157 results on '"Deborah M. Sloboda"'
Search Results
2. Associations of Adverse Childhood Experiences with Frailty in Older Adults: A Cross-Sectional Analysis of Data from the Canadian Longitudinal Study on Aging
- Author
-
Oxana Mian, Laura N. Anderson, Daniel W. Belsky, Andrea Gonzalez, Jinhui Ma, Deborah M. Sloboda, Dawn M.E. Bowdish, and Chris P. Verschoor
- Subjects
Male ,Canada ,Aging ,Cross-Sectional Studies ,Frailty ,Adverse Childhood Experiences ,Activities of Daily Living ,Humans ,Female ,Longitudinal Studies ,Parental Death ,Geriatrics and Gerontology ,Aged - Abstract
Introduction: Frailty in older adults, characterized by a decline in multiple physiological systems and increasing vulnerability to loss of independence, disability, and death, is a public health priority in developed countries. Etiology of frailty extends across the lifespan and may begin in early life, but empirical evidence for this association is scarce. In this study, we examined whether adverse childhood experiences (ACEs) are associated with frailty in later life. Methods: We conducted a cross-sectional analysis of data for a population-based sample of 27,748 adults aged 45–85 years from the Canadian Longitudinal Study on Aging. The frailty index (FI) was computed with 76 health-related characteristics of physical and cognitive performance, self-rated health, chronic conditions, visual and hearing ability, activities of daily living, and well-being. Self-reported exposure to ACEs included physical, emotional, and sexual abuse, neglect, and witnessing intimate partner violence prior age of 16 and parental death, divorce, and living with a family member with mental illness prior age of 18. Generalized linear regression models with gamma error distribution and identity link function, adjusted for age and sex, were used to examine associations of each ACE type and the number of ACE types (0, 1, 2, or 3+) reported by an individual with FI. All models were adjusted for income, education, smoking, and alcohol consumption in sensitivity analysis. Results: Individuals exposed to ACEs had elevated levels of FI (mean = 0.13, SD = 0.09) than those unexposed, with the largest difference observed for neglect (B [95% CI]: 0.05 [0.04, 0.06]) and the smallest for parental death and divorce (0.015 [0.01,0.02]). The ACE count was associated with frailty in a graded manner, with the FI difference reaching 0.04 [0.037, 0.044] for participants exposed to 3+ ACE types. The association between ACEs and frailty tended to be stronger for women than men and for men aged 45–64 years than older men. Conclusions: Our study supports previous studies showing that exposure to ACEs is associated with frailty in adults. Our findings suggest that screening for ACEs involving childhood maltreatment may be useful for identifying individuals at risk of frailty and prevention of ACEs may have long-term benefits for healthy aging.
- Published
- 2021
- Full Text
- View/download PDF
3. Early Life Adversity and Female Reproductive Outcomes
- Author
-
Patrycja A. Jazwiec and Deborah M. Sloboda
- Published
- 2022
- Full Text
- View/download PDF
4. Diet-induced obesity alters intestinal monocyte-derived and tissue-resident macrophages in female mice independent of TNF
- Author
-
Jessica A. Breznik, Jennifer Jury, Elena F. Verdú, Deborah M. Sloboda, and Dawn M. E. Bowdish
- Abstract
Macrophages are essential for homeostatic maintenance of the anti-inflammatory and tolerogenic intestinal environment, yet monocyte-derived macrophages can promote local inflammation. Pro-inflammatory macrophage accumulation within the intestines may contribute to the development of systemic chronic inflammation and immunometabolic dysfunction in obesity. Using a model of high fat diet-induced obesity in C57BL/6J female mice, we assessed intestinal permeability by in vitro and in vivo assays, and quantitated intestinal macrophages in ileum and colon tissues by multicolour flow cytometry after short (6 weeks), intermediate (12 weeks), and prolonged (18 weeks) diet allocation. We characterized monocyte-derived CD4−TIM4− and CD4+TIM4− macrophages, as well as tissue-resident CD4+TIM4+ macrophages. Diet-induced obesity had tissue and time-dependent effects on intestinal permeability, as well as monocyte and macrophage numbers, surface phenotype, and intracellular production of the cytokines IL-10 and TNF. We found that obese mice had increased paracellular permeability, in particular within the ileum, but this did not elicit recruitment of monocytes, nor a local pro-inflammatory response by monocyte-derived or tissue-resident macrophages, in either the ileum or colon. Proliferation of monocyte-derived and tissue-resident macrophages was also unchanged. Wildtype and TNF−/- littermate mice had similar intestinal permeability and macrophage population characteristics in response to diet-induced obesity. These data are unique from reported effects of diet-induced obesity on macrophages in metabolic tissues, as well as outcomes of acute inflammation within the intestines, and collectively indicate that TNF does not mediate effects of diet-induced obesity on intestinal monocyte-derived and tissue-resident intestinal macrophages in young female mice.Abstract Figure
- Published
- 2022
- Full Text
- View/download PDF
5. Parity modulates impact of BMI and Gestational Weight Gain on gut microbiota in human pregnancy
- Author
-
Katherine M. Kennedy, Andreas Plagemann, Julia Sommer, Marie Hofmann, Wolfgang Henrich, Jon F.R. Barrett, Michael G. Surette, Stephanie Atkinson, Thorsten Braun, and Deborah M. Sloboda
- Abstract
Pregnancy requires maternal adaptations to support fetal growth: whether these adaptations include temporal shifts in the gut microbiome is still unclear. We investigated the maternal gut microbiome longitudinally over the course of pregnancy and the impact of pre-pregnancy BMI (pBMI) and GWG. We also determined whether parity modulated observed associations. We show that the gut microbiota of participants with higher pBMI changed less over the course of pregnancy in primiparous, but not multiparous participants. This suggests that previous pregnancies may have persistent impacts on maternal adaptations to pregnancy. This ecological memory appears to be passed to the next generation, as parity modulated the impact of maternal GWG on the infant gut microbiome. This work supports a role for the gut microbiome in maternal adaptations to pregnancy and highlights the need for longitudinal sampling and accounting for parity as key considerations for studies of the microbiome in pregnancy and infants.
- Published
- 2022
- Full Text
- View/download PDF
6. Paternal obesity alters the sperm epigenome and is associated with changes in the placental transcriptome and cellular composition
- Author
-
Anne-Sophie Pépin, Patrycja A. Jazwiec, Vanessa Dumeaux, Deborah M. Sloboda, and Sarah Kimmins
- Abstract
SummaryPaternal obesity has been implicated in adult-onset metabolic disease in offspring. However, the molecular mechanisms driving these paternal effects and the developmental processes involved remain poorly understood. One underexplored possibility is the role of paternally driven gene expression in placenta function. To address this, we investigated paternal high-fat diet-induced obesity in relation to sperm epigenetic signatures, the placenta transcriptome and cellular composition. C57BL6/J males were fed either a control or high-fat diet for 10 weeks beginning at 6 weeks of age. Males were timed-mated with control-fed C57BL6/J females to generate pregnancies, followed by collection of sperm, and placentas at embryonic day (E)14.5. Chromatin immunoprecipitation targeting histone H3 lysine 4 tri-methylation (H3K4me3) followed by sequencing (ChIP-seq) was performed on sperm to define obesity-associated changes in enrichment. Paternal obesity corresponded with altered sperm H3K4me3 enrichment at imprinted genes, and at promoters of genes involved in metabolism and development. Notably, sperm altered H3K4me3 was localized at placental enhancers and genes implicated in placental development and function. Bulk RNA-sequencing on placentas detected paternal obesity-induced sex-specific changes in gene expression associated with hypoxic processes such as angiogenesis, nutrient transport and imprinted genes. Paternal obesity was also linked to placenta development; specifically, a deconvolution analysis revealed altered trophoblast cell lineage specification. These findings implicate paternal obesity-effects on placenta development and function as one mechanism underlying offspring metabolic disease.Summary sentencePaternal obesity impacts the sperm epigenome at genes implicated in placenta development and is associated with an altered placenta transcriptome and trophoblast cell lineage specification.
- Published
- 2022
- Full Text
- View/download PDF
7. Intestinal permeability and peripheral immune cell composition are altered by pregnancy and adiposity at mid- and late-gestation in the mouse
- Author
-
Tatiane A. Ribeiro, Jessica A. Breznik, Katherine M. Kennedy, Erica Yeo, Brianna K. E. Kennelly, Patrycja A. Jazwiec, Violet S. Patterson, Christian J. Bellissimo, Fernando F. Anhê, Jonathan D. Schertzer, Dawn M. E. Bowdish, and Deborah M. Sloboda
- Abstract
It is clear that the gastrointestinal tract influences metabolism and immune function. Most studies to date have used male test subjects, with a focus on effects of obesity and dietary challenges. Despite significant physiological adaptations that occur across gestation, relatively few studies have examined pregnancy-related gut function. In this study, we investigated the impacts of pregnancy and adiposity on maternal intestinal epithelium morphology, in vivo intestinal permeability, and peripheral blood immunophenotype, using control (CTL) and high-fat (HF) fed non-pregnant female mice and pregnant mice at mid-(embryonic day (E)14.5) and late (E18.5) gestation. We found that small intestine length increased between non-pregnant mice and dams at late-gestation, but ileum villus length, and ileum and colon crypt depths and goblet cell numbers remained similar. Compared to CTL-fed mice, HF-fed mice had reduced small intestine length, ileum crypt depth and villus length. Goblet cell numbers were only consistently reduced in HF-fed non-pregnant mice. Pregnancy increased in vivo gut permeability, with a greater effect at mid-versus late-gestation. Non-pregnant HF-fed mice had greater gut permeability, and permeability was also increased in HF-fed pregnant dams at mid but not late-gestation. The impaired maternal gut barrier in HF-fed dams at mid-gestation coincided with changes in maternal blood and bone marrow immune cell composition, including an expansion of circulating inflammatory Ly6Chigh monocytes. In summary, pregnancy has temporal effects on maternal intestinal structure and barrier function, and on peripheral immunophenotype, which are further modified by HF diet-induced maternal adiposity.
- Published
- 2022
- Full Text
- View/download PDF
8. Fetal meconium does not have a detectable microbiota before birth
- Author
-
Katherine M. Kennedy, Deborah M. Sloboda, Michael G. Surette, Laura Rossi, Max J. Gerlach, Markus M. Heimesaat, Thorsten Braun, and Thomas Adam
- Subjects
Male ,Meconium ,Microbiology (medical) ,congenital, hereditary, and neonatal diseases and abnormalities ,medicine.drug_class ,Immunology ,Antibiotics ,Physiology ,Gestational Age ,Applied Microbiology and Biotechnology ,Microbiology ,Feces ,03 medical and health sciences ,Fetus ,fluids and secretions ,Pregnancy ,Staphylococcus epidermidis ,Immunity ,Genetics ,medicine ,Humans ,Colonization ,reproductive and urinary physiology ,030304 developmental biology ,0303 health sciences ,Bacteria ,biology ,Cesarean Section ,030306 microbiology ,business.industry ,Microbiota ,Infant, Newborn ,Rectum ,Infant ,Gestational age ,Cell Biology ,medicine.disease ,biology.organism_classification ,female genital diseases and pregnancy complications ,embryonic structures ,Female ,business - Abstract
Microbial colonization of the human intestine impacts host metabolism and immunity; however, exactly when colonization occurs is unclear. Although many studies have reported bacterial DNA in first-pass meconium samples, these samples are typically collected hours to days after birth. Here, we investigated whether bacteria could be detected in meconium before birth. Fetal meconium (n = 20) was collected by rectal swab during elective breech caesarean deliveries without labour and before antibiotics and compared to technical and procedural controls (n = 5), first-pass meconium (neonatal meconium; n = 14) and infant stool (n = 25). Unlike first-pass meconium, no microbial signal distinct from negative controls was detected in fetal meconium by 16S ribosomal RNA gene sequencing. Additionally, positive aerobic (n = 10 of 20) and anaerobic (n = 12 of 20) clinical cultures of fetal meconium (13 of 20 samples positive in at least one culture) were identified as likely skin contaminants, most frequently Staphylococcus epidermidis, and not detected by sequencing in most samples (same genera detected by culture and sequencing in 2 of 13 samples with positive culture). We conclude that fetal gut colonization of healthy term infants does not occur before birth and that microbial profiles of neonatal meconium reflect populations acquired during and after birth. Fetal meconium does not have a detectable microbiota, as shown using 16S rRNA sequencing and culture of rectal swabs collected during elective breech caesarean sections without labour and before antibiotics, indicating that colonization occurs during and after birth.
