Your browser doesn't support javascript.
loading
Cerebrum, liver, and muscle regulatory networks uncover maternal nutrition effects in developmental programming of beef cattle during early pregnancy.
Diniz, Wellison J S; Crouse, Matthew S; Cushman, Robert A; McLean, Kyle J; Caton, Joel S; Dahlen, Carl R; Reynolds, Lawrence P; Ward, Alison K.
Afiliación
  • Diniz WJS; Department of Animal Sciences, Center for Nutrition and Pregnancy, North Dakota State University, Fargo, ND, USA. w.dasilvadiniz@ndsu.edu.
  • Crouse MS; USDA, ARS, U.S. Meat Animal Research Center, Clay Center, NE, USA.
  • Cushman RA; USDA, ARS, U.S. Meat Animal Research Center, Clay Center, NE, USA.
  • McLean KJ; Department of Animal Science, University of Tennessee, Knoxville, TN, USA.
  • Caton JS; Department of Animal Sciences, Center for Nutrition and Pregnancy, North Dakota State University, Fargo, ND, USA.
  • Dahlen CR; Department of Animal Sciences, Center for Nutrition and Pregnancy, North Dakota State University, Fargo, ND, USA.
  • Reynolds LP; Department of Animal Sciences, Center for Nutrition and Pregnancy, North Dakota State University, Fargo, ND, USA.
  • Ward AK; Department of Animal Sciences, Center for Nutrition and Pregnancy, North Dakota State University, Fargo, ND, USA.
Sci Rep ; 11(1): 2771, 2021 02 02.
Article en En | MEDLINE | ID: mdl-33531552
ABSTRACT
The molecular basis underlying fetal programming in response to maternal nutrition remains unclear. Herein, we investigated the regulatory relationships between genes in fetal cerebrum, liver, and muscle tissues to shed light on the putative mechanisms that underlie the effects of early maternal nutrient restriction on bovine developmental programming. To this end, cerebrum, liver, and muscle gene expression were measured with RNA-Seq in 14 fetuses collected on day 50 of gestation from dams fed a diet initiated at breeding to either achieve 60% (RES, n = 7) or 100% (CON, n = 7) of energy requirements. To build a tissue-to-tissue gene network, we prioritized tissue-specific genes, transcription factors, and differentially expressed genes. Furthermore, we built condition-specific networks to identify differentially co-expressed or connected genes. Nutrient restriction led to differential tissue regulation between the treatments. Myogenic factors differentially regulated by ZBTB33 and ZNF131 may negatively affect myogenesis. Additionally, nutrient-sensing pathways, such as mTOR and PI3K/Akt, were affected by gene expression changes in response to nutrient restriction. By unveiling the network properties, we identified major regulators driving gene expression. However, further research is still needed to determine the impact of early maternal nutrition and strategic supplementation on pre- and post-natal performance.
Asunto(s)

Texto completo: 1 Colección: 01-internacional Base de datos: MEDLINE Asunto principal: Fenómenos Fisiologicos de la Nutrición Prenatal / Desarrollo Fetal / Feto / Fenómenos Fisiológicos Nutricionales de los Animales Tipo de estudio: Prognostic_studies Límite: Animals / Pregnancy Idioma: En Revista: Sci Rep Año: 2021 Tipo del documento: Article País de afiliación: Estados Unidos

Texto completo: 1 Colección: 01-internacional Base de datos: MEDLINE Asunto principal: Fenómenos Fisiologicos de la Nutrición Prenatal / Desarrollo Fetal / Feto / Fenómenos Fisiológicos Nutricionales de los Animales Tipo de estudio: Prognostic_studies Límite: Animals / Pregnancy Idioma: En Revista: Sci Rep Año: 2021 Tipo del documento: Article País de afiliación: Estados Unidos