2020 - 07/08/2021

Participants

1. Yuan-Yu Lin, National Taiwan University
2. Chao-Wei Huang, National Pingtung University
3. Theo van Kempen, Nutreco, Netherlands
4. WooKyun Kim, University of Georgia
5. Derris Devost-Burnett, Mississippi State University
6. Kichoon Lee, The Ohio State University
7. Sheila Jacobi, The Ohio State University
8. Jim Kinder, The Ohio State University
9. Kola Ajuwon, Purdue University
10. Enkai Li, Purdue University
11. Tobi Ogunribido, Purdue University
12. Sean Adams, UC Davis
13. Min Du, Washington State University
14. Werner G. Bergen, Auburn University
15. Steve Smith, Texas A & M
16. Don Beitz, Iowa State University
17. Minjeong Kim, University of Tennessee, Knoxville
18. U-Suk Jung, University of Tennessee, Knoxville

Brief Summary of Minutes

The meeting this year was held virtually on Zoom platform on July 7, 2021. Kichoon Lee of The Ohio State University was teh convener, assisted by Kola Ajuwon of Purdue University. We discussed how to increase teh membership. We also discussed the format of our future meetings. We had several presentations from multiple stations. The agenda is in the attached file. The presentations were lively, engaging and highly informative. Members agreed to reconvene next year, again virtually.

Accomplishments

<ul><br /> <li><strong>Short-term Outcomes:</strong>&nbsp;Increased knowledge of adipose tissue development and role in the regulation of animal growth, animal and human health.</li><br /> <li><strong>Outputs:</strong>&nbsp;Members published several excellent quality publications in the reporting year.</li><br /> <li><strong>Activities:</strong>&nbsp;Members had an annual meeting where latest research in the area were discussed. Members also attended multiple scientific meetings in the past year.</li><br /> <li><strong>Milestones:</strong>&nbsp;Members made significant progress in ther research areas and we continue to meet annually.&nbsp;</li><br /> </ul>

Publications

Impact Statements

  1. Activities of the NCCC210 have contributed significantly to knowledge of adipose biology in farm animals that have helped to increase efficiency of animal growth, bringing increased profitability to animal producers. Activities conducted by members of NNNNc210 have helped to train multiple scholars that are conducting impactful research. Our efforts have also contributed to better understanding of obesity etiology.
Back to top

Date of Annual Report: 11/15/2022

Report Information

Annual Meeting Dates: 11/14/2022 - 11/14/2022
Period the Report Covers: 07/09/2021 - 11/15/2022

Participants

1. Kim Barnes, West Virginia University; 2. Sheila Jacobi, Ohio State University; 3. Rui Zheng, Ohio State University; 4. Rebecca Brown, Ohio State University; 3. Theo van Kempen, North Carolina State University; 4. WooKyun Kim, University of Georgia; 5. Kichoon Lee, The Ohio State University; 6. Joonbum Lee, The Ohio State University; 7. Donghwan Kim, The Ohio State University; 8. Kola Ajuwon, Purdue University; 9. Enkai Li, Purdue University; 10. Yan Huang, University of Arkansas; 11. Sesha Reddy, University of Georgia; 12. Min Du, Washington State University; 13. Steve Smith, Texas A & M; 14. Don Beitz, Iowa State University; 15. Brynn Voy, University of Tennessee, Knoxville; 16. Minjeong Kim, University of Tennessee, Knoxville; 17. U-Suk Jung, University of Tennessee, Knoxville

Brief Summary of Minutes

The meeting this year was held virtually on Zoom platform on November 14, 2022. Sheila Jacobi of The Ohio State University convened the meeting at 10:00AM EST. Kim Barnes gave an administrative update for the NCCC210 group. We discussed the format of future meetings and that zoom had allowed us to connect easier since many of us go to several different professional meetings. We had ten presentations representing multiple stations. The group presented data of multiple species of food production animals related to growth and lipid metabolism in health and disease (see attached agenda). The presentations were informative and engaging with scientific discussions around the areas of research. Providing feedback and elaborating on potential collaboration. At the end of the presentations administrative round up of increasing numbers at meetings, the discussion of a summer 2023



Webinar for the group (Committee Steve Smith, Kola Ajuwon, and Sheila Jacobi),the station reports should be submitted to Sheila Jacobi by December 14th for summary into final report, and Yan Huang, Assistant Professor at the University of Arkansas, agreed to the chair for the NCCC210 Annual Meeting in 2023.


2023 Webinar Committee will be reaching out in early 2023 to collect feedback on webinar topics.

Accomplishments

<ul><br /> <li><strong>Short-term outcomes: </strong>Increased the knowledge of adipose development, lipid metabolism and regulation of food animal growth and health. Develop of further knowledge around large animal models of human physiology.</li><br /> <li><strong>Outputs: </strong>Members published several excellent quality publications in the reporting year. Graduate students training was enriched by presentations and scientific conversations and feedback.</li><br /> <li><strong>Activities</strong>: Members were actively engaged with their graduate student and postdoctoral trainees in the annual meeting to discuss the latest research. Further, members are have attended several scientific meetings throughout the past year.</li><br /> <li><strong>Milestones</strong>: Members made significant progress in their research endeavors, developed strong collaborations, and we will continue to meet annually. Additionally, in 2023 the NCCC210 will be planning a Webinar focused around a to-be-determined area within the group.</li><br /> </ul>

Publications

Impact Statements

  1. 1. Activities of the NCCC210 group have contributed significantly to the knowledge of adipose tissue and lipid biology in multiple food production animals. The research is important in efficiency of animal growth to improve producer profitability and generating safe and affordable protein for human nutrition. Members of the NCCC210 have been instrumental in training graduate student and postdoctoral scholars who contribute to the generation of impactful data from their research. Further, the large animal models of adipose and lipid metabolism generate meaningful data for understanding human biology and disease.
Back to top
Log Out ?

Are you sure you want to log out?

Press No if you want to continue work. Press Yes to logout current user.

Report a Bug
Report a Bug

Describe your bug clearly, including the steps you used to create it.