Acessibilidade / Reportar erro

Prenatal origins of productivity and quality of beef

ABSTRACT

The productive traits of beef cattle are orchestrated by their genetics, post-natal environmental conditions, and also by the intrauterine background. Both under- or overnutrition, as specific dietary components, are able to promote persistent effects on the offspring. This occurs because dietary factors act not only affecting the availability of substrates for fetal anabolism and oxidative metabolism, but also as signals that regulate several events toward fetal development. Therefore, this study aimed to summarize the gestational nutrition effects on the offspring performance and meat quality in a long term. Overall, studies have shown that many of these alterations are under the control of epigenetic mechanisms, as DNA methylation, histones modification, and non-coding RNA. The current knowledge has indicated that the fetal programming responses are dependent on the window of fetal development in which the dietary treatment is applied, the intensity of maternal nutritional stimuli, and the treatment application length. Collectively, studies demonstrated that muscle cell hyperplasia is impaired when maternal requirements were not achieved in the second third of gestation, which limits the formation of a greater number of muscle fibers and the offspring growth potential in a long term. Changes in muscle fibers metabolism and in collagen content were also reported as consequence of a dietary perturbation during pregnancy. In contrast, a maternal overnutrition during the late pregnancy has been associated with beneficial responses on meat quality. In summary, ensuring an adequate maternal environment during the fetal development is crucial to enhance the productive responses in beef cattle operations.

adipogenesis; bovine; fibrogenesis; maternal nutrition; myogenesis; progenitor cells

1. Introduction

Fetal programming is the response of an organism to an environmental challenge during a critical period of intrauterine development, which leads to persistent changes (Nathanielsz et al., 2007Nathanielsz, P. W.; Poston, L. and Taylor, P. D. 2007. In utero exposure to maternal obesity and diabetes: animal models that identify and characterize implications for future health. Obstetrics and Gynecology Clinics of North America 34:201-212. https://doi.org/10.1016/j.ogc.2007.03.006
https://doi.org/10.1016/j.ogc.2007.03.00...
). Both maternal under- and overnutrition can trigger changes in the development, metabolism, and physiology of the offspring (Nissen et al., 2003Nissen, P. M.; Danielsen, V. O.; Jorgensen, P. F. and Oksbjerg, N. 2003. Increased maternal nutrition of sows has no beneficial effects on muscle fiber number or postnatal growth and has no impact on the meat quality of the offspring. Journal of Animal Science 81:3018-3027. https://doi.org/10.2527/2003.81123018x
https://doi.org/10.2527/2003.81123018x...
; Greenwood and Cafe, 2007Greenwood, P. L. and Cafe, L. M. 2007. Prenatal and pre-weaning growth and nutrition of cattle: long-term consequences for beef production. Animal 1:1283-1296. https://doi.org/10.1017/S175173110700050X
https://doi.org/10.1017/S175173110700050...
; Duarte et al., 2014Duarte, M. S.; Gionbelli, M. P.; Paulino, P. V. R.; Serão, N. V. L.; Nascimento, C. S.; Botelho, M. E.; Martins, T. S.; Filho, S. C. V.; Dodson, M. V.; Guimarães, S. E. F. and Du, M. 2014. Maternal overnutrition enhances mRNA expression of adipogenic markers and collagen deposition in skeletal muscle of beef cattle fetuses. Journal of Animal Science 92:3846-3854. https://doi.org/10.2527/jas.2014-7568
https://doi.org/10.2527/jas.2014-7568...
; Gionbelli et al., 2018Gionbelli, T. R. S.; Veloso, C. M.; Rotta, P. P.; Valadares Filho, S. C.; Carvalho, B. C.; Marcondes, M. I.; Cunha, C. S.; Novaes, M. A. S.; Prezotto, L. D.; Duarte, M. S. and Gionbelli, M. P. 2018. Foetal development of skeletal muscle in bovines as a function of maternal nutrition, foetal sex and gestational age. Journal of Animal Physiology and Animal Nutrition 102:545-556. https://doi.org/10.1111/jpn.12786
https://doi.org/10.1111/jpn.12786...
; Costa et al., 2021a). Such modifications are under the control of epigenetics, which act as a memory of the environment exposure (Wu et al., 2006Wu, G.; Bazer, F. W.; Wallace, J. M. and Spencer, T. E. 2006. Board-invited review: Intrauterine growth retardation: Implications for the animal sciences. Journal of Animal Science 84:2316-2337. https://doi.org/10.2527/jas.2006-156
https://doi.org/10.2527/jas.2006-156...
; Sinclair et al., 2016Sinclair, K. D.; Rutherford, K. M. D.; Wallace, J. M.; Brameld, J. M.; Stöger, R.; Alberio, R.; Sweetman, D.; Gardner, D. S.; Perry, V. E. A.; Adam, C. L.; Ashworth, C. J.; Robinson, J. E. and Dwyer, C. M. 2016. Epigenetics and developmental programming of welfare and production traits in farm animals. Reproduction, Fertility and Development 28:1443-1478. https://doi.org/10.1071/RD16102
https://doi.org/10.1071/RD16102...
; Paradis et al., 2017Paradis, F.; Wood, K. M.; Swanson, K. C.; Miller, S. P.; McBride, B. W. and Fitzsimmons, C. 2017. Maternal nutrient restriction in mid-to-late gestation influences fetal mRNA expression in muscle tissues in beef cattle. BMC Genomics 18:632. https://doi.org/10.1186/s12864-017-4051-5
https://doi.org/10.1186/s12864-017-4051-...
; Batistel et al., 2019Batistel, F.; Alharthi, A. S.; Yambao, R. R. C.; Elolimy, A. A.; Pan, Y. X.; Parys, C. and Loor, J. J. 2019. Methionine supply during late-gestation triggers offspring sex-specific divergent changes in metabolic and epigenetic signatures in bovine placenta. The Journal of Nutrition 149:6-17. https://doi.org/10.1093/jn/nxy240
https://doi.org/10.1093/jn/nxy240...
).

Dietetic manipulations over pregnancy are an opportunity to improve the offspring performance and meat quality, but at the same time, is a way to compromise these characteristics irreversibly. The basic structure of skeletal muscle tissue is composed by muscle fibers, adipocytes, and connective tissue, all derived from the mesenchymal stem cells of mesoderm (Du et al., 2013Du, M.; Huang, Y.; Das, A. K.; Yang, Q.; Duarte, M. S.; Dodson, M. V. and Zhu, M. J. 2013. Meat Science and Muscle Biology Symposium: Manipulating mesenchymal progenitor cell differentiation to optimize performance and carcass value of beef cattle. Journal of Animal Science 91:1419-1427. https://doi.org/10.2527/jas.2012-5670
https://doi.org/10.2527/jas.2012-5670...
) (Figure 1). In this sense, maternal nutrition acts to control the fate steam cell in the different lineages, regulating the balance between myogenesis, adipogenesis, and fibrogenesis (Du et al., 2013Du, M.; Huang, Y.; Das, A. K.; Yang, Q.; Duarte, M. S.; Dodson, M. V. and Zhu, M. J. 2013. Meat Science and Muscle Biology Symposium: Manipulating mesenchymal progenitor cell differentiation to optimize performance and carcass value of beef cattle. Journal of Animal Science 91:1419-1427. https://doi.org/10.2527/jas.2012-5670
https://doi.org/10.2527/jas.2012-5670...
; Blair et al., 2021Blair, A. D.; Gubbels, E. R.; Block, J. J.; Olson, K. C.; Grubbs, J. K. and Underwood, K. R. 2021. Maternal nutrition and meat quality of progeny. Meat and Muscle Biology 5:1-9. https://doi.org/10.22175/mmb.12990
https://doi.org/10.22175/mmb.12990...
).

Figure 1
Mesenchymal progenitor cells differentiate into myogenic and fibroadipogenic cells during fetal muscle development in beef cattle.

