Skip to main content
Top
Published in: European Journal of Nutrition 4/2016

01-06-2016 | Original Contribution

Moderately increased maternal dietary energy intake delays foetal skeletal muscle differentiation and maturity in pigs

Authors: Tiande Zou, Dongting He, Bing Yu, Jie Yu, Xiangbing Mao, Ping Zheng, Jun He, Zhiqing Huang, Yan Shu, Yue Liu, Daiwen Chen

Published in: European Journal of Nutrition | Issue 4/2016

Login to get access

Abstract

Objectives

This study aimed to evaluate the effects of moderately increased maternal dietary energy intake during gestation on foetal skeletal muscle development and metabolism with pig as a model.

Methods

Twelve primiparous purebred Large White sows (initial body weight 135.5 ± 1.6 kg) were allocated to one of two energy intake treatments: normal-energy-intake group (Con, 30.96 MJ DE/day) as recommended by the National Research Council (NRC; 2012) and high-energy-intake group (HE, 34.15 MJ DE/day). The nutritional treatments were introduced from mating to day 90 of gestation. On day 90 of gestation, foetuses were examined by morphological, biochemical and molecular analysis of the longissimus muscle. Umbilical vein serum hormones were measured.

Results

Sow body weight was increased in HE group compared with Con group (P < 0.05), whereas foetal myofibre density was decreased (P < 0.05). Meanwhile, protein concentration, creatine kinase and lactate dehydrogenase activities and umbilical vein serum triiodothyronine (T3) concentration were decreased in HE foetuses (P < 0.05). Maternal HE diets decreased the mRNA abundance of muscle growth-related genes, myosin heavy-chain (MYH/MyHC) genes (MYH2 and MYH1) and insulin-like growth factor 1 and insulin growth factor-binding protein 5 (P < 0.05). Furthermore, the protein expressions of myogenic differentiation factor 1, myogenin and fast-MyHC isoforms were reduced in HE foetuses (P < 0.05).

Conclusion

Our results suggest that moderately increased maternal dietary energy intake delays the differentiation and maturation in skeletal muscle of the foetus on day 90 of gestation.
Literature
1.
go back to reference Godfrey KM (1998) Maternal regulation of fetal development and health in adult life. Eur J Obstet Gyn Reprod Biol 78:141–150CrossRef Godfrey KM (1998) Maternal regulation of fetal development and health in adult life. Eur J Obstet Gyn Reprod Biol 78:141–150CrossRef
2.
go back to reference Wu G, Bazer FW, Cudd TA et al (2004) Maternal nutrition and fetal development. J Nutr 134:2169–2172 Wu G, Bazer FW, Cudd TA et al (2004) Maternal nutrition and fetal development. J Nutr 134:2169–2172
3.
go back to reference Metges CC, Görs S, Lang IS et al (2014) Low and high dietary protein: carbohydrate ratios during pregnancy affect materno-fetal glucose metabolism in pigs. J Nutr 144:155–163CrossRef Metges CC, Görs S, Lang IS et al (2014) Low and high dietary protein: carbohydrate ratios during pregnancy affect materno-fetal glucose metabolism in pigs. J Nutr 144:155–163CrossRef
4.
go back to reference Drake AJ, Reynolds RM (2010) Impact of maternal obesity on offspring obesity and cardiometabolic disease risk. Reproduction 140:387–398CrossRef Drake AJ, Reynolds RM (2010) Impact of maternal obesity on offspring obesity and cardiometabolic disease risk. Reproduction 140:387–398CrossRef
5.
go back to reference Du M, Yan X, Tong JF et al (2010) Maternal obesity, inflammation, and fetal skeletal muscle development. Biol Reprod 82:4–12CrossRef Du M, Yan X, Tong JF et al (2010) Maternal obesity, inflammation, and fetal skeletal muscle development. Biol Reprod 82:4–12CrossRef
6.
go back to reference Harding JE (2001) The nutritional basis of the fetal origins of adult disease. Int J Epidemiol 30:15–23CrossRef Harding JE (2001) The nutritional basis of the fetal origins of adult disease. Int J Epidemiol 30:15–23CrossRef
7.
go back to reference Vernarelli JA, Mitchell DC, Rolls BJ et al (2014) Dietary energy density is associated with obesity and other biomarkers of chronic disease in US adults. Eur J Nutr 54:59–65CrossRef Vernarelli JA, Mitchell DC, Rolls BJ et al (2014) Dietary energy density is associated with obesity and other biomarkers of chronic disease in US adults. Eur J Nutr 54:59–65CrossRef
8.
go back to reference Flegal KM, Carroll MD, Kit BK et al (2012) Prevalence of obesity and trends in the distribution of body mass index among US adults, 1999–2010. JAMA 307:491–497CrossRef Flegal KM, Carroll MD, Kit BK et al (2012) Prevalence of obesity and trends in the distribution of body mass index among US adults, 1999–2010. JAMA 307:491–497CrossRef
9.
go back to reference Bruce KD, Hanson MA (2010) The developmental origins, mechanisms, and implications of metabolic syndrome. J Nutr 140:648–652CrossRef Bruce KD, Hanson MA (2010) The developmental origins, mechanisms, and implications of metabolic syndrome. J Nutr 140:648–652CrossRef
10.
go back to reference Heerwagen MJ, Miller MR, Barbour LA et al (2010) Maternal obesity and fetal metabolic programming: a fertile epigenetic soil. Am J Physiol-Regul Integr Comp Physiol 299:R711–R722CrossRef Heerwagen MJ, Miller MR, Barbour LA et al (2010) Maternal obesity and fetal metabolic programming: a fertile epigenetic soil. Am J Physiol-Regul Integr Comp Physiol 299:R711–R722CrossRef
11.
go back to reference Rkhzay-Jaf J, O’Dowd JF, Stocker CJ (2012) Maternal obesity and the fetal origins of the metabolic syndrome. Curr Cardiovasc Risk Rep 6:487–495CrossRef Rkhzay-Jaf J, O’Dowd JF, Stocker CJ (2012) Maternal obesity and the fetal origins of the metabolic syndrome. Curr Cardiovasc Risk Rep 6:487–495CrossRef
12.
go back to reference Schlabritz-Loutsevitch N, Ballesteros B, Dudley C et al (2007) Moderate maternal nutrient restriction, but not glucocorticoid administration, leads to placental morphological changes in the baboon (Papio sp.). Placenta 28:783–793CrossRef Schlabritz-Loutsevitch N, Ballesteros B, Dudley C et al (2007) Moderate maternal nutrient restriction, but not glucocorticoid administration, leads to placental morphological changes in the baboon (Papio sp.). Placenta 28:783–793CrossRef
13.
go back to reference Samuelsson A-M, Matthews PA, Argenton M et al (2008) Diet-induced obesity in female mice leads to offspring hyperphagia, adiposity, hypertension, and insulin resistance a novel murine model of developmental programming. Hypertension 51:383–392CrossRef Samuelsson A-M, Matthews PA, Argenton M et al (2008) Diet-induced obesity in female mice leads to offspring hyperphagia, adiposity, hypertension, and insulin resistance a novel murine model of developmental programming. Hypertension 51:383–392CrossRef
14.
go back to reference Zhu M, Du M, Nathanielsz P et al (2010) Maternal obesity up-regulates inflammatory signaling pathways and enhances cytokine expression in the mid-gestation sheep placenta. Placenta 31:387–391CrossRef Zhu M, Du M, Nathanielsz P et al (2010) Maternal obesity up-regulates inflammatory signaling pathways and enhances cytokine expression in the mid-gestation sheep placenta. Placenta 31:387–391CrossRef
