NCCC_old210: Regulation of Adipose Tissue Accretion in Meat-Producing Animals (NCCC210)

(Multistate Research Coordinating Committee and Information Exchange Group)

Status: Inactive/Terminating

NCCC_old210: Regulation of Adipose Tissue Accretion in Meat-Producing Animals (NCCC210)

Duration: 10/01/2014 to 09/30/2019

Administrative Advisor(s):


NIFA Reps:


Non-Technical Summary

Statement of Issues and Justification

NCCC210, Regulation of Adipose Tissue Growth in Meat Animals, meets the needs of several stakeholder communities, namely, consumers, livestock producers, and scientists. The committee is composed of scientists with broad interests and expertise in factors that affect the development and composition of adipose tissue in meat that is an important component of human diets. The need to control excessive adipose tissue growth, to identify the fatty acid composition of this tissue, and understand the underpinnings of lipid metabolism in adipose and muscle will provide consumers with meat and meat products with enhanced nutritional value. Meat animal producers will have greater information to make informed decisions about production practices that will enhance meat animal productive efficiency, sensory characteristics, and advance nutritional contributions of meat in human diets. Most importantly, scientists will benefit from data exchange, developments in techniques and methodology that arise from the project through advancing the quest for knowledge of adipose tissue biology. Indeed, adipose tissue presents a dynamic metabolic paradigm as knowledge of its composition, biochemistry, molecular biology, endocrinology, and morphology rapidly progresses.

Importance of the work

The Project Committee meets annually to report and discuss research conducted by individual members, and to advance collaborations among members. The importance of the work conducted by the Project members in the context of the annual meeting and year-round regular correspondence is best summed up as development of ideas and strategies, and critique of planned studies, so that subsequent research is conducted to provide data of importance to advancement of adipose tissue biology of meat animals. In the light of exploding understanding of adipose biology there is a critical need to further continue this research with meat animal species and comparative animal models. If the research of this Project is not conducted, or if the group does not meet to advance the research, then knowledge and understanding of current research technologies will be stifled through the void in scientific interchange. The centrality of adipose tissues in whole body metabolism puts this Committee's expertise and work at the epicenter of meat animal lipid research. Continuing this project group will result in further enhancement of biological understanding that is relevant to meat production strategy, dietary qualities of meat products for use in human diets. Because the project's science focuses in part in energy partitioning and lipid trafficking, it has implications for milk production and quality, as well. In addition to clear agricultural relevance, the project members' efforts in elucidating adipose biology and fat metabolism also contribute to knowledge important to public health, e.g. obesity and related metabolic maladies.

NCCC210 and its predecessors have been at the forefront of animal fat synthesis and deposition related research for well over 60 years. Initially the committee had a strong meat science orientation which over the years expanded also into areas of growth and development, nutrition and the biology of obesity. While adipose tissue is necessary for survival to supply and store energy and as an endocrine organ, excess fat is expensive to produce and is unwanted by consumers of lean meat. However, the deposition of intramuscular (marbling) adipose tissue must be promoted to achieve maximum carcass/product quality (for the producer) as well as often times a desired eating experience (for consumers). Animal growth can be modified to decrease fat production by nutritional, pharmacological, and physiological control of intrinsic or extrinsic processes, but this must be balanced with maintaining marbling deposition. Project Committee members continue to be the primary multi-state and Land Grant Universities contributors in the area of adipose tissue biology.

Over the last 40+ years the committee has been closely aligned with NC1184 (a national scope multi-state committee) which was organized to concentrate on meat animal protein metabolism and muscle biology. Predecessors of NCCC210 were initially a major contributor to solving the then critical issues in meat animal fat metabolism including such topics as soft pork. Committee members were also keenly interested in comparative lipid metabolism and were among the first to establish the sites and mechanisms of de novo fatty acid synthesis across different species. Aside from many peer-reviewed publications in animal fat biology, a landmark publication at the time was Biology of Fat in Meat Animals, published by the Project Committee members in 1976 (College of Agriculture, University of Wisconsin).


Members have and continue to focus on nutritional/ pharmacological treatments to change food animals to a more lean body composition. The utility of dietary conjugated linoleic acids in regulating fat deposition and the potential use of thiazolidinediones (human diabetes medicine) in marbling has been pursued more recently. Over the last 30 years mechanisms controlling adipocyte hyperplasia, differentiation, and growth (hypertrophy) and effects of somatotropin and beta-adrenergic agonists on body composition were also emphasized. An important outcome of the latter research was publication of The Biology of Fat in Meat Animals: Current Advances, by Project Committee members in 1995 (ASAS Press). Committee members also spawned the emergence of growth biology in University (Animal Sciences) curricula and in the establishment of the Growth and Biology section in the Journal of Animal Science.


The present scope of NCCC210 (NCR 97) Project Committee members is national and includes academic and USDA/ARS participants from throughout the USA. The impact of NCCC210 is not only felt in U.S. academia and government laboratories, but also in the food production and harvesting sector both in the U.S. and on the international stage. Different industry representatives, keynote speakers, and ad hoc participants are invited to each meeting to provide a unique perspective on adipose tissue biology that complements Committee member expertise. As an example, Dr. Shih-Torng Ding, National Taiwan University Professor and former postdoctoral research associate of Dr. Harry Mersmann (retired NCCC-097/210 member), continues to provide the committee with an international perspective through his guest participation. Dr. Sean Adams (USDA Western Human Nutrition Research Center), Research Leader for the Obesity and Metabolism Research Unit, helps link Committee findings and expertise to nutrition and human metabolic health application.


Currently, NCCC210 members address problems in adipose tissue biology by using techniques in molecular biology/gene expression, metabolite and fatty acid quantification and immunology. One continuing theme in the committee efforts has been to develop, perfect and utilize cutting-edge experimental systems to address contemporary issues in what is now often referred to as lipidology. Thus, members are now planning to adopt high-throughput procedures in metabolomics, gene expression, micro-array analyses, new PCR technologies, next generation sequencing (DNA) and RNA sequencing. Developing broad use of these methods for meat animal lipid metabolism and associated areas as obesity by a group of lipid experts on the technical committee from various institutions (bio-medicine and agriculture) will assure (and has) adoption of such experimental approaches by colleagues working in growth and development, fat metabolism, meat sciences and nutrition at various land-grant institutions. Due to the species-independent nature of modern omics research analytical tools, the Committee has a unique opportunity to leverage these approaches to push the field of meat animal metabolism to new heights.


Before about 1990, basic mechanisms of fat synthesis and oxidation were typically studied using key enzymes in fat-related metabolic pathways. In the biomedical community, similar such work was proceeding, as well as early development of adipocyte tissue culture. A major impetus into studying molecular regulation of adipocyte differentiation and lipid metabolism in food animal species was the work of Goldstein and Brown (1) and Spiegelman (2). They discovered and delineated the roles of specific transcription factors in molecular regulation of de novo lipogenesis and cholesterol metabolism. Emergence of these hitherto unknown new paradigms in molecular regulation of cell differentiation and fat metabolism gave great impetus to similar studies with food animal species by NCCC210 committee members at participating state institutions and USDA laboratories as well as the biomedical community at large. Earlier studies by NCCC210 participants have now been extended to studies of gene expression and genomic approaches to even further delineate the intricacies of molecular regulation of lipid metabolism and energy balance. For example, utilizing microarray and RNA sequence analysis and targeted gene expression facilitated by laser capture micro-dissection and real-time PCR on serial tissue samples from cattle and pigs, workers at the Alabama Station (Bergen, Brandebourg: Auburn Univ.) are obtaining new data on potential physiological factors related to residual feed intake/feed efficiency, heat tolerance and the molecular regulation of adipose depot development as related to marbling. Other participating stations have also utilized these approaches to study the role of genetics/genomics in fatty acid composition in beef cattle (Beitz: Iowa State Univ.), and other transcription factors and gene cascades (Lee: Ohio State Univ.) as well as micro-array based gene profiling of either fasted or insulin-deprived chicken (Voy, Univ. of Tennessee) and pig (Hausman: Univ. of Georgia) adipose tissues. Research by various members of the Committee is also proceeding to identify additional transcription factors critical in regulating proliferation and differentiation of adipocytes. Among these are zinc-finger protein transcription factors which play a pivotal role in adipocyte differentiation (Du: Washington State Univ.).


While most of the Committee members have continued to focus on farm animal systems in their research, present efforts in molecular regulation of adipocyte differentiation and overall molecular regulation of fat metabolism is closely aligned with work done principally with laboratory rodents and transformed fat cells in culture. This clearly denotes that the committee membership and ad hoc participants are involved in cutting-edge research in lipid metabolism principally oriented toward animals for food use.


Not all research by the committee over the last 10 years is solely focused on gene-level regulation of lipid metabolism. Several project committee members have studied mechanisms for conjugated linoleic acid modification of growth and fat deposition (Barnes: W. Virginia Univ.), while another member studies development of omega-3 fatty acids in adipocytes of grass-fed beef. Stearoyl CoA desaturase (SCD) has a significant role in lipid metabolism in humans and food animals. It is a key enzyme that imparts some softness in animal fats by converting stearic acid to the monounsaturated oleic acid (18:1, n9) and SCD is transcriptionally controlled. Other research has focused on how to increase understanding of the role of the extracelluar matrix and dietary fatty acids on regulation of inflamation in adipocytes (Ajuwon: Purdue). The role of fatty acid oxidation in neonatal piglet survival and postnatal growth continues is a principal area of study by the North Carolina Station (Odle: NC State Univ.). Finally, there have been studies among several stations exploring the role and benefits of omega 3 highly unsaturated fatty acids (HUFA) primarily in rodent or swine lipid metabolism. While important for many aspects in meat production, such findings also are useful as animal model studies to human nutrition and medicine. Application of metabolomics approaches to questions of fuel trafficking and obesity-associated phenotypes has been a focus of some Committee member efforts (Adams: USDA WHNRC, Davis). These studies have, for instance, highlighted interactions between perturbations in branched chain amino acid metabolism, changes in fat oxidation and metabolic disease outcomes (3, 4). This may be applicable to animal food production in light of leucine's known role in triggering protein synthesis, since a goal of meat production is to obtain desired lean/fat ratios and fatty acid ratios (omega 3/omega 6). These efforts are an example of significant interactions between the technical committee and human nutrition and lipid metabolism scientists elsewhere and indicate a working relationship between NCCC210 and scientists in related areas of biomedicine and human nutrition.


In the last few years there has been increasing interest in utilizing stromal vascular cells (pre-lipid) and dedifferentiated fat cells as a source of stem cells for research and therapeutic purposes. This is a novel extension of the plethora of work with cell cultures with primary fat cells or specific cell lines. Dr. Shih-Torng Ding of the National University at Taiwan (an international ad hoc regular participant in NCCC210) has made some interesting advances here while committee associated scientists at Washington State, Georgia and Auburn have recognized the possibilities of stem cell production from adipose cells and have written a timely review paper (5).


Further the deliverables from the committee include extensive explorations of comparative lipid metabolism and application of data to human nutrition and lipid metabolism issues. Another influence of the Committee on agricultural and life sciences research has through timely publications and books. During the 2004-2009 project period, nine members of the Committee published a paper entitled The Biology and Regulation of Preadipocytes and Adipocytes in Meat Animals (6). This paper is now among highly cited articles in the Journal of Animal Science. More recently, based on their wide experience in lipid metabolism and nutrition, Committee members from Washington State University, University of Georgia and Auburn University have published a paper entitled A long journey to effective obesity treatments: Is there light at the end of the tunnel? (7). Committee members were invited to speak at a 2011 joint conference (Agri-Medical Research: Providing Dual Benefit for Agriculture and Human Health) sponsored by American Society for Nutrition (ASN), American Society of Animal Science (ASAS), and American Dairy Science Association (ADSA) in New Orleans. Committee members initiated a NCCC210-sponsored American Society for Nutrition symposium entitled Adipose and Lipid Biology: Crossing Taxonomic Boundaries for which speakers submitted follow-up articles to the journal Advances in Nutrition (8). Thus, the Committee has remained very active in knowledge dissemination to the animal science and nutrition communities through publication and conference participation.