Overall, studies have shown that inadequate maternal diets in beef cattle have negative consequences such as a lower population of muscle fibers (Marquez et al., 2017Marquez, D. C.; Paulino, M. F.; Rennó, L. N.; Villadiego, F. C.; Ortega, R. M.; Moreno, D. S.; Martins, L. S.; De Almeida, D. M.; Gionbelli, M. P.; Manso, M. R.; Melo, L. P.; Moura, F. H. and Duarte, M. S. 2017. Supplementation of grazing beef cows during gestation as a strategy to improve skeletal muscle development of the offspring. Animal 11:2184-2192. https://doi.org/10.1017/S1751731117000982
https://doi.org/10.1017/S175173111700098...
; Costa et al., 2021a), due to changes in the mRNA abundance of myogenic regulatory factors (Jennings et al., 2016Jennings, T. D.; Gonda, M. G.; Underwood, K. R.; Wertz-Lutz, A. E. and Blair, A. D. 2016. The influence of maternal nutrition on expression of genes responsible for adipogenesis and myogenesis in the bovine fetus. Animal 10:1697-1705. https://doi.org/10.1017/S1751731116000665
https://doi.org/10.1017/S175173111600066...
) involved with cell determination, proliferation, and differentiation. Those changes lead to a lower muscle growth potential (Costa et al., 2021b), which affects the whole-body energy expenditure in the post-natal life, once skeletal muscle is the major glucose utilization site (Mohammadabadi et al., 2021Mohammadabadi, M.; Bordbar, F.; Jensen, J.; Du, M. and Guo, W. 2021. Key genes regulating skeletal muscle development and growth in farm animals. Animals 11:835. https://doi.org/10.3390/ani11030835
https://doi.org/10.3390/ani11030835...
). Less muscle fiber hyperplasia can be replaced by intramuscular collagen deposits (Costa et al., 2021a), which may contribute to increase meat toughness (Fontes et al., 2021Fontes, M. M. S.; Costa, T. C.; Lopes, M. M.; Spuza, R. O.; Carneiro, L. S.; Paulino, P. V. R.; Chizzotti, M. L.; Silva, F. F.; Serão, N. V. L. and Duarte, M. S. 2021. Intramuscular collagen characteristics and expression of related genes in skeletal muscle of cull cows receiving a high-energy diet. Meat Science 177:108495. https://doi.org/10.1016/j.meatsci.2021.108495
https://doi.org/10.1016/j.meatsci.2021.1...
). Moreover, prenatal nutritional insults can also cause changes in muscle fiber metabolism regulated by transcription factors (Ramírez-Zamudio et al., 2022Ramírez-Zamudio, G. D.; Cruz, W. F. G.; Schoonmaker, J. P.; Resende, F. D.; Siqueira, G. R.; Machado Neto, O. R.; Gionbelli, T. R. S.; Teixeira, P. D.; Rodrigues, L. M.; Gionbelli, M. P. and Ladeira, M. M. 2022. Effect of rumen-protected fat on performance, carcass characteristics and beef quality of the progeny from Nellore cows fed by different planes of nutrition during gestation. Livestock Science 258:104851. https://doi.org/10.1016/j.livsci.2022.104851
https://doi.org/10.1016/j.livsci.2022.10...
) known as skeletal muscle metabolic plasticity (Aragão et al., 2014Aragão, R. S.; Guzmãn-Quevedo, O.; Pérez-García, G.; Manhães-de-Castro, R. and Bolaños-Jiménez, F. 2014. Maternal protein restriction impairs the transcriptional metabolic flexibility of skeletal muscle in adult rat offspring. British Journal of Nutrition 112:328-337. https://doi.org/10.1017/S0007114514000865
https://doi.org/10.1017/S000711451400086...
), which can negatively affect marbling deposition (Marquez et al., 2017Marquez, D. C.; Paulino, M. F.; Rennó, L. N.; Villadiego, F. C.; Ortega, R. M.; Moreno, D. S.; Martins, L. S.; De Almeida, D. M.; Gionbelli, M. P.; Manso, M. R.; Melo, L. P.; Moura, F. H. and Duarte, M. S. 2017. Supplementation of grazing beef cows during gestation as a strategy to improve skeletal muscle development of the offspring. Animal 11:2184-2192. https://doi.org/10.1017/S1751731117000982
https://doi.org/10.1017/S175173111700098...
). Therefore, monitoring the gestational environment is crucial to enhance the efficiency of meat production.

This comprehensive review aimed to highlight the effects of maternal nutrition on the offspring performance and meat quality, once the identification of these responses plays a central role in the global beef satisfying demand. Here, we first discuss the maternal and placental metabolism changes in response to the availability of nutrients over gestation, as well as the underlying mechanisms involved with phenotypic alterations observed in a long term. Then, we summarized the main effects of maternal nutrition on productive traits of beef cattle.

2. Changes in maternal and placental metabolism in response to nutritional challenges during gestation

During gestation, females from all species undergo homeorhesis, in which several physiological changes occur to ensure the continuous supply of essential metabolites to support fetal growth and development (Redmer et al., 2004Redmer, D. A.; Wallace, J. M. and Reynolds, L. P. 2004. Effect of nutrient intake during pregnancy on fetal and placental growth and vascular development. Domestic Animal Endocrinology 27:199-217. https://doi.org/10.1016/j.domaniend.2004.06.006
https://doi.org/10.1016/j.domaniend.2004...
). Thus, when fetal development is critical, due to nutrient deficiency, the mother tends to favor the fetal system, with coordinated changes in her own tissue metabolism that regulate the nutrient partitioning needed to supply the fetus (Bauman and Currie, 1980Bauman, D. E. and Currie, W. B. 1980. Partitioning of nutrients during pregnancy and lactation: a review of mechanisms involving homeostasis and homeorhesis. Journal of Dairy Science 63:1514-1529. https://doi.org/10.3168/jds.S0022-0302(80)83111-0
https://doi.org/10.3168/jds.S0022-0302(8...
).

In addition to the utilization of propionate for glucose production during periods of low availability or high demand for glucose, energy reserves may also be mobilized and used as a gluconeogenic precursor (Funston et al., 2010a). Under conditions of nutrient deficiency, the amino acids provided by the mobilization of maternal skeletal muscle are used to improve fetal access to amino acids (Bell et al., 2005Bell, A. W.; Ferrell, C. L. and Freetly, H. C. 2005. Pregnancy and fetal metabolism. p.523-550. In: Quantitative aspects of ruminant digestion and metabolism. 2nd ed. Dijkstra, J.; Forbes, J. M. and France, J., eds. CABI Publishing, Wallingford, UK.) or may also be used in maternal gluconeogenesis. The utilization of long-chain fatty acids, non-esterified fatty acids (NEFA), or ketoacids by the fetuses is limited due to the low placental ability to transport these substrates (Bell et al., 2005Bell, A. W.; Ferrell, C. L. and Freetly, H. C. 2005. Pregnancy and fetal metabolism. p.523-550. In: Quantitative aspects of ruminant digestion and metabolism. 2nd ed. Dijkstra, J.; Forbes, J. M. and France, J., eds. CABI Publishing, Wallingford, UK.). However, although NEFA seem to be not utilized by the fetus as a carbon source for energy production, this substrate supplies the pregnant dam with substrates for their own maintenance, and thus it indirectly contributes to spare glucose and amino acids to supply fetal requirements (Bell and Ehrhardt, 2000Bell, A. W. and Ehrhardt, R. A. 2000. Regulation of macronutrient partitioning between maternal and conceptus tissues in the pregnant ruminant. p.275-293. In: Ruminant physiology: Digestion, metabolism, growth and reproduction. Cronjé, P. B., ed. CABI Publishing, Wallingford, UK.).

Maternal tissue mobilization or deposition occurs as a function of dietary substrate supplies (McNeill et al., 1997McNeill, D. M.; Slepetis, R.; Ehrhardt, R. A.; Smith, D. M. and Bell, A. W. 1997. Protein requirements of sheep in late pregnancy: partitioning of nitrogen between gravid uterus and maternal tissues. Journal of Animal Science 75:809-816. https://doi.org/10.2527/1997.753809x
https://doi.org/10.2527/1997.753809x...
). Thus, nutritional adjustments for pregnant cows undergoing nutritional restrictions have been the subject of studies (Lopes et al., 2020Lopes, R. C.; Sampaio, C. B.; Trece, A. S.; Teixeira, P. D.; Gionbelli, T. R. S.; Santos, L. R.; Costa, T. C.; Duarte, M. S. and Gionbelli, M. P. 2020. Impacts of protein supplementation during late gestation of beef cows on maternal skeletal muscle and liver tissues metabolism. Animal 14:1867-1875. https://doi.org/10.1017/S1751731120000336
https://doi.org/10.1017/S175173112000033...
), which, in general, aimed to establish nutritional management that minimizes lean tissue catabolism and the negative effects on the fetus. For instance, Lopes et al. (2020)Lopes, R. C.; Sampaio, C. B.; Trece, A. S.; Teixeira, P. D.; Gionbelli, T. R. S.; Santos, L. R.; Costa, T. C.; Duarte, M. S. and Gionbelli, M. P. 2020. Impacts of protein supplementation during late gestation of beef cows on maternal skeletal muscle and liver tissues metabolism. Animal 14:1867-1875. https://doi.org/10.1017/S1751731120000336
https://doi.org/10.1017/S175173112000033...
showed the importance of supplementation for undernourished beef cows and reported a tendency toward greater mRNA expression of skeletal muscle synthesis markers in cows that received protein supplementation during late gestation. Such results likely demonstrate that a consequence of protein supplementation during gestation is a reduction in the intensity of lean tissue mobilization.