15.
go back to reference Yan X, Huang Y, Zhao J-X et al (2011) Maternal obesity-impaired insulin signaling in sheep and induced lipid accumulation and fibrosis in skeletal muscle of offspring. Bio Reprod 85:172–178CrossRef Yan X, Huang Y, Zhao J-X et al (2011) Maternal obesity-impaired insulin signaling in sheep and induced lipid accumulation and fibrosis in skeletal muscle of offspring. Bio Reprod 85:172–178CrossRef
16.
go back to reference Rooyackers OE, Nair KS (1997) Hormonal regulation of human muscle protein metabolism. Annu Rev Nutr 17:457–485CrossRef Rooyackers OE, Nair KS (1997) Hormonal regulation of human muscle protein metabolism. Annu Rev Nutr 17:457–485CrossRef
17.
go back to reference Rehfeldt C, Stickland NC, Fiedler I et al (1999) Environmental and genetic factors as sources of variation in skeletal muscle fibre number. Basic Appl Myol 9:235–254 Rehfeldt C, Stickland NC, Fiedler I et al (1999) Environmental and genetic factors as sources of variation in skeletal muscle fibre number. Basic Appl Myol 9:235–254
18.
go back to reference Nissen P, Danielsen V, Jorgensen P et al (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. J Anim Sci 81:3018–3027 Nissen P, Danielsen V, Jorgensen P et al (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. J Anim Sci 81:3018–3027
19.
go back to reference Du M, Zhao JX, Yan X et al (2011) Fetal muscle development, mesenchymal multipotent cell differentiation, and associated signaling pathways. J Anim Sci 89:583–590CrossRef Du M, Zhao JX, Yan X et al (2011) Fetal muscle development, mesenchymal multipotent cell differentiation, and associated signaling pathways. J Anim Sci 89:583–590CrossRef
20.
go back to reference Miller E, Ullrey D (1987) The pig as a model for human nutrition. Annu Rev Nutr 7:361–382CrossRef Miller E, Ullrey D (1987) The pig as a model for human nutrition. Annu Rev Nutr 7:361–382CrossRef
21.
go back to reference Humphray SJ, Scott CE, Clark R et al (2007) A high utility integrated map of the pig genome. Genome Biol 8:R139CrossRef Humphray SJ, Scott CE, Clark R et al (2007) A high utility integrated map of the pig genome. Genome Biol 8:R139CrossRef
22.
go back to reference Spurlock ME, Gabler NK (2008) The development of porcine models of obesity and the metabolic syndrome. J Nutr 138:397–402 Spurlock ME, Gabler NK (2008) The development of porcine models of obesity and the metabolic syndrome. J Nutr 138:397–402
23.
go back to reference Wigmore P, Stickland N (1983) Muscle development in large and small pig fetuses. J Anat 137:235 Wigmore P, Stickland N (1983) Muscle development in large and small pig fetuses. J Anat 137:235
24.
go back to reference Dwyer C, Stickland N, Fletcher J (1994) The influence of maternal nutrition on muscle fiber number development in the porcine fetus and on subsequent postnatal growth. J Anim Sci 72:911–917 Dwyer C, Stickland N, Fletcher J (1994) The influence of maternal nutrition on muscle fiber number development in the porcine fetus and on subsequent postnatal growth. J Anim Sci 72:911–917
25.
go back to reference Rehfeldt C, Walther K (1997) A combined assay for DNA, protein, and the incorporation of [3H] label in cultured muscle cells. Anal Biochem 251:294–297CrossRef Rehfeldt C, Walther K (1997) A combined assay for DNA, protein, and the incorporation of [3H] label in cultured muscle cells. Anal Biochem 251:294–297CrossRef
26.
go back to reference Smith P, Krohn RI, Hermanson G et al (1985) Measurement of protein using bicinchoninic acid. Anal Biochem 150:76–85CrossRef Smith P, Krohn RI, Hermanson G et al (1985) Measurement of protein using bicinchoninic acid. Anal Biochem 150:76–85CrossRef
27.
go back to reference Livak KJ, Schmittgen TD (2001) Analysis of relative gene expression data using real-time quantitative PCR and the 2−ΔΔCT method. Methods 25:402–408CrossRef Livak KJ, Schmittgen TD (2001) Analysis of relative gene expression data using real-time quantitative PCR and the 2−ΔΔCT method. Methods 25:402–408CrossRef
28.
go back to reference Zhu MJ, Ford SP, Means WJ et al (2006) Maternal nutrient restriction affects properties of skeletal muscle in offspring. J Physiol 575:241–250CrossRef Zhu MJ, Ford SP, Means WJ et al (2006) Maternal nutrient restriction affects properties of skeletal muscle in offspring. J Physiol 575:241–250CrossRef
29.
go back to reference Bee G (2004) Effect of early gestation feeding, birth weight, and gender of progeny on muscle fiber characteristics of pigs at slaughter. J Anim Sci 82:826–836 Bee G (2004) Effect of early gestation feeding, birth weight, and gender of progeny on muscle fiber characteristics of pigs at slaughter. J Anim Sci 82:826–836
30.
go back to reference Gatford KL, Ekert JE, Blackmore K et al (2003) Variable maternal nutrition and growth hormone treatment in the second quarter of pregnancy in pigs alter semitendinosus muscle in adolescent progeny. Brit J Nutr 90:283–293CrossRef Gatford KL, Ekert JE, Blackmore K et al (2003) Variable maternal nutrition and growth hormone treatment in the second quarter of pregnancy in pigs alter semitendinosus muscle in adolescent progeny. Brit J Nutr 90:283–293CrossRef