Members have hundreds of thousands of dollars in grants each year from USDA/AFRI, commodity groups, industry, and state funds. They serve on multiple journal editorial boards (J. Anim. Sci., J. Nutr., Dom. Anim. Endocrin., Exp. Biol. Med., Adv. Nutr.), serve as Editor and section editors for J. Anim. Sci., review multiple grants, present numerous seminars and symposium talks, and organize and chair meeting sessions and symposia.

Technical feasibility of the plans and research

The research of Committee individuals is greatly enhanced because of the interaction at the meeting with detailed discussion, critique and trouble-shooting of experimental design, methods, results and interpretation. Members with less expertise in a specific area or sub-discipline have access to input by other members with demonstrated expertise. Thus, detailed presentation and discussion of research design, methodologies and results at the meeting greatly increases the feasibility of the projects and the probability of success. As research approaches and methodologies continue to advance and evolve, Program Committee member contributions are markedly improved because of these interactions.

Advantages of doing the work as a multi-state effort


Advantages of collaboration as a multi-state effort are best defined as diversity of efforts emphasized by members from the individual states. The diversity includes species, discipline, and technical emphasis. This allows the Committee to span needs of consumers and industry more completely and to leverage the most precise and advanced technologies. Species diversity is partially dictated by inventories of these food producing animals in specific regions of the country. For example, faculty at the University of Wyoming focus on sheep and cattle, those in Texas and University of California-Davis focus primarily on beef cattle, and faculty working with swine are represented by a large number of Committee members. Moreover, other members of the Committee emphasize cell and tissue culture to accomplish goals consistent with those of the NCCC210 project.

Likely impacts of successful collaboration

Outcomes of collaboration among Project Committee members are not limited to the confines of the annual meeting. Individuals and collaborators who are a part of this Project Committee have been and continue to be successful in obtaining extramural research grants because of the research conducted, in part, in conjunction with input and involvement of Project Committee members. The Project Committee members continue to conduct symposia focusing specifically on regulation of adipose tissue accretion in food producing animals at national scientific meetings, as well as publishing proceedings and books on this topic. Advancements realized by Project Committee members also positively impact the biomedical community because of the direct link to human nutrition and human adipose tissue biology that has relevance to disease states such as obesity, inflammation, and the metabolic syndrome. The greatest impacts of the NCCC210 multi-state project have been: 1) Enhanced quality of research in making the whole greater than the sum of the parts through collegial collaborations, particularly at the annual meeting that is held preceding the Experimental Biology meetings; 2) The broad utilization of the knowledge base that is communicated through Project Committee member publications that result under the auspices of the NCCC210 Multistate Project Committee; and 3) Future agricultural science leadership development; the NCC210 Project Committee meeting always includes participation and speaking opportunities for graduate students and postdoctoral fellows, and an unparalleled networking opportunity. Several Committee Members were previous students and postdoctoral associates who now lead academic and government laboratory groups.



Objectives

  1. At annual NCCC210 committee meetings members will share and critique new techniques, experimental designs, and in progress-recent data regarding adipocyte biology and animal lipid metabolism to enhance innovative research, experimental approaches, methodologies and data interpretation for applications ranging from livestock and meat production to human health and nutrition.
  2. Plan to increasingly implement newly emerging high through-put omics based technologies and experimental methodologies to further refine our understanding of adipose tissue biology and its role in regulation of whole body metabolism in farm animals, animal models and humans.
  3. Initiate and sponsor two symposia during the next five years at Experiential Biology (EB) and American Society of Animal Science (ASAS) meetings. One symposium will be targeted on comparative animals lipid biology and adipocyte function (at EB) while the second will focus on lipid metabolism in livestock species (at ASAS).
  4. Publish peer-reviewed scientific reviews based on contemporary issues and controversies in lipid biology and obesity, on impact of climate change on adipose deposition and growth of meat animals and the potential role of adipocyte culture in stem cells production. The expertise of the committee will serve an educational/broadening function for students and workers in the field.
  5. Summarize preliminary results from common areas of work from a number of committee participants to develop and submit joint research projects. Members of the Project Committee plan to develop research proposal(s) for coordinated study in lipid biology with application to animal agriculture, human obesity, as well as appropriate in vitro models for use in clinical applications of tissue regeneration /reconstruction. The annual interaction by all members, as well as more frequent interactions of select members will enhance the opportunities for extramural funding shared between our institutions to further research and discovery in the biology of lipids, especially utilizing emerging research methodologies for adipose tissue in meat animals and humans.

Procedures and Activities

NCCCC210's renewal will continue to provide expertise and national leadership for research in modern concepts of adipocyte biology, in vivo fat deposition, intramuscular/intra-fascicular adipose development and animal growth. This is expressed in the contribution of member's expertise to review panels such as USDA NRI Animal Growth and Nutrient Utilization grant programs. In addition members have been on the joint NIH/USDA panel on agricultural animal research as a research model for biomedical issues. A significant crossover from animal to human lipidology research will be a main theme of the committee's activities in this renewal period (2014-2019).

While adoption of new experimental strategies and equipment has been a hallmark of this committee, the areas of comprehensive lipidomics, high throughput gene expression based on RNA sequencing and utilization of bioinformatics in our research will be much more evident during this renewal period. This will allow the committee to focus on within species lipidology for specific understanding in enhanced detail and through the use of bioinformatics will enhance the impact of comparative lipidology.

Share and summarize preliminary data from various stations to develop multi-institutional grants in adipocyte biology. An emerging area here is the use of adipose tissues for stem cell production. In addition develop avenues to share unique infrastructures/equipment from different representative institution. Such may include advanced ultra-sensitive chromatography equipment for lipid metabolomics, mass spectrometer and sequencing /bioinformatics technology not available to all committee members.

Plan two symposia programs to be held at annual meetings of Experimental Biology (EB/ASN) and American Society of Animal Sciences (ASAS). This will include identifying timely topics and speakers. As indicated in objective 3, the plan is to utilize the extensive expertise of committee members. The EB symposia will be targeted toward comparative lipid biology and adipocyte cell function. For ASAS the symposium will be targeted to food animal lipidology, myocyte-adipocyte cross talk and regulation of adipocyte differentiation. In addition the committee desires to write additional review articles for major publications emphasizing lipid biology and obesity and role of climate change on adipose deposition and animal growth (see objective 3).

Within the committee membership, adipocyte culture and differentiation mechanisms remain key components of their research. This research is likely to be expanded to attempts to systemize production of stem cells from adipose tissues.


The committee will maintain its influence by continuing interactions with other scientists in the lipid metabolism area in the meat sciences, nutrition and biochemistry communities by active participation in activities of these groups and learned societies. Above all however the principal activities of the committee will revolve around the annual meetings.

Expected Outcomes and Impacts

  • Exchange of ideas, information, techniques, experimental designs, and data.The open and extensive exchange of information and ideas at the annual Project Committee meeting provides important suggestions to investigators about design of experiments and new approaches not previously considered. Input from multiple persons with expertise about experiments and data interpretation heightens the quality of scientific endeavors of Project Committee members. The annual Project Committee meeting that is held at the same venue and immediately preceding the Experimental Biology meetings is the primary and in many cases only venue through which all members of the committee work with each other in an interactive forum.
  • Initiate new and relevant individual and collaborative research projects. Although much of the experimentation by members precludes exchange of materials because of sample fragility, co-operative projects arise from the high quality and thought provoking research discussions between members at the annual meeting.
  • Publication of original research and review articles pertaining to adipocyte biology and lipid metabolism, including joint publications with other members. Members continue to be at the forefront of modern approaches to the biology of adipocyte growth, with emphasis on meat producing species.
  • Authoritative presentation of concepts of adipocyte biology and lipid metabolism at lectures, symposia, and conferences. The publication record of NCCC210 has been superlative over the last 40 years. As the original members retire, they are replaced by equally prolific scientists. The publication of journal articles, symposium proceedings, reviews, book chapters, and books about adipocyte growth by members is expected to continue in the future.
  • Continue to provide expertise and national leadership for research in modern concepts of adipocyte biology and animal growth. This is expressed in the contribution of members to review panels such as the USDA NRI Animal Growth and Nutrient Utilization grant programs.
  • Project Milestones and Dates for Completion of the Objectives: Members of this committee conduct research in the areas described within the Objectives. Progress within any specific Objective is directly associated with the amount of funding each member obtains to develop appropriate experiments. There is no possible means to establish timelines or milestones because the funding sources are inconsistent and grants are obtained by individuals rather than the committee as a group, although a multi-institutional grant proposal is anticipated. It is our ongoing goal to develop collaborative research; however, the uncertainty with which funding occurs under our Committees established framework does not allow for discrete timelines. The continuation of activities under objective 1 is a constant goal and an important benchmark for each year of the project duration. For service/education oriented activities such as objectives 3 and 4 will be initiated in 2014 with actual symposia during 2016 to 2018. Reviews and other scholarly writings will be submitted as suitable during the timespan of the project.

Projected Participation

View Appendix E: Participation

Educational Plan


The annual Project Committee meeting is enhanced by attendance of graduate students and postdoctoral fellows associated with members of the Committee. The gathering is very informal around a large table. These young scientists are encouraged to actively participate and to present results to the group and several have been employed to tenure track positions and now contribute as state representative Project Committee members. Typically, a representative from industry is invited to participate/present at the meeting allowing members to make contacts and discuss funding opportunities. New members are solicited, as current members become aware of scientists doing research on adipose tissue growth. The results from the research of Project Committee members are disseminated in a multiplicity of reviewed journal articles coupled with numerous published symposia proceedings and book chapters.

Organization/Governance

The Governance for NCCC210 includes the election of a Chair, a Chair-elect, and Secretary. All officers are elected for one year. The Chair-elect arranges the meeting for the subsequent year. The annual meeting is held in junction with Experimental Biology (Federation of American Societies of Experimental Biology). Continuity is provided by the stable membership and excellent attendance. Administrative guidance is provided by an assigned Administrative Advisor and a CSREES Representative.

Literature Cited

1.Horton JD, Goldstein JL, Brown MS: SREBPs: activators of the complete program of cholesterol and fatty acid synthesis in the liver. J Clin Invest. 2002 May; 109(9):1125-31


2.Tontonoz P, Kim JB, Graves RA, Spiegelman BM ADD1: a novel helix-loop-helix transcription factor associated with adipocyte determination and differentiation.Mol Cell Biol. 1993 Aug; 13(8):4753-9.


3.She P, Olson KC, Kadota Y, Inukai A, Shimomura Y, Hoppel CL, Adams SH, Kawamata Y, Matsumoto H, Sakai R, Lang CH, Lynch CJ: Leucine and protein metabolism in obese Zucker rats. PLoS One. 2013;8(3):e59443. doi: 10.1371/journal.pone.0059443. Epub 2013 Mar 20.


4. Lackey DE, Lynch CJ, Olson KC, Mostaedi R, Ali M, Smith WH, Karpe F, Humphreys S, Bedinger DH, Dunn TN, Thomas AP, Oort PJ, Kieffer DA, Amin R, Bettaieb A, Haj FG, Permana P, Anthony TG, Adams SH. Regulation of adipose branched-chain amino acid catabolism enzyme expression and cross-adipose amino acid flux in human obesity. Am J Physiol Endocrinol Metab. 2013, 304:E1175-87.



5. Wei S, Zhang L, Zhou X, Du M, Jiang Z, Hausman GJ, Bergen WG, Zan L, Dodson MV: Emerging roles of zinc finger proteins in regulating adipogenesis.
Cell Mol Life Sci. 2013 Jun 13. [Epub ahead of print]



6. Hausman GJ, Dodson MV, Ajuwon K, Azain M, Barnes KM, Guan LL, Jiang Z, Poulos SP, Sainz RD, Smith S, Spurlock M, Novakofski J, Fernyhough ME, Bergen WG. Board-invited review: the biology and regulation of preadipocytes and adipocytes in meat animals. J Anim Sci. 2009 Apr; 87(4):1218-46.