Under conditions of low nutrient availability causing intrauterine growth restriction, an additional compensatory mechanism involving the placenta may occur (Redmer et al., 2004Redmer, D. A.; Wallace, J. M. and Reynolds, L. P. 2004. Effect of nutrient intake during pregnancy on fetal and placental growth and vascular development. Domestic Animal Endocrinology 27:199-217. https://doi.org/10.1016/j.domaniend.2004.06.006
https://doi.org/10.1016/j.domaniend.2004...
). Borowicz et al. (2007)Borowicz, P. P.; Arnold, D. R.; Johnson, M. L.; Grazul-Bilska, A. T.; Redmer, D. A. and Reynolds, L. P. 2007. Placental growth throughout the last two-thirds of pregnancy in sheep: vascular development and angiogenic factor expression. Biology and Reproduction 76:259-267. https://doi.org/10.1095/biolreprod.106.054684
https://doi.org/10.1095/biolreprod.106.0...
reported that when metabolizable protein is reduced to 60% of requirements in sheep, uterine blood flow increased, indicating an adaptation of placental vasculature. Therefore, it is possible that nutrient deprivation due to inadequate placenta size and function affects fetuses from well-nourished dams. Additionally, fetuses from undernourished dams may not have difficulty meeting their nutrient requirements due to compensatory mechanisms in the placental system (Redmer et al., 2004Redmer, D. A.; Wallace, J. M. and Reynolds, L. P. 2004. Effect of nutrient intake during pregnancy on fetal and placental growth and vascular development. Domestic Animal Endocrinology 27:199-217. https://doi.org/10.1016/j.domaniend.2004.06.006
https://doi.org/10.1016/j.domaniend.2004...
). For instance, Vonnahme et al. (2007)Vonnahme, K. A.; Zhu, M. J.; Borowicz, P. P.; Geary, T. W.; Hess, B. W.; Reynolds, L. P.; Caton, J. S.; Means, W. J. and Ford, S. P. 2007. Effect of early gestational undernutrition on angiogenic factor expression and vascularity in the bovine placentome. Journal of Animal Science 85:2464-2472. https://doi.org/10.2527/jas.2006-805
https://doi.org/10.2527/jas.2006-805...
showed that nutrient restriction from 30 to 125 days of gestation in bovine increased placental mRNA concentrations of placental growth factor, improving fetal weight due to a greater transfer of nutrients through the placenta. Under a moderate nutritional restriction, the placenta may contribute to an increase in the abundance of Glucose transporter 3 (GLUT-3) as an attempt to increase its ability to glucose transfer (Bell and Ehrhardt, 2000Bell, A. W. and Ehrhardt, R. A. 2000. Regulation of macronutrient partitioning between maternal and conceptus tissues in the pregnant ruminant. p.275-293. In: Ruminant physiology: Digestion, metabolism, growth and reproduction. Cronjé, P. B., ed. CABI Publishing, Wallingford, UK.). However, under severe and prolonged nutrient restriction, the placenta may reduce glucose uptake and use glucose for its own demands (Bell and Ehrhardt, 2000Bell, A. W. and Ehrhardt, R. A. 2000. Regulation of macronutrient partitioning between maternal and conceptus tissues in the pregnant ruminant. p.275-293. In: Ruminant physiology: Digestion, metabolism, growth and reproduction. Cronjé, P. B., ed. CABI Publishing, Wallingford, UK.). McCrabb et al. (1992)McCrabb, G. J.; Egan, A. R. and Hosking, B. J. 1992. Maternal undernutrition during mid-pregnancy in sheep: variable effects on placental growth. The Journal of Agricultural Science 118:127-132. https://doi.org/10.1017/S002185960006809X
https://doi.org/10.1017/S002185960006809...
showed that pregnant sheep subjected to nutrient restriction in mid-gestation presented a decrease in placenta size without changing the number of individual placentomes or the fetal weight and dimensions. In contrast, Zhang et al. (2016)Zhang, H.; Sun, L. W.; Wang, Z. Y.; Deng, M. T.; Zhang, G. M.; Guo, R. H.; Ma, T. W. and Wang, F. 2016. Dietary N-carbamylglutamate and rumen-protected L-arginine supplementation ameliorate fetal growth restriction in undernourished ewes. Journal of Animal Science 94:2072-2085. https://doi.org/10.2527/jas.2015-9587
https://doi.org/10.2527/jas.2015-9587...
observed that undernourished animals presented lower concentrations of serum total polyamines in the uterine artery, fetal umbilical vein, and amniotic and allantoic fluids, which are crucial mediators of placental growth and angiogenesis, of fetal cellular function and synthesis of DNA and protein (Zhang et al., 2016Zhang, H.; Sun, L. W.; Wang, Z. Y.; Deng, M. T.; Zhang, G. M.; Guo, R. H.; Ma, T. W. and Wang, F. 2016. Dietary N-carbamylglutamate and rumen-protected L-arginine supplementation ameliorate fetal growth restriction in undernourished ewes. Journal of Animal Science 94:2072-2085. https://doi.org/10.2527/jas.2015-9587
https://doi.org/10.2527/jas.2015-9587...
).

Therefore, the compensatory mechanisms related to placental functioning may occur under conditions of nutritional restriction by the pregnant dam during gestation in attempt to mitigate the effects on fetal development, which may buffer the negative effects on the development of fetuses.

3. Maternal nutrition effects on epigenetic mechanisms underlying the skeletal muscle development

It is well established that among omics extracts (transcripts, proteins, and metabolites), a set of regulations and interactions generates a specific response according to the environment. These modulations may be explored through epigenetic analysis and systems biology approaches. Epigenetics explains how gene expression might be altered without affecting the nucleotide sequence (Feil, 2006Feil, R. 2006. Environmental and nutritional effects on the epigenetic regulation of genes. Mutation Research - Fundamental and Molecular Mechanisms of Mutagenesis 600:46-57. https://doi.org/10.1016/j.mrfmmm.2006.05.029
https://doi.org/10.1016/j.mrfmmm.2006.05...
). Moreover, this set of mechanisms is transferred between cell generations, constituting epigenetic memory. Of the epigenetic modifications, DNA methylation, chromatin remodeling, and noncoding RNA are relevant mechanisms for maternal nutrition and fetal programming.

DNA methylation is related to gene silencing, since the inclusion of a methyl group at the 5’ position of the cytosine residues located in the CpG islands in the promoter region of a gene inhibits the interaction between the transcriptional machinery complex and the target gene (Osorio et al., 2017Osorio, J. S.; Vailati-Riboni, M.; Palladino, A.; Luo, J. and Loor, J. J. 2017. Application of nutrigenomics in small ruminants: Lactation, growth, and beyond. Small Ruminant Research 154:29-44. https://doi.org/10.1016/j.smallrumres.2017.06.021
https://doi.org/10.1016/j.smallrumres.20...
). This process is widely influenced by dietary precursors, which are responsible for donating chemical groups to positively or negatively regulate DNA methylation (Osorio et al., 2017Osorio, J. S.; Vailati-Riboni, M.; Palladino, A.; Luo, J. and Loor, J. J. 2017. Application of nutrigenomics in small ruminants: Lactation, growth, and beyond. Small Ruminant Research 154:29-44. https://doi.org/10.1016/j.smallrumres.2017.06.021
https://doi.org/10.1016/j.smallrumres.20...
). The methyl donor S-adenosylmethionine (SAM), synthesized in the methionine cycle, is transferred to DNA through DNA methyltransferases (DNMT) (Triantaphyllopoulos et al., 2016Triantaphyllopoulos, K. A.; Ikonomopoulos, I. and Bannister, A. J. 2016. Epigenetics and inheritance of phenotype variation in livestock. Epigenetics and Chromatin 9:31. https://doi.org/10.1186/s13072-016-0081-5
https://doi.org/10.1186/s13072-016-0081-...
). Demethylation and, consequently, the reversion of gene silencing are catalyzed by the α-ketoglutarate (α-KG)-dependent ten-eleven translocation (TET) family of proteins (Ito et al., 2010Ito, S.; D’Alessio, A. C.; Taranova, O. V.; Hong, K.; Sowers, L. C. and Zhang, Y. 2010. Role of tet proteins in 5mC to 5hmC conversion, ES-cell self-renewal and inner cell mass specification. Nature 466:1129-1133. https://doi.org/10.1038/nature09303
https://doi.org/10.1038/nature09303...
). At the transcriptional level, energy restriction during late gestation has been found to alter the skeletal muscle and blood transcriptome of calves; specifically, genes related to energy metabolism and muscle development are downregulated in muscle cells, accompanied by a decrease in the expression of genes associated with the immune response (Sanglard et al., 2018Sanglard, L. P.; Nascimento, M.; Moriel, P.; Sommer, J.; Ashwell, M.; Poore, M. H.; Duarte, M. S. and Serão, N. V. L. 2018. Impact of energy restriction during late gestation on the muscle and blood transcriptome of beef calves after preconditioning. BMC Genomics 19:702. https://doi.org/10.1186/s12864-018-5089-8
https://doi.org/10.1186/s12864-018-5089-...
). While evaluating the DNA methylation level of some important gene inducers of cell differentiation, Paradis et al. (2017)Paradis, F.; Wood, K. M.; Swanson, K. C.; Miller, S. P.; McBride, B. W. and Fitzsimmons, C. 2017. Maternal nutrient restriction in mid-to-late gestation influences fetal mRNA expression in muscle tissues in beef cattle. BMC Genomics 18:632. https://doi.org/10.1186/s12864-017-4051-5
https://doi.org/10.1186/s12864-017-4051-...
observed hypermethylation in the promoter region of IGF2 in fetal skeletal muscle of offspring born from cows that were nutrient-restricted during mid- to late gestation, emphasizing the interaction between the nutritional plan and changes in gene expression.

Chromatin remodeling is mediated by histone post-translational modification (PTM), which involves the inclusion of a set of chemical or protein groups (e.g., methyl, acetyl, phosphate, and ubiquitin) to the histone tails (Triantaphyllopoulos et al., 2016Triantaphyllopoulos, K. A.; Ikonomopoulos, I. and Bannister, A. J. 2016. Epigenetics and inheritance of phenotype variation in livestock. Epigenetics and Chromatin 9:31. https://doi.org/10.1186/s13072-016-0081-5
https://doi.org/10.1186/s13072-016-0081-...
). The combination of different PTM in a specific histone is called the histone code (Jenuwein and Allis, 2001Jenuwein, T. and Allis, C. D. 2001. Translating the histone code. Science 293:1074-1080.). Depending on the histone code, chromatin may assume the structure of heterochromatin (compacted) or euchromatin (relaxed), which are associated with the repression or activation of gene expression, respectively (Jenuwein and Allis, 2001Jenuwein, T. and Allis, C. D. 2001. Translating the histone code. Science 293:1074-1080.). As an example, a decrease in the histone code H3K27me3 (histone 3 lysine 27 trimethylation) marker of gene silencing promoted an increase in overall adipogenesis in fetal mice from obese mothers (Yang et al., 2013Yang, Q. Y.; Liang, J. F.; Rogers, C. J.; Zhao, J. X.; Zhu, M. J. and Du, M. 2013. Maternal obesity induces epigenetic modifications to facilitate Zfp423 expression and enhance adipogenic differentiation in fetal mice. Diabetes 62:3727-3735. https://doi.org/10.2337/db13-0433
https://doi.org/10.2337/db13-0433...
). In contrast, the increase in the histone codes H3K9Ac (histone 3 lysine 9 acetylation) and H3K4me3 (histone 3 lysine 4 trimethylation) markers of gene activation, in the promoter region of myostatin, resulted in the reduction in muscle mass of piglets born from sows fed low-protein diets during pregnancy and lactation (Jia et al., 2016Jia, Y.; Gao, G.; Song, H.; Cai, D.; Yang, X. and Zhao, R. 2016. Low-protein diet fed to crossbred sows during pregnancy and lactation enhances myostatin gene expression through epigenetic regulation in skeletal muscle of weaning piglets. European Journal of Nutrition 55:1307-1314. https://doi.org/10.1007/s00394-015-0949-3
https://doi.org/10.1007/s00394-015-0949-...
).

