Sean W Limesand

Sean W Limesand

Professor, Animal and Comparative Biomedical Sciences
Professor, Physiological Sciences - GIDP
Director, Agriculture Research Complex
Professor, Obstetrics and Gynecology
Chair, Institutional Animal Care-USE Committee
Professor, BIO5 Institute
Department Affiliations
Contact
(520) 626-8903

Work Summary

Our current research program use an integrative approach at the whole animal, isolated organ, cellular and molecular levels to investigate developmental adaptations in pancreatic β-cells and insulin sensitivity that result from early life risk factors, such as intrauterine growth restriction, and increase risk of glucose intolerance and Diabetes in later life.

Research Interest

Sean W. Limesand, PhD, is an Associate Professor in the School of Animal and Comparative Biomedical Sciences at the University of Arizona in the College of Agriculture and Life Sciences. He is also a member of the UA’s BIO5 Institute and Department of Obstetrics and Gynecology. Dr. Limesand is nationally and internationally recognized for his work studying fetal endocrinology and metabolism in pregnancy and in pregnancies compromised by pathology such as intrauterine growth restriction and diabetes. His research is focused on defining developmental consequences resulting from a compromised intrauterine environment. Specifically, he is focused on fetal adaptations in insulin secretion and action that when altered in utero create lifelong metabolic complications. Dr. Limesand has lead the charge on prenatal origins of –cell dysfunction as the Principal Investigator for a number of federal and foundation grant awards and published more than 40 peer-reviewed articles on topics related to this research. Keywords: Diabetes, Pregnancy, Perinatal Biology

Publications

Yates, D. T., Green, A. S., & Limesand, S. W. (2011). Catecholamines mediate multiple fetal adaptations during placental insufficiency that contribute to intrauterine growth restriction: lessons from hyperthermic sheep.. Journal of pregnancy, 2011, 740408-.

PMID: 21773031;PMCID: PMC3135098;Abstract:

Placental insufficiency (PI) prevents adequate delivery of nutrients to the developing fetus and creates a chronic state of hypoxemia and hypoglycemia. In response, the malnourished fetus develops a series of stress hormone-mediated metabolic adaptations to preserve glucose for vital tissues at the expense of somatic growth. Catecholamines suppress insulin secretion to promote glucose sparing for insulin-independent tissues (brain, nerves) over insulin-dependent tissues (skeletal muscle, liver, and adipose). Likewise, premature induction of hepatic gluconeogenesis helps maintain fetal glucose and appears to be stimulated by both norepinephrine and glucagon. Reduced glucose oxidation rate in PI fetuses creates a surplus of glycolysis-derived lactate that serves as substrate for hepatic gluconeogenesis. These adrenergically influenced adaptive responses promote in utero survival but also cause asymmetric intrauterine growth restriction and small-for-gestational-age infants that are at greater risk for serious metabolic disorders throughout postnatal life, including obesity and type II diabetes.

Penrod, L. V., Allen, R. E., Turner, J. L., Limesand, S. W., & Arns, M. J. (2013). Effects of oxytocin, lipopolysaccharide (LPS), and polyunsaturated fatty acids on prostaglandin secretion and gene expression in equine endometrial explant cultures. Domestic animal endocrinology, 44(1).

Increased secretion of prostaglandin F(2)α (PGF(2)α) within the uterus because of uterine inflammation can cause luteolysis and result in early embryonic loss. Supplementation with polyunsaturated fatty acids (PUFAs) has been shown to influence PG production in many species, although the effects on the mare remain unknown. The present study aimed to determine fatty acid uptake in equine endometrial explants and evaluate their influence on PG secretion and expression of enzymes involved in PG synthesis in vitro. Equine endometrial explants were treated with 100 μM arachidonic acid, eicosapentaenoic acid, or docosahexaenoic acid and then challenged with oxytocin (250 nM) or lipopolysaccharide (LPS; 1 μg/mL). Production of PGF(2)α and PG E(2) (PGE(2)) was measured, and mRNA expression of enzymes involved in PG synthesis was determined with quantitative real-time PCR. Media concentrations of PGF(2)α and PGE(2) were higher (P 0.0001) from endometrial explants challenged with oxytocin or LPS compared with controls despite which fatty acid was added. Only DHA lowered (P 0.0001) media concentrations of PGF(2)α and PGE(2) from explants. Endometrial explants stimulated with oxytocin had increased expression of PG-endoperoxide synthase 1 (PTGS1; P 0.02), PG-endoperoxide synthase 2 (PTGS2; P 0.001), PG F(2)α synthase (PGFS; P 0.01), PG E(2) synthase (PGES; P 0.01), and phospholipase A(2) (PLA(2); P 0.005) compared with controls and regardless of fatty acid treatment; whereas stimulation with LPS increased expression of PTGS2 (P 0.004), PGFS (P 0.03), PGES (P 0.01), and PLA(2) (P 0.01) compared with controls and regardless of fatty acid treatment. Treatment with PUFAs, specifically DHA, can influence PG secretion in vitro through mechanisms other than enzyme expression.