7. Dodson MV, Boudina S, Albrecht E, Bucci L, Culver MF, Wei S, Bergen WG, Amaral AJ, Moustaid-Moussa N, Poulos S, Hausman GJ: A long journey to effective obesity treatments: is there light at the end of the tunnel? Exp Biol Med (Maywood). 2013 May; 238(5):491-501.


8.Adams SH, Barnes KM, Odle J.Comparative metabolic physiology in the 'omics' era: a call to arms, paws, flippers, and claws. Adv Nutr. 2013 Sep 1;4(5):568-9 doi: 10.3945/an.113.004374


Publications of NCCC210 2008-20012


2009

Ashwell, M.S., RP. Ceddia, R.L. House, J.P. Cassady, E.J. Eisen, T.E. Eling, J.B. Collins, S.F. Grissom, and J. Odle. 2009. J. Nutr. Biochem. http://dx.doi.org/10.1016/j.jnutbio.2009.06.013

Barnes, K.M. and J.L. Miner. 2009. The role of resistin in insulin sensitivity in rodents and humans. Curr. Prot. Pept. Sci. 10:96-107.

Barnes, K.M., J.K. Evenson, A.M. Raines, and R.A. Sunde. 2009. Selenium regulation of the selenoproteome in rats. J. Nutr. 139:199-206.

Basu, U., M. Taniguchi, Y. Zhao, M.V. Dodson and L.L. Guan. 2010. Application of 'omics' technologies on improvement of meat quality in livestock species. In Nutritional Biochemistry: Genomics, Metabolomics and Food Supply. Nova Science Publishers, Inc. Hauppauge, NY. Chapter 4. (ISBN 978-60741-916-7), 30 pages

Bergen WG and G Wu. 2009. Intestinal nitrogen recycling and utilization in health and disease. J Nutr139: 821-825.

Bergen WG. 2009. Milk fat depression and lipid repartitioning in lactating cows. J Nutr 139: 826-827.

Broughton, K. S., D. C. Rule, Y. Ye, X. Zhang, M. Driscoll, and B. W. Culver. 2009. Dietary omega-3 fatty acids differentially influence ova release and ovarian cyclooxygenase-1 and cyclooxygenase-2 expression in rats. Nutr. Res. 29:197-205.

Brown K.R., R.D. Goodband, M.D. Tokach, S. S. Dritz, J.L. Nelssen, J.E. Minton, J.J. Higgins, X. Lin, J. Odle, J.C. Woodworth, and B.J. Johnson. 2008. Effects of feeding L-carnitine to gilts through day 70 of gestation on litter traits and the expression of insulin-like growth factor system components and L-carnitine concentration in foetal tissues. J. Anim. Physiol. Anim. Nutr. 92:660-667.

Chen, J., M.V. Dodson and Z. Jiang. 2010. Cellular and molecular comparison of redifferentiation of intramuscular- and subcutaneous- adipocyte derived progeny cells. International Journal of Biological Science 6(1):80-88

Chen, J., M. Guridi, M.E. Fernyhough, Z. Jiang, L.L. Guan, G.J. Hausman and M.V. Dodson. 2009. Clonal mature adipocyte production of proliferative-competent daughter cells requires lipid export prior to cell division. International Journal of Stem Cells 2:76-79

Corl, B.A., J. Odle, X. Niu, A.J. Moeser, L.A. Gatlin, O.T. Phillips, A.T. Blikslager, and J. M. Rhoads. 2008. Arginine activates intestinal p70S6k and protein synthesis in piglet rotavirus enteritis. J. Nutr. 2008;138 24-29.

Corl, B.A., S.A. Mathews Oliver, X. Lin, W.T. Oliver, Y. Ma, R.J. Harrell, and J. Odle 2008. Conjugated linoleic acid reduces body fat accretion and lipogenic gene expression in neonatal pigs fed low- or high-fat formulas. J. Nutr. 138:449-454.

Davidson, S., B.A. Hopkins, J. Odle, C. Brownie, V. Fellner and L.W. Whitlow, 2008. Supplementing limited methionine diets with rumen-protected methionine, betaine, and choline in early lactation holstein cows. J. Dairy Sci. 91:1552-1559.


Dodson, M.V., L.L. Guan, M.E. Fernyhough, P.S. Mir, L. Bucci, D.C. McFarland, J. Novakofsky, J.M. Reecy, K.M Ajuwon, D.P. Thompson, G.J. Hausman, W.G. Bergen, M. Benson and Z. Jiang. 2010. Perspectives on the formation of an interdisciplinary research team. Biochemical Biophysical Research Communications 391:1155-1157

Dodson, M. V., Z. Jiang, J. Chen, G. J. Hausman, l. L. Guan, J. Novakofski, D. P. Thompson, C. L. Lorenzen, M. E. Fernyhough, P. S. Mir, and J. M. Reecy. 2010. Allied industry approaches to alter intramuscular fat content and composition in beef animals. J Food Sci 75:R1-R8.

Dodson, M. V., J. L. Vierck, G. J. Hausman, L. L. Guan, M. E. Fernyhough, S. P. Poulos, P. S. Mir, and Z. Jiang. 2010. Examination of adipose depot-specific PPAR moieties. Biochem. Biophys. Res. Commun. 394:241-242.

Dong, F., Y. Hua, P. Zhao, J. Ren, M. Du, and N. Sreejayan. (2009). Chromium inhibits skeletal muscle atrophy in hindlimb suspended mice. Journal of Nutritional Biochemistry, 20: 992-999.


Du, M., X. Yan, J. F. Tong, J. X. Zhao, and M. J. Zhu. (2010). Maternal obesity, inflammation and fetal skeletal muscle development. Biology of Reproduction, 82: 4-12.



Hausman, G. J., M. V. Dodson, K. Ajuwon, M. Azain, K. M. Barnes, L. L. Guan, Z. Jiang, S. P. Poulos, R. D. Sainz, S. Smith, M. Spurlock, J. Novakofski, M. E. Fernyhough, and W. G. Bergen. 2009. BOARD-INVITED REVIEW: The biology and regulation of preadipocytes and adipocytes in meat animals. Journal of Animal Science 87:1218-1246.

Hausman, G. J. and C. R. Barb. 2010. Adipose Tissue and the Reproductive Axis: Biological Aspects. Endocr. Dev. 19:31-44
.
He, M.L., R. Sharma, P. Mir, E. Okine and M.V. Dodson. 2010. Feed withdrawal regimens can abate lipodystrophy, but glucose tolerance is associated with appropriate diameter of retroperitoneal adipocytes in rats. Nutrition Research 30:125-133


Hess, H.A., B.A. Corl, X. Lin, S.K. Jacobi, R.J. Harrell, A.T. Blikslager and J. Odle. 2008. Enrichment of intestinal mucosal phospholipids with arachidonic and eicosapentaenoic acids fed to suckling piglets is dose and time dependent. J. Nutr. 138:2164-2171.


Herfel, T.M., S.K. Jacobi, X. Lin, D.C. Walker, Z.E. Jouni, and J. Odle. 2009. Safety evaluation of polydextrose in infant formula using a suckling piglet model. Food Chem. Tox. 47:1530-1537



Knotts,T., H.W. Lee, J.B. Kim, P.J. Oort, R. McPherson, R. Dent, K. Tachibana, T. Doi, S. Yu, J.K. Reddy, K. Uno, H. Katagiri, M. Pasarica, S.R. Smith, D.D. Sears, M. Grino, and S.H. Adams. Molecular characterization of the tumor suppressor candidate 5 gene: regulation by PPAR³ and identification of TUSC5 coding variants in lean and obese humans. PPAR Research, vol. 2009, Article ID 867678, 13 pages, doi:10.1155/2009/867678, 2009


Li B, J Shin, and K Lee. Interferon-stimulated gene ISG12b1 inhibits adipogenic differentiation and mitochondrial biogenesis in 3T3-L1 cells. Endocrinology. 2009. 150(3):1217-1224.

Liu LF, A Purushotham, AA Wendel, K Koba, J Deiuliis, K Lee, and MA Belury. Regulation of adipose triglyceride lipase by rosiglitazone. Diabetes Obes. Metab. 2009.11(2):131-142.


Lee K, J Shin, JD Latshaw, Y Suh, and J Serr. Cloning of adipose triglyceride lipase complementary deoxyribonucleic acid in poultry and expression of adipose triglyceride lipase during development of adipose in chickens. Poult. Sci. 2009. 88(3):620-


Lkhagvadorj, S., L. Qu, W. Cai, O. P. Couture, C. R. Barb, G. J. Hausman, D. Nettleton, L. L. Anderson, J. C. Dekkers, and C. K. Tuggle. 2010. Gene expression profiling of the short-term adaptive response to acute caloric restriction in liver and adipose tissues of pigs differing in feed efficiency. Am. J Physiol Regul. Integr. Comp Physiol 298:R494-R507.


Liu, F., J. Yin, M. Du, P. Yan, J. Xu, X. Zhu, and J. Yu. (2009). Heat stress induced damage to porcine small intestinal epithelium associated with down-regulation of epithelial growth factor (EGF) signaling. Journal of Animal Science, 87: 1941-1949.


Ludden, P. A., O. Kucuk, D. C. Rule, and B. W. Hess. 2009. Growth and carcass fatty acid composition of beef steers fed soybean oil for increasing duration before slaughter. Meat Sci. 82:185-192.


Mack,C.M., C.J. Soares, J.K. Wilson, J.R. Athanacio, V.F. Turik, J.L. Trevaskis, J.D. Roth, P.A. Smith, B. Gedulin, C.M. Jodka, B.L. Roland, S.H. Adams, A. Lwin, J. Herich, K.D. Laugero, C. Vu, R. Pittner, J.R. Paterniti Jr., M. Hanley, S. Ghosh, D.G. Parkes. Davalintide (AC2307), a novel amylin-mimetic peptide: enhanced pharmacological properties over native amylin to reduce food intake and body weight. Int. J. Obesity 2009 Nov 24.


McPhee, M. J., J. W. Oltjen, J. G. Fadel, D. G. Mayer, and R. D. Sainz. 2009. Parameter estimation and sensitivity analysis of fat deposition models in beef steers using acsl. Xtreme. Mathematics and Computers in Simulation 79: 2701-2712.

Murrieta, C.M., B. W. Hess, S. L. Lake, E. J. Scholljegerdes, and D. C. Rule. 2010. Body condition score and day of lactation regulate fatty acid metabolism in milk somatic cells and adipose tissue of beef cow. Livestock Sci. doi:10.1016/jlivesci.2010.02.028.

Poulos, S. P., D. B. Hausman, and G. J. Hausman. 2010. The development and endocrine functions of adipose tissue. Mol. Cell Endocrinol, 323:20-34.


Rule, Daniel C. 2009. Lipids in Muscle. Structure, composition, and metabolism. In (M. Du and R. J. McCormick, eds.) Applied Muscle Biology and Meat Science, pp. 247-273. CRC Press, Boca Raton, Fl.


Schriever, SC, KM Barnes, JK Evenson, AM Raines, and RA Sunde. 2009. Selenium
requirements are higher for glutathione peroxidase-1 mRNA than Gpx 1 activity in rat
testis. Exp. Biol. Med. 234:513-521.



Seifert,E., O. Fiehn, V. Bezaire, D.B. Bickel, G. Wohlgemuth, S.H. Adams, and M-E. Harper. Long-chain fatty acid combustion rate is associated with unique metabolite profiles in skeletal muscle mitochondria. (co-corresponding authors) PLoS One 5(3): e9834, 2010


Shin S, JY Han, and K Lee. Cloning of Avian delta-like 1 homolog (Dlk1) gene: The biallelic expression of Dlk1 in avian species. Poult Sci. 2010. 89(5):948-955.


Serr J, Y Suh, and K Lee. Regulation of adipose triglyceride lipase by fasting and re-feeding in avian species. Poult Sci. 2009. 88(12):2585-2591.


Shin J, B Li, ME Davis, Y Suh, and K Lee. Comparative analysis of fatty acid binding protein 4 promoters: Conservation of PPAR binding sites. J. Anim. Sci. 2009. 87(12):3923-3934.