When the effects of maternal feed restriction during different stages of gestation on the newborn goat skeletal muscle transcriptome (Costa et al., 2021c) and proteome (Costa et al., 2022Costa, T. C.; Dutra, L. L.; Mendes, T. A. O.; Santos, M. M.; Veroneze, R.; Gionbelli, M. P. and Duarte, M. S. 2022. Impact of maternal feed restriction at different stages of gestation on the proteomic profile of the newborn skeletal muscle. Animals 12:1011. https://doi.org/10.3390/ani12081011
https://doi.org/10.3390/ani12081011...
) were evaluated, it was observed that proteins exclusively expressed in each treatment (feed restriction in the first vs. last half of gestation) were present in both treatments at the transcriptional level. This suggested possible posttranscriptional regulation that repressed a set of genes in one of the treatments. The mechanism of post-transcriptional regulation may be mediated by noncoding RNA, called microRNA (miRNA). The inhibitory role of miRNA involves base-pairing with the target mRNA, which promotes repression (Wang et al., 2013Wang, X.; Gu, Z. and Jiang, H. 2013. MicroRNAs in farm animals. Animal 7:1567-1575. https://doi.org/10.1017/S1751731113001183
https://doi.org/10.1017/S175173111300118...
). Imperfect base-pairing with the target mRNA inhibits translations and, consequently, protein synthesis, while perfect complementation causes the degradation of the target mRNA (Wang et al., 2013Wang, X.; Gu, Z. and Jiang, H. 2013. MicroRNAs in farm animals. Animal 7:1567-1575. https://doi.org/10.1017/S1751731113001183
https://doi.org/10.1017/S175173111300118...
).

Therefore, maternal nutrition directly affects fetal metabolism through the pool of available nutrients, which mediate epigenetic mechanisms. The integration of omics data using a systems biology approach, combined with epigenetic analysis, may contribute valuable information on the effects of maternal nutrition on offspring skeletal muscle development and metabolism at the cellular level, which is reflected in the skeletal muscle growth and development and may cause changes in the quality traits of meat.

4. The impact of maternal nutrition on the performance, carcass, and meat quality traits of the offspring

In tropical and subtropical regions, forages are the main components of the diet in most cow-calf herds (Bell and Greenwood, 2013Bell, A. W. and Greenwood, P. L. 2013. Optimizing maternal cow, grower and finisher performance in beef production systems. p.45-66. In: Optimization of feed use efficiency in ruminant production systems. Makkar, H. P. S. and Beever, D., eds. FAO Animal Production and Health Proceedings, No. 16. FAO and Asian-Australasian Association of Animal Production Societies, Rome.). Such a scenario promotes variation in pasture availability and quality throughout the year, which is insufficient to meet the nutritional requirements of pregnant cows, mainly during mid- to late gestation (Lemos et al., 2012Lemos, B. J. M.; Souza, F. M.; Moreira, K. K. G.; Guimarães, T. P.; Pereira, M. L. R.; Ferreira, S. F. and Silva, R. M. 2012. Suplementação de bovinos de corte em pastejo. PUBVET 6:Art-1457.). Therefore, maternal restriction during critical periods of fetal skeletal muscle and adipose tissue development may compromise the performance and meat quality of the offspring (Figure 2).

Figure 2
The impact of maternal nutrition on the performance and carcass characteristics of the offspring.

During the dry season, pastures are deficient in proteins; thus, the restriction of energy and other nutrients in pregnant cows is also observed. In fact, the reduction of protein intake affects ruminal microorganism growth, which is responsible for the degradation of dietary fibers, causing a limitation of energy and dry matter intake (DMI) by cows (Sampaio et al., 2010Sampaio, C. B.; Detmann, E.; Paulino, M. F.; Valadares Filho, S. C.; Souza, M. A.; Lazzarini, I.; Paulino, P. V. R. and Queiroz, A. C. 2010. Intake and digestibility in cattle fed low-quality tropical forage and supplemented with nitrogenous compounds. Tropical Animal Health and Production 42:1471-1479. https://doi.org/10.1007/s11250-010-9581-7
https://doi.org/10.1007/s11250-010-9581-...
). Therefore, the use of nutritional strategies that increase the protein intake of pregnant cows improves the digestibility of low-quality fibers and, consequently, enhances maternal-fetal nutrient flow (Marquez et al., 2017Marquez, D. C.; Paulino, M. F.; Rennó, L. N.; Villadiego, F. C.; Ortega, R. M.; Moreno, D. S.; Martins, L. S.; De Almeida, D. M.; Gionbelli, M. P.; Manso, M. R.; Melo, L. P.; Moura, F. H. and Duarte, M. S. 2017. Supplementation of grazing beef cows during gestation as a strategy to improve skeletal muscle development of the offspring. Animal 11:2184-2192. https://doi.org/10.1017/S1751731117000982
https://doi.org/10.1017/S175173111700098...
). However, studies examining the effects of maternal nutritional strategies on offspring performance and carcass characteristics have had variable results (Tables 1a and 1b).

Table 1a
Effects of nutritional management of pregnant cows on performance and carcass characteristics of the offspring
Table 1b
Effects of nutritional management of pregnant cows on performance and carcass characteristics of the offspring

Underwood et al. (2010)Underwood, K. R.; Tong, J. F.; Price, P. L.; Roberts, A. J.; Grings, E. E.; Hess, B. W.; Means, W. J. and Du, M. 2010. Nutrition during mid to late gestation affects growth, adipose tissue deposition, and tenderness in cross-bred beef steers. Meat Science 86:588-593. https://doi.org/10.1016/j.meatsci.2010.04.008
https://doi.org/10.1016/j.meatsci.2010.0...
showed that cows fed improved pasture for 30 days at mid-gestation exhibited an increase of 10% in the offspring weaning and feedlot weight compared with the offspring resulting from cows fed native range pasture (~6% crude protein). In addition, an increase of approximately 19 kg carcass and 13.6% subcutaneous fat was observed in the resulting offspring (Underwood et al., 2010Underwood, K. R.; Tong, J. F.; Price, P. L.; Roberts, A. J.; Grings, E. E.; Hess, B. W.; Means, W. J. and Du, M. 2010. Nutrition during mid to late gestation affects growth, adipose tissue deposition, and tenderness in cross-bred beef steers. Meat Science 86:588-593. https://doi.org/10.1016/j.meatsci.2010.04.008
https://doi.org/10.1016/j.meatsci.2010.0...
). These results may suggest that outcomes of fetal programming through maternal nutrition may have an indirect effect on meat quality traits. The increased subcutaneous fat thickness in the carcass of offspring born from dams in better nutritional conditions likely help to prevent the rapid decline in temperature during the transformation process of muscle to meat, avoiding cold-shortening, which contributes to meat toughness (Ockerman and Basu, 2014Ockerman, H. W. and Basu, L. 2014. Carcass chilling and boning. p.142-147. In: Encyclopedia of meat science. 2nd ed. Dikeman, M. and Devine, C., eds. Elsevier.). Moreover, the heaviest carcasses in progeny born from dams that received adequate nutrition during gestation may result from greater muscle fiber development during the fetal stage. Costa et al. (2021a) showed that maternal protein restriction during mid-gestation reduces the number of muscle fibers in offspring.