- Published
- 2021
- Full Text
- View/download PDF
9. Exposure to high fructose corn syrup during adolescence in the mouse alters hepatic metabolism and the microbiome in a sex‐specific manner
- Author
-
Deborah M. Sloboda, Katherine M. Kennedy, Cole R. McCourt, Rebecca A. Simmons, Sara E. Pinney, Miles A. Mundy, Shazia Bhat, and Michael G. Surette
- Subjects
Male ,0301 basic medicine ,medicine.medical_specialty ,Physiology ,Fructose ,Biology ,Impaired glucose tolerance ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Non-alcoholic Fatty Liver Disease ,Internal medicine ,Diabetes mellitus ,medicine ,Animals ,Microbiome ,Carbohydrate-responsive element-binding protein ,High-fructose corn syrup ,Microbiota ,Fatty liver ,Metabolism ,Lipid Metabolism ,medicine.disease ,030104 developmental biology ,Endocrinology ,Lipogenesis ,Female ,High Fructose Corn Syrup ,030217 neurology & neurosurgery - Abstract
Key points The prevalence of obesity and non-alcoholic fatty liver disease in children is dramatically increasing at the same time as consumption of foods with a high sugar content. Intake of high fructose corn syrup (HFCS) is a possible etiology as it is thought to be more lipogenic than glucose. In a mouse model, HFCS intake during adolescence increased fat mass and hepatic lipid levels in male and female mice. However, only males showed impaired glucose tolerance. Multiple metabolites including lipids, bile acids, carbohydrates, and amino acids were altered in liver in a sex-specific manner at 6 weeks of age. Some of these changes were also present in adulthood even though HFCS exposure ended at 6 weeks. HFCS significantly altered the gut microbiome which was associated with changes in key microbial metabolites. These results suggest that HFCS intake during adolescence has profound metabolic changes that are linked to changes in the microbiome and these changes are sex-specific. Abstract The rapid increase in obesity, diabetes and fatty liver disease in children over the past 20 years has been linked to increased high fructose corn syrup (HFCS) consumption making it essential to determine the short and long-term effects of HFCS during this vulnerable developmental window. We hypothesized that HFCS exposure during adolescence significantly impairs hepatic metabolic signaling pathways and alters gut microbial composition contributing to changes in energy metabolism with sex-specific effects. C57bl/6J mice with free access to HFCS during adolescence (3-6 weeks of age) underwent glucose tolerance and body composition testing and hepatic metabolomics, gene expression and triglyceride content analysis at 6 and 30 weeks of age (n = 6-8 per sex). At 6 weeks HFCS-exposed mice had significant increases in fat mass, glucose intolerance, hepatic triglycerides (females) and de novo lipogenesis gene expression (ACC, DGAT, FAS, ChREBP, SCD, SREBP, CPT and PPARα) with sex-specific effects. At 30 weeks HFCS-exposed mice also had abnormalities in glucose tolerance (males) and fat mass (females). HFCS exposure enriched carbohydrate, amino acid, long chain fatty acid and secondary bile acid metabolism at 6 weeks with changes in secondary bile metabolism at 6 and 30 weeks. Microbiome studies performed immediately before and after HFCS exposure identified profound shifts of microbial species in male mice only. In summary, a short-term HFCS exposure during adolescence induces fatty liver, alters important metabolic pathways, some of which continue to be altered in adulthood, and changes the microbiome in a sex-specific manner. This article is protected by copyright. All rights reserved.
- Published
- 2021
- Full Text
- View/download PDF
10. Placental Metabolomics for Assessment of Sex-specific Differences in Fetal Development During Normal Gestation
- Author
-
Michelle Saoi, Deborah M. Sloboda, Wajiha Gohir, Philip Britz-McKibbin, and Katherine M. Kennedy
- Subjects
0301 basic medicine ,Male ,Placenta ,Physiology ,lcsh:Medicine ,Biology ,01 natural sciences ,Article ,Fetal Development ,03 medical and health sciences ,chemistry.chemical_compound ,Mice ,Metabolomics ,Fetus ,Pregnancy ,medicine ,Placental Extracts ,Animals ,lcsh:Science ,Sex Characteristics ,Multidisciplinary ,010401 analytical chemistry ,Body Weight ,Fatty Acids ,lcsh:R ,Bioanalytical chemistry ,medicine.disease ,0104 chemical sciences ,Mice, Inbred C57BL ,030104 developmental biology ,medicine.anatomical_structure ,chemistry ,In utero ,Purines ,Metabolome ,Gestation ,Uric acid ,Female ,lcsh:Q - Abstract
The placenta is a metabolically active interfacial organ that plays crucial roles in fetal nutrient delivery, gas exchange and waste removal reflecting dynamic maternal and fetal interactions during gestation. There is growing evidence that the sex of the placenta influences fetal responses to external stimuli in utero, such as changes in maternal nutrition and exposure to environmental stressors. However, the exact biochemical mechanisms associated with sex-specific metabolic adaptations during pregnancy and its link to placental function and fetal development remain poorly understood. Herein, multisegment injection-capillary electrophoresis-mass spectrometry is used as a high throughput metabolomics platform to characterize lyophilized placental tissue (~2 mg dried weight) from C57BL/6J mice fed a standardized diet. Over 130 authentic metabolites were consistently measured from placental extracts when using a nontargeted metabolomics workflow with stringent quality control and robust batch correction. Our work revealed distinct metabolic phenotype differences that exist between male (n = 14) and female (n = 14) placentae collected at embryonic day E18.5. Intracellular metabolites associated with fatty acid oxidation and purine degradation were found to be elevated in females as compared to male placentae (p 0.40), including uric acid, valerylcarnitine, hexanoylcarnitine, and 3-hydroxyhexanolycarnitine. This murine model sheds new insights into sex-specific differences in placental mitochondrial function and protective mechanisms against deleterious oxidative stress that may impact fetal growth and birth outcomes later in life.
- Published
- 2020
- Full Text
- View/download PDF
11. Maternal undernutrition during pregnancy and lactation affects testicular morphology, the stages of spermatogenic cycle, and the testicular IGF-I system in adult offspring
- Author
-
Paula Lombide, Mark H. Vickers, Deborah M. Sloboda, Daniel Cavestany, Helen Viotti, Graeme Martin, and Graciela Pedrana
- Subjects
Male ,Offspring ,media_common.quotation_subject ,Medicine (miscellaneous) ,Biology ,Andrology ,Pregnancy ,Lactation ,Testis ,medicine ,Animals ,Humans ,Weaning ,Insulin-Like Growth Factor I ,Rats, Wistar ,Spermatogenesis ,Maternal-Fetal Exchange ,media_common ,Malnutrition ,Maternal Nutritional Physiological Phenomena ,Sertoli cell ,medicine.disease ,Rats ,Disease Models, Animal ,Seminiferous tubule ,medicine.anatomical_structure ,Prenatal Exposure Delayed Effects ,Female ,Reproduction - Abstract
Maternal undernutrition decreases sperm production in male offspring, possibly through insulin-like growth factor (IGF-I). To test this hypothesis, we fed pregnant Wistar rats ad libitum with a standard diet (CONTROL) or fed 50% of CONTROL intake, either throughout pregnancy (UNP), lactation (UNL, or both (UNPL). After weaning, male offspring (n = 10 per treatment) were fed a standard diet until postnatal day 160, when testes process for histological and molecular analyses. IGF-I immunostaining area and intensity in the testis were greater (P = 0.003) in the UNPL group compared to CONTROL, but lower in the UNP group (P < 0.0001). Levels of IGF-I receptor transcript were lower in the UNPL and UNL groups, compared to CONTROL. There were more Ki-67-positive germ and Sertoli cells, in all underfed groups than in CONTROL. Compared to CONTROL, frequency of spermatogenic cycle stage VII was lower in all underfed groups, and seminiferous tubule diameter was smaller in UNP and UNPL. Plasma FSH concentrations were greater in UNP male offspring compared to all groups (P = 0.05), whereas inhibin B concentrations were greater in UNP (P = 0.01) and UNL (P = 0.003) than in CONTROL or UNPL. Thus, prenatal undernutrition leads to a decrease in testicular IGF-I levels, whereas of pre- and postnatal undernutrition increased testicular IGF-I levels and decreased amounts of IGF-I receptor mRNA in adult offspring. We conclude that maternal undernutrition during pregnancy and lactation leads to long-lasting effects on adult male offspring testicular morphology, spermatogenesis, and IGF-I testicular system.
- Published
- 2020
- Full Text
- View/download PDF
12. The intestine and the microbiota in maternal glucose homeostasis during pregnancy
- Author
-
Erica Yeo, Patricia L Brubaker, and Deborah M Sloboda
- Subjects
Intestines ,Endocrinology ,Glucose ,Pregnancy ,Endocrinology, Diabetes and Metabolism ,Microbiota ,Homeostasis ,Humans ,Female ,Incretins - Abstract
It is now well established that, beyond its role in nutrient processing and absorption, the intestine and its accompanying gut microbiome constitute a major site of immunological and endocrine regulation that mediates whole-body metabolism. Despite the growing field of host-microbe research, few studies explore what mechanisms govern this relationship in the context of pregnancy. During pregnancy, significant maternal metabolic adaptations are made to accommodate the additional energy demands of the developing fetus and to prevent adverse pregnancy outcomes. Recent data suggest that the maternal gut microbiota may play a role in these adaptations, but changes to maternal gut physiology and the underlying intestinal mechanisms remain unclear. In this review, we discuss selective aspects of intestinal physiology including the role of the incretin hormone, glucagon-like peptide 1 (GLP-1), and the role of the maternal gut microbiome in the maternal metabolic adaptations to pregnancy. Specifically, we discuss how bacterial components and metabolites could mediate the effects of the microbiota on host physiology, including nutrient absorption and GLP-1 secretion and action, and whether these mechanisms may change maternal insulin sensitivity and secretion during pregnancy. Finally, we discuss how these pathways could be altered in disease states during pregnancy including maternal obesity and diabetes.
- Published
- 2022
13. Paternal obesity induces placental hypoxia and sex-specific impairments in placental vascularization and offspring metabolism†
- Author
-
Patrycja A Jazwiec, Violet S Patterson, Tatiane A Ribeiro, Erica Yeo, Katherine M Kennedy, Paulo C F Mathias, Jim J Petrik, and Deborah M Sloboda
- Subjects
Male ,Placenta ,Cell Biology ,General Medicine ,Diet, High-Fat ,Placentation ,Fathers ,Mice ,Glucose ,Reproductive Medicine ,Pregnancy ,Animals ,Humans ,Female ,Obesity ,Hypoxia - Abstract
Paternal obesity predisposes offspring to metabolic dysfunction, but the underlying mechanisms remain unclear. We investigated whether this metabolic dysfunction is associated with changes in placental vascular development and is fueled by endoplasmic reticulum (ER) stress-mediated changes in fetal hepatic development. We also determined whether paternal obesity indirectly affects the in utero environment by disrupting maternal metabolic adaptations to pregnancy. Male mice fed a standard chow or high fat diet (60%kcal fat) for 8–10 weeks were time-mated with female mice to generate pregnancies and offspring. Glucose tolerance was evaluated in dams at mid-gestation (embryonic day (E) 14.5) and late gestation (E18.5). Hypoxia, angiogenesis, endocrine function, macronutrient transport, and ER stress markers were evaluated in E14.5 and E18.5 placentae and/or fetal livers. Maternal glucose tolerance was assessed at E14.5 and E18.5. Metabolic parameters were assessed in offspring at ~60 days of age. Paternal obesity did not alter maternal glucose tolerance but induced placental hypoxia and altered placental angiogenic markers, with the most pronounced effects in female placentae. Paternal obesity increased ER stress-related protein levels (ATF6 and PERK) in the fetal liver and altered hepatic expression of gluconeogenic factors at E18.5. Offspring of obese fathers were glucose intolerant and had impaired whole-body energy metabolism, with more pronounced effects in female offspring. Metabolic deficits in offspring due to paternal obesity may be mediated by sex-specific changes in placental vessel structure and integrity that contribute to placental hypoxia and may lead to poor fetal oxygenation and impairments in fetal metabolic signaling pathways in the liver.
- Published
- 2021
14. Gut microbiota-based vaccination engages innate immunity to improve blood glucose control in obese mice
- Author
-
Nicole G. Barra, Gabriella Paniccia, Fernando F. Anhê, Brittany M. Duggan, Deborah M. Sloboda, Jessica G. Wallace, Matthew S. Miller, Jonathan D. Schertzer, Akhilesh K. Tamrakar, and Hannah D. Stacey
- Subjects
Innate immune system ,biology ,business.industry ,Ileum ,Gut flora ,medicine.disease ,biology.organism_classification ,digestive system ,Vaccination ,Subcutaneous injection ,medicine.anatomical_structure ,Antigen ,Diabetes mellitus ,Immunology ,medicine ,biology.protein ,Antibody ,business - Abstract
Obesity and diabetes increase circulating levels of microbial components derived from the gut microbiota. Individual bacterial factors (i.e., postbiotics) can have opposing effects on metabolic inflammation and blood glucose control. We tested the net effect of gut bacterial extracts on blood glucose using a microbiota-based vaccination strategy in mice. Male and female mice had improved insulin sensitivity and blood glucose control five weeks after a single subcutaneous injection of a specific dose of a bacterial extract obtained from the luminal contents of the proximal gut. Injection of mice with proximal gut extracts from germ-free mice revealed that bacteria were required for a microbiota-based vaccination to improve blood glucose control. Vaccination ofNod1−/−,Nod2−/−, andRipk2−/−mice showed that each of these innate immune proteins was required for bacterial extract injection to improve blood glucose control. A microbiota-based vaccination promoted a proximal gut immunoglobulin-G (IgG) response directed against bacterial extract antigens, where subcutaneous injection of mice with the luminal contents of the ileum elicited a bacterial extract-specific IgG response that is compartmentalized to the ileum of vaccinated mice. A microbiota-based vaccination was associated with an altered the microbiota composition in the ileum and colon of mice. Lean mice required a single injection of proximal gut bacterial extracts, but high fat diet (HFD)-fed, obese mice required prime-boost bacterial extract injections for improvements in blood glucose control. These data show that, upon subversion of the gut barrier, vaccination with proximal gut bacterial extracts engages innate immunity to promote long-lasting improvements in blood glucose control in a dose-dependent manner.