Shin J, DR Bae, JD Latshaw, MP Wick, JM Reddish, and K Lee. Technical note: A gene delivery system in the embryonic cells of avian species using a human adenoviral vector. J. Anim. Sci. 2009. 87:2791-2795.


Shin J, SG Velleman, JD Latshaw, MP Wick, Y Suh, and K Lee. The ontogeny of Delta-Like Protein 1 mRNA expression during muscle development and regeneration: Comparison of broiler and leghorn chickens. Poult Sci. 2009. 88:1427-1437.


Su, S.Y., M.V. Dodson, X.-B. Li, Q.F. Li, H.W. Wang and Z. Xie. 2009. The effects of dietary betaine supplementation on fatty liver performance, serum parameters, histological changes, methylation status and the mRNA expression level of spot14± in Landes goose fatty liver. Comparative Biochemistry and Physiology (Part A; Molecular and Integrative Physiology) 154:308-314

Tong, J. F., X. Yan, M. J. Zhu, and M. Du. (2009). AMP-activated protein kinase enhances the expression of muscle-specific ubiquitin ligases despite its activation of IGF-1/Akt signaling in C2C12 myotubes. Journal of Cellular Biochemistry, 108: 458-468.


Tong, J. F., X. Yan, M. J. Zhu, S. P. Ford, P. W. Nathanielsz, and M. Du. (2009). Maternal obesity down-regulates myogenesis and ²-catenin signaling in fetal skeletal muscle. American Journal of Physiology-Endocrinology and Metabolism, 296: E917-924.


Xi, L., K. Brown, J. Woodworth, K. Shim, B. Johnson and J. Odle. 2008. Maternal dietary L-carnitine supplementation influences fetal carnitine status and stimulates carnitine palmitoyltransferase and pyruvate dehydrogenase complex activities in swine. J. Nutr. 138:2356-2362.


Yan, X., M. J. Zhu, W. Xu, J. F. Tong, S. P. Ford, P. W. Nathanielsz, and M. Du. (2010). Up-regulation of TLR4/NF-ºB signaling is associated with enhanced adipogenesis and insulin resistance in fetal skeletal muscle of obese sheep at late gestation. Endocrinology, 151: 380-387.


Yue, T., J. Yin, F. Li, D. Li, M. Du. (2010). High glucose induces differentiation and adipogenesis in porcine muscle satellite cells via mTOR. BMB Reports, 43: 140-145.

Zhao, J.X. , X. Yan, J.F. Tong, W.J. Means, R. J. McCormick, M.J. Zhu, and M. Du. (2010). Mouse AMPKg3 R225Q mutation affecting its growth performance when fed a high energy diet. Journal of Animal Science, 88: 1332-1340.


Zhao, Y.M., U. Basu, M. Zhou, M.V. Dodson, J.A. Basarb and L.L. Guan. 2010. Proteome difference associated with different fat formation in the bovine subcutaneous fat tissues. BMC Proteome Science 8:14

2010


Adapala V.J., K. K. Buhman and K.M. Ajuwon. 2011. Novel anti-inflammatory role of SLPI in adipose tissue and its regulation by high fat diet. J Inflamm (Lond). 8:5.

Adapala, V.J., S.A. Adedokun, R.V. Considine and K.M. Ajuwon. 2011. Acute Inflammation Plays a Limited Role in the Regulation of Adipose Tissue COL1A1 Protein Abundance. J. Nutr. Biochem. In press.


Beitz, D.C., S. Zhang, T.J. Knight,and J.M. Reecy. 2011. Genetic markers in fatty acid synthase for identification of meat product fatty acid content in cattle. U.S. Patent No. US 7,910,308 B1.


Chu, P.Y., T.K. Li, S.T. Ding, I.R. Lai, and T.L. Shen. 2010. EGF-induced Grb7 recruits and promotes Ras activity essential for the tumorigenicity of Sk-Br3 breast cancer cells J. Biol. Chem. 285: 29279-85.



Deiuliis J, Shin J, Murphy E, Kronberg SL, Eastridge ML, Suh Y, Yoon JT, K Lee. Bovine adipose triglyceride lipase is not altered and adipocyte fatty acid-binding protein is increased by dietary flaxseed. 2010. Lipids. 45:963973.


Deiuliis JA, Liu LF, Belury MA, Rim JS, Shin S, and K Lee. Beta(3)-adrenergic signaling acutely down regulates adipose triglyceride lipase in brown adipocytes. Lipids. 2010. 45:479-489.


Dodson, M.V., G.J. Hausman, L.L. Guan, M. Du, T.P. Rasmussen, S.P Poulos, P. Mir, W.G. Bergen, M.E. Fernyhough, D.C. McFarland, R.P. Rhoads, B. Soret, J.M. Reecy, S.G. Velleman and Z. Jiang. 2010. Lipid metabolism, adipocyte depot physiology and utilization of meat animals as experimental models for metabolic research. International Journal of Biological Science 6(7):682-690


Dodson, M.V., J.L. Vierck, G.J. Hausman, L.L. Guan, M.E. Fernyhough, S.P. Poulos, P. Mir, and Z. Jiang. 2010. Examination of adipose depot-specific PPAR moieties. Biochemical Biophysical Research Communications 349:241-242


Dodson, M.V. G.J. Hausman, L.L. Guan, M. Du, T.P. Rasmussen, S.P. Poulos, P. Mir, W.G. Bergen, M.E. Fernyhough, D.C. McFarland, R.P. Rhoads, B. Soret, J.M. Reecy, S.G. Velleman and Z. Jiang. 2010. Skeletal muscle stem cells from animals I. Basic cell biology. International Journal of Biological Science 6(5):465-474


Dodson MV, GJ Hausman, GL Juan, M Du, TP Rasmussen, SP Poulos, P Mir WG Bergen,ME Fernyhough et al. 2010. Lipid metabolism, adipocyte depot physiology and utilization of meat animals as experimental models for metabolic research. Int J Biol Sci 6: 691-699.


Dodson MV, Guan LL, Fernyhough ME, Mir PS, McFarland DC, Novakofski, Reecy JM, Ajuwon KM, Thompson DP, Hausman GJ, Benson M, Bergen WG. and Jiang Z. 2010. Perspectives on the formationof an interdisciplinary research team. Biochem Biophys Res Comm 391: 1155-1157.


Du, M., J. X. Zhao, X. Yan, Y. Huang, L. V. Nicodemus, W. Yue, R. J. McCormick, and M. J. Zhu. (2011). Fetal muscle development, mesenchymal multipotent cell differentiation and associated signaling pathways. Journal of Animal Science, 89: 583-590.

Du, M., J. D. Yin, and M. J. Zhu. (2010). Cellular signaling pathways regulating adipogenesis and marbling of skeletal muscle. Meat Science, 86:103-109.



Du, M., J. F. Tong, J. X. Zhao, K. R. Underwood, M. J. Zhu, S. P. Ford, and P. W. Nathanielsz. (2010). Fetal programming of skeletal muscle development in ruminant animals. Journal of Animal Science, 88: E51-60.


Duan, Q., J. Reecy, R. Tait, Q. Liu, A. van Eenennaam, R. Mateescu, A. Garmyn, and D.C. Beitz. 2010. Genetic polymorphisms in bovine ferroportin are associated with longissimus dorsi muscle iron content. Abstr. No. 229.2 FASEB J. 24.


Fernyhough, M.E., L.R. Bucci, J. Feliciano and M.V. Dodson. 2010. The effect of nutritional supplements on the proliferation and differentiation of muscle-derived stem cells. International Journal of Stem Cells 3:63-67



Hausman, G. J., M. V. Dodson, K. Ajuwon, M. Azain, K. M. Barnes, L. L. Guan, Z. Jiang, S. P. Poulos, R. D. Sainz, S. Smith, M. Spurlock, J. Novakofski, M. E. Fernyhough, and W. G. Bergen. 2009. Board Invited Review: The biology and regulation of preadipocytes and adipocytes in meat animals. J. Anim Sci. 87: 1218-1246.


Huang, Y., X. Yan, J. X. Zhao, M. J. Zhu, R. J. McCormick, S. P. Ford, P. W. Nathanielsz, J. Ren, and M. Du. (2010). Maternal obesity induces fibrosis in fetal myocardium of sheep. American Journal of Physiology-Endocrinology and Metabolism, 298: E1254-1260.


Jang HJ, Choi JW, Kim YM, Shin SS, Lee K, Han JY. Reactivation of Transgene Expression by Alleviating CpG Methylation of the Rous sarcoma virus Promoter in Transgenic Quail Cells. 2011. Mol. Biotechnol. DOI 10.1007/s12033-011-9393-7.


Jin, W., M.V. Dodson, S.S. Moore, J.A. Basarb and L.L. Guan. 2010. Characterization of microRNA expression in bovine subcutaneous fat tissues: A potential regulatory mechanism of subcutaneous adipose tissue development. BMC Molecular Biology 11:29-37


Jin, W.W., J.M. Romao, M.V. Dodson, G.J. Hausman,S.S. Moore and L.L. Guan. submitted. MicroRNA regulation of mammalian adipogenesis. Experimental Biology and Medicine


Li, Q., J. Wang, D. Bu, Khas-Erdene, K. Liu, H. Wel, L. Zhou, and D.C. Beitz. 2010. Influence of linolenic acid content on the oxidation of milk fat. J. Agr. Food Chem. 58:3741-3746.


Lin, E.C. Lin, H. A. Hsu, M. C. Chao, F. T. Chan, L. M. Wang, H. S. Tsao, C. H. Chang, P. Y. Lin, B. J. Wang, H. W. Yuan, S. T. Ding, and P. H. Wang. 2011. Application of CHD1 gene and EE0.6 sequences to identify sexes of several protected bird species in Taiwan. Taiwania (accepted)


Liu, L.R., S.P. Lin, C.C. Chen, Y.J. Chen, T.L. Shen and S.T. Ding 2011. Serum amyloid A induces lipolysis by down-regulating perilipin through ERK1/2 and PKA signaling pathways. Obesity. (In press).



Meers, S. A., T. D. Pringle, R. D. Jones, and M. J. Azain. 2010. Effect of body composition on diet selection in finishing pigs. J. Anim. Sci. 88:1733-1740.


Michelizzi, V.N., M.V. Dodson, Z. Pan, J.J. Michal, D.J. McLean, J.E. Womack and Z. Jiang. 2010. Water buffalo genome science comes of age. International Journal of Biological Science 6:333-349


Michelizzi, V.N., X. Wu, M.V. Dodson, J.J. Michal, J. Jorge-Zambrano-Varon, D.J. McLean and Z. Jiang. 2011. A global view of 54,001 single nucleotide polymorphisms (SNPs) on the Illumina Bovine SNP-50 Bead Chip and their transferability to water buffalo. International Journal Biological Science 7(1):18-27


Oh S, Suh Y, Pang MG, K Lee. Cloning of avian G(0)/G(1) switch gene 2 (G0S2) genes and developmental and nutritional regulation of G0S2 in chicken adipose tissue. 2011. J. Anim Sci. 89(2):367-75.


Murrieta, C.M., B. W. Hess, S. L. Lake, E. J. Scholljegerdes, and D. C. Rule. 2010. Body condition score and day of lactation regulate fatty acid metabolism in milk somatic cells and adipose tissue of beef cow. Livestock Sci. 131:65-72.



Poulos, S.P., M.V. Dodson and G.J. Hausman. 2010. Cell line models of preadipocytes and adipocytes. Experimental Biology and Medicine 235:1185-1193



Relling AE, Lee K, Loerch SC, Reynolds CK. Effects of Glucose, Propionate and Splanchnic Hormones on Neuropeptide mRNA concentrations in the Ovine Hypothalamus. 2011. Journal of Animal Physiology and Animal Nutrition. (Accepted)


Schoonmaker, J.P., A.H. Trenkle, and D.C. Beitz. 2010. Effect of feeding wet distillers grains on performance, marbling deposition, and fatty acid content of beef from steers fed low- or high-forage diets. J. Anim. Sci. 88:3657-3665.