In general, progeny from dams that receive an adequate nutritional plan shows better performance during the initial stages of life (Stalker et al., 2006Stalker, L. A.; Adams, D. C.; Klopfenstein, T. J.; Feuz, D. M. and Funston, R. N. 2006. Effects of pre- and postpartum nutrition on reproduction in spring calving cows and calf feedlot performance. Journal of Animal Science 84:2582-2589. https://doi.org/10.2527/jas.2005-640
https://doi.org/10.2527/jas.2005-640...
; Stalker et al., 2007Stalker, L. A.; Ciminski, L. A.; Adams, D. C.; Klopfenstein, T. J. and Clark, R. T. 2007. Effects of weaning date and prepartum protein supplementation on cow performance and calf growth. Rangeland Ecology and Management 60:578-587. https://doi.org/10.2111/06-082R1.1
https://doi.org/10.2111/06-082R1.1...
; Funston et al., 2010a; Funston et al., 2010b; Rodrigues et al., 2020Rodrigues, L. M.; Schoonmaker, J. P. S.; Resende, F. D.; Siqueira, G. R.; Machado Neto, O. R.; Gionbelli, M. P.; Gionbelli, T. R. S. and Ladeira, M. M. 2020. Effects of protein supplementation on Nellore cows’ reproductive performance, growth, myogenesis, lipogenesis and intestine development of the progeny. Animal Production Science 61:371-380. https://doi.org/10.1071/AN20498
https://doi.org/10.1071/AN20498...
), while few studies have reported significant gains throughout the production cycle (Stalker et al., 2007Stalker, L. A.; Ciminski, L. A.; Adams, D. C.; Klopfenstein, T. J. and Clark, R. T. 2007. Effects of weaning date and prepartum protein supplementation on cow performance and calf growth. Rangeland Ecology and Management 60:578-587. https://doi.org/10.2111/06-082R1.1
https://doi.org/10.2111/06-082R1.1...
; Underwood et al., 2010Underwood, K. R.; Tong, J. F.; Price, P. L.; Roberts, A. J.; Grings, E. E.; Hess, B. W.; Means, W. J. and Du, M. 2010. Nutrition during mid to late gestation affects growth, adipose tissue deposition, and tenderness in cross-bred beef steers. Meat Science 86:588-593. https://doi.org/10.1016/j.meatsci.2010.04.008
https://doi.org/10.1016/j.meatsci.2010.0...
). In contrast, other studies have failed to find an effect of adequate nutrition during mid- to late gestation on offspring performance throughout the production cycle or on carcass characteristics (Larson et al., 2009Larson, D. M.; Martin, J. L.; Adams, D. C. and Funston, R. N. 2009. Winter grazing system and supplementation during late gestation influence performance of beef cows and steer progeny. Journal of Animal Science 87:1147-1155. https://doi.org/10.2527/jas.2008-1323
https://doi.org/10.2527/jas.2008-1323...
; Mulliniks et al., 2012Mulliniks, J. T.; Sawyer, J. E.; Mathis, C. P.; Cox, S. H. and Petersen, M. K. 2012. Winter protein management during late gestation alters range cow and steer progeny performance. Journal of Animal Science 90:5099-5106. https://doi.org/10.2527/jas.2012-5535
https://doi.org/10.2527/jas.2012-5535...
; Mulliniks et al., 2013Mulliniks, J. T.; Mathis, C. P.; Cox, S. H. and Petersen, M. K. 2013. Supplementation strategy during late gestation alters steer progeny health in the feedlot without affecting cow performance. Animal Feed Science and Technology 185:126-132. https://doi.org/10.1016/j.anifeedsci.2013.07.006
https://doi.org/10.1016/j.anifeedsci.201...
). Such variations in the phenotypic responses of progeny affected by maternal nutrition depend on multiple factors, such as herd management during the production phases, genetic composition, maternal nutritional history, and adaptability to the environment (Broadhead et al., 2019Broadhead, D.; Mulliniks, J. T. and Funston, R. N. 2019. Developmental programming in a beef production system. Veterinary Clinics of North America: Food Animal Practice 35:379-390. https://doi.org/10.1016/j.cvfa.2019.02.011
https://doi.org/10.1016/j.cvfa.2019.02.0...
).

Maternal nutritional status during gestation may impact the qualitative properties of the meat from offspring (Alvarenga et al., 2016Alvarenga, T. I. R. C.; Copping, K. J.; Han, X.; Clayton, E. H.; Meyer, R. J.; Rodgers, R. J.; McMillen, I. C.; Perry, V. E. A. and Geesink, G. 2016. The influence of peri-conception and first trimester dietary restriction of protein in cattle on meat quality traits of entire male progeny. Meat Science 121:141-147. https://doi.org/10.1016/j.meatsci.2016.06.006
https://doi.org/10.1016/j.meatsci.2016.0...
; Maresca et al., 2019Maresca, S.; Valiente, S. L.; Rodriguez, A. M.; Testa, L. M.; Long, N. M.; Quintans, G. I. and Pavan, E. 2019. The influence of protein restriction during mid- to late gestation on beef offspring growth, carcass characteristic and meat quality. Meat Science 153:103-108. https://doi.org/10.1016/j.meatsci.2019.03.014
https://doi.org/10.1016/j.meatsci.2019.0...
; Webb et al., 2019Webb, M. J.; Block, J. J.; Funston, R. N.; Underwood, K. R.; Legako, J. F.; Harty, A. A.; Salverson, R. R.; Olson, K. C. and Blair, A. D. 2019. Influence of maternal protein restriction in primiparous heifers during mid- and/or late-gestation on meat quality and fatty acid profile of progeny. Meat Science 152:31-37. https://doi.org/10.1016/j.meatsci.2019.02.006
https://doi.org/10.1016/j.meatsci.2019.0...
). For instance, the meat of steers born from dams raised under improved pastures for 30 days at mid-gestation was more tender than that of steers born from dams fed native pasture (~6% crude protein) (Underwood et al., 2010Underwood, K. R.; Tong, J. F.; Price, P. L.; Roberts, A. J.; Grings, E. E.; Hess, B. W.; Means, W. J. and Du, M. 2010. Nutrition during mid to late gestation affects growth, adipose tissue deposition, and tenderness in cross-bred beef steers. Meat Science 86:588-593. https://doi.org/10.1016/j.meatsci.2010.04.008
https://doi.org/10.1016/j.meatsci.2010.0...
). In addition to tenderness, the pH, color, water-holding capacity, and marbling of the meat may be affected by maternal nutrition due to alterations in the metabolic characteristics of muscle fibers (Fahey et al., 2005Fahey, A. J.; Brameld, J. M.; Parr, T. and Buttery, P. J. 2005. The effect of maternal undernutrition before muscle differentiation on the muscle fiber development of the newborn lamb. Journal of Animal Science 83:2564-2571. https://doi.org/10.2527/2005.83112564x
https://doi.org/10.2527/2005.83112564x...
; Picard and Gagaoua, 2020Picard, B. and Gagaoua, M. 2020. Muscle fiber properties in cattle and their relationships with meat qualities: an overview. Journal of Agricultural and Food Chemistry 68:6021-6039. https://doi.org/10.1021/acs.jafc.0c02086
https://doi.org/10.1021/acs.jafc.0c02086...
), as well as the proportion of muscle, adipose, and connective tissue formed during the prenatal phase (Duarte et al., 2014Duarte, M. S.; Gionbelli, M. P.; Paulino, P. V. R.; Serão, N. V. L.; Nascimento, C. S.; Botelho, M. E.; Martins, T. S.; Filho, S. C. V.; Dodson, M. V.; Guimarães, S. E. F. and Du, M. 2014. Maternal overnutrition enhances mRNA expression of adipogenic markers and collagen deposition in skeletal muscle of beef cattle fetuses. Journal of Animal Science 92:3846-3854. https://doi.org/10.2527/jas.2014-7568
https://doi.org/10.2527/jas.2014-7568...
; Du et al., 2015Du, M.; Wang, B.; Fu, X.; Yang, Q. and Zhu, M. J. 2015. Fetal programming in meat production. Meat Science 109:40-47. https://doi.org/10.1016/j.meatsci.2015.04.010
https://doi.org/10.1016/j.meatsci.2015.0...
). For example, a 50% nutrient restriction in sheep during the first 30 days of gestation enhanced the proportion of muscle fibers with the characteristics of slow contraction and oxidative metabolism in offspring (Fahey et al., 2005Fahey, A. J.; Brameld, J. M.; Parr, T. and Buttery, P. J. 2005. The effect of maternal undernutrition before muscle differentiation on the muscle fiber development of the newborn lamb. Journal of Animal Science 83:2564-2571. https://doi.org/10.2527/2005.83112564x
https://doi.org/10.2527/2005.83112564x...
). Muscles with a greater proportion of slow-twitch fibers and oxidative metabolism show a low rate of postmortem pH decline due to low glycogen storage, resulting in an elevated final pH of the meat (Kim et al., 2016Kim, G. D.; Yang, H. S. and Jeong, J. Y. 2016. Comparison of characteristics of myosin heavy chain-based fiber and meat quality among four bovine skeletal muscles. Korean Journal for Food Science of Animal Resources 36:819-828. https://doi.org/10.5851/kosfa.2016.36.6.819
https://doi.org/10.5851/kosfa.2016.36.6....
). When the pH is higher than 5.6, there is a change in the negative charge and structures of the muscular matrix, which results in greater intracellular water retention, negatively affecting meat color (Ramanathan et al., 2020Ramanathan, R.; Hunt, M. C.; Mancini, R. A.; Nair, M. N.; Denzer, M. L.; Suman, S. P. and Mafi, G. G. 2020. Recent updates in meat color research: integrating traditional and high-throughput approaches. Meat and Muscle Biology 4(2). https://doi.org/10.22175/mmb.9598
https://doi.org/10.22175/mmb.9598...
). Moreover, changes in the final pH interfere with the activity of proteolytic enzymes, which are responsible for tenderness (Matarneh et al., 2017Matarneh, S. K.; England, E. M.; Scheffler, T. L. and Gerrard, D. E. 2017. The conversion of muscle to meat. p.159-185. In: Lawrie´ s meat science. Elsevier.).

However, some changes may occur at the molecular level (Table 2) without resulting in phenotypic changes. Jennings et al. (2016)Jennings, T. D.; Gonda, M. G.; Underwood, K. R.; Wertz-Lutz, A. E. and Blair, A. D. 2016. The influence of maternal nutrition on expression of genes responsible for adipogenesis and myogenesis in the bovine fetus. Animal 10:1697-1705. https://doi.org/10.1017/S1751731116000665
https://doi.org/10.1017/S175173111600066...
, evaluated the effects of energy levels [72, 87, or 146% of net energy for maintenance (NEm) requirements] during early to mid-gestation and did not find effects of maternal nutrition on muscle histology characteristics (fiber area, diameter, and number), despite the effects on mRNA expression in skeletal muscle. In this study, myogenin was upregulated in the skeletal muscle of fetuses from cows fed at 72% NEm compared with those from cows fed at 87% NEm, indicating a potential reduction in myoblast differentiation, followed by an earlier fusion of these cells in fetuses exposed to undernutrition.