- Published
- 2021
- Full Text
- View/download PDF
15. Long-term effects of a maternal high-fat: high-fructose diet on offspring growth and metabolism and impact of maternal taurine supplementation
- Author
-
Clare M. Reynolds, Deborah M. Sloboda, Mark H. Vickers, Clint Gray, Rachna Patel, and Minglan Li
- Subjects
Male ,0301 basic medicine ,medicine.medical_specialty ,Taurine ,Offspring ,medicine.medical_treatment ,Medicine (miscellaneous) ,Adipose tissue ,Fructose ,Diet, High-Fat ,Obesity, Maternal ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Metabolic Diseases ,Pregnancy ,Internal medicine ,Lactation ,Animals ,Medicine ,030212 general & internal medicine ,Rats, Wistar ,business.industry ,Insulin ,Leptin ,medicine.disease ,Obesity ,Rats ,030104 developmental biology ,Endocrinology ,medicine.anatomical_structure ,Animals, Newborn ,chemistry ,Prenatal Exposure Delayed Effects ,Dietary Supplements ,Female ,business - Abstract
Objective:Maternal obesity is associated with obesity and metabolic disorders in offspring. However, there remains a paucity of data on strategies to reverse the effects of maternal obesity on maternal and offspring health. With maternal undernutrition, taurine supplementation improves outcomes in offspring mediated in part via improved glucose–insulin homeostasis. The efficacy of taurine supplementation in the setting of maternal obesity on health and well-being of offspring is unknown. We examined the effects of taurine supplementation on outcomes related to growth and metabolism in offspring in a rat model of maternal obesity.Design:Wistar rats were randomised to: 1) control diet during pregnancy and lactation (CON); 2) CON with 1.5% taurine in drinking water (CT); 3) maternal obesogenic diet (MO); or 4) MO with taurine (MOT). Offspring were weaned onto the control diet for the remainder of the study.Results:At day 150, offspring body weights and adipose tissue weights were increased in MO groups compared to CON. Adipose tissue weights were reduced in MOT versus MO males but not females. Plasma fasting leptin and insulin were increased in MO offspring groups but were not altered by maternal taurine supplementation. Plasma homocysteine concentrations were reduced in all maternal taurine-supplemented offspring groups. There were significant interactions across maternal diet, taurine supplementation and sex for response to an oral glucose tolerance test , a high-fat dietary preference test and pubertal onset in offspring.Conclusions:These results demonstrate that maternal taurine supplementation can partially ameliorate adverse developmental programming effects in offspring in a sex-specific manner.
- Published
- 2019
- Full Text
- View/download PDF
16. Obesity during pregnancy results in maternal intestinal inflammation, placental hypoxia, and alters fetal glucose metabolism at mid-gestation
- Author
-
Jim Petrik, Michael G. Surette, Deborah M. Sloboda, Erica Yeo, Yu Fei Xia, Dawn M. E. Bowdish, Christian J. Bellissimo, and Jessica G. Wallace
- Subjects
0301 basic medicine ,beta-Defensins ,Placenta ,lcsh:Medicine ,Feces ,Mice ,0302 clinical medicine ,Pregnancy ,lcsh:Science ,2. Zero hunger ,Multidisciplinary ,Toll-Like Receptors ,Intrauterine growth ,Cell Hypoxia ,3. Good health ,Intestines ,Butyrates ,Cytokines ,Female ,medicine.symptom ,medicine.medical_specialty ,Offspring ,Reproductive biology ,030209 endocrinology & metabolism ,Inflammation ,Gestational Age ,Butyrate ,Carbohydrate metabolism ,Diet, High-Fat ,Article ,03 medical and health sciences ,Fetus ,Internal medicine ,medicine ,Animals ,Obesity ,business.industry ,Macrophages ,Lachnospiraceae ,lcsh:R ,Gluconeogenesis ,Hypoxia (medical) ,medicine.disease ,Fatty Acids, Volatile ,Gastrointestinal Microbiome ,Pregnancy Complications ,030104 developmental biology ,Endocrinology ,Glucose ,Gene Expression Regulation ,lcsh:Q ,business ,Carrier Proteins - Abstract
We investigated whether diet-induced changes in the maternal intestinal microbiota were associated with changes in bacterial metabolites and their receptors, intestinal inflammation, and placental inflammation at mid-gestation (E14.5) in female mice fed a control (17% kcal fat, n = 7) or a high-fat diet (HFD 60% kcal fat, n = 9; ad libitum) before and during pregnancy. Maternal diet-induced obesity (mDIO) resulted in a reduction in maternal fecal short-chain fatty acid producing Lachnospiraceae, lower cecal butyrate, intestinal antimicrobial peptide levels, and intestinal SCFA receptor Ffar3, Ffar2 and Hcar2 transcript levels. mDIO increased maternal intestinal pro-inflammatory NFκB activity, colonic CD3+ T cell number, and placental inflammation. Maternal obesity was associated with placental hypoxia, increased angiogenesis, and increased transcript levels of glucose and amino acid transporters. Maternal and fetal markers of gluconeogenic capacity were decreased in pregnancies complicated by obesity. We show that mDIO impairs bacterial metabolite signaling pathways in the mother at mid-gestation, which was associated with significant structural changes in placental blood vessels, likely as a result of placental hypoxia. It is likely that maternal intestinal changes contribute to adverse maternal and placental adaptations that, via alterations in fetal hepatic glucose handling, may impart increased risk of metabolic dysfunction in offspring.
- Published
- 2019
- Full Text
- View/download PDF
17. Associations between exposure to adverse childhood experiences and biological aging: Evidence from the Canadian Longitudinal Study on Aging
- Author
-
Oxana, Mian, Daniel W, Belsky, Alan A, Cohen, Laura N, Anderson, Andrea, Gonzalez, Jinhui, Ma, Deborah M, Sloboda, Dawn Me, Bowdish, and Chris P, Verschoor
- Subjects
Adult ,Male ,Aging ,Canada ,Endocrine and Autonomic Systems ,Endocrinology, Diabetes and Metabolism ,Psychiatry and Mental health ,Endocrinology ,Adverse Childhood Experiences ,Humans ,Female ,Longitudinal Studies ,Prospective Studies ,Biological Psychiatry - Abstract
People exposed to adverse childhood experiences (ACEs) suffer from an increased risk of chronic disease and shorter lifespan. These individuals also tend to exhibit accelerated reproductive development and show signs of advanced cellular aging as early as childhood. These observations suggest that ACEs may accelerate biological processes of aging through direct or indirect mechanisms; however, few population-based studies have data to test this hypothesis. We analysed ACEs and biological aging data from the Canadian Longitudinal Study on Aging (CLSA; n = 23,354 adults aged 45-85) and used the BioAge R package to compute three indices of biological aging from blood-chemistry and organ-function data: Klemera-Doubal method (KDM) biological age, phenotypic age (PA), and homeostatic dysregulation (HD). Adults with ACEs tended to be biologically older than those with no ACEs, although the observed effect-sizes were small (Cohen's d0.15), with the exception of neglect (d=0.35 for KDM and PA). Associations were similar for men and women and tended to be smaller for older as compared to midlife participants. Subtypes of ACEs perceived as being more severe (e.g., being pushed or kicked, experiencing forced sexual activity, witnessing physical violence) and more frequent and diverse exposures were associated with relatively larger effect-sizes. These findings support the hypothesis that ACEs contribute to accelerated biological aging, although replication is needed in studies with access to prospective records of ACEs and cellular-level measurements of biological aging. Furthermore, future work to better understand the degree to which associations between ACEs and biological aging are moderated by specific life-course pathways, and mediated by lifestyle and socioeconomic factors is warranted.
- Published
- 2022
- Full Text
- View/download PDF
18. Over-celling fetal microbial exposure
- Author
-
Laura Rossi, Jon Barrett, Christian J. Bellissimo, Jessica A. Breznik, Markus M. Heimesaat, Samuel Parry, Frederic D. Bushman, Liza Konnikova, Marcus C. de Goffau, Michal A. Elovitz, Katherine M. Kennedy, Thorsten Braun, Omry Koren, Nicola Segata, Michael G. Surette, Jens Walter, Rebecca A. Simmons, Deborah M. Sloboda, Experimental Vascular Medicine, Vascular Medicine, and ACS - Diabetes & metabolism
- Subjects
Fetus ,Pregnancy ,Placenta ,MEDLINE ,Physiology ,Humans ,Female ,Biology ,General Biochemistry, Genetics and Molecular Biology - Published
- 2021
19. Effects of Obesity-Associated Chronic Inflammation on Peripheral Blood Immunophenotype Are Not Mediated by TNF in Female C57BL/6J Mice
- Author
-
Kevin P. Foley, Jonathan D. Schertzer, Dhanyasri Maddiboina, Deborah M. Sloboda, Jessica A. Breznik, and Dawn M. E. Bowdish
- Subjects
Male ,Cellular immunity ,Immunology ,Inflammation ,CCL2 ,Systemic inflammation ,Diet, High-Fat ,Proinflammatory cytokine ,Immunophenotyping ,Mice ,Immune system ,Sex Factors ,Immunology and Allergy ,Medicine ,Animals ,Antigens, Ly ,Humans ,Obesity ,Adiposity ,Mice, Knockout ,Immunity, Cellular ,business.industry ,Tumor Necrosis Factor-alpha ,Monocyte ,General Medicine ,Mice, Inbred C57BL ,Disease Models, Animal ,medicine.anatomical_structure ,Adipose Tissue ,Chronic Disease ,Leukocytes, Mononuclear ,Tumor necrosis factor alpha ,Female ,medicine.symptom ,business - Abstract
Chronic low-grade systemic inflammation in obesity contributes to the development and progression of aspects of metabolic syndrome. In obese male mice, expanded adipose tissue releases proinflammatory cytokines, including TNF, which promotes an increase in immature, proinflammatory, peripheral blood Ly-6Chigh monocytes. The aim of this study was to characterize how TNF alters circulating cellular immunity in female mice with diet-induced obesity. We initially quantified peripheral blood immune cells by flow cytometry in female wild-type C57BL/6J mice after 3–30 wk of allocation to a high-fat (HF) or standard chow diet. We assessed effects of diet and time on neutrophil, monocyte, B cell, NK cell, CD4+ T cell, and CD8+ T cell populations. There was a significant interaction of the effects of diet type and time on the numbers and prevalence of circulating total monocytes and Ly-6Chigh, Ly-6Clow, and Ly-6C− subsets. Circulating monocytes, in particular Ly-6Chigh monocytes, were increased in HF-fed mice compared with chow-fed mice. Ly-6Chigh monocytes from HF-fed mice also had a more immature phenotype yet were highly responsive to the chemotactic ligand CCL2 and had greater intracellular production of TNF. Comparisons of the effects of HF diet feeding in littermate wild-type (TNF+/+) and TNF−/− female mice showed that genetic ablation of TNF did not protect from higher adiposity or an increase in circulating, immature, proinflammatory Ly-6Chigh monocytes during HF diet–induced obesity. These data emphasize the importance of considering biological sex when determining the mechanisms of TNF action in obesity-induced cellular inflammation and in other chronic inflammatory conditions.