Serr J, Suh Y, K Lee Cloning of comparative gene identification-58 in avian species and investigation of its developmental and nutritional regulation in chicken adipose tissue. 2011. J. Anim Sci. doi:10.2527/jas.2011-3897

Serr J, Y. Suh, S.A. Oh, S. Shin, MS. Kim, J.D. Latshaw and K. Lee. 2011. Acute up-regulation of adipose triglyceride lipase and release of non-esterified fatty acids by dexamethasone in chicken adipose tissue. Lipids. (Accepted)

Shin S, JY Han, and K Lee. Cloning of Avian delta-like 1 homolog (Dlk1) gene: The biallelic expression of Dlk1 in avian species. 2010. Poult Sci. 89(5):948-955.


Tai, C. C. and S.T. Ding 2010. N-3 polyunsaturated fatty acids regulate lipid metabolism through several inflammation mediators: mechanisms and implications for obesity prevention. J. Nutr. Biochem. 21:357-363.


Tseng, Y.W., P.H. Wang, H.S. Lee, B.H. Liu, H.J. Mersmann, E.C. Lin, and S.T. Ding 2010. Regulation of the expression of angiotensin converting enzyme 2 by polyunsaturated fatty acids in porcine adipocytes. J. Anim. Sci. 88: 3563-3567.


Tu, P.A., E.C. Lin, J.W. Shiau, Y.T. Chen, S.T. Ding, F.Y. Lai, and P.H. Wang. 2011. Genetic variation and polymorphisms for the promoter region of the porcine myostatin gene. J. Agri. Assoc. Taiwan 12: 105-125.


Wang, Y.C. W.H. Kuo, C.Y. Chen, H.Y. Lin, H.T. Wu, B.H. Liu, C.H. Chen, H.J. Mersmann, K.J. Chang, and S.T. Ding 2010. Docosahexaenoic acid regulates serum amyloid A protein to promote lipolysis through down regulation of perilipin. J. Nutr. Biochem. 21:317-324.


Ward, M. and K.M. Ajuwon. 2011. Regulation of Pre-adipocyte Proliferation and Apoptosis by the Small Leucine Rich Proteoglycans, Biglycan and Decorin. Cell Proliferation, 44: 343-351.


Yan, X., M. J. Zhu, W. Xu, J. F. Tong, S. P. Ford, P. W. Nathanielsz, and M. Du. (2010). Up-regulation of TLR4/NF-ºB signaling is associated with enhanced adipogenesis and insulin resistance in fetal skeletal muscle of obese sheep at late gestation. Endocrinology, 151: 380-387.

Yang, C.C., S.J. Deng, C.C. Hsu, B.H. Liu, E.C. Lin, W.T.K. Cheng, P.H. Wang, and S.T. Ding 2010. Visfatin regulates genes related to lipid metabolism in porcine adipocytes. J. Anim. Sci. 88: 3233-3241.


Yu, Y. H., S. C. Wu, W. T. K. Cheng, H. J. Mersmann, T. L. Shen and S. T. Ding 2011. The function of porcine PPAR³ and dietary fish oil effect on the expression of lipid and glucose metabolism related genes. J. Nutri. Biochem. 22:179-186.


Yu, Y., P.H. Wang, H., W. T. K. Cheng, H. J. Mersmann, S. C. Wu, and S. T. Ding 2010. Porcine peroxisome proliferator-activated receptor ´ mediates the lipolytic effects of dietary fish oil to reduce body fat deposition. J. Anim. Sci. 88:2009-2018.

Zhang, S., T.J. Knight, J.M. Reecy, T.L. Wheeler, S.D. Shackelford, L.V. Cundiff, and D.C. Beitz. 2010. Associations of polymorphisms in the promoter I of bovine acetyl-CoA carboxylase-alpha gene with beef fatty acid composition. Anim. Gen. 41:417-420.



Zhou, J. X., J. Hu, M. J. Zhu, and M. Du. (2011). Trenbolone enhances myogenic differentiation by enhancing b-catenin signaling in muscle-derived stem cells of cattle. Domestic Animal Endocrinology, In


2011

Adedokun S.A., K.M. Ajuwon, L.F. Romero and O. Adeola. 2012. Ileal endogenous amino acid losses: Response of broiler chickens to fiber and mild coccidial vaccine challenge. Poult Sci. 91:899-907.
Adeola, O., A.P. Schinckel, K.M. Ajuwon and M.E. Einstein. 2012. Accretion rates and efficiency of amino acid utilization during the twenty-nine days post-hatch period in male ducks. J. Poult. Sci. 49: 79-85.



Alexander L.S., A. Mahajan, J. Odle, K.L. Flann, R.P. Rhoads, and C.H. Stahl. 2010. Dietary phosphate restriction decreases stem cell proliferation and subsequent growth potential in neonatal pigs. J. Nutr. 140:477-482.



Bai, S.P., L. Lu, R.L. Wang, L. Xi, L.Y. Zhang, and X.G. Luo. 2011. Manganese source affects manganese transport and gene expression of divalent metal transporter 1 in the small intestine of broilers. Br J Nutr. 15:1-10. [Epub ahead of print]



Bai, X.M., Q.G. Ma, L.H. Zhao, L. Xi, and C. Ji. 2011. Effects of alpha-lipoic acid supplementation on antioxidative ability and performance of sows and nursing piglets. J Anim Physiol Anim Nutr (Berl). 10.1111/j.1439-0396.2011.01205.x. [Epub ahead of print]


Banz, W.J., D. April D. Strader, K.M. Ajuwon, Y. Hou, C. Y. Meyers and J. Davis. 2012. (+)-Z-Bisdehydrodoisynolic acid enhances basal metabolism and fatty acid oxidation in female obese Zucker rats. J. Obesity. Vol. 2012 (2012), Article ID 154145,doi:10.1155/2012/154145.


Barnes, KM, NR Winslow, AG Shelton, KC Hlusko, and MJ Azain. 2012. Effect of dietary conjugated linoleic acid on marbling and intramuscular adipocytes in pork. J. Anim. Sci. 90:1142-1149.


Bergen WG, Burnett DD. 2012. Topics in transcriptional control of lipid metabolism: from transcription factors to gene-promoter polymorphisms. J Genomics 1:3-21.


Bost, K.L., L. Hudson, J. Odle, K.J. Piller, and C. Stahl. 2011. Acute, systemic cytokine responses in swine following non-lethal staphylococcal enterotoxin B challenge. Swine in Biomedical Research Conference. Chicago, IL. S4-2, page 63.


Brown, C., X. Lin, E. van Heugten, R. Harrell, D. Hanson, and J. Odle. 2010. Production of value-added pork by enrichment with omega-3 fatty acids. Midwest Regional Animal Science Meetings, Des Moines, IA. J. Anim. Sci. 88(E-Suppl. 3): 72.


Cabrera R.A., R.D. Boyd, S.B. Jungst, E.R. Wilson, M.E. Johnston, J.L. Vignes JL, and J. Odle. 2010. Impact of lactation length and piglet weaning weight on long-term growth and viability of the progeny. J. Anim. Sci. 88:2265-2276.


.
.
Campbell, J., S Jacobi, Y. Liu, K. Hard Robertson, J. Drayton, I. Medina, J. Polo, J. Crenshaw, and J. Odle. 2012. Evaluation of immunoglobulin absorption from colostrum supplements gavaged to newborn piglets. 12th International Symposium on Digestive Physiology of Pigs. Keystone, CO.


Dodson, M.V. and G.J. Hausman. 2011. Metabolic syndromes: Resolving a malady that involves numerous tissues, cells, regulators and regulatory pathways. Metabolic Syndrome 1(1):1

Dodson, M.V., G.J. Hausman, L.L. Guan, M. Du, and Z. Jiang. 2011. Potential impact of mature adipocyte dedifferentiation in terms of cell numbers. International Journal of Stem Cells 4:76-78


Dodson, M.V., P. Mir, G.J. Hausman, L.L. Guan, M. Du, M.E. Fernyhough and W.G. Bergen. 2011. Obesity, metabolic syndrome and adipocytes. Journal of Lipids 721686:1-5


Dodson, M.V., S. Wei, M. Duarte, M. Du, Z. Jiang, G.J. Hausman and W.G. Bergen. 2012. Cell supermarket: Adipose tissue as a source of stem cells. Journal of Genomics 1(1):39-44


Dodson, M.V., Z. Jiang, M. Du and G.J Hausman. 2012. Adipogenesis: It is not just lipid that comprises adipose tissue. Journal of Genomics 1(1):1-4


Du, M., and K. M. Carlin. (2012). Meat Science and Muscle Biology Symposium: extracellular matrix in skeletal muscle development and meat quality. Journal of Animal Science, 90: 922-923.


Garmyn, A.J., G.G. Hilton, R.G. Mateescu, J.B. Morgan, J.M. Reecy, R.G. Tait, Jr., D.C. Beitz, Q. Duan, J.P. Schoonmaker, M.S. Mayes, M.E. Drewnoski, Q. Liu, and D.L. VanOverbeke. 2011. Estimation of relationships between mineral concentration and fatty acid composition of longissimus muscle and beef palatability traits. J. Anim. Sci. 89:2849-2858.


Halsey CHC, Weber PS, Reiter SS, Stronach BN, Bartosh JL, and Bergen WG. 2011. The effect of ractopamine hydrochloride on gene expression in adipose tissues of finishing pigs J. Anim Sci. 89: 1011-1019.


Harris, G.K, M.A. Lila, D.K. Larick, S.H. Zeisel, and J. Odle. 2010. Establishing a transdisciplinary graduate program in functional foods, bioactive food components, and human health. USDA-AFRI Project Directo


Hausman, G.J. and M.V. Dodson. 2012. Stromal vascular cells and adipogenesis: Cells within adipose depots regulate adipogenesis. Journal of Genomics 1(1):56-66


He, M.L., P.S. Mir, R. Sharma, K. Schwartzkopf-Genswein, T. Entz, G. Travis, M.E.R. Dugan, D. Rolland, E.K. Okine and M.V. Dodson. 2011. Effect of supplementation of beef cattle diets with oil containing n6 and n3 fatty acids and subjecting the steers to 48h feed withdrawal treatments on animal productivity, carcass characteristics and fatty acid composition. Livestock Science 142:253-263


Herfel, T.M. S.K. Jacobi, X. Lin, V. Fellner, D.C. Walker, Z.E. Jouni and J. Odle. 2011. Polydextrose enrichment of infant formula demonstrates prebiotic characteristics by altering intestinal microbiota, organic acid concentrations, and cytokine expression in suckling piglets. J. Nutr. 141:2139-2145.


Huang, J.X., L. Lu, L. Xi, X.G. Luo, and B. Liu. 2010. Effects of age and strain on the expression of leptin receptor, neuropeptide Y and pro-opiomelanocortin in the hypothalamus of young chickens. Br Poult Sci. 51(5):696-702.


Huang, Y., J. X. Zhao, X. Yan, M. J. Zhu, N. M. Long, R. J. McCormick, S. P. Ford, P. W. Nathannielsz, and M. Du. 2012. Maternal obesity enhances collagen accumulation and cross-linking in skeletal muscle of ovine offspring. PLOS one, 7, e31691.


Inskeep, T.K., C. Stahl, J. Odle, J. Oakes, L. Hudson, K.L. Bost, and K.J. Piller. 2010. Oral vaccine formulations stimulate mucosal and systemic antibody responses against staphylococcal enterotoxin B in a piglet model. Clin. Vaccine. Immunol. 17:1163-1169.


Ippagunta, S, T.J. Hadenfeldt, JL Miner, and KM Barnes. 2011. Dietary conjugated linoleic acid induces lipolysis in adipose tissue of coconut oil-fed mice but not soy oil-fed mice. Lipids 46:821-830.



Jacobi, S.K., X. Lin, B.A. Corl, H.A. Hess, and J. Odle. 2011. Dietary arachidonate alters ontogeny of the desaturase-elongase pathway in liver and intestine of suckling pigs. J. Nutr. 141:458-553.


Jin, W.W., J.M. Romao, M.V. Dodson, G.J. Hausman, S.S. Moore and L.L. Guan. 2011. MicroRNA regulation of mammalian adipogenesis. Experimental Biology and Medicine 236:997-1004


Kanosky, KM, S Ippagunta, and KM Barnes. 2012. Effects of dietary conjugated linoleic acid and coconut oil on muscle lipid content in mice. Scientifica (In Review).