Table 2
Effects of prenatal nutrition on gene expression and characteristics of skeletal muscle in cattle

Rodrigues et al. (2020)Rodrigues, L. M.; Schoonmaker, J. P. S.; Resende, F. D.; Siqueira, G. R.; Machado Neto, O. R.; Gionbelli, M. P.; Gionbelli, T. R. S. and Ladeira, M. M. 2020. Effects of protein supplementation on Nellore cows’ reproductive performance, growth, myogenesis, lipogenesis and intestine development of the progeny. Animal Production Science 61:371-380. https://doi.org/10.1071/AN20498
https://doi.org/10.1071/AN20498...
investigated the effects of protein supplementation during mid- to late gestation in grazing beef cows with moderate nutritional restriction on performance and molecular markers in offspring (Table 2). Protein supplementation of the dams did not affect the expression of myogenic genes. However, a downregulation of C/EBPA and FABP4 was observed in 11-day-old calves from supplemented dams. These findings indicate that offspring from non-supplemented cows showed early adipogenic differentiation, which may impair the proliferation of intramuscular adipocytes. In summary, maternal restriction during critical periods of fetal skeletal muscle and adipose tissue development may compromise the performance and meat quality of the offspring; however, the use of maternal nutritional strategies shows better performance and carcass characteristics on offspring.

5. Summary and future perspectives

Maternal nutrition affects the skeletal muscle development of the fetus, exerting long-term effects on offspring performance and growth. Maternal undernutrition during fetal development reduces the number of muscle fibers, alters muscle fiber composition, and impacts fetal adipogenesis. However, adequate supplementation with nutrients improves skeletal muscle development and adipogenesis, increasing marbling in offspring. Thus, understanding the effects of maternal supplementation during different gestational periods on the performance and final carcass composition of the progeny may help improve meat production and carcass and meat quality traits.

Acknowledgments

We thank the Conselho Nacional de Desenvolvimento Científico e Tecnológico (CNPq, grant 313858/2021-7), Fundação de Amparo à Pesquisa do Estado de Minas Gerais (FAPEMIG, grant #APQ-02496), Instituto Nacional de Ciência e Tecnologia Ciência Animal (INCT-CA, grant #465377/2014-9), Coordenação de Aperfeiçoamento de Pessoal de Nível Superior (CAPES, grant #001), Cargill, and Trouw Nutrition for the financial support of fetal programing research in our lab.