- Published
- 2021
20. Paternal obesity results in placental hypoxia and sex-specific impairments in placental vascularization and offspring metabolic function
- Author
-
Jim Petrik, Erica Yeo, Katherine M. Kennedy, Patrycja A. Jazwiec, Deborah M. Sloboda, Tatiane A. Ribeiro, Paulo Cezar de Freitas Mathias, and Violet S. Patterson
- Subjects
medicine.medical_specialty ,Fetus ,Pregnancy ,ATF6 ,Offspring ,Hypoxia (medical) ,Biology ,medicine.disease ,Endocrinology ,In utero ,Internal medicine ,medicine ,Unfolded protein response ,medicine.symptom ,Placental lactogen - Abstract
Paternal obesity predisposes offspring to metabolic dysfunction, but the underlying mechanisms remain unclear. We investigated whether paternal obesity-induced offspring metabolic dysfunction is associated with placental endoplasmic reticulum (ER) stress and impaired vascular development. We determined whether offspring glucose intolerance is fueled by ER stress-mediated changes in fetal hepatic development. Furthermore, we also determined whether paternal obesity may indirectly affect in utero development by disrupting maternal metabolic adaptations to pregnancy. Male mice fed a standard chow diet (CON; 17% kcal fat) or high fat diet (PHF; 60% kcal fat) for 8-10 weeks were time-mated with control female mice to generate pregnancies and offspring. Glucose tolerance in pregnant females was evaluated at mid-gestation (embryonic day (E) 14.5) and term gestation (E18.5). At E14.5 and E18.5, fetal liver and placentae were collected, and markers of hypoxia, angiogenesis, endocrine function, and macronutrient transport, and unfolded protein response (UPR) regulators were evaluated to assess ER stress. Young adult offspring glucose tolerance and metabolic parameters were assessed at ∼60 days of age. Paternal obesity did not alter maternal glucose tolerance or placental lactogen in pregnancy but did induce placental hypoxia, ER stress, and altered placental angiogenesis. This effect was most pronounced in placentae associated with female fetuses. Consistent with this, paternal obesity also activated the ATF6 and PERK branches of the UPR in fetal liver and altered hepatic expression of gluconeogenic factors at E18.5. Adult offspring of obese fathers showed glucose intolerance and impaired whole-body energy metabolism, particularly in female offspring. Thus, paternal obesity programs sex-specific adverse placental structural and functional adaptations and alters fetal hepatic development via ER stress-induced pathways. These changes likely underpin metabolic deficits in adult offspring.Summary SentencePaternal obesity alters placental vascular structures and is associated with sex-specific compromises in glucose tolerance and metabolism in young offspring
- Published
- 2021
- Full Text
- View/download PDF
21. Investigating the normalization and normative views of gestational weight gain: Balancing recommendations with the promotion and support of healthy pregnancy diets
- Author
-
Sarah Oresnik, Mary Barker, Stephanie A. Atkinson, Tina Moffat, Sarah D. McDonald, Deborah M. Sloboda, Luseadra McKerracher, and Beth Murray-Davis
- Subjects
Gerontology ,Adult ,Canada ,Health Knowledge, Attitudes, Practice ,media_common.quotation_subject ,030209 endocrinology & metabolism ,03 medical and health sciences ,Young Adult ,0302 clinical medicine ,Promotion (rank) ,Pregnancy ,Genetics ,medicine ,Humans ,0601 history and archaeology ,Socioeconomic status ,Ecology, Evolution, Behavior and Systematics ,media_common ,Ontario ,060101 anthropology ,Descriptive statistics ,06 humanities and the arts ,medicine.disease ,Focus group ,Gestational Weight Gain ,United States ,3. Good health ,Diet ,Anthropology ,Normative ,Household income ,Female ,Anatomy ,Worry ,Psychology - Abstract
Objectives Gestational weight gain (GWG) is increasingly monitored in the United States and Canada. While promoting healthy GWG offers benefits, there may be costs with over-surveillance. We aimed to explore these costs/benefits. Methods Quantitative data from 350 pregnant survey respondents and qualitative focus group data from 43 pregnant/post-partum and care-provider participants were collected in the Mothers to Babies (M2B) study in Hamilton, Canada. We report descriptive statistics and discussion themes on GWG trajectories, advice, knowledge, perceptions, and pregnancy diet. Relationships between GWG monitoring/normalization and worry, knowledge, diet quality, and sociodemographics-namely low-income and racialization-were assessed using χ2 tests and a linear regression model and contextualized with focus group data. Results Most survey respondents reported GWG outside recommended ranges but rejected the mid-20th century cultural norm of "eating for two"; many worried about gaining excessively. Conversely, respondents living in very low-income households were more likely to be gaining less than recommended GWG and to worry about gaining too little. A majority had received advice about GWG, yet half were unable to identify the range recommended for their prepregnancy BMI. This proportion was even lower for racialized respondents. Pregnancy diet quality was associated with household income, but not with receipt or understanding of GWG guidance. Care-providers encouraged normalized GWG, while worrying about the consequences of pathologizing "abnormal" GWG. Conclusions Translation of GWG recommendations should be done with a critical understanding of GWG biological normalcy. Supportive GWG monitoring and counseling should consider clinical, socioeconomic, and community contexts.
- Published
- 2021
22. Fetal gut colonization: meconium does not have a detectable microbiota before birth
- Author
-
Deborah M. Sloboda, Thomas Adam, Katherine M. Kennedy, Michael G. Surette, Max J. Gerlach, Laura Rossi, Markus M. Heimesaat, and Thorsten Braun
- Subjects
Fetus ,Gut colonization ,biology ,medicine.drug_class ,Antibiotics ,Physiology ,biology.organism_classification ,female genital diseases and pregnancy complications ,fluids and secretions ,Meconium ,Staphylococcus epidermidis ,Immunity ,embryonic structures ,medicine ,Colonization ,reproductive and urinary physiology ,Bacteria - Abstract
Microbial colonization of the human intestine impacts host metabolism and immunity, however when colonization occurs is unclear. Although numerous studies have reported bacterial DNA in first-pass meconium samples, these samples are collected hours to days after birth. We investigated whether bacteria could be detected in meconium prior to birth. Fetal meconium (n = 20) was collected by rectal swab during elective breech Cesarean sections without labour prior to antibiotics and compared to technical and procedural controls (n = 5), first-pass meconium (neonatal meconium; n = 14), and infant stool (n = 25). Unlike first-pass meconium, no microbial signal distinct from negative controls was detected in fetal meconium by 16S rRNA gene sequencing. Additionally, positive aerobic (n = 10 of 20) and anaerobic (n = 12 of 20) clinical cultures of fetal meconium (13 of 20 samples positive in at least one culture) were identified as likely skin contaminants, most frequently Staphylococcus epidermidis, and not detected by sequencing in most samples (same genera detected by culture and sequencing in 2 of 13 samples with positive culture). We conclude that fetal gut colonization does not occur before birth, and that microbial profiles of neonatal meconium reflect populations acquired during and after birth.
- Published
- 2021
- Full Text
- View/download PDF
23. Re: 'Invited Commentary: The Disillusionment of Developmental Origins of Health and Disease (DOHaD) Epidemiology'
- Author
-
Hazel Inskip, Mark A. Hanson, Cyrus Cooper, Keith M. Godfrey, Lucilla Poston, Caroline H.D. Fall, Mary Barker, Deborah M. Sloboda, and Janis Baird
- Subjects
Gerontology ,medicine.medical_specialty ,Epidemiology ,business.industry ,MEDLINE ,Disease ,Pregnancy ,Prenatal Exposure Delayed Effects ,Humans ,Medicine ,Female ,AcademicSubjects/MED00860 ,Letters to the Editor ,business - Published
- 2021
24. Addressing embodied inequities in health: how do we enable improvement in women's diet in pregnancy?
- Author
-
Deborah M. Sloboda, L Zalot, Beth Murray-Davis, D Williams, Tina Moffat, J Vickers-Manzin, Sarah Oresnik, Mary Barker, and Luseadra McKerracher
- Subjects
Gerontology ,Canada ,media_common.quotation_subject ,Psychological intervention ,Medicine (miscellaneous) ,Stakeholder engagement ,Pregnancy ,Humans ,Social determinants of health ,media_common ,Community resilience ,Nutrition and Dietetics ,Community engagement ,Postpartum Period ,Public Health, Environmental and Occupational Health ,Pregnancy Outcome ,Developmental origins ,Social Support ,Prenatal Care ,Focus Groups ,Focus group ,Health equity ,Dietary inequities ,Diet ,Socioeconomic Factors ,Female ,Health inequities ,Psychological resilience ,Preconception Care ,Psychology ,Research Paper - Abstract
Objective:To disrupt cycles of health inequity, traceable to dietary inequities in the earliest stages of life, public health interventions should target improving nutritional wellbeing in preconception/pregnancy environments. This requires a deep engagement with pregnant/postpartum people (PPP) and their communities (including their health and social care providers, HSCP). We sought to understand the factors that influence diet during pregnancy from the perspectives of PPP and HSCP, and to outline intervention priorities.Design:We carried out thematic network analyses of transcripts from ten focus group discussions (FGD) and one stakeholder engagement meeting with PPP and HSCP in a Canadian city. Identified themes were developed into conceptual maps, highlighting local priorities for pregnancy nutrition and intervention development.Setting:FGD and the stakeholder meeting were run in predominantly lower socioeconomic position (SEP) neighbourhoods in the sociodemographically diverse city of Hamilton, Canada.Participants:All local, comprising twenty-two lower SEP PPP and forty-three HSCP.Results:Salient themes were resilience, resources, relationships and the embodied experience of pregnancy. Both PPP and HSCP underscored that socioeconomic-political forces operating at multiple levels largely determined the availability of individual and relational resources constraining diet during pregnancy. Intervention proposals focused on cultivating individual and community resilience to improve early-life nutritional environments. Participants called for better-integrated services, greater income supports and strengthened support programmes.Conclusions:Hamilton stakeholders foregrounded social determinants of inequity as main factors influencing pregnancy diet. They further indicated a need to develop interventions that build resilience and redistribute resources at multiple levels, from the household to the state.
- Published
- 2020
- Full Text
- View/download PDF
25. Maternal undernutrition during pregnancy and lactation increases transcription factors, ETV5 and GDNF, and alters regulation of apoptosis and heat shock proteins in the testis of adult offspring in the rat
- Author
-
Graeme Martin, Mark H. Vickers, Paula Lombide, Helen Viotti, Camila Larrañaga, Daniel Cavestany, Alejandra Diaz, Deborah M. Sloboda, and Graciela Pedrana
- Subjects
Male ,Apoptosis ,Reproductive technology ,0302 clinical medicine ,Endocrinology ,Pregnancy ,Lactation ,Testis ,Glial cell line-derived neurotrophic factor ,0303 health sciences ,030219 obstetrics & reproductive medicine ,Sertoli cell ,Spermatozoa ,Up-Regulation ,DNA-Binding Proteins ,Meiosis ,medicine.anatomical_structure ,Prenatal Exposure Delayed Effects ,Animal Nutritional Physiological Phenomena ,Female ,Germ cell ,Biotechnology ,Offspring ,Nutritional Status ,Biology ,Andrology ,03 medical and health sciences ,Genetics ,medicine ,Animals ,HSP70 Heat-Shock Proteins ,Glial Cell Line-Derived Neurotrophic Factor ,HSP90 Heat-Shock Proteins ,Rats, Wistar ,Spermatogenesis ,Molecular Biology ,030304 developmental biology ,Sertoli Cells ,Malnutrition ,Maternal Nutritional Physiological Phenomena ,medicine.disease ,Hsp70 ,Disease Models, Animal ,Reproductive Medicine ,biology.protein ,Animal Science and Zoology ,Apoptosis Regulatory Proteins ,Developmental Biology ,Transcription Factors - Abstract
We tested whether changes in Sertoli cell transcription factors and germ cell heat shock proteins (HSPs) are linked to the effects of maternal undernutrition on male offspring fertility. Rats were fed ad libitum with a standard diet (CONTROL) throughout pregnancy and lactation or with 50% of CONTROL intake throughout pregnancy (UNP) or lactation (UNL) or both periods (UNPL). After postnatal Day 21, 10 male pups per group were fed a standard diet ad libitum until postnatal Day 160 when testes were processed for histological, mRNA and immunohistochemical analyses. Compared with CONTROL: caspase-3 was increased in UNP and UNPL (P = 0.001); Bax was increased in UNL (P = 0.002); Bcl-2 (P
- Published
- 2020
26. Biological sex, not reproductive cycle, influences peripheral blood immune cell prevalence in mice
- Author
-
Jessica A. Breznik, Christian Schulz, Jinhui Ma, Dawn M. E. Bowdish, and Deborah M. Sloboda
- Subjects
0301 basic medicine ,Male ,Physiology ,Cell ,Disease ,Biology ,CD8-Positive T-Lymphocytes ,Flow cytometry ,Immunophenotyping ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Immune system ,medicine ,Prevalence ,Animals ,medicine.diagnostic_test ,Laboratory mouse ,Flow Cytometry ,Peripheral ,Killer Cells, Natural ,Mice, Inbred C57BL ,030104 developmental biology ,medicine.anatomical_structure ,Female ,030217 neurology & neurosurgery ,CD8 - Abstract
Key points Traditionally the female sex, compared with the male sex, has been perceived as having greater variability in many physiological traits, including within the immune system. We investigated effects of biological sex and the female reproductive cycle on numbers of circulating leukocytes in C57BL/6J mice. We show that biological sex, but not female reproductive cyclicity, has a significant effect on peripheral blood immune cell prevalence and variability, and that sex differences were not consistent amongst common inbred laboratory mouse strains. We found that male C57BL/6J mice, compared with female mice, have greater variability in peripheral blood immunophenotype, and that this was influenced by body weight. We created summary tables for researchers to facilitate experiment planning and sample size calculations for peripheral immune cells that consider the effects of biological sex. Abstract Immunophenotyping (i.e. quantifying the number and types of circulating leukocytes) is used to characterize immune changes during health and disease, and in response to pharmacological and other interventions. Despite the importance of biological sex in immune function, there is considerable uncertainty amongst researchers as to the extent to which biological sex or the female reproductive cycle influence blood immunophenotype. We quantified circulating leukocytes by multicolour flow cytometry in young C57BL/6J mice and assessed the effects of the reproductive cycle, biological sex, and other experimental and biological factors on data variability. We found that there are no significant effects of the female reproductive cycle on the prevalence of peripheral blood B cells, NK cells, CD4+ T cells, CD8+ T cells, monocytes, or neutrophils. Immunophenotype composition and variability do not significantly change between stages of the female reproductive cycle. There are, however, sex-specific differences in immune cell prevalence, with fewer monocytes, neutrophils, and NK cells in female mice. Surprisingly, immunophenotype is more variable in male mice, and weight is a significant contributing factor. We provide tools for researchers to perform a priori sample size calculations for two-group and factorial analyses. We show that immunophenotype varies between inbred mouse strains, and that using equal sample sizes of male and female mice is not always appropriate for within-sex evaluations of immune cell populations in peripheral blood.