Kelly AC, Mateus-Pinilla NE, Douglas M, Douglas M, Shelton P, Novakofski J. 2011. Microsatellites behaving badly: empirical evaluation of genotyping errors and subsequent impacts on population studies. Genet Mol Res. 10(4):2534-53.


Lawrence, TLJ, VR Fowler, JE Novakofski. 2012. Growth of Farm Animals, 3rd edition. CABI Publishing. In Proof.


Lin X, K. Shim K, and J. Odle. 2010. Carnitine palmitoyltransferase I control of acetogenesis, the major pathway of fatty acid beta-oxidation in liver of neonatal swine. Am. J. Physiol. Regul. Integr. Comp. Physiol. 298: R1435 - R1443.


Lin, X., J. Bo, S.A. Mathews Oliver, B.A. Corl, S.K. Jacobi, W.T. Oliver, R.J. Harrell, and J. Odle. 2011. Dietary conjugated linoleic acid alters long chain polyunsaturated fatty acid metabolism in brain and liver of neonatal pigs. J. Nutr. Biochem. 22:1047-1054.


Lin, X., M. Azain and J. Odle. 2012. Lipid nutrition and metabolism in swine. In: Sustainable swine nutrition. (L.I. Chiba, editor). Wiley-Blackwell. (in press).


Liu, Z.H., L. Lu, S.F. Li, L.Y. Zhang, L. Xi, K.Y. Zhang, and X.G. Luo. 2011. Effects of supplemental zinc source and level on growth performance, carcass traits, and meat quality of broilers. Poult Sci. 90(8):1782-90.


Long, N. M., C. B. Tousley, K. R. Underwood, W. J. Means, B. W. Hess, M. Du. And S. P. Ford, 2012. Effects of early to mid-gestational undernutrition on offspring growth, carcass characteristics and histology of adipose tissue in beef cattle. Journal of Animal Science, 90: 197-206.


Mahajan, A., L.S. Alexander, B.S. Seabolt, D.E. Catrambone, J.P. McClung, J. Odle, T.W. Pfeiler, E. G. Loboa, and C.H. Stahl. 2011. Dietary calcium restriction affects mesenchymal stem cell activity and bone development in neonatal pigs. J. Nutr. 141:373-379.


Mahajan, A., L.S. Alexander, B.S. Seabolt, J. Odle, E.G. Loboa, and C.H. Stahl. 2010. Dietary calcium affects the differentiation potential of mesenchymal stem cells. FASEB J. 24:545.1
Mao, G., G.A. Kraus, I. Kim, M.E. Spurlock, T.B. Bailey, and D.C. Beitz. 2011. Effect of a mitochondria-targeted vitamin E derivative on mitochondrial alteration and systemic oxidative
stress in mice. Br. J. Nutr. 106:87-95.


Mao, G., G.A. Kraus, I. Kim, M.E. Spurlock, T.B. Bailey, Q. Zhang, and D.C. Beitz. 2010. A mitochondria-targeted vitamin E derivative decreases hepatic oxidative stress and inhibits fat deposition in mice. J. Nutr. 140: 1425-1431.


Mir, P.S., M.L. He, G. Travis, T. Entz, T. McAllister, S. Marchand, A. Schaefer, J. Meadus, P. LaPage, E. Okine and M.V. Dodson. 2012. Periodic 48 hr feed withdrawl improves glucose tolerance in growing pigs by influencing adipogenesis and lipogenesis. Nutrition and Metabolism 9:10-22


Mir, P.S., M.L. He, K. Schwartzkopf-Genswein, R.O. Lemieux, T. Entz, M.E.R. Dugan, E. Okine, M.V. Dodson, F.A. Brown and G. Travis. 2012. Effect of supplementation of beef cattle diets with oil containing n6 and n3 fatty acids and subjecting the steers to 48h feed withdrawal treatments on plasma hormone profiles and adipose tissue cellularity. Livestock Science doi: 10.1016/J.livsci.2012.03.001


Sheilds, M.C., E. van Heugten, X. Lin, J. Odle and C.S. Stark. 2011. Evaluation of the nutritional value of glycerol for nursery pigs. J. Anim. Sci. 89:2145-2153.


Shelton, VJ, AG Shelton, MJ Azain, and KM Hargrave-Barnes. 2012. Short-term feeding and tissue incorporation of conjugated linoleic acid reduced feed intake and body fat in mice. Nutr. Res. (In Review).


Shen, Y. B., G. Voilqué, J. Odle, and S. W. Kim. 2012. Effects of elevating Trp intake on growth and physiological changes in nursery pigs. J. Anim. Sci. (in press)


Tong, J. F., X. Yan, J. X. Zhao, M. J. Zhu, P. W. Nathanielsz, and M. Du. 2011. Metformin mitigates the impaired development of skeletal muscle in the offspring of obese mice. Nutrition and Diabetes, 1: e7.


Turdi, S., M. R. Kandadi, J. X. Zhao, A. F. Huff, M. Du, and J. Ren. 2011. Deficiency in AMP-activated protein kinase exaggerates high fat diet-induced cardiac hypertrophy and contractile dysfunction. Journal of Molecular and Cellular Cardiology. 50: 712-722.


Wei, S., M. Duarte, L. Zan, M. Du, Z. Jiang, L.L. Guan, J. Chen, G.J. Hausman and M.V. Dodson. 2012. Cellular and molecular implications of mature adipocyte dedifferentiation. Journal of Genomics 1:5-12


Xu, Y., M.-J. Zhu, M.V. Dodson and M. Du. 2012. Developmental programming of fetal skeletal muscle and adipose tissue development. Journal of Genomics 1:29-38


Yan, X., M. J. Zhu, M. V. Dodson, and M. Du. (2012). Developmental programming of fetal skeletal muscle and adipose tissue development. Journal of Genomics, 1, 29-38.


Yan, X., Y. Huang, J. X. Zhao, N. M. Long, A. B. Uthlaut, M. J. Zhu, S. P. Ford, P. W. Nathanielsz, and M. Du. 2011. Maternal obesity impaired insulin signaling and induced lipid accumulation and fibrosis in skeletal muscle of offspring. Biology of Reproduction, 85: 172-178.

Yu, Y., L. Lu, R.L. Wang, L. Xi, X.G. Luo, and B. Liu. 2010. Effects of zinc source and phytate on zinc absorption by in situ ligated intestinal loops of broilers. Poult Sci. 89(10):2157-65


Zhang, S., T. Knight, J.M. Reecy, and D. Beitz. 2011. Genetic markers in fatty acid synthase for identification of meat product fatty acid content in cattle. U.S. Patent #7,910,308B1.


Zhao, J. X., J. Hu, M. J. Zhu, and M. Du. 2011. Trenbolone enhances myogenic differentiation by enhancing ²-catenin signaling in muscle-derived stem cells of cattle. Domestic Animal Endocrinology, 40: 222-229.


Zhao, J. X., W. F. Yue, M. J. Zhu, and M. Du. 2011. AMP-activated protein kinase regulates ²-catenin transcription via histone deacetylase 5. Journal of Biological Chemistry, 286: 16426-16434.


2012 (By stations)


University of California-Davis/USDA Western Regional Nutrition Research Center


1. A.P. Thomas,T.N. Dunn, J.B. Drayton, P.J. Oort, and S.H. Adams. A high
calcium diet containing nonfat dry milk reduces weight gain and associated adipose tissue inflammation in diet-induced obese mice when compared to high calcium alone. Nutrition & Metabolism 9:3, 2012


2. J.A. Viscarra, J.P. Vázquez-Medina, R. Rodriguez, C.D. Champagne, S.H. Adams, D.E. Crocker, R.M. Ortiz. Decreased expression of adipose CD36 and FATP1 are associated with increased plasma nonesterified fatty acids during prolonged fasting in northern elephant seal pups (Mirounga angustirostris). J. Exp. Biol. 215(Pt 14):2455-64, 2012


3. S. Huang, J.M. Rutkowsky, R.G. Snodgrass, K.D. Ono-Moore, D.A. Schneider, J.W. Newman, S.H. Adams, D.H. Hwang. Saturated fatty acids activate TLR-mediated pro-inflammatory signaling pathways. J. Lipid Res. 53(9):2002-13, 2012


4. A.P. Thomas,T.N. Dunn, J.B. Drayton, P.J. Oort, S.H. Adams. A dairy-based high calcium diet improves glucose homeostasis and reduces further weight gain in high fat fed mice in the context of pre-existing obesity. 2012 Obesity, Sep 18. doi: 10.1002/oby.20039 [Epub ahead of print).


5. D. Grapov, S.H. Adams, T.L. Pedersen, W.T. Garvey, K.H. Lok, J.W. Newman. Type 2 diabetes associated changes in the plasma non-esterified fatty acids, oxylipins and endocannabinoids. PLoS ONE 7(11): e48852, 2012


6. S. Millership, N. Ninkina, I. Guschina, J. Norton, R. Brambilla, P.J. Oort, S.H. Adams, R.J. Dennis, P.J. Voshol, J.J. Rochford, V.L. Buchman. Increased lipolysis and altered lipid homeostasis protect ³-synuclein null mutant mice from diet-induced obesity. PNAS 109(51):20943-8, 2012


7. M.G. Witbracht, M. Van Loan, S.H. Adams, N.L. Keim, K.D. Laugero. Dairy food consumption and meal-induced cortisol response interacted to influence weight loss in overweight women undergoing a 12-week meal-controlled weight loss intervention. J. Nutrition 143(1):46-52, 2013


8. P. She, K.C. Olson, Y. Kadota, A. Inukai, Y. Shimomura, C.L. Hoppel, S.H. Adams, Y. Kawamata, H. Matsumoto, R. Sakai, C.H. Lang, C.J. Lynch. Leucine and protein metabolism in obese Zucker rats. PLoS ONE 8(3): e59443, 2013


9. T.P. Garcia, S. Müller, R.J. Carroll, T.N. Dunn, A.P. Thomas, S.H. Adams, S.D. Pillai, R.L. Walzem. Structured variable selection with q-values. 2013 Apr 10 [Epub ahead of print], Biostatistics


10. D.E. Lackey, C.J. Lynch, K.C. Olson, R. Mostaedi, M. Ali, W.H. Smith, F. Karpe, S. Humphreys, D.H. Bedinger, T.N. Dunn, A.P. Thomas, P.J. Oort, D.A. Kieffer, R. Amin, A. Bettaieb, F.G. Haj, P. Permana, T.G. Anthony, S.H. Adams. Regulation of adipose branched chain amino acid catabolism enzyme expression and cross-adipose amino acid flux in human obesity. 2013 Am. J. Physiol: Endocrinol. Metab, Mar 19 [Epub ahead of print],.


11. S.H. Adams, K.M. Barnes, and J. Odle. Comparative Metabolic Physiology in the 'omics' Era: A Call to Arms, Paws, Flippers, and Claws (intro to symposium Adipose and Lipid Biology: Crossing Taxonomic Boundaries held at the Experimental Biology 2013 meeting, 4/19/2013 in Boston, MA). submitted, Advances in Nutrition


The Ohio State University

1.Md. J. Alam, C. D. Jeong, L. L. Mamuad, H. G. Sung, D. W. Kim, S. B. Cho, K. Lee, C. O. Jeon, Sang S. Lee. 2012. Bacterial Community Dynamics during Swine In vitro Fermentation Using Starch as a Substrate with Different Feed Additives for Odor Reduction. Asian-Aust. J. Anim. Sci. 25(5): 690-700.


2. Relling AE, Lee K, Loerch SC, Reynolds CK. 2012. Effects of Glucose, Propionate and Splanchnic Hormones on Neuropeptide mRNA concentrations in the Ovine Hypothalamus. Journal of Animal Physiology and Animal Nutrition. Vol. 96 No.4:656-662.


3. Park YJ, Mohamed ES, Oh SA, Yoon SJ, Kwon WS, Kim HR, Lee MS, Lee K, Pang MG. 2012. Sperm Penetration Assay as an Indicator of Bull Fertility. J Reprod Dev. 2012. 58(4):461-6.