References

  • Alvarenga, T. I. R. C.; Copping, K. J.; Han, X.; Clayton, E. H.; Meyer, R. J.; Rodgers, R. J.; McMillen, I. C.; Perry, V. E. A. and Geesink, G. 2016. The influence of peri-conception and first trimester dietary restriction of protein in cattle on meat quality traits of entire male progeny. Meat Science 121:141-147. https://doi.org/10.1016/j.meatsci.2016.06.006
    » https://doi.org/10.1016/j.meatsci.2016.06.006
  • Aragão, R. S.; Guzmãn-Quevedo, O.; Pérez-García, G.; Manhães-de-Castro, R. and Bolaños-Jiménez, F. 2014. Maternal protein restriction impairs the transcriptional metabolic flexibility of skeletal muscle in adult rat offspring. British Journal of Nutrition 112:328-337. https://doi.org/10.1017/S0007114514000865
    » https://doi.org/10.1017/S0007114514000865
  • Batistel, F.; Alharthi, A. S.; Yambao, R. R. C.; Elolimy, A. A.; Pan, Y. X.; Parys, C. and Loor, J. J. 2019. Methionine supply during late-gestation triggers offspring sex-specific divergent changes in metabolic and epigenetic signatures in bovine placenta. The Journal of Nutrition 149:6-17. https://doi.org/10.1093/jn/nxy240
    » https://doi.org/10.1093/jn/nxy240
  • Bauman, D. E. and Currie, W. B. 1980. Partitioning of nutrients during pregnancy and lactation: a review of mechanisms involving homeostasis and homeorhesis. Journal of Dairy Science 63:1514-1529. https://doi.org/10.3168/jds.S0022-0302(80)83111-0
    » https://doi.org/10.3168/jds.S0022-0302(80)83111-0
  • Bell, A. W. and Ehrhardt, R. A. 2000. Regulation of macronutrient partitioning between maternal and conceptus tissues in the pregnant ruminant. p.275-293. In: Ruminant physiology: Digestion, metabolism, growth and reproduction. Cronjé, P. B., ed. CABI Publishing, Wallingford, UK.
  • Bell, A. W.; Ferrell, C. L. and Freetly, H. C. 2005. Pregnancy and fetal metabolism. p.523-550. In: Quantitative aspects of ruminant digestion and metabolism. 2nd ed. Dijkstra, J.; Forbes, J. M. and France, J., eds. CABI Publishing, Wallingford, UK.
  • Bell, A. W. and Greenwood, P. L. 2013. Optimizing maternal cow, grower and finisher performance in beef production systems. p.45-66. In: Optimization of feed use efficiency in ruminant production systems. Makkar, H. P. S. and Beever, D., eds. FAO Animal Production and Health Proceedings, No. 16. FAO and Asian-Australasian Association of Animal Production Societies, Rome.
  • Blair, A. D.; Gubbels, E. R.; Block, J. J.; Olson, K. C.; Grubbs, J. K. and Underwood, K. R. 2021. Maternal nutrition and meat quality of progeny. Meat and Muscle Biology 5:1-9. https://doi.org/10.22175/mmb.12990
    » https://doi.org/10.22175/mmb.12990
  • Bonnet, M.; Cassar-Malek, I.; Chilliard, Y. and Picard, B. 2010. Ontogenesis of muscle and adipose tissues and their interactions in ruminants and other species. Animal 4:1093-1109. https://doi.org/10.1017/S1751731110000601
    » https://doi.org/10.1017/S1751731110000601
  • Borowicz, P. P.; Arnold, D. R.; Johnson, M. L.; Grazul-Bilska, A. T.; Redmer, D. A. and Reynolds, L. P. 2007. Placental growth throughout the last two-thirds of pregnancy in sheep: vascular development and angiogenic factor expression. Biology and Reproduction 76:259-267. https://doi.org/10.1095/biolreprod.106.054684
    » https://doi.org/10.1095/biolreprod.106.054684
  • Broadhead, D.; Mulliniks, J. T. and Funston, R. N. 2019. Developmental programming in a beef production system. Veterinary Clinics of North America: Food Animal Practice 35:379-390. https://doi.org/10.1016/j.cvfa.2019.02.011
    » https://doi.org/10.1016/j.cvfa.2019.02.011
  • Costa, T. C.; Dutra, L. L.; Mendes, T. A. O.; Santos, M. M.; Veroneze, R.; Gionbelli, M. P. and Duarte, M. S. 2022. Impact of maternal feed restriction at different stages of gestation on the proteomic profile of the newborn skeletal muscle. Animals 12:1011. https://doi.org/10.3390/ani12081011
    » https://doi.org/10.3390/ani12081011
  • Costa, T. C.; Du, M.; Nascimento, K. B.; Galvão, M. C.; Meneses, J. A. M.; Schultz, E. B.; Gionbelli, M. P. and Duarte, M. S. 2021a. Skeletal muscle development in postnatal beef cattle resulting from maternal protein restriction during mid-gestation. Animals 11:860. https://doi.org/10.3390/ani11030860
    » https://doi.org/10.3390/ani11030860
  • Costa, T. C.; Gionbelli, M. P. and Duarte, M. S. 2021b. Fetal programming in ruminant animals: understanding the skeletal muscle development to improve meat quality. Animal Frontiers 11:66-73. https://doi.org/10.1093/af/vfab061
    » https://doi.org/10.1093/af/vfab061
  • Costa, T. C.; Mendes, T. A. O.; Fontes, M. M. S.; Lopes, M. M.; Du, M.; Serão, N. V. L.; Sanglard, L. M. P.; Bertolini, F.; Rothschild, M. F.; Silva, F. F.; Gionbelli, M. P. and Duarte, M. S. 2021c. Transcriptome changes in newborn goats’ skeletal muscle as a result of maternal feed restriction at different stages of gestation. Livestock Science 248:104503. https://doi.org/10.1016/j.livsci.2021.104503
    » https://doi.org/10.1016/j.livsci.2021.104503
  • Du, M.; Huang, Y.; Das, A. K.; Yang, Q.; Duarte, M. S.; Dodson, M. V. and Zhu, M. J. 2013. Meat Science and Muscle Biology Symposium: Manipulating mesenchymal progenitor cell differentiation to optimize performance and carcass value of beef cattle. Journal of Animal Science 91:1419-1427. https://doi.org/10.2527/jas.2012-5670
    » https://doi.org/10.2527/jas.2012-5670
  • Du, M.; Wang, B.; Fu, X.; Yang, Q. and Zhu, M. J. 2015. Fetal programming in meat production. Meat Science 109:40-47. https://doi.org/10.1016/j.meatsci.2015.04.010
    » https://doi.org/10.1016/j.meatsci.2015.04.010
  • Duarte, M. S.; Gionbelli, M. P.; Paulino, P. V. R.; Serão, N. V. L.; Nascimento, C. S.; Botelho, M. E.; Martins, T. S.; Filho, S. C. V.; Dodson, M. V.; Guimarães, S. E. F. and Du, M. 2014. Maternal overnutrition enhances mRNA expression of adipogenic markers and collagen deposition in skeletal muscle of beef cattle fetuses. Journal of Animal Science 92:3846-3854. https://doi.org/10.2527/jas.2014-7568
    » https://doi.org/10.2527/jas.2014-7568
  • Fahey, A. J.; Brameld, J. M.; Parr, T. and Buttery, P. J. 2005. The effect of maternal undernutrition before muscle differentiation on the muscle fiber development of the newborn lamb. Journal of Animal Science 83:2564-2571. https://doi.org/10.2527/2005.83112564x
    » https://doi.org/10.2527/2005.83112564x
  • Feil, R. 2006. Environmental and nutritional effects on the epigenetic regulation of genes. Mutation Research - Fundamental and Molecular Mechanisms of Mutagenesis 600:46-57. https://doi.org/10.1016/j.mrfmmm.2006.05.029
    » https://doi.org/10.1016/j.mrfmmm.2006.05.029
  • Fontes, M. M. S.; Costa, T. C.; Lopes, M. M.; Spuza, R. O.; Carneiro, L. S.; Paulino, P. V. R.; Chizzotti, M. L.; Silva, F. F.; Serão, N. V. L. and Duarte, M. S. 2021. Intramuscular collagen characteristics and expression of related genes in skeletal muscle of cull cows receiving a high-energy diet. Meat Science 177:108495. https://doi.org/10.1016/j.meatsci.2021.108495
    » https://doi.org/10.1016/j.meatsci.2021.108495
  • Funston, R. N.; Larson, D. M. and Vonnahme, K. A. 2010a. Effects of maternal nutrition on conceptus growth and offspring performance: implications for beef cattle production. Journal of Animal Science 88:E205-E215. https://doi.org/10.2527/jas.2009-2351
    » https://doi.org/10.2527/jas.2009-2351
  • Funston, R. N.; Martin, J. L.; Adams, D. C. and Larson, D. M. 2010b. Winter grazing system and supplementation of beef cows during late gestation influence heifer progeny. Journal of Animal Science 88:4094-4101. https://doi.org/10.2527/jas.2010-3039
    » https://doi.org/10.2527/jas.2010-3039
  • Gionbelli, T. R. S.; Veloso, C. M.; Rotta, P. P.; Valadares Filho, S. C.; Carvalho, B. C.; Marcondes, M. I.; Cunha, C. S.; Novaes, M. A. S.; Prezotto, L. D.; Duarte, M. S. and Gionbelli, M. P. 2018. Foetal development of skeletal muscle in bovines as a function of maternal nutrition, foetal sex and gestational age. Journal of Animal Physiology and Animal Nutrition 102:545-556. https://doi.org/10.1111/jpn.12786
    » https://doi.org/10.1111/jpn.12786
  • Greenwood, P. L. and Cafe, L. M. 2007. Prenatal and pre-weaning growth and nutrition of cattle: long-term consequences for beef production. Animal 1:1283-1296. https://doi.org/10.1017/S175173110700050X
    » https://doi.org/10.1017/S175173110700050X
  • Ito, S.; D’Alessio, A. C.; Taranova, O. V.; Hong, K.; Sowers, L. C. and Zhang, Y. 2010. Role of tet proteins in 5mC to 5hmC conversion, ES-cell self-renewal and inner cell mass specification. Nature 466:1129-1133. https://doi.org/10.1038/nature09303
    » https://doi.org/10.1038/nature09303
  • Jennings, T. D.; Gonda, M. G.; Underwood, K. R.; Wertz-Lutz, A. E. and Blair, A. D. 2016. The influence of maternal nutrition on expression of genes responsible for adipogenesis and myogenesis in the bovine fetus. Animal 10:1697-1705. https://doi.org/10.1017/S1751731116000665
    » https://doi.org/10.1017/S1751731116000665
  • Jenuwein, T. and Allis, C. D. 2001. Translating the histone code. Science 293:1074-1080.
  • Jia, Y.; Gao, G.; Song, H.; Cai, D.; Yang, X. and Zhao, R. 2016. Low-protein diet fed to crossbred sows during pregnancy and lactation enhances myostatin gene expression through epigenetic regulation in skeletal muscle of weaning piglets. European Journal of Nutrition 55:1307-1314. https://doi.org/10.1007/s00394-015-0949-3
    » https://doi.org/10.1007/s00394-015-0949-3
  • Kim, G. D.; Yang, H. S. and Jeong, J. Y. 2016. Comparison of characteristics of myosin heavy chain-based fiber and meat quality among four bovine skeletal muscles. Korean Journal for Food Science of Animal Resources 36:819-828. https://doi.org/10.5851/kosfa.2016.36.6.819
    » https://doi.org/10.5851/kosfa.2016.36.6.819
  • Larson, D. M.; Martin, J. L.; Adams, D. C. and Funston, R. N. 2009. Winter grazing system and supplementation during late gestation influence performance of beef cows and steer progeny. Journal of Animal Science 87:1147-1155. https://doi.org/10.2527/jas.2008-1323
    » https://doi.org/10.2527/jas.2008-1323
  • Lemos, B. J. M.; Souza, F. M.; Moreira, K. K. G.; Guimarães, T. P.; Pereira, M. L. R.; Ferreira, S. F. and Silva, R. M. 2012. Suplementação de bovinos de corte em pastejo. PUBVET 6:Art-1457.
  • Lopes, R. C.; Sampaio, C. B.; Trece, A. S.; Teixeira, P. D.; Gionbelli, T. R. S.; Santos, L. R.; Costa, T. C.; Duarte, M. S. and Gionbelli, M. P. 2020. Impacts of protein supplementation during late gestation of beef cows on maternal skeletal muscle and liver tissues metabolism. Animal 14:1867-1875. https://doi.org/10.1017/S1751731120000336
    » https://doi.org/10.1017/S1751731120000336
  • Maresca, S.; Valiente, S. L.; Rodriguez, A. M.; Testa, L. M.; Long, N. M.; Quintans, G. I. and Pavan, E. 2019. The influence of protein restriction during mid- to late gestation on beef offspring growth, carcass characteristic and meat quality. Meat Science 153:103-108. https://doi.org/10.1016/j.meatsci.2019.03.014
    » https://doi.org/10.1016/j.meatsci.2019.03.014