- Published
- 2020
27. Behaviour change interventions: getting in touch with individual differences, values and emotions
- Author
-
Polly Hardy-Johnson, Wendy Lawrence, David J. Farrell, Janis Baird, Leanne Morrison, Christina Vogel, Sarah Jenner, Kathryn Woods-Townsend, Hazel Inskip, Mary Barker, Sofia Strömmer, Sarah Shaw, Sara Correia Simao, Deborah M. Sloboda, and Millie Barrett
- Subjects
Behavior Control ,05 social sciences ,Emotions ,Health Behavior ,Appeal ,Psychological intervention ,Individuality ,Medicine (miscellaneous) ,050109 social psychology ,Rationality ,Cognition ,Health Promotion ,Variety (cybernetics) ,03 medical and health sciences ,0302 clinical medicine ,Systematic review ,Treatment Outcome ,Behaviour change interventions ,Humans ,0501 psychology and cognitive sciences ,030212 general & internal medicine ,Psychology ,Games for Health ,Social psychology - Abstract
Systematic reviews and meta-analyses suggest that behaviour change interventions have modest effect sizes, struggle to demonstrate effect in the long term and that there is high heterogeneity between studies. Such interventions take huge effort to design and run for relatively small returns in terms of changes to behaviour.So why do behaviour change interventions not work and how can we make them more effective? This article offers some ideas about what may underpin the failure of behaviour change interventions. We propose three main reasons that may explain why our current methods of conducting behaviour change interventions struggle to achieve the changes we expect: 1) our current model for testing the efficacy or effectiveness of interventions tends to a mean effect size. This ignores individual differences in response to interventions; 2) our interventions tend to assume that everyone values health in the way we do as health professionals; and 3) the great majority of our interventions focus on addressing cognitions as mechanisms of change. We appeal to people’s logic and rationality rather than recognising that much of what we do and how we behave, including our health behaviours, is governed as much by how we feel and how engaged we are emotionally as it is with what we plan and intend to do.Drawing on our team’s experience of developing multiple interventions to promote and support health behaviour change with a variety of populations in different global contexts, this article explores strategies with potential to address these issues.
- Published
- 2020
28. Gut microbiota-based vaccination engages innate immunity to improve blood glucose control in obese mice
- Author
-
Gabriella Paniccia, Hannah D. Stacey, Brittany M. Duggan, Deborah M. Sloboda, Jessica G. Wallace, Michael G. Surette, Matthew S. Miller, Akhilesh K. Tamrakar, Fernando F. Anhê, Nicole G. Barra, and Jonathan D. Schertzer
- Subjects
Blood Glucose ,Male ,PRR, pattern recognition receptor ,Mice, Obese ,MDP, muramyl dipeptide ,Gut flora ,Mice ,Cecum ,Subcutaneous injection ,0302 clinical medicine ,HFD, high fat diet ,Medicine ,IgG, immunoglobin-G ,Internal medicine ,2. Zero hunger ,0303 health sciences ,PGN, peptidoglycan ,biology ,Microbiota ,Vaccination ,Diabetes ,3. Good health ,medicine.anatomical_structure ,FMT, fecal microbial transfer ,LPS, lipopolysaccharide ,GTT, glucose tolerance test ,OGSIS, oral glucose-stimulated insulin secretion ,Female ,Original Article ,CD, control diet ,Glycemic Control ,Diet, High-Fat ,03 medical and health sciences ,Insulin resistance ,RED, reciprocal endpoint dilution ,Diabetes mellitus ,Diabetes Mellitus ,Animals ,Obesity ,Molecular Biology ,ITT, insulin tolerance test ,030304 developmental biology ,Immunometabolism ,Innate immune system ,business.industry ,meso-DAP, meso-diaminopimelic acid ,Cell Biology ,biology.organism_classification ,medicine.disease ,RC31-1245 ,Immunity, Innate ,Small intestine ,Gastrointestinal Microbiome ,Mice, Inbred C57BL ,Glucose ,Immunology ,T2D, Type 2 Diabetes ,business ,030217 neurology & neurosurgery - Abstract
Objective Obesity and diabetes increase circulating levels of microbial components derived from the gut microbiota. Individual bacterial factors (i.e., postbiotics) can have opposing effects on blood glucose. Methods We tested the net effect of gut bacterial extracts on blood glucose in mice using a microbiota-based vaccination strategy. Results Male and female mice had improved glucose and insulin tolerance five weeks after a single subcutaneous injection of a specific dose of a bacterial extract obtained from the luminal contents of the upper small intestine (SI), lower SI, or cecum. Injection of mice with intestinal extracts from germ-free mice revealed that bacteria were required for a microbiota-based vaccination to improve blood glucose control. Vaccination of Nod1−/−, Nod2−/−, and Ripk2−/− mice showed that each of these innate immune proteins was required for bacterial extract injection to improve blood glucose control. A microbiota-based vaccination promoted an immunoglobulin-G (IgG) response directed against bacterial extract antigens, where subcutaneous injection of mice with the luminal contents of the lower SI elicited a bacterial extract-specific IgG response that is compartmentalized to the lower SI of vaccinated mice. A microbiota-based vaccination was associated with an altered microbiota composition in the lower SI and colon of mice. Lean mice only required a single injection of small intestinal-derived bacterial extract, but high fat diet (HFD)-fed, obese mice required prime-boost bacterial extract injections for improvements in blood glucose control. Conclusions Subversion of the gut barrier by vaccination with a microbiota-based extract engages innate immunity to promote long-lasting improvements in blood glucose control in a dose-dependent manner., Graphical abstract Image 1, Highlights • Subcutaneous injection of gut bacterial extracts improved blood glucose control in mice. • Microbiota-based vaccination engaged NOD1-NOD2-RIPK2 to alter blood glucose. • Microbiota-based vaccination promoted a proximal gut IgG response. • Microbiota-based vaccination altered the composition of the gut microbiome. • Obese mice required prime-boost injections to improve blood glucose control.
- Published
- 2022
- Full Text
- View/download PDF
29. Histomorphologic Analysis of the Late-term Rat Fetus and Placenta
- Author
-
Pauline M. Herst, Phanie L. Charest, Maryse Lessard, Julius Haruna, Mathieu Dalvai, Marie-Odile Benoit-Biancamano, Janice L. Bailey, Deborah M. Sloboda, and Vanessa Vrolyk
- Subjects
0301 basic medicine ,Pathology ,medicine.medical_specialty ,040301 veterinary sciences ,Placenta ,Dissection (medical) ,Toxicology ,Pathology and Forensic Medicine ,Rats, Sprague-Dawley ,0403 veterinary science ,Gross examination ,03 medical and health sciences ,Basal (phylogenetics) ,Fetus ,Rat fetus ,Pregnancy ,medicine ,Animals ,Molecular Biology ,reproductive and urinary physiology ,Histocytological Preparation Techniques ,business.industry ,Histology ,04 agricultural and veterinary sciences ,Cell Biology ,medicine.disease ,Rats ,030104 developmental biology ,medicine.anatomical_structure ,embryonic structures ,Gestation ,Female ,business - Abstract
Histological examination of the rat placenta and fetus is uncommon. Toxicological studies mainly rely on gross examination of the fetus and on fetal and placental weights. These are often insufficient to assess the fetal and placental toxicity of xenobiotics. The small size of the fetus makes its dissection labor-intensive. Thus, our objective was to develop a simple and accurate technique to evaluate the rat fetus and placenta. Sprague-Dawley rat fetuses at gestational day 19.5 ( n = 18) and their placentas ( n = 32) were fixed in formalin. Placentas were cut transversally in the center. Fetuses were cut following a freehand whole-body serial sectioning diagram adapted from Wilson’s method. Sections were stained with hematoxylin–eosin–phloxine–saffron, and histomorphometry was used to measure the area of the fetal placental region (27.2 ± 1.7 mm2), including the labyrinth (22.2 ± 1.0 mm2) and the basal zone (4.8 ± 0.8 mm2). Our whole-fetus serial sectioning technique resulted in 12 precise cutting planes that fit on 3 histological slides, enabling the examination of most organs without labor-intensive dissection. Quantitative analysis of placental areas improves the understanding of the pathogenesis of treatment-related changes. This technique provides a standardized method for future research in pertinent fields such as developmental biology and toxicology.
- Published
- 2018
- Full Text
- View/download PDF
30. Paternal Obesity is Associated with Endoplasmic Reticulum Stress-Induced Placental Hypoxia and Altered Vascularization Without Changes in Maternal Glucose Tolerance in Pregnancy
- Author
-
Tatiane Aparecida Ribeiro, Paulo Cezar de Freitas Mathias, Jim Petrik, Deborah M. Sloboda, Violet S. Patterson, and Patrycja A. Jazwiec
- Subjects
medicine.medical_specialty ,Pregnancy ,business.industry ,Endoplasmic reticulum ,Stress induced ,Obstetrics and Gynecology ,Hypoxia (medical) ,medicine.disease ,Obesity ,Endocrinology ,Reproductive Medicine ,Internal medicine ,medicine ,medicine.symptom ,business ,Developmental Biology - Published
- 2021
- Full Text
- View/download PDF
31. Knowledge about the Developmental Origins of Health and Disease is independently associated with variation in diet quality during pregnancy
- Author
-
Luseadra McKerracher, Meghan McConnell, Sarah D. McDonald, Beth Murray-Davis, Stephanie A. Atkinson, Mary Barker, Deborah M. Sloboda, and Tina Moffat
- Subjects
0301 basic medicine ,Adult ,knowledge translation ,Canada ,Health Knowledge, Attitudes, Practice ,Pregnancy Nutrition ,Mixed regression ,Nutritional Status ,Disease ,03 medical and health sciences ,0302 clinical medicine ,Pregnancy ,Knowledge translation ,health inequities ,medicine ,Humans ,030212 general & internal medicine ,Eating behaviour ,030109 nutrition & dietetics ,Nutrition and Dietetics ,business.industry ,Public Health, Environmental and Occupational Health ,Obstetrics and Gynecology ,Original Articles ,developmental origins ,diet quality ,Maternal Nutritional Physiological Phenomena ,medicine.disease ,3. Good health ,Diet ,Pregnancy Complications ,Variation (linguistics) ,nutrition ,Diet quality ,Pediatrics, Perinatology and Child Health ,Original Article ,Female ,pregnancy ,business ,Demography - Abstract
Environmental factors affecting development through embryogenesis, pregnancy, and infancy impact health through all subsequent stages of life. Known as the Developmental Origins of Health and Disease (DOHaD) hypothesis, this concept is widely accepted among health and social scientists. However, it is unclear whether DOHaD-based ideas are reaching the general public and/or influencing behaviour. This study thus investigated whether and under what circumstances pregnant people in Canada are familiar with DOHaD, and if DOHaD familiarity relates to eating behaviour. Survey responses from pregnant people from Hamilton, Canada, were used to assess respondents' knowledge of DOHaD (hereafter, DOHaDKNOWLEDGE) compared with their knowledge of more general pregnancy health recommendations (Pregnancy GuidelineKNOWLEDGE). The survey also characterized respondents' pregnancy diet quality and sociodemographic profiles. We fit two multiple, linear, mixed regression models to the data, one with DOHaDKNOWLEDGE score as the dependent variable and the other with diet quality score as the dependent. In both models, responses were clustered by respondents' neighbourhoods. Complete, internally consistent responses were available for 330 study-eligible respondents. Relative to Pregnancy GuidelineKNOWLEDGE, respondents had lower, more variable DOHaDKNOWLEDGE scores. Additionally, higher DOHaDKNOWLEDGE was associated with higher socio-economic position, older age, and lower parity, independent of Pregnancy GuidelineKNOWLEDGE. Diet quality during pregnancy was positively associated with DOHaDKNOWLEDGE, adjusting for sociodemographic factors. A subset of relatively high socio-economic position respondents was familiar with DOHaD. Greater familiarity with DOHaD was associated with better pregnancy diet quality, hinting that translating DOHaD knowledge to pregnant people may motivate improved pregnancy nutrition and thus later-life health for developing babies.