4. Lee. A, Suh Y, MP Wick, and K Lee. 2012. Temporal Myosin Heavy Chain Isoform Expression Transitions Faster in Broiler Chickens When Compared to Single Comb White Leghorns. Poultry Sci. 2012. 91(11):2872-6.


5. Li X, Suh Y, Kim BR, Moeller SJ, Lee K. 2012. Alternative splicing and developmental and hormonal regulation of porcine comparative gene identification-58 (CGI-58) mRNA. J Anim Sci. 90(12):4346-54.


6. Yasmeen R, Reichert B, Deiuliis J, Yang F, Lynch A, Meyers J, Sharlach M, Shin S, Volz KS, Green KB, Lee K, Alder H, Duester G, Zechner R, Rajagopalan S, Ziouzenkova O. 2013. Autocrine Function of Aldehyde Dehydrogenase 1 as a Determinant of Diet- and Sex-Specific Differences in Visceral Adiposity. Diabetes. 62(1):124-36.

7. Yang S, Suh Y, Choi YM, Shin S, Han JY, Lee K. 2013. Loss of Fat with Increased Adipose Triglyceride Lipase-Mediated Lipolysis in Adipose Tissue during Laying Stages in Quail. Lipids. 48(1):13-21.


8. Ahn J, Oh SA, Suh Y, Moeller SJ, Lee K. 2013. Porcine G(0)/G (1) Switch Gene 2 (G0S2) Expression is Regulated During Adipogenesis and Short-Term In-Vivo Nutritional Interventions. Lipids. 48(3):209-18.


9. Alam MJ, Mamuad LL, Kim SH, Jeong CD, Sung HG, Cho SB, Jeon CO, Lee K, Lee SS. 2013. Effect of Phytogenic Feed Additives in Soybean Meal on In vitro Swine Fermentation for Odor Reduction and Bacterial Community Comparison. Asian-Aust. J. Anim. Sci. 26(2):266-74.


10. Choi YM, Nam KW, Choe JH, Ryu YC, Wick MP, Lee K, Kim BC. 2013. Growth, carcass, fiber type, and meat quality characteristics in Large White pigs with different live weights. http://dx.doi.org/10.1016/j.livsci.2013.02.009.


11. Choi Y. M., D. Sarah, S. Shin, M. P. Wick, B. C. Kim, and K. Lee. Comparative growth performance in different Japanese quail lines: The effect of muscle fiber DNA content and morphology. Poultry Sci. (In Press).


12. Song Y., J. Ahn, Y. Suh, M. E. Davis, and K. Lee. Identification of Novel Tissue-Specific Genes by Analysis of Microarray Databases: A Human and Mouse model. PLoS One (Minor Revision).


13. Choi Y. M., S. Shin, M. P. Wick, J. H. Choe, and K. Lee. Comparative growth performance in different Japanese quail lines: The importance of muscle fiber hypertrophy. Animal. (Submitted).


14 Serr J., X. Li, and K. Lee. The regulation of lipolysis in food animals. J. Anim. Sci. Technol. (Invited review).


Washington State University


1. Das, A. K., Q. Y. Yang, X. Fu, J. F. Liang, M. S. Duarte, M. J. Zhu, G. D. Trobridge, and M. Du. (2012). AMP-activated protein kinase stimulates myostatin expression in C2C12 cells. Biochemical and Biophysical Research Communications, 427: 36-40.


2. Dodson, M.V., S. Boudina, E. Albrecht, L. Bucci, M. Fernyhough-Culver, S. Wei, W.G. Bergen, A.J. Amaral, N. Moustaid-Moussa, S.Poulos and G.J. Hausman. 2013. A long journey to effective obesity treatments: Is there light at the end of the tunnel? Experimental Biology and Medicine doi: 10.1177/1535370213477603


3. Du, M., and K. M. Carlin. (2012). Meat Science and Muscle Biology Symposium: extracellular matrix in skeletal muscle development and meat quality. Journal of Animal Science, 90: 922-923.


4. Du, M., Y. Huang, A.K. Das, Q. Yang, M.S. Duarte, M.V. Dodson and M-Y. Zhu. 2013. Manipulating mesenchymal progenitor cell differentiation to optimize performance and carcass value of beef cattle. Journal of Animal Science doi: 10.2527/jas.2012-5670


5. Duarte, M.S., M.P. Gionbelli, P.V.R. Paulino, N.V.L. Serao, R. Mezzomo, M.V. Dodson, M. Du, J. Busboom and S.E.F. Guimaraes. 2013. Effects of pregnancy and feeding level on carcass and meat quality traits of Nellore cows. Meat Science 94:139-144


6. Duarte, M.S., M.P. Gionbelli, P.V.R. Paulino, N.V.L. Serao, T.S. Martins, P.I.S. Totaro, S.C. Valdares-Filho, M.V. Dodson and M. Du. 2013. Effects of maternal nutrition on development of gastrointestinal tract of bovine fetus. Livestock Science doi: 10.1016.livsci.2013.01.006


7. Duarte, M.S., P.V.R. Paulino, A. Das, S. Wei, N.V.L. Serao, X. Fu, S. Harris, M.V. Dodson and M. Du. 2013. Enhancement of adipogenesis and fibrogenesis in skeletal muscle of Wagyu compared to Angus cattle. Journal of Animal Science doi: 10.2527/jas.2012-5892


8. Duarte, M.S., S. Wei, M. Du, Z. Jiang, P.V.R. Paulino, L. Zan, G.J. Hausman and M.V. Dodson. 2012. Isolation of mature adipocytes and stromal vascular cells under adverse sampling conditions. Journal of Metabolic Syndrome 1(4):112-116


9. Huang, Y., J. X. Zhao, X. Yan, M. J. Zhu, N. M. Long, R. J. McCormick, S. P. Ford, P. W. Nathannielsz, and M. Du. (2012). Maternal obesity enhances collagen accumulation and cross-linking in skeletal muscle of ovine offspring. PLOS one, 7, e31691.


10. Huang, Y., A. K. Das, Q. Y. Yang, M. J. Zhu, and M. Du. (2012). Zfp423 promotes adipogenic differentiation of bovine stromal vascular cells. PLOS one, 7(10): e47496.


11. Jellyman, J. K., M. S. Martin-Gronert, Cripps, R. L., Giussani, D. A., Ozanne S. E., Q. W. Shen, M. Du, A. L. Fowden, and A. J. Forhead. (2012). Effects of cortisol and dexamethasone on insulin signaling pathways in skeletal muscle of the ovine fetus during late gestation. PLOS one, 7: e52363.


12. Yan, X.,Y. Huang, J. X. Zhao, C. J. Rogers, M. J. Zhu, S. P. Ford, P. W. Nathanielsz, and M. Du. (2012). Maternal obesity down-regulates microRNA (miRNA) let-7g expression, a possible mechanism for enhanced adipogenesis during ovine fetal skeletal muscle development. International Journal of Obesity, 37: 568-575.


13. Wei, S., L. Zan, G.J. Hausman, T.P. Rasmussen, W.G. Bergen and M.V. Dodson. 2013. Dedifferentiated adipocyte-derived progeny cells [DFAT cells]: Stem cells of adipose tissue. Adipocyte 2(3):1-6


14. Wei, S., L. Zan, H. Wang, G. Cheng, M. Du, Z. Jiang, G.J. Hausman, D.C. McFarland and M.V. Dodson. 2013. FABP4 regulates ADIPOQ, LEP and LEPR expression in bovine preadipocytes. Genetics and Molecular Research 12(1):494-505


15. Wei, S., M. Du, Z. Jiang, M.S. Duarte, M. Fernyhough-Culver, E. Albrecht, K. Will, L. Zan, G.J. Hausman, E. Elabd, W.G. Bergen, U. Basu and M.V. Dodson. 2013. Bovine dedifferentiated adipose tissue [DFAT] cells: DFAT cell isolation. Adipocyte, 1:148-159.


16. Wei, S., M.S. Duarte, M. Du, P.V.R. Paulino, Z. Jiang, E. Albrecht, M. Fernyhough-Culver, L. Zan and M.V. Dodson. 2012. Bovine mature adipocytes readily return to a proliferative state. Tissue & Cell 44:385-390


University of Missouri

1. JA Fletcher, JW Perfield II, JP Thyfault, and RS Rector. The second meal effect and its influence on glycemia. J of Nutrition Disorder & Ther. 2012, 2:108. doi:10.4172/jndt.1000108


2. EM Morris, JA Fletcher, JP Thyfault, RS Rector. The Role of Angiotensin II in Nonalcoholic Steatohepatitis. Molecular and Cellular Endocrinology. 2012 May 11. PMID: 22579612.


3. JA Fletcher, GM Meers, MH Laughlin, JA Ibdah, JP Thyfault, and RS Rector. Modulating Fibroblast Growth Factor-21 in Hyperphagic OLETF Rats with Daily Exercise and Caloric Restriction. Appl Physiol Nutr Metabol. 37(6):1054-62, 2012. PMID: 22891896


4. CR Mikus, BT Roseguini, GM Meers, EM Morris, RS Rector, JL Libla, DJ Oberlin, SJ Borengasser, AM Taylor, JA Ibdah, MH Laughlin, and JP Thyfault. Voluntary wheel running selectively augments insulin-stimulated vasodilation in arterioles from white skeletal muscle of insulin resistant rats. Microcirculation. 19(8):729-38, 2012. PMID: 22804760


5. J Martin, J Padilla, N Jenkins, J Crissey, S Bender, RS Rector, JP Thyfault, and MH Laughlin. Functional adaptations in the skeletal muscle microvasculature to endurance and interval sprint training in the type 2 diabetic OLETF rat. J Appl Physiol . 113(8): 1223-32, 2012. PMID: 22923508


6. NT Jenkins, J Padilla, AA Arce-Esquivel, DS Bayless, JS Martin, HJ Leidy, FW Booth, RS Rector, and MH Laughlin. Effects of Endurance Exercise Training, Metformin, and their Combination on Adipose Tissue Leptin and IL-10 Secretion in OLETF Rats. J Appl Physiol.113(12):1873-83, 2012. PMID 23019312


7. EA Dannecker, Y Liu, RS Rector, TR Thomas, RB Fillingim, and ME Robinson. Sex Differences in Exercise-Induced Pain and Muscle Damage. J Pain. 13(12):1242-49, 2012. PMID: 23182229


8. LC Ortinau, RT Pickering, KJ Nickelson, KL Stromsdorfer, CY Naik, RA Haynes, DE Bauman, RS Rector, KL Fritsche, JW Perfield II. Sterculic oil, a natural SCD1 inhibitor, improves glucose homeostasis in obese ob/ob mice. ISRN Endocrinology. 2012:947323, 2012. PMID: 23209931


9. EA Dannecker, Y Liu, RS Rector, TR Thomas, SP Sayers, C Leeuwenburgh, BK Ray. The effects of fasting on indicators of muscle damage. Exp Gerontol. 2012 Dec 22. doi:pii: S0531-5565(12)00319-1. PMID: 23266375


10. JW Perfield II, LC Ortinau, RT Pickering, ML Ruebel, GM Meers, and RS Rector. Altered Hepatic Lipid Metabolism Contributes to Nonalcoholic Fatty Liver Disease in Leptin-Deficient Ob/Ob Mice. J Obesity, 2013; 2013:296537. PMID: 23401753


11. RS Rector, EM Morris, S Ridenhour, GM Meers, FF Hsu, J Turk, and JA Ibdah. Selective hepatic insulin resistance in mice heterozygous for a mitochondrial trifunctional protein defect. Hepatology. Jan 2013, In Press. PMID: 23359250


12. MD Roberts, DS Bayless, TE Childs, NT Jenkins, J Padilla, JS Martin, LP Oberle, JM Company, VJ Dalbo3, FW Booth, RS Rector, MH Laughlin. Elevated skeletal muscle irisin precursor FNDC5 mRNA in obese OLETF rats. Metabolism 2013. Mar 13.62: 1052-1056. PMID: 23498898


13. Padilla J, Jenkins NT, Lee S, Zhang H, Cui J, Zuidema MY, Zhang C, Hill MA, Perfield JW 2nd, Ibdah JA, Booth FW, Davis JW, Laughlin MH, Rector RS. Vascular transcriptional alterations produced by juvenile obesity in Ossabaw swine. Physiol Genomics. 2013 Apr 16. PMID: 23592636


14. MA Linden, GM Meers, ML Ruebel, NT Jenkins, FW Booth, ML Laughlin, JA Ibdah, JP Thyfault, and RS Rector. Hepatic Steatosis Development with Four Weeks of Physical Inactivity in Previously Active, Hyperphagic OLETF Rats. Am J Physiol Regul Integr Comp Physiol. 2013 Mar 6. PMID: 23467323

University of Tennessee



1. Fletcher SJ, Kalupahana NS, Soltani-Bejnood M, Kim JH, Saxton AM, Wasserman DH, De Taeye B, Voy BH, Quignard-Boulange A, Moustaid-Moussa N. Transgenic mice overexpressing renin exhibit glucose intolerance and diet-genotype interactions.
Front Endocrinol (Lausanne). 2012;3:166. doi: 10.3389/fendo.2012.00166. Epub 2013 Jan 7


2. MetabR: an R script for linear model analysis of quantitative metabolomic data.
Ernest B, Gooding JR, Campagna SR, Saxton AM, Voy BH. BMC Res Notes. 2012 Oct 30;5:596. doi: 10.1186/1756-0500-5-596.