31.
go back to reference Cerisuelo A, Baucells M, Gasa J et al (2009) Increased sow nutrition during midgestation affects muscle fiber development and meat quality, with no consequences on growth performance. J Anim Sci 87:729–739CrossRef Cerisuelo A, Baucells M, Gasa J et al (2009) Increased sow nutrition during midgestation affects muscle fiber development and meat quality, with no consequences on growth performance. J Anim Sci 87:729–739CrossRef
32.
go back to reference Heyer A, Andersson H, Lindberg J et al (2004) Effect of extra maternal feed supply in early gestation on sow and piglet performance and production and meat quality of growing/finishing pigs. Acta Agr Scand A-Anim Sci 54:44–55 Heyer A, Andersson H, Lindberg J et al (2004) Effect of extra maternal feed supply in early gestation on sow and piglet performance and production and meat quality of growing/finishing pigs. Acta Agr Scand A-Anim Sci 54:44–55
33.
go back to reference Musser R, Davis D, Tokach M et al (2006) Effects of high feed intake during early gestation on sow performance and offspring growth and carcass characteristics. Anim Feed Sci Tech 127:187–199CrossRef Musser R, Davis D, Tokach M et al (2006) Effects of high feed intake during early gestation on sow performance and offspring growth and carcass characteristics. Anim Feed Sci Tech 127:187–199CrossRef
34.
go back to reference Hines E, Coffey J, Starkey C et al (2013) Improvement of maternal vitamin D status with 25-hydroxycholecalciferol positively impacts porcine fetal skeletal muscle development and myoblast activity. J Anim Sci 91:4116–4122CrossRef Hines E, Coffey J, Starkey C et al (2013) Improvement of maternal vitamin D status with 25-hydroxycholecalciferol positively impacts porcine fetal skeletal muscle development and myoblast activity. J Anim Sci 91:4116–4122CrossRef
35.
go back to reference Bérard J, Bee G (2010) Effects of dietary l-arginine supplementation to gilts during early gestation on foetal survival, growth and myofiber formation. Animal 4:1680–1687CrossRef Bérard J, Bee G (2010) Effects of dietary l-arginine supplementation to gilts during early gestation on foetal survival, growth and myofiber formation. Animal 4:1680–1687CrossRef
36.
go back to reference Rehfeldt C, Kuhn G, Vanselow J et al (2001) Maternal treatment with somatotropin during early gestation affects basic events of myogenesis in pigs. Cell Tissue Res 306:429–440CrossRef Rehfeldt C, Kuhn G, Vanselow J et al (2001) Maternal treatment with somatotropin during early gestation affects basic events of myogenesis in pigs. Cell Tissue Res 306:429–440CrossRef
37.
go back to reference Rehfeldt C, Nissen PM, Kuhn G et al (2004) Effects of maternal nutrition and porcine growth hormone (pGH) treatment during gestation on endocrine and metabolic factors in sows, fetuses and pigs, skeletal muscle development, and postnatal growth. Domest Anim Endocrin 27:267–285CrossRef Rehfeldt C, Nissen PM, Kuhn G et al (2004) Effects of maternal nutrition and porcine growth hormone (pGH) treatment during gestation on endocrine and metabolic factors in sows, fetuses and pigs, skeletal muscle development, and postnatal growth. Domest Anim Endocrin 27:267–285CrossRef
38.
go back to reference Tong JF, Yan X, Zhu MJ et al (2009) Maternal obesity downregulates myogenesis and β-catenin signaling in fetal skeletal muscle. Am J Physiol Endocrinol Metab 296:E917–E924CrossRef Tong JF, Yan X, Zhu MJ et al (2009) Maternal obesity downregulates myogenesis and β-catenin signaling in fetal skeletal muscle. Am J Physiol Endocrinol Metab 296:E917–E924CrossRef
39.
go back to reference Simar D, Chen H, Lambert K et al (2012) Interaction between maternal obesity and post-natal over-nutrition on skeletal muscle metabolism. Nutr Metab Cardiovas 22:269–276CrossRef Simar D, Chen H, Lambert K et al (2012) Interaction between maternal obesity and post-natal over-nutrition on skeletal muscle metabolism. Nutr Metab Cardiovas 22:269–276CrossRef
40.
go back to reference Lefaucheur L, Ecolan P, Barzic Y-M et al (2003) Early postnatal food intake alters myofiber maturation in pig skeletal muscle. J Nutr 133:140–147 Lefaucheur L, Ecolan P, Barzic Y-M et al (2003) Early postnatal food intake alters myofiber maturation in pig skeletal muscle. J Nutr 133:140–147
41.
go back to reference Kalbe C, Bérard J, Porm M et al (2013) Maternal l-arginine supplementation during early gestation affects foetal skeletal myogenesis in pigs. Livest Sci 157:322–329CrossRef Kalbe C, Bérard J, Porm M et al (2013) Maternal l-arginine supplementation during early gestation affects foetal skeletal myogenesis in pigs. Livest Sci 157:322–329CrossRef
42.
go back to reference Schiaffino S, Reggiani C (2011) Fiber types in mammalian skeletal muscles. Physiol Rev 91:1447–1531CrossRef Schiaffino S, Reggiani C (2011) Fiber types in mammalian skeletal muscles. Physiol Rev 91:1447–1531CrossRef
43.
go back to reference Lefaucheur L, Edom F, Ecolan P et al (1995) Pattern of muscle fiber type formation in the pig. Dev Dynam 203:27–41CrossRef Lefaucheur L, Edom F, Ecolan P et al (1995) Pattern of muscle fiber type formation in the pig. Dev Dynam 203:27–41CrossRef
44.
go back to reference Da Costa N, McGillivray C, Chang KC (2003) Postnatal myosin heavy chain isoforms in prenatal porcine skeletal muscles: insights into temporal regulation. Anat Rec Part A 273:731–740CrossRef Da Costa N, McGillivray C, Chang KC (2003) Postnatal myosin heavy chain isoforms in prenatal porcine skeletal muscles: insights into temporal regulation. Anat Rec Part A 273:731–740CrossRef
45.
go back to reference Florini JR, Ewton DZ, Magri KA (1991) Hormones, growth factors, and myogenic differentiation. Annu Rev Physiol 53:201–216CrossRef Florini JR, Ewton DZ, Magri KA (1991) Hormones, growth factors, and myogenic differentiation. Annu Rev Physiol 53:201–216CrossRef
46.
go back to reference Finkelstein DI, Andrianakis P, Luff AR et al (1991) Effects of thyroidectomy on development of skeletal muscle in fetal sheep. Am J Physiol Regul Integr Comp Physiol 261:R1300–R1306 Finkelstein DI, Andrianakis P, Luff AR et al (1991) Effects of thyroidectomy on development of skeletal muscle in fetal sheep. Am J Physiol Regul Integr Comp Physiol 261:R1300–R1306
47.
go back to reference Zhang D, Wang X, Li Y et al (2014) Thyroid hormone regulates muscle fiber type conversion via miR-133a1. J Cell Biol 207:753–766CrossRef Zhang D, Wang X, Li Y et al (2014) Thyroid hormone regulates muscle fiber type conversion via miR-133a1. J Cell Biol 207:753–766CrossRef
48.
go back to reference Charge SB, Rudnicki MA (2004) Cellular and molecular regulation of muscle regeneration. Physiol Rev 84:209–238CrossRef Charge SB, Rudnicki MA (2004) Cellular and molecular regulation of muscle regeneration. Physiol Rev 84:209–238CrossRef
49.
go back to reference Pownall ME, Gustafsson MK, Emerson CP Jr (2002) Myogenic regulatory factors and the specification of muscle progenitors in vertebrate embryos. Annu Rev Cell Dev Biol 18:747–783CrossRef Pownall ME, Gustafsson MK, Emerson CP Jr (2002) Myogenic regulatory factors and the specification of muscle progenitors in vertebrate embryos. Annu Rev Cell Dev Biol 18:747–783CrossRef
50.
go back to reference Relaix F, Rocancourt D, Mansouri A et al (2005) A Pax3/Pax7-dependent population of skeletal muscle progenitor cells. Nature 435:948–953CrossRef Relaix F, Rocancourt D, Mansouri A et al (2005) A Pax3/Pax7-dependent population of skeletal muscle progenitor cells. Nature 435:948–953CrossRef
51.
go back to reference Sabourin LA, Rudnicki MA (2000) The molecular regulation of myogenesis. Clin Genet 57:16–25CrossRef Sabourin LA, Rudnicki MA (2000) The molecular regulation of myogenesis. Clin Genet 57:16–25CrossRef
52.
go back to reference Zhao J, Schmieg FI, Simmons DT et al (1994) Mouse p53 represses the rat brain creatine kinase gene but activates the rat muscle creatine kinase gene. Mol Cell Biol 14:8483–8492CrossRef Zhao J, Schmieg FI, Simmons DT et al (1994) Mouse p53 represses the rat brain creatine kinase gene but activates the rat muscle creatine kinase gene. Mol Cell Biol 14:8483–8492CrossRef
53.
go back to reference Collett-Solberg PF, Cohen P (2000) Genetics, chemistry, and function of the IGF/IGFBP system. Endocrine 12:121–136CrossRef Collett-Solberg PF, Cohen P (2000) Genetics, chemistry, and function of the IGF/IGFBP system. Endocrine 12:121–136CrossRef
54.
go back to reference Duan C, Ren H, Gao S (2010) Insulin-like growth factors (IGFs), IGF receptors, and IGF-binding proteins: roles in skeletal muscle growth and differentiation. Gen Comp Endocr 167:344–351CrossRef Duan C, Ren H, Gao S (2010) Insulin-like growth factors (IGFs), IGF receptors, and IGF-binding proteins: roles in skeletal muscle growth and differentiation. Gen Comp Endocr 167:344–351CrossRef
55.
go back to reference Ren H, Yin P, Duan C (2008) IGFBP-5 regulates muscle cell differentiation by binding to IGF-II and switching on the IGF-II auto-regulation loop. J Cell Biol 182:979–991CrossRef Ren H, Yin P, Duan C (2008) IGFBP-5 regulates muscle cell differentiation by binding to IGF-II and switching on the IGF-II auto-regulation loop. J Cell Biol 182:979–991CrossRef
56.
go back to reference Maak S, Wicke M, Swalve HH (2005) Analysis of gene expression in specific muscles of swine and turkey. Arch Tierz Dummerstorf 48:135–140 Maak S, Wicke M, Swalve HH (2005) Analysis of gene expression in specific muscles of swine and turkey. Arch Tierz Dummerstorf 48:135–140
Metadata
Title
Moderately increased maternal dietary energy intake delays foetal skeletal muscle differentiation and maturity in pigs
Authors
Tiande Zou
Dongting He
Bing Yu
Jie Yu
Xiangbing Mao
Ping Zheng
Jun He
Zhiqing Huang
Yan Shu
Yue Liu
Daiwen Chen
Publication date
01-06-2016
Publisher
Springer Berlin Heidelberg
Published in
European Journal of Nutrition / Issue 4/2016
Print ISSN: 1436-6207
Electronic ISSN: 1436-6215
DOI
https://doi.org/10.1007/s00394-015-0996-9

Other articles of this Issue 4/2016

European Journal of Nutrition 4/2016 Go to the issue
Live Webinar | 27-06-2024 | 18:00 (CEST)

Keynote webinar | Spotlight on medication adherence

Live: Thursday 27th June 2024, 18:00-19:30 (CEST)

WHO estimates that half of all patients worldwide are non-adherent to their prescribed medication. The consequences of poor adherence can be catastrophic, on both the individual and population level.

Join our expert panel to discover why you need to understand the drivers of non-adherence in your patients, and how you can optimize medication adherence in your clinics to drastically improve patient outcomes.

Prof. Kevin Dolgin
Prof. Florian Limbourg
Prof. Anoop Chauhan
Developed by: Springer Medicine
Obesity Clinical Trial Summary

At a glance: The STEP trials

A round-up of the STEP phase 3 clinical trials evaluating semaglutide for weight loss in people with overweight or obesity.

Developed by: Springer Medicine

Highlights from the ACC 2024 Congress

Year in Review: Pediatric cardiology

Watch Dr. Anne Marie Valente present the last year's highlights in pediatric and congenital heart disease in the official ACC.24 Year in Review session.

Year in Review: Pulmonary vascular disease

The last year's highlights in pulmonary vascular disease are presented by Dr. Jane Leopold in this official video from ACC.24.

Year in Review: Valvular heart disease

Watch Prof. William Zoghbi present the last year's highlights in valvular heart disease from the official ACC.24 Year in Review session.

Year in Review: Heart failure and cardiomyopathies

Watch this official video from ACC.24. Dr. Biykem Bozkurt discusses last year's major advances in heart failure and cardiomyopathies.