Chen, X., Green, A. S., Macko, A. R., Yates, D. T., Kelly, A. C., & Limesand, S. W. (2013). Enhanced Insulin Responsiveness and Islet Adrenergic Desensitization after Discontinuing Chronic Norepinephrine Suppression in Fetal Sheep. American journal of physiology. Endocrinology and metabolism.

Intrauterine growth restricted (IUGR) fetuses experience prolonged hypoxemia, hypoglycemia, and elevated norepinephrine (NE) concentrations, resulting in hypoinsulinemia and β-cell dysfunction. We previously showed that acute adrenergic blockade revealed enhanced insulin secretion responsiveness in the IUGR fetus. To determine if chronic exposure to NE alone enhances β-cell responsiveness afterward, we continuously infused NE into fetal sheep for 7 days and, after terminating the infusion, evaluated glucose-stimulated insulin secretion (GSIS) and glucose-potentiated arginine-induced insulin secretion (GPAIS). During treatment, NE-infused fetuses had greater (P 0.05) plasma NE concentrations, and exhibited hyperglycemia (P 0.01) and hypoinsulinemia (P 0.01) compared to controls. GSIS during the NE infusion also was reduced (P 0.05) compared to pre-treatment values. GSIS and GPAIS were ~4-fold greater (P 0.01) in NE fetuses three hours after discontinuing the 7 days NE infusion compared to age-matched controls or pre-treatment GSIS and GPAIS values of NE fetuses. In isolated pancreatic islets from NE fetuses, mRNA concentrations of adrenergic receptor isoforms (α1D, α2A, α2C, and β1), G-protein subunit αi-2, and uncoupling protein 2 were lower (P 0.05) compared to controls, but β-cell regulatory genes were not different. Our findings indicate that chronic exposure to elevated NE persistently suppresses insulin secretion. After removal, NE fetuses demonstrated a compensatory enhancement in insulin secretion that was associated with adrenergic desensitization and greater stimulus-secretion coupling in pancreatic islets.

Wallace, J. M., Regnault, T. R., Limesand, S. W., Hay Jr., W. W., & Anthony, R. V. (2005). Investigating the causes of low birth weight in contrasting ovine paradigms. Journal of Physiology, 565(1), 19-26.

PMID: 15774527;PMCID: PMC1464509;Abstract:

Intrauterine growth restriction (IUGR) still accounts for a large incidence of infant mortality and morbity worldwide. Many of the circulatory and transport properties of the sheep placenta are similar to those of the human placenta and as such, the pregnant sheep offers an excellent model in which to study the development of IUGR. Two natural models of ovine IUGR are those of hyperthermic exposure during pregnancy, and adolescent overfeeding, also during pregnancy. Both models yield significantly reduced placental weights and an asymmetrically growth-restricted fetus, and display altered maternal hormone concentrations, indicative of an impaired trophoblast capacity. Additionally, impaired placental angiogenesis and uteroplacental blood flow appears to be an early defect in both the hyperthermic and adolescent paradigms. The effects of these alterations in placental functional development appear to be irreversible. IUGR fetuses are both hypoxic and hypoglycaemic, and have reduced insulin and insulin-like growth factor-1 (IGF-1), and elevated concentrations of lactate. However, fetal utilization of oxygen and glucose, on a weight basis, remain constant compared with control pregnancies. Maintained utilization of these substrates, in a substrate-deficient environment, suggests increased sensitivities to metabolic signals, which may play a role in the development of metabolic diseases in later adult life. © The Physiological Society 2005.

Hay, W. W., Brown, L. D., Rozance, P. J., Wesolowski, S. R., & Limesand, S. W. (2016). Challenges in nourishing the intrauterine growth-restricted foetus - Lessons learned from studies in the intrauterine growth-restricted foetal sheep. Acta paediatrica (Oslo, Norway : 1992), 105(8), 881-9.

Previous attempts to improve growth and development of the intrauterine growth-restricted (IUGR) foetus during pregnancy have not worked or caused harm. Our research identifies tissue-specific mechanisms underlying foetal growth restriction and then tests strategies to improve growth and ameliorate many of the metabolic problems before the infant is born. The goal of our studies is to reduce the impact of foetal growth restriction at critical stages of development on the lifelong complications of IUGR offspring.