3. Applications of subsurface microscopy.
Tetard L, Passian A, Farahi RH, Voy BH, Thundat T. Methods Mol Biol. 2012;926:331-43. doi: 10.1007/978-1-62703-002-1_21.


4. Transcriptomic and metabolomic profiling of chicken adipose tissue in response to insulin neutralization and fasting. Ji B, Ernest B, Gooding JR, Das S, Saxton AM, Simon J, Dupont J, Métayer-Coustard S, Campagna SR, Voy BH. BMC Genomics. 2012 Aug 31;13:441. doi: 10.1186/1471-2164-13-441.


5. A systematic comparison of genome-scale clustering algorithms. Jay JJ, Eblen JD, Zhang Y, Benson M, Perkins AD, Saxton AM, Voy BH, Chesler EJ, Langston MA. BMC Bioinformatics. 2012 Jun 25;13 Suppl 10:S7. doi: 10.1186/1471-2105-13-S10-S7.


Auburn University

1. Wei S, Bergen WG, Hausman GJ, Zan L and Dodson MV. 2013. Cell culture purity issues and
DFAT cells. Biochem Biophys Res Comm, doi 10.1016/j.bbrc.2013.03.006


2. Wei S, Zan L, Hausman GJ, Rasmussen TP, Bergen WG and Dodson MV. 2013.
Dedifferentiated adipocyte-derived progeny cells (DFAT cells). Potential stem cells of adipose
tissue. Adipocyte; dx.doi.org/10.4161/adip.23784


3. Dodson MV, Boudina S, Albrecht E, Bucci L, Fernyhough-Culver M, Wei S, Bergen WG, Amaral A, Moustaid-Moussa N, Poulos S and Hausman GJ. 2013. A long journey to effective obesity treatments: is there light at the end of the tunnel? 2013. Exp Biol Med, doi:
10.1177/1535370213477603



4. Wei S, Du M , Jiang Z , Duarte M, Fernyhough-Culver M , Albrecht E, Will K , Zan L, Hausman GJ, Elabd E , Bergen WG and Basu U. 2013. Bovine dedifferentiated adipose tissue [DFAT] cells: DFAT cell isolation. Adipocyte, 2: 148-159



5. Key, C.N, S.D. Perkins, C. L. Bratcher, L. A. Kriese-Anderson, and T. D. Brandebourg. 2013. Grain feeding coordinately alters expression patterns of transcription factor and metabolic genes
in subcutaneous adipose tissue of crossbred heifers. J. of Anim. Sci. 91:2616-2627.




6. Brandebourg, T.D. and C.Y. Hu. 2013. Feeding retinoic acid reduces total carcass fat in growing broiler chickens. Poultry, Fisheries & Wildlife Sci. (accepted)


7. Brandebourg, T.D., Wolfe D.F., and C.D. Foradori. 2013. U.S. beef industry: a sustainable
success story, challenges and priorities. J Fisheries Livest Prod 1:1 Accessed
http://dx.doi.org/10.4172/jflp.1000102


8. Merkin G.V., Pittman K., and T.D. Brandebourg. 2013. Bridging the gap to sustainable salmon farming: overcoming the gaping problem.. J Fisheries Livest Prod. (accepted)


9. Brandebourg, T.D. 2013. Drawing the line: animal agriculture, animal welfare, and the need to feed the world. Poultry, Fisheries & Wildlife Sci. (accepted)


10. . Cannon, C.V., Irwin, M.H., Kohn M.C., Brandebourg T.D., Ðikic D., and C. A. Pinkert. 2012. Mitochondrial DNA sequence and phylogenetic evaluation of geographically disparate Sus
scrofa breeds. Animal Biotechnology. (submitted)



West Virginia University

1. Barnes, KM, NR Winslow, AG Shelton, KC Hlusko, and MJ Azain. 2012. Effect of dietary
conjugated linoleic acid on marbling and intramuscular adipocytes in pork. J. Anim. Sci.
90:1142-1149.


2. Shelton, VJ, AG Shelton, MJ Azain, and KM Hargrave-Barnes. 2012. Short-term feeding and
tissue incorporation of conjugated linoleic acid reduced feed intake and body fat in mice.
Nutr. Res. 32:827-836.


3. Gatrell, S.K., L.E. Berg, J.T. Barnard, J.G. Grimmett, K.M. Barnes, and K.P. Blemings. 2013. Lysine catabolism in pig tissues, tissue distribution of indices of lysine catabolism in growing swine. J. Anim. Sci. 91:238-247.


4. Matak, K.E., K.H. Maditz, K.M. Barnes, S.K. Beamer, and P.B. Kenney. 2013. Effect of dietary inclusion of conjugated linoleic acid on quality indicators and sensory characteristics of aged pork loin. J. Ag. Sci. (In Press).


5. Kanosky, KM, S Ippagunta, and KM Barnes. 2013. Mice do not accumulate muscle lipid in response to dietary conjugated linoleic acid. J. Anim. Sci. 91:4705-4712.


6. Ippagunta, S., Z. Angius, M. Sanda, and K.M. Barnes. 2013. Time-dependent effect of CLA in
coconut oil fed mice on lipolysis and lipid metabolism. Lipids, 48: 1145-1155..



Purdue University

1) Adapala, V.J., S.A. Adedokun, R.V. Considine and K.M. Ajuwon. 2012. Acute inflammation plays a limited role in the regulation of adipose tissue col1a1 protein abundance. J Nutr Biochem. 23:567-72.



2) Adedokun, S.A., K.M. Ajuwon, L.F. Romero and O. Adeola. 2012. Ileal endogenous amino acid losses: Response of broiler chickens to fiber and mild coccidial vaccine challenge. Poult. Sci. 91:899-907.


3) Atkinson, R.L., P.M. Walker, S.W. Reader, J.M. Carmack, K.M. Ajuwon, S.L. Lake, B.R. Wiegand, and L.A. Foster. 2012. Effect of low-fat corn distillers grains fed at 40 and 70% inclusion on growth performance and meat quality of steers. Professional Animal Scientist. 28:41-55.


4) Banz, W.J., D. A. Strader, K.M. Ajuwon, Y. Hou, C. Y. Meyers and J. Davis. 2012. (+)-Z-Bisdehydrodoisynolic acid enhances basal metabolism and fatty acid oxidation in female obese Zucker rats. J. Obesity. 2012:154145. doi: 10.1155/2012/154145 (5 pages).


5) Adeola, O., A.P. Schinckel, K.M. Ajuwon and M.E. Einstein. 2012. Accretion rates and efficiency of amino acid utilization during the twenty-nine days post-hatch period in male ducks. J. Poult. Sci. 49: 79-85.


6) Adapala, V.J., M. Ward and K.M. Ajuwon. 2012. Adipose tissue biglycan as a potential antiinflammatory target of sodium salicylate in mice fed a high fat diet. J Inflamm (Lond). 9:15. doi: 10.1186/1476-9255-9-15. (7 pages).


7) Ward, M.G and K.M. Ajuwon. 2012. Biglycan deletion alters adiponectin expression in murine adipose tissue and 3T3-L1 adipocytes. PLoS One. 7(11):e50554. doi: 10.1371/journal.pone.0050554. (10 pages).


National Taiwan University

1. Hsu, C.C., E.C. Lin, S.C. Chen, S.C. Huang, B.H. Liu, Y.H. Yu, C.C. Chen, C.C. Yang, C.Y. Lien., Y.H. Wang, C.W. Liu, H.J. Mersmann, W.T.K. Cheng, and S.T. Ding (coresponding). 2012. Differential gene expression between the porcine morula and blastocyst. Reprod. Domest. Anim. 47:69-81. (SCI, AGRICULTURE, DAIRY & ANIMAL SCIENCE Top 30% 15/55), IF:1.356.


2. Chen, Y.J., C.C. Chen, T.K. Li, P.H. Wang, L.R. Liu, F.Y. Chang, Y.C. Wang, Y.H. Yu, S.P. Lin, H.J. Mersmann, S.T. Ding (coresponding), 2012. Docosahexaenoic acid suppresses the expression of FoxO and its target genes. J. Nutri. Biochem.23:1609-1616. (SCI, NUTRITION & DIETETICS Top 15% IF: 3.891, 11/72)


3. Chang, Chia-Ching, Chih-Chien Chen and Shih-Torng Ding (corresponding author). 2012. Amelioration of obesity-associated inflammation by n-3 PUFA. Chin. Anim. Sci. 41: 1-14.


4. Tu, P. A., S. T. Ding, E.-C. Lin, M. C. Wu, and P. H. Wang. 2012. The association of genetic variations in the promoter region of myostatin gene with growth traits in Duroc pigs. Anim. Biotechnol. (SCI, IF=1.102, Ranking=22/55) (accepted)


5. Chang, Y. Y., M. C. Chao, E. -C. Lin, S. T. Ding, H. S. Tsao, H. W. Yuan, and P. H. Wang. 2012. Development of microsatellite markers in ungulate mammal Formosan serow (Capricornis swinhoei). Conservation Genet. Resour. 4: 755-757. (SCI, IF=0.485, Biodiversity conservation, Rank=27/35)


6. Chang, Y. C., S.T. Ding (Equal contribution), Y.H. Lee, Y.C. Wang, M.F. Huang and I.H. Liu. 2013. Taurine homeostasis requires de novo synthesis via cysteine sulfinic acid decarboxylase during zebrafish early embryogenesis. Amino Acids. 44:615-629. (SCI, BIOCHEMISTRY & MOLECULAR BIOLOGY Top 35%, 115/289 IF=3.248)


7. Wang, P.H., Hsu, H.A., M.C. Chao, F.T. Chan, L.M. Wang, P.I. Lin, C.H. Chang, H.W. Yuan, C.C. Chen, and S.T. Ding (corresponding). 2013. Sex identification in the Collared Scops Owl (Otus bakkamoena) with novel markers generated by random amplified polymorphic DNA. Cons. Genet. Res. 5:239-242. (SCI, IF=0.485, Biodiversity conservation, Rank=27/35)


8. Lin, Y.Y, Y.C. Chen, Y. Lin, Y. P. Chiu, C. C. Chen, B. H. Liu, H. J. Mersmann, S. C. Wu, and S. T. Ding (corresponding). Modulation of glucose and lipid metabolism by porcine adiponectin receptor 1-transgenic mesenchymal stem cells in diet-induced obese mice. Cytotherapy. In Press. (SCI)







Attachments

Land Grant Participating States/Institutions

AL, GA, HI, IA, IN, MS, NC, OH, TN, TX, VT, WA, WV, WY

Non Land Grant Participating States/Institutions

University of Arkansas, University of Arkansas for Medical Sciences, University of Missouri - Columbia
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.