  • Marquez, D. C.; Paulino, M. F.; Rennó, L. N.; Villadiego, F. C.; Ortega, R. M.; Moreno, D. S.; Martins, L. S.; De Almeida, D. M.; Gionbelli, M. P.; Manso, M. R.; Melo, L. P.; Moura, F. H. and Duarte, M. S. 2017. Supplementation of grazing beef cows during gestation as a strategy to improve skeletal muscle development of the offspring. Animal 11:2184-2192. https://doi.org/10.1017/S1751731117000982
    » https://doi.org/10.1017/S1751731117000982
  • Matarneh, S. K.; England, E. M.; Scheffler, T. L. and Gerrard, D. E. 2017. The conversion of muscle to meat. p.159-185. In: Lawrie´ s meat science. Elsevier.
  • McCrabb, G. J.; Egan, A. R. and Hosking, B. J. 1992. Maternal undernutrition during mid-pregnancy in sheep: variable effects on placental growth. The Journal of Agricultural Science 118:127-132. https://doi.org/10.1017/S002185960006809X
    » https://doi.org/10.1017/S002185960006809X
  • McLean, K. J.; Boehmer, B. H.; Spicer, L. J. and Wettemann, R. P. 2018. The effects of protein supplementation of fall calving beef cows on pre-and postpartum plasma insulin, glucose and IGF-I, and postnatal growth and plasma insulin and IGF-I of calves. Journal of Animal Science 96:2629-2639. https://doi.org/10.1093/jas/sky173
    » https://doi.org/10.1093/jas/sky173
  • McNeill, D. M.; Slepetis, R.; Ehrhardt, R. A.; Smith, D. M. and Bell, A. W. 1997. Protein requirements of sheep in late pregnancy: partitioning of nitrogen between gravid uterus and maternal tissues. Journal of Animal Science 75:809-816. https://doi.org/10.2527/1997.753809x
    » https://doi.org/10.2527/1997.753809x
  • Mohammadabadi, M.; Bordbar, F.; Jensen, J.; Du, M. and Guo, W. 2021. Key genes regulating skeletal muscle development and growth in farm animals. Animals 11:835. https://doi.org/10.3390/ani11030835
    » https://doi.org/10.3390/ani11030835
  • Mulliniks, J. T.; Mathis, C. P.; Cox, S. H. and Petersen, M. K. 2013. Supplementation strategy during late gestation alters steer progeny health in the feedlot without affecting cow performance. Animal Feed Science and Technology 185:126-132. https://doi.org/10.1016/j.anifeedsci.2013.07.006
    » https://doi.org/10.1016/j.anifeedsci.2013.07.006
  • Mulliniks, J. T.; Sawyer, J. E.; Mathis, C. P.; Cox, S. H. and Petersen, M. K. 2012. Winter protein management during late gestation alters range cow and steer progeny performance. Journal of Animal Science 90:5099-5106. https://doi.org/10.2527/jas.2012-5535
    » https://doi.org/10.2527/jas.2012-5535
  • Nathanielsz, P. W.; Poston, L. and Taylor, P. D. 2007. In utero exposure to maternal obesity and diabetes: animal models that identify and characterize implications for future health. Obstetrics and Gynecology Clinics of North America 34:201-212. https://doi.org/10.1016/j.ogc.2007.03.006
    » https://doi.org/10.1016/j.ogc.2007.03.006
  • Nissen, P. M.; Danielsen, V. O.; Jorgensen, P. F. and Oksbjerg, N. 2003. Increased maternal nutrition of sows has no beneficial effects on muscle fiber number or postnatal growth and has no impact on the meat quality of the offspring. Journal of Animal Science 81:3018-3027. https://doi.org/10.2527/2003.81123018x
    » https://doi.org/10.2527/2003.81123018x
  • Ockerman, H. W. and Basu, L. 2014. Carcass chilling and boning. p.142-147. In: Encyclopedia of meat science. 2nd ed. Dikeman, M. and Devine, C., eds. Elsevier.
  • Osorio, J. S.; Vailati-Riboni, M.; Palladino, A.; Luo, J. and Loor, J. J. 2017. Application of nutrigenomics in small ruminants: Lactation, growth, and beyond. Small Ruminant Research 154:29-44. https://doi.org/10.1016/j.smallrumres.2017.06.021
    » https://doi.org/10.1016/j.smallrumres.2017.06.021
  • Paradis, F.; Wood, K. M.; Swanson, K. C.; Miller, S. P.; McBride, B. W. and Fitzsimmons, C. 2017. Maternal nutrient restriction in mid-to-late gestation influences fetal mRNA expression in muscle tissues in beef cattle. BMC Genomics 18:632. https://doi.org/10.1186/s12864-017-4051-5
    » https://doi.org/10.1186/s12864-017-4051-5
  • Picard, B.; Lefaucheur, L.; Berri, C. and Duclos, M. J. 2002. Muscle fibre ontogenesis in farm animal species. Reproduction Nutrition Development 42:415-431. https://doi.org/10.1051/rnd:2002035
    » https://doi.org/10.1051/rnd:2002035
  • Picard, B. and Gagaoua, M. 2020. Muscle fiber properties in cattle and their relationships with meat qualities: an overview. Journal of Agricultural and Food Chemistry 68:6021-6039. https://doi.org/10.1021/acs.jafc.0c02086
    » https://doi.org/10.1021/acs.jafc.0c02086
  • Ramanathan, R.; Hunt, M. C.; Mancini, R. A.; Nair, M. N.; Denzer, M. L.; Suman, S. P. and Mafi, G. G. 2020. Recent updates in meat color research: integrating traditional and high-throughput approaches. Meat and Muscle Biology 4(2). https://doi.org/10.22175/mmb.9598
    » https://doi.org/10.22175/mmb.9598
  • Ramírez-Zamudio, G. D.; Cruz, W. F. G.; Schoonmaker, J. P.; Resende, F. D.; Siqueira, G. R.; Machado Neto, O. R.; Gionbelli, T. R. S.; Teixeira, P. D.; Rodrigues, L. M.; Gionbelli, M. P. and Ladeira, M. M. 2022. Effect of rumen-protected fat on performance, carcass characteristics and beef quality of the progeny from Nellore cows fed by different planes of nutrition during gestation. Livestock Science 258:104851. https://doi.org/10.1016/j.livsci.2022.104851
    » https://doi.org/10.1016/j.livsci.2022.104851
  • Redmer, D. A.; Wallace, J. M. and Reynolds, L. P. 2004. Effect of nutrient intake during pregnancy on fetal and placental growth and vascular development. Domestic Animal Endocrinology 27:199-217. https://doi.org/10.1016/j.domaniend.2004.06.006
    » https://doi.org/10.1016/j.domaniend.2004.06.006
  • Rodrigues, L. M.; Schoonmaker, J. P. S.; Resende, F. D.; Siqueira, G. R.; Machado Neto, O. R.; Gionbelli, M. P.; Gionbelli, T. R. S. and Ladeira, M. M. 2020. Effects of protein supplementation on Nellore cows’ reproductive performance, growth, myogenesis, lipogenesis and intestine development of the progeny. Animal Production Science 61:371-380. https://doi.org/10.1071/AN20498
    » https://doi.org/10.1071/AN20498
  • Sampaio, C. B.; Detmann, E.; Paulino, M. F.; Valadares Filho, S. C.; Souza, M. A.; Lazzarini, I.; Paulino, P. V. R. and Queiroz, A. C. 2010. Intake and digestibility in cattle fed low-quality tropical forage and supplemented with nitrogenous compounds. Tropical Animal Health and Production 42:1471-1479. https://doi.org/10.1007/s11250-010-9581-7
    » https://doi.org/10.1007/s11250-010-9581-7
  • Sanglard, L. P.; Nascimento, M.; Moriel, P.; Sommer, J.; Ashwell, M.; Poore, M. H.; Duarte, M. S. and Serão, N. V. L. 2018. Impact of energy restriction during late gestation on the muscle and blood transcriptome of beef calves after preconditioning. BMC Genomics 19:702. https://doi.org/10.1186/s12864-018-5089-8
    » https://doi.org/10.1186/s12864-018-5089-8
  • Sinclair, K. D.; Rutherford, K. M. D.; Wallace, J. M.; Brameld, J. M.; Stöger, R.; Alberio, R.; Sweetman, D.; Gardner, D. S.; Perry, V. E. A.; Adam, C. L.; Ashworth, C. J.; Robinson, J. E. and Dwyer, C. M. 2016. Epigenetics and developmental programming of welfare and production traits in farm animals. Reproduction, Fertility and Development 28:1443-1478. https://doi.org/10.1071/RD16102
    » https://doi.org/10.1071/RD16102
  • Stalker, L. A.; Adams, D. C.; Klopfenstein, T. J.; Feuz, D. M. and Funston, R. N. 2006. Effects of pre- and postpartum nutrition on reproduction in spring calving cows and calf feedlot performance. Journal of Animal Science 84:2582-2589. https://doi.org/10.2527/jas.2005-640
    » https://doi.org/10.2527/jas.2005-640
  • Stalker, L. A.; Ciminski, L. A.; Adams, D. C.; Klopfenstein, T. J. and Clark, R. T. 2007. Effects of weaning date and prepartum protein supplementation on cow performance and calf growth. Rangeland Ecology and Management 60:578-587. https://doi.org/10.2111/06-082R1.1
    » https://doi.org/10.2111/06-082R1.1
  • Triantaphyllopoulos, K. A.; Ikonomopoulos, I. and Bannister, A. J. 2016. Epigenetics and inheritance of phenotype variation in livestock. Epigenetics and Chromatin 9:31. https://doi.org/10.1186/s13072-016-0081-5
    » https://doi.org/10.1186/s13072-016-0081-5
  • Underwood, K. R.; Tong, J. F.; Price, P. L.; Roberts, A. J.; Grings, E. E.; Hess, B. W.; Means, W. J. and Du, M. 2010. Nutrition during mid to late gestation affects growth, adipose tissue deposition, and tenderness in cross-bred beef steers. Meat Science 86:588-593. https://doi.org/10.1016/j.meatsci.2010.04.008
    » https://doi.org/10.1016/j.meatsci.2010.04.008
  • Vonnahme, K. A.; Zhu, M. J.; Borowicz, P. P.; Geary, T. W.; Hess, B. W.; Reynolds, L. P.; Caton, J. S.; Means, W. J. and Ford, S. P. 2007. Effect of early gestational undernutrition on angiogenic factor expression and vascularity in the bovine placentome. Journal of Animal Science 85:2464-2472. https://doi.org/10.2527/jas.2006-805
    » https://doi.org/10.2527/jas.2006-805
  • Wang, X.; Gu, Z. and Jiang, H. 2013. MicroRNAs in farm animals. Animal 7:1567-1575. https://doi.org/10.1017/S1751731113001183
    » https://doi.org/10.1017/S1751731113001183
  • Wang, B.; Yang, Q.; Harris, C. L.; Nelson, M. L.; Busboom, J. R.; Zhu, M. and Du, M. 2016. Nutrigenomic regulation of adipose tissue development — role of retinoic acid: A review. Meat Science 120:100-106. https://doi.org/10.1016/j.meatsci.2016.04.003
    » https://doi.org/10.1016/j.meatsci.2016.04.003
  • Webb, M. J.; Block, J. J.; Funston, R. N.; Underwood, K. R.; Legako, J. F.; Harty, A. A.; Salverson, R. R.; Olson, K. C. and Blair, A. D. 2019. Influence of maternal protein restriction in primiparous heifers during mid- and/or late-gestation on meat quality and fatty acid profile of progeny. Meat Science 152:31-37. https://doi.org/10.1016/j.meatsci.2019.02.006
    » https://doi.org/10.1016/j.meatsci.2019.02.006
  • Wu, G.; Bazer, F. W.; Wallace, J. M. and Spencer, T. E. 2006. Board-invited review: Intrauterine growth retardation: Implications for the animal sciences. Journal of Animal Science 84:2316-2337. https://doi.org/10.2527/jas.2006-156
    » https://doi.org/10.2527/jas.2006-156
  • Yang, Q. Y.; Liang, J. F.; Rogers, C. J.; Zhao, J. X.; Zhu, M. J. and Du, M. 2013. Maternal obesity induces epigenetic modifications to facilitate Zfp423 expression and enhance adipogenic differentiation in fetal mice. Diabetes 62:3727-3735. https://doi.org/10.2337/db13-0433
    » https://doi.org/10.2337/db13-0433
  • Zhang, H.; Sun, L. W.; Wang, Z. Y.; Deng, M. T.; Zhang, G. M.; Guo, R. H.; Ma, T. W. and Wang, F. 2016. Dietary N-carbamylglutamate and rumen-protected L-arginine supplementation ameliorate fetal growth restriction in undernourished ewes. Journal of Animal Science 94:2072-2085. https://doi.org/10.2527/jas.2015-9587
    » https://doi.org/10.2527/jas.2015-9587

Publication Dates

  • Publication in this collection
    04 Nov 2022
  • Date of issue
    2022

History

  • Received
    14 Apr 2022
  • Accepted
    23 June 2022
Sociedade Brasileira de Zootecnia Universidade Federal de Viçosa / Departamento de Zootecnia, 36570-900 Viçosa MG Brazil, Tel.: +55 31 3612-4602, +55 31 3612-4612 - Viçosa - MG - Brazil
E-mail: rbz@sbz.org.br