- Published
- 2019
- Full Text
- View/download PDF
32. Translating the Developmental Origins of Health and Disease concept to improve the nutritional environment for our next generations:A call for a reflexive, positive, multi-level approach
- Author
-
Luseadra McKerracher, Mary Barker, Tina Moffat, D Williams, and Deborah M. Sloboda
- Subjects
blame ,media_common.quotation_subject ,Nutritional Status ,Medicine (miscellaneous) ,030209 endocrinology & metabolism ,Health Promotion ,Disease ,Knowledge Translation (KT) ,Blame ,03 medical and health sciences ,0302 clinical medicine ,Knowledge translation ,Reflexivity ,Humans ,Conversation ,Child ,Life Style ,non-communicable diseases (NCDs) ,media_common ,Schools ,030219 obstetrics & reproductive medicine ,business.industry ,DOHaD ,Feeding Behavior ,Public relations ,Solidarity ,Diet ,mothers ,Disease concept ,business ,Psychology ,Inclusion (education) - Abstract
Evidence supporting the Developmental Origins of Health and Disease (DOHaD) hypothesis indicates that improving early life environments can reduce non-communicable disease risks and improve health over the lifecourse. A widespread understanding of this evidence may help to reshape structures, guidelines and individual behaviors to better the developmental conditions for the next generations. Yet, few efforts have yet been made to translate the DOHaD concept beyond the research community. To understand why, and to identify priorities for DOHaD Knowledge Translation (KT) programs, we review here a portion of published descriptions of DOHaD KT efforts and critiques thereof. We focus on KT targeting people equipped to apply DOHaD knowledge to their everyday home or work lives. We identified 17 reports of direct-to-public DOHaD KT that met our inclusion criteria. Relevant KT programs have been or are being initiated in nine countries, most focusing on secondary school students or care-workers-in-training; few target parents-to-be. Early indicators suggest that such programs can empower participants. Main critiques of DOHaD KT suggest it may overburden mothers with responsibility for children’s health and health environments, minimizing the roles of other people and institutions. Simultaneously, though, many mothers-to-be seek reliable guidance on prenatal health and nutrition, and would likely benefit from engagement with DOHaD KT. We thus recommend emphasizing solidarity, and bringing together people likely to one day become parents (youth), people planning pregnancies, expecting couples, care workers and policymakers into empowering conversation about DOHaD and about the importance and complexity of early life environments.
- Published
- 2019
- Full Text
- View/download PDF
33. Nutritional adversity, sex and reproduction: 30 years of DOHaD and what have we learned?
- Author
-
Deborah M. Sloboda and Patrycja A. Jazwiec
- Subjects
0301 basic medicine ,medicine.medical_specialty ,Offspring ,Endocrinology, Diabetes and Metabolism ,Nutritional Status ,Disease ,Biology ,Developmental psychology ,03 medical and health sciences ,0302 clinical medicine ,Endocrinology ,Risk Factors ,Internal medicine ,medicine ,Animals ,Humans ,Obesity ,Organism ,030219 obstetrics & reproductive medicine ,Reproductive function ,Reproduction ,Inheritance (genetic algorithm) ,medicine.disease ,Malnutrition ,030104 developmental biology ,Female ,Reprogramming - Abstract
It is well established that early life environmental signals, including nutrition, set the stage for long-term health and disease risk – effects that span multiple generations. This relationship begins early, in the periconceptional period and extends into embryonic, fetal and early infant phases of life. Now known as the Developmental Origins of Health and Disease (DOHaD), this concept describes the adaptations that a developing organism makes in response to early life cues, resulting in adjustments in homeostatic systems that may prove maladaptive in postnatal life, leading to an increased risk of chronic disease and/or the inheritance of risk factors across generations. Reproductive maturation and function is similarly influenced by early life events. This should not be surprising, since primordial germ cells are established early in life and thus vulnerable to early life adversity. A multitude of ‘modifying’ cues inducing developmental adaptations have been identified that result in changes in reproductive development and impairments in reproductive function. Many types of nutritional challenges including caloric restriction, macronutrient excess and micronutrient insufficiencies have been shown to induce early life adaptations that produce long-term reproductive dysfunction. Many pathways have been suggested to underpin these associations, including epigenetic reprogramming of germ cells. While the mechanisms still remain to be fully investigated, it is clear that a lifecourse approach to understanding lifetime reproductive function is necessary. Furthermore, investigations of the impacts of early life adversity must be extended to include the paternal environment, especially in epidemiological and clinical studies of offspring reproductive function.
- Published
- 2019
34. Fetal Growth Restriction Is Associated With Decreased Number of Ovarian Follicles and Impaired Follicle Growth in Young Adult Guinea Pig Offspring
- Author
-
Brad Matushewski, Deborah M. Sloboda, Xinglin Li, Bryan S. Richardson, and Patrycja A. Jazwiec
- Subjects
0301 basic medicine ,Offspring ,Guinea Pigs ,Growth Differentiation Factor 9 ,Ovary ,Biology ,Guinea pig ,Andrology ,03 medical and health sciences ,Follicle ,0302 clinical medicine ,Growth factor receptor ,Ovarian Follicle ,Pregnancy ,Follicular phase ,medicine ,Animals ,Caloric Restriction ,030219 obstetrics & reproductive medicine ,Fetal Growth Retardation ,Obstetrics and Gynecology ,Receptors, Somatomedin ,Maternal Nutritional Physiological Phenomena ,Antral follicle ,030104 developmental biology ,medicine.anatomical_structure ,In utero ,Prenatal Exposure Delayed Effects ,Female - Abstract
Background: The mechanisms mediating the impacts of fetal growth restriction (FGR) on follicular development are commonly studied in mouse/rat models, where ovarian development occurs largely during the early postnatal period. These models have shown that FGR is associated with premature follicle loss, early pubertal onset, and accelerated ovarian aging. Whether the same occurs in precocious species is unknown. Objective: Since guinea pig follicle development occurs in utero in a manner consistent with human ovarian development, we sought to determine whether FGR had similar impacts on guinea pig ovarian development. Methods: Dunkin-Hartley guinea pig dams were randomized to receive a control (CON) or a nutrient-restricted diet (FGR) prior to conception until weaning. Offspring ovaries were collected at prepubertal (postnatal day [P] 25) and young adult (P110) time points. Results: Prepubertal offspring exposed to FGR showed little differences in ovarian transcript levels and follicle counts. Young adult FGR offspring, however, showed reductions in the number of transitioning, primary, and antral follicles, as well as corpora lutea. This loss in follicles was associated with reduced insulin-like growth factor receptor and growth differentiation factor-9 messenger RNA levels in FGR P110 offspring compared to CON. Conclusion: We demonstrate that FGR in guinea pigs is accompanied by perturbations in signaling pathways essential for proper follicle growth and manifests as reductions in growing follicles in offspring, but these changes do not manifest until postpuberty. These data support the fact that accelerated reproductive maturation/aging is a conserved phenotype that is associated with in utero nutritional adversity.
- Published
- 2019
35. Acephate exposure during a perinatal life program to type 2 diabetes
- Author
-
Elaine Vieira, Audrei Pavanello, Tatiane Aparecida Ribeiro, Paulo Cezar de Freitas Mathias, Isabela Peixoto Martins, Kelly Valério Prates, Deborah M. Sloboda, Claudinéia Conationi da Silva Franco, Sandra da Silva Silveira, Flávio Andrade Francisco, Veridiana Mota Moreira, Rosiane Aparecida Miranda, Laize Peron Tófolo, Luiz Felipe Barella, Ananda Malta, Júlio Cezar de Oliveira, Kesia Palma-Rigo, Gabriel Sergio Fabricio, Rodrigo Mello Gomes, and Vander Silva Alves
- Subjects
0301 basic medicine ,Insecticides ,medicine.medical_specialty ,Offspring ,Physiology ,Type 2 diabetes ,010501 environmental sciences ,Biology ,Toxicology ,01 natural sciences ,03 medical and health sciences ,chemistry.chemical_compound ,Insulin resistance ,Pregnancy ,Internal medicine ,Lactation ,medicine ,Animals ,Humans ,Rats, Wistar ,Acephate ,0105 earth and related environmental sciences ,Mortality rate ,Organothiophosphorus Compounds ,Lipid metabolism ,medicine.disease ,Rats ,030104 developmental biology ,medicine.anatomical_structure ,Endocrinology ,Animals, Newborn ,Diabetes Mellitus, Type 2 ,chemistry ,Phosphoramides ,Female - Abstract
Acephate has been used extensively as an insecticide in agriculture. Its downstream sequelae are associated with hyperglycemia, lipid metabolism dysfunction, DNA damage, and cancer, which are rapidly growing epidemics and which lead to increased morbidity and mortality rates and soaring health-care costs. Developing interventions will require a comprehensive understanding of which excess insecticides during perinatal life can cause insulin resistance and type 2 diabetes. A Wistar rat animal model suggests that acephate exposure during pregnancy and lactation causes alterations in maternal glucose metabolism and programs the offspring to be susceptible to type 2 diabetes at adulthood. Therapeutic approaches based on preventive actions to food contaminated with insecticides during pregnancy and lactation could prevent new cases of type 2 diabetes.
- Published
- 2016
- Full Text
- View/download PDF
36. Fetal and neonatal outcomes after term and preterm delivery following betamethasone administration in twin pregnancies
- Author
-
Boris Tutschek, Hannah C. Gil, Andreas Plagemann, Joachim W. Dudenhausen, Wolfgang Henrich, Deborah M. Sloboda, Thorsten Braun, Alexander Weichert, and Thomas Harder
- Subjects
Adult ,medicine.medical_specialty ,Birth weight ,Gestational Age ,Betamethasone ,Cohort Studies ,Fetal Development ,03 medical and health sciences ,0302 clinical medicine ,Pregnancy ,Germany ,medicine ,Humans ,030212 general & internal medicine ,Twin Pregnancy ,Retrospective Studies ,Asphyxia ,030219 obstetrics & reproductive medicine ,Anthropometry ,Dose-Response Relationship, Drug ,Obstetrics ,business.industry ,Pregnancy Outcome ,Obstetrics and Gynecology ,Gestational age ,General Medicine ,Delivery, Obstetric ,medicine.disease ,Premature birth ,Pregnancy, Twin ,Premature Birth ,Female ,medicine.symptom ,business ,Premature rupture of membranes ,medicine.drug - Abstract
To investigate effects of betamethasone on fetal growth and neonatal outcomes in twins.A retrospective cohort study was conducted of twins delivered at one center in Berlin, Germany, between 1993 and 2011. The betamethasone group included twin pregnancies with preterm labor, cervical shortening, preterm premature rupture of membranes, or vaginal bleeding, and exposure to betamethasone between 23(+5) and 33(+6) weeks. The control group included twin pregnancies with no betamethasone exposure matched for length at delivery. Fetal growth and neonatal anthropometric data were analyzed by twin-pair structure, dose, and gestational age (linear mixed model).Overall, 1922 live-born twin pairs (653 betamethasone group, 1269 controls) were included. Compared with controls, late-preterm twins exposed to betamethasone were lighter (mean difference -126g), had a smaller head circumference (-0.4cm), and a shorter body length (-0.8cm) after adjustment for confounders (P0.05). Female neonates from mixed or same-sex twin pairs had a lower birth weight than controls (betamethasone ≤16mg: -114g; betamethasone 24mg: -124g; betamethasone24mg: -187g), with no detectable improvement in neonatal morbidity (hyperbilirubinemia, respiratory distress, asphyxia) or mortality.Betamethasone reduced birth weight, head circumference, and length of female preterm neonates in twin pairs in a dose-dependent manner. The neonatal mortality and morbidity were not improved by betamethasone.
- Published
- 2016
- Full Text
- View/download PDF
37. Adverse metabolic phenotype of adolescent girls with non-alcoholic fatty liver disease plus polycystic ovary syndrome compared with other girls and boys
- Author
-
Roger Hart, Oyekoya T. Ayonrinde, Leon A. Adams, Lawrence J. Beilin, Dorota A. Doherty, Martha Hickey, Wendy H. Oddy, Deborah M. Sloboda, John K. Olynyk, and Trevor A. Mori
- Subjects
medicine.medical_specialty ,endocrine system diseases ,030209 endocrinology & metabolism ,digestive system ,03 medical and health sciences ,0302 clinical medicine ,Sex hormone-binding globulin ,Insulin resistance ,Internal medicine ,medicine ,030219 obstetrics & reproductive medicine ,Hepatology ,biology ,Adiponectin ,business.industry ,Fatty liver ,Gastroenterology ,Case-control study ,nutritional and metabolic diseases ,Odds ratio ,medicine.disease ,Polycystic ovary ,female genital diseases and pregnancy complications ,digestive system diseases ,Endocrinology ,biology.protein ,business ,Body mass index - Abstract
Background and Aims: Non-alcoholic fatty liver disease (NAFLD) and polycystic ovary syndrome (PCOS) share risk associations of adiposity and insulin resistance. We examined the impact of a PCOS diagnosis on the metabolic phenotype of adolescent girls with NAFLD and compared this to girls without PCOS or NAFLD and to age-matched boys. Methods: Community-based adolescents from the Raine Cohort participated in assessments for NAFLD (572 girls and 592 boys) and PCOS (244 girls). One hundred and ninety-nine girls attended both assessments. Results: Amongst the 199 girls, PCOS was diagnosed in 16.1% and NAFLD in 18.6%. NAFLD was diagnosed in 10.1% of the boys. NAFLD was more prevalent in girls with PCOS than girls without PCOS (37.5% vs 15.1%, P = 0.003). Girls with NAFLD plus PCOS had greater adiposity (waist circumference, body mass index, suprailiac skinfold thickness [SST], serum androgens, high-sensitivity C-reactive protein, ferritin, homeostasis model assessment for insulin resistance (HOMA-IR), and lower serum sex hormone binding globulin levels than girls with NAFLD without a PCOS diagnosis (all P
- Published
- 2016
- Full Text
- View/download PDF
38. In uterobetamethasone affects 3β-hydroxysteroid dehydrogenase and inhibin-α immunoexpression during testis development
- Author
-
Paula Lombide, Graeme Martin, Helen Viotti, Deborah M. Sloboda, C. Pino, Daniel Cavestany, Graciela Pedrana, and G. Sanguinetti
- Subjects
Male ,0301 basic medicine ,endocrine system ,medicine.medical_specialty ,17-Hydroxysteroid Dehydrogenases ,Offspring ,Medicine (miscellaneous) ,Gestational Age ,030209 endocrinology & metabolism ,Betamethasone ,Immunoenzyme Techniques ,03 medical and health sciences ,Fetus ,0302 clinical medicine ,Pregnancy ,Internal medicine ,Testis ,Animals ,Medicine ,Inhibins ,Sheep ,business.industry ,030104 developmental biology ,Endocrinology ,Gene Expression Regulation ,In utero ,Gestation ,Female ,business ,Spermatogenesis ,Glucocorticoid ,Immunostaining ,medicine.drug - Abstract
Prenatal glucocorticoids, commonly used in women at risk of preterm delivery, can predispose the newborn to disease in later life. Since male reproductive function is likely to reflect testis development during fetal life, we studied the effects of prenatal glucocorticoids on two key intra-testicular factors that play roles in cellular proliferation and differentiation, 3β-hydroxysteroid dehydrogenase (3β-HSD) and inhibin-α. Pregnant sheep (n=42) were treated with betamethasone (0.5 mg/kg) or saline (control) at 104, 111 and 118 days of gestation (DG). Testicular tissue was sampled from fetuses at 121 and 132DG, and from lambs at 45 and 90 postnatal days (PD). Within the betamethasone treated group, 3β-HSD immunostaining area was greater at 121DG than at 90PD (P=0.04), but the intensity of immunostaining was higher at 90PD than at 121DG (P=0.04), 132DG (P=0.04) and 45PD (P=0.03). Control animals showed no changes in 3β-HSD area or intensity of immunostaining. No significant differences were observed between treated and control animals in immunostaining area, but immunostaining was more intense in the treated group than in the control group at 90PD (P=0.03). For inhibin-α, the proportion of immunostaining area declined in treated offspring from 121DG to 45PD, in contrast to control values, but recovered fully by 90PD, concomitantly with the onset of spermatogenesis. In conclusion, prenatal betamethasone increased the postnatal testicular expression of inhibin-α but reduced the expression of 3β-HSD. These effects could compromise androgen-mediated testicular development and therefore adult capacity for spermatogenesis.
- Published
- 2016
- Full Text
- View/download PDF
39. TNF, but not hyperinsulinemia or hyperglycemia, is a key driver of obesity‐induced monocytosis revealing that inflammatory monocytes correlate with insulin in obese male mice
- Author
-
Deborah M. Sloboda, Trevor C. Lau, Dessi Loukov, Jessica A. Breznik, Kevin P. Foley, Avee Naidoo, Christian Schulz, Jonathan D. Schertzer, and Dawn M. E. Bowdish
- Subjects
0301 basic medicine ,Blood Glucose ,Male ,medicine.medical_specialty ,insulin ,obesity ,Physiology ,Leukocytosis ,medicine.medical_treatment ,Immunology ,TNF ,Adipose tissue ,Inflammation ,Type 2 diabetes ,Monocytes ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Insulin resistance ,Monocytosis ,Physiology (medical) ,Internal medicine ,medicine ,Hyperinsulinemia ,Animals ,Antigens, Ly ,Original Research ,business.industry ,Tumor Necrosis Factor-alpha ,Monocyte ,Insulin ,medicine.disease ,Endocrinology and Metabolism ,Mice, Inbred C57BL ,030104 developmental biology ,medicine.anatomical_structure ,Endocrinology ,inflammation ,monocyte ,medicine.symptom ,business ,030217 neurology & neurosurgery - Abstract
Inflammation contributes to obesity‐related hyperinsulinemia and insulin resistance, which often precede type 2 diabetes. Inflammation is one way that obesity can promote insulin resistance. It is not clear if the extent of obesity, hyperinsulinemia, or hyperglycemia, underpins changes in cellular immunity during diet‐induced obesity. In particular, the requirement for obesity or directionality in the relationship between insulin resistance and monocyte characteristics is poorly defined. Inflammatory cytokines such as tumor necrosis factor (TNF) can contribute to insulin resistance. It is unclear if TNF alters monocytosis or specific markers of cellular immunity in the context of obesity. We measured bone marrow and blood monocyte characteristics in WT and TNF −/− mice that were fed obesogenic, high fat (HF) diets. We also used hyperglycemic Akita mice and mice implanted with insulin pellets in order to determine if glucose or insulin were sufficient to alter monocyte characteristics. We found that diet‐induced obesity in male mice increased the total number of monocytes in blood, but not in bone marrow. Immature, inflammatory (Ly6Chigh) monocytes decreased within the bone marrow and increased within peripheral blood of HF‐fed mice. We found that neither hyperinsulinemia nor hyperglycemia was sufficient to induce the observed changes in circulating monocytes in the absence of diet‐induced obesity. In obese HF‐fed mice, antibiotic treatment lowered insulin and insulin resistance, but did not alter circulating monocyte characteristics. Fewer Ly6Chigh monocytes were present within the blood of HF‐fed TNF −/− mice in comparison to HF‐fed wild‐type (WT) mice. The prevalence of immature Ly6Chigh monocytes in the blood correlated with serum insulin and insulin resistance irrespective of the magnitude of adipocyte or adipose tissue hypertrophy in obese mice. These data suggest that diet‐induced obesity instigates a TNF‐dependent increase in circulating inflammatory monocytes, which predicts increased blood insulin and insulin resistance independently from markers of adiposity or adipose tissue expansion.
- Published
- 2018
40. High-fat diet intake modulates maternal intestinal adaptations to pregnancy and results in placental hypoxia, as well as altered fetal gut barrier proteins and immune markers
- Author
-
Michael G. Surette, Michelle Saoi, Jim Petrik, Christian J. Bellissimo, Deborah M. Sloboda, Philip Britz-McKibbin, Katherine M. Kennedy, Wajiha Gohir, and Jessica G. Wallace
- Subjects
0301 basic medicine ,Lipopolysaccharides ,Male ,medicine.medical_specialty ,Physiology ,Placenta ,Inflammation ,Biology ,Gut flora ,Diet, High-Fat ,Proinflammatory cytokine ,Fetal Development ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Fetus ,Pregnancy ,Internal medicine ,medicine ,Animals ,Obesity ,Intestinal Mucosa ,Hypoxia ,Barrier function ,digestive, oral, and skin physiology ,medicine.disease ,biology.organism_classification ,Gastrointestinal Microbiome ,Intestines ,Mice, Inbred C57BL ,030104 developmental biology ,Endocrinology ,medicine.anatomical_structure ,Female ,medicine.symptom ,030217 neurology & neurosurgery - Abstract
Key points Maternal obesity has been associated with shifts in intestinal microbiota, which may contribute to impaired barrier function Impaired barrier function may expose the placenta and fetus to pro-inflammatory mediators We investigated the impacts of diet-induced obesity in mice on maternal and fetal intestinal structure and placental vascularization Diet-induced obesity decreased maternal intestinal short chain fatty acids and their receptors, impaired gut barrier integrity and was associated with fetal intestinal inflammation. Placenta from obese mothers showed blood vessel immaturity, hypoxia, increased transcript levels of inflammation, autophagy and altered levels of endoplasmic reticulum stress markers. These data suggest that maternal intestinal changes probably contribute to adverse placental adaptations and also impart an increased risk of obesity in the offspring via alterations in fetal gut development. Abstract Shifts in maternal intestinal microbiota have been implicated in metabolic adaptations to pregnancy. In the present study, we generated cohorts of female C57BL/6J mice fed a control (17% kcal fat, n = 10-14) or a high-fat diet (HFD 60% kcal from fat, n = 10-14; ad libitum) aiming to investigate the impact on the maternal gut microbiota, intestinal inflammation and gut barrier integrity, placental inflammation and fetal intestinal development at embryonic day 18.5. HFD was associated with decreased relative abundances of short-chain fatty acid (SCFA) producing genera during pregnancy. These diet-induced shifts paralleled decreased maternal intestinal mRNA levels of SCFA receptor Gpr41, modestly decreased cecal butyrate, and altered mRNA levels of inflammatory cytokines and immune cell markers in the maternal intestine. Maternal HFD resulted in impaired gut barrier integrity, with corresponding increases in circulating maternal levels of lipopolysaccharide (LPS) and tumour necrosis factor. Placentas from HFD dams demonstrated blood vessel immaturity and hypoxia; decreased free carnitine, acylcarnitine derivatives and trimethylamine-N-oxide; and altered mRNA levels of inflammation, autophagy, and ER stress markers. HFD exposed fetuses had increased activation of nuclear factor-kappa B and inhibition of the unfolded protein response in the developing intestine. Taken together, these data suggest that HFD intake prior to and during pregnancy shifts the composition of the maternal gut microbiota and impairs gut barrier integrity, resulting in increased maternal circulating LPS, which may ultimate contribute to changes in placental vascularization and fetal gut development.
- Published
- 2018
41. High-fat diet intake modulates maternal intestinal adaptations to pregnancy, and results in placental hypoxia and impaired fetal gut development
- Author
-
Wajiha Gohir, Katherine M. Kennedy, Jessica G. Wallace, Michelle Saoi, Christian J. Bellissimo, Philip Britz-McKibbin, Jim J. Petrik, Michael G. Surette, and Deborah M. Sloboda
- Subjects
2. Zero hunger ,0303 health sciences ,medicine.medical_specialty ,Pregnancy ,Fetus ,biology ,Inflammation ,Butyrate ,Hypoxia (medical) ,Gut flora ,medicine.disease ,biology.organism_classification ,3. Good health ,Proinflammatory cytokine ,03 medical and health sciences ,0302 clinical medicine ,Endocrinology ,Immune system ,030220 oncology & carcinogenesis ,Internal medicine ,medicine ,medicine.symptom ,030304 developmental biology - Abstract
Shifts in maternal intestinal microbiota have been implicated in metabolic adaptations to pregnancy. In this study we investigated how high-fat diet intake impacts the maternal gut microbiota, intestinal inflammation and gut barrier integrity, placental inflammation, and fetal intestinal development at E18.5. High-fat diet (HFD) was associated with decreased relative abundancesof SCFA producing genera during pregnancy. These diet-induced shifts paralleled decreased maternal intestinal mRNA levels of SCFA receptor Gpr41, modestly decreased cecal butyrate, and altered mRNA levels of inflammatory cytokines and immune cell markers in the maternal intestine. Maternal HFD resulted inimpaired gut barrier integrity, with corresponding increases in circulating maternal levels of LPS and TNF.Placentafromhigh-fat fed damsdemonstrated blood vessel immatu-rityand hypoxia, decreased freecarnitine, acylcarnitine derivatives, TMAO, as well as altered mRNA levels of inflammation, autophagy and ER stress markers. HFD exposed fetuses had increased activation of NF-κB and inhibition of the unfolded protein response in the developing intestine. Together, these data suggest that high-fat diet intake prior to and during pregnancy shifts the composition of the maternal gut microbiota and impairs gut barrier integrity, resulting in increased maternal circulating LPS, which may ultimate contribute to changes in placental vasculariza-tion and fetal gut development.Funding informationFarncombe Family Digestive Health Research Institute (KMK); Canadian Institute of Health Research (CJB); Canada Research Chairs Program (MGS, DMS); Natural Sciences and Engineering Research Council of Canada, Genome Canada (PBM).
- Published
- 2018
- Full Text
- View/download PDF
42. Sex-specific and lasting effects of a single course of antenatal betamethasone treatment on human placental 11β-HSD2
- Author
-
A.K. Hardt, F Braun, Deborah M. Sloboda, Wolfgang Henrich, Andreas Plagemann, L. Ehrlich, John Challis, and Thorsten Braun
- Subjects
0301 basic medicine ,Male ,medicine.medical_specialty ,Hydrocortisone ,Birth weight ,Placenta ,Umbilical cord ,Betamethasone ,Fetal Development ,03 medical and health sciences ,0302 clinical medicine ,Sex Factors ,Adrenocorticotropic Hormone ,Pregnancy ,Internal medicine ,11-beta-Hydroxysteroid Dehydrogenase Type 2 ,medicine ,Humans ,Glucocorticoids ,Fetus ,030219 obstetrics & reproductive medicine ,Anthropometry ,business.industry ,Obstetrics and Gynecology ,Transplacental ,Gestational age ,Organ Size ,030104 developmental biology ,Endocrinology ,medicine.anatomical_structure ,Reproductive Medicine ,Gestation ,Female ,business ,Head ,Developmental Biology ,medicine.drug - Abstract
Introduction We have previously shown that even a single course of antenatal betamethasone (BET) as an inductor for lung maturity reduces birth weight and head circumference. Moreover, animal studies link BET administration to alterations of the hypothalamic-pituitary-adrenal-gland-axis (HPA). The unhindered development of the fetal HPA axis is dependent on the function and activity of 11β-hydroxysteroiddehydrogenase type 2 (11β-HSD2), a transplacental cortisol barrier. Therefore, we investigated the effects of BET on this transplacental barrier and fetal growth. Methods Pregnant women treated with a single course of BET between 23 + 5 to 34 + 0 weeks of gestation were compared to gestational-age-matched controls. Placental size and neonatal anthropometrics were taken. Cortisol and ACTH levels were measured in maternal and umbilical cord blood samples. Placental 11β-hydroxysteroiddehydrogenase type 1 (11β-HSD1) protein levels and 11β-HSD2 protein and activity levels were determined. Parameters were analyzed independent of sex, and in subgroups divided by gender and gestational age. Results In term born females, BET administration was associated with reduced head circumference and decreased 11β-HSD2 protein levels and enzyme activity. Males treated with BET, especially those born prematurely, showed increased 11β-HSD2 protein levels. Conclusion A single course of BET alters placental glucocorticoid metabolism in a sex-specific manner. Decreased 11β-HSD2 levels in term born females may lead to an increased placental transfer of maternal cortisol and therefore result in a reduced head circumference and a higher risk for altered stress response in adulthood. Further research is needed to conclude the significance of increased 11β-HSD2 levels in males.
- Published
- 2018
43. Maternal nutrient restriction impairs young adult offspring ovarian signaling resulting in reproductive dysfunction and follicle loss†
- Author
-
Jim Petrik, Deborah M. Sloboda, Wajiha Gohir, Kaitlyn A. Chan, and Patrycja A. Jazwiec
- Subjects
0301 basic medicine ,endocrine system ,Receptors, Peptide ,Offspring ,Apoptosis ,Estrous Cycle ,Biology ,Andrology ,Phosphatidylinositol 3-Kinases ,03 medical and health sciences ,Follicle ,Ovarian Follicle ,Pregnancy ,Lactation ,medicine ,Animals ,Phosphorylation ,Rats, Wistar ,Ovarian follicle ,Caloric Restriction ,Estrous cycle ,Prenatal nutrition ,Caspase 3 ,Reproduction ,Ovary ,Maternal Nutritional Physiological Phenomena ,Cell Biology ,General Medicine ,medicine.disease ,Antral follicle ,Rats ,030104 developmental biology ,medicine.anatomical_structure ,Reproductive Medicine ,Prenatal Exposure Delayed Effects ,Receptors, FSH ,Female ,Follicle Stimulating Hormone ,Proto-Oncogene Proteins c-akt ,Receptors, Transforming Growth Factor beta ,Signal Transduction - Abstract
Reproductive abnormalities are included as health complications in offspring exposed to poor prenatal nutrition. We have previously shown in a rodent model that offspring born to nutrient restriction during pregnancy are born small, enter puberty early, and display characteristics of early ovarian aging as adults. The present study investigated whether key proteins involved in follicle recruitment and growth mediate ovarian follicle loss. Pregnant rats were randomized to a standard diet throughout pregnancy and lactation (CON), or a calorie-restricted (50% of control) diet (UN) during pregnancy. Offspring reproductive phenotype was investigated at postnatal days 4, 27, and 65. Maternal UN resulted in young adult (P65) irregular estrous cyclicity due to persistent estrus, a significant loss of antral follicles, corpora lutea, and an increase in atretic follicles. This decrease in growing follicles in UN offspring appears to be due to increased apoptosis as seen by immunopositive staining of pro-apoptotic factor CASP3 (caspase 3) in ovaries of young adult offspring. UN prepubertal offspring had reduced expression levels of Fshr in antral follicles, which may contribute to a decrease in PI3K/AKT activation evident as a decrease in pAKT immunolocalization in prepubertal antral follicles. Moreover, neonatal ovaries of UN offspring show decreased levels of immunopositive staining for AMHR2 (anti-mullerian hormone receptor 2). Collectively, these data demonstrate that maternal UN during pregnancy impacts ovarian function in offspring as early as P65 and provides a model for understanding the mechanisms driving early life UN-induced follicle loss and reproductive dysfunction.
- Published
- 2018
- Full Text
- View/download PDF
44. The murine female intestinal microbiota does not shift throughout the estrous cycle
- Author
-
Jessica G. Wallace, Jake C. Szamosi, Deborah M. Sloboda, Michael G. Surette, and Ryan Potts
- Subjects
0301 basic medicine ,Physiology ,Maternal Health ,lcsh:Medicine ,Biochemistry ,Mice ,Pregnancy ,RNA, Ribosomal, 16S ,Medicine and Health Sciences ,lcsh:Science ,Data Management ,Multidisciplinary ,Obstetrics and Gynecology ,Genomics ,Intestines ,RNA, Bacterial ,Medical Microbiology ,Gestation ,Female ,Anatomy ,Research Article ,Microbial Taxonomy ,Computer and Information Sciences ,Estrous Cycle ,Microbial Genomics ,Biology ,Microbiology ,03 medical and health sciences ,Immune system ,Genetics ,Sex Hormones ,medicine ,Animals ,Endocrine system ,Microbiome ,Feces ,Taxonomy ,Estrous cycle ,Fetus ,Bacteria ,lcsh:R ,Organisms ,Biology and Life Sciences ,Estrogens ,medicine.disease ,Hormones ,Gastrointestinal Microbiome ,Gastrointestinal Tract ,030104 developmental biology ,Women's Health ,lcsh:Q ,Physiological Processes ,Digestive System - Abstract
Pregnancy is accompanied by maternal physiological adaptations including metabolic, endocrine, immune, cardiovascular, skeletomuscular and neurological modifications that facilitate fetal and placental growth and development. Emerging evidence suggests that the maternal intestinal microbiota is modified over the course of healthy pregnancy. We have recently identified a maternal intestinal microbial shift within hours of conception; a shift that continued with advancing gestation. It is possible that maternal gut bacterial profiles might be associated with the known endocrine changes that accompany the female reproductive (estrous) cycle. Methods: To determine whether the estrous cycle influenced the shifts in the maternal intestinal microbiota, time-matched fecal pellets were collected daily for 3 consecutive estrous cycles from individually housed, non-pregnant female C57BL/6J mice (n = 10) fed a control diet. Estrous stage was identified by cell type predominance in vaginal cytological samples. The corresponding fecal pellets for each estrous stage were processed for bacterial 16S rRNA sequencing of the variable 3 (V3) region. Results: Estrous cycle stage accounted for a very small and not statistically significant proportion of the variation in the fecal microbiota according to PERMANOVA testing performed on Bray-Curtis dissimilarity scores. These values displayed no significant clustering of fecal microbial communities by estrous stage. Conclusion: The estrous cycle does not result in any significant shift in the intestinal microbial community in the reproductively mature, regularly cycling female mouse.
- Published
- 2018
45. Adolescents as agents of healthful change through scientific literacy development: A school-university partnership program in New Zealand
- Author
-
Helen A. Mora, Deborah M. Sloboda, Mark H. Vickers, Susan M. B. Morton, and Jacquie L. Bay
- Subjects
lcsh:LC8-6691 ,Medical education ,lcsh:Special aspects of education ,Research ,Behavior change ,Nature of Science ,030209 endocrinology & metabolism ,Health literacy ,Science education ,lcsh:Education (General) ,lcsh:LB5-3640 ,Education ,Scientific evidence ,lcsh:Theory and practice of education ,03 medical and health sciences ,0302 clinical medicine ,Scientific literacy ,Health promotion ,Knowledge translation ,Pedagogy ,030212 general & internal medicine ,lcsh:L ,lcsh:L7-991 ,Psychology ,lcsh:Education - Abstract
Background Scientific literacy development is widely emphasized as the overarching goal of science education. It encompasses development of understanding of the nature of science as well as knowledge, attitudes, and values that contribute to empowering adolescents to engage with and make evidence-based decisions about socioscientific issues. Scientific literacy development is enhanced when learning is contextualized in exploration of socioscientific issues. Noncommunicable diseases (NCDs) associated with a combination of obesity and adverse environmental exposures are examples of pressing health-related SSIs facing the world today. Evidence emerging from the field of Developmental Origins of Health and Disease (DOHaD) has identified adolescence as a key life-phase where population-wide education-based interventions that empower teens to engage in science-based health-promoting behaviors could significantly change the course of this epidemic. To achieve this, learning resources that support scientific and health literacy development contextualized in issues linking NCD risk and DOHaD are required. The Healthy Start to Life Education for Adolescents Project is a school-university partnership program designed to support scientific and health literacy development, knowledge translation, and participant-led actions relating to NCD risk prevention. This study assesses the impact of program participation in a cohort of 11–14-year-olds in New Zealand. Evaluation comprised analysis of individually matched questionnaires, pre-, 3 months, and 12 months post-intervention (n = 201) and 6 months post-intervention interviews (n = 40). Results Positive engagement in science learning occurred. Positive changes in health-related awareness and attitudes 3 months post-intervention were sustained to 12 months. Adolescents reporting pre-intervention dietary behaviors associated with increased obesity risk reported sustained positive behavior changes (p
- Published
- 2017
- Full Text
- View/download PDF
46. The influence of antenatal exposure to phthalates on subsequent female reproductive development in adolescence: a pilot study
- Author
-
Jeffrey A. Keelan, Hanne Frederiksen, Martha Hickey, Deborah M. Sloboda, N E Skakkebaek, Dorota A. Doherty, Craig E. Pennell, K M Main, Roger Hart, and John P. Newnham
- Subjects
Embryology ,medicine.medical_specialty ,Adolescent ,Phthalic Acids ,Physiology ,Pilot Projects ,Cohort Studies ,chemistry.chemical_compound ,Endocrinology ,Sex hormone-binding globulin ,Pregnancy ,medicine ,Humans ,Androstenedione ,Child ,Testosterone ,Gynecology ,biology ,business.industry ,Reproduction ,Puberty ,Phthalate ,Infant ,Obstetrics and Gynecology ,Anti-Müllerian hormone ,Cell Biology ,Adolescent Development ,medicine.disease ,Reproductive Medicine ,chemistry ,Child, Preschool ,Prenatal Exposure Delayed Effects ,biology.protein ,Menarche ,Gestation ,Female ,business - Abstract
We hypothesised that antenatal exposure to ubiquitous phthalates may lead to an earlier menarche and a lower prevalence of polycystic ovarian syndrome (PCOS) and polycystic ovarian morphology (PCO) in adolescence. The Western Australian Pregnancy Cohort (Raine) Study recruited 3000 women at 18 weeks of gestation in 1989–1991, 1377 had antenatal serum stored without thawing at −80 °C. An unselected subset was evaluated in the early follicular phase for PCO and PCOS by ultrasound and serum evaluation in adolescence. Serum was analysed for anti-Müllerian hormone (AMH), inhibin B, sex hormone binding globulin (SHBG), testosterone, androstenedione and DHEAS. Four hundred microlitres of the frozen maternal serum underwent isotope-diluted liquid chromatography–tandem mass spectrometry, with preceding enzymatic deconjugation followed by solid-phase extraction to determine phthalate exposure. Two hundred and forty four girls attended assessment and most common phthalate metabolites were detectable in the majority of the 123 samples available. Several phthalates were negatively associated with maternal SHBG, and associations with maternal androgens were less consistent. The sum of the metabolites of di-(2-ethylhexyl) phthalate was associated with a non-significant tendency towards an earlier age at menarche (P=0.069). Uterine volume was positively associated with mono-(carboxy-iso-octyl) phthalate (P=0.018). Exposure to monoethyl phthalate (MEP) and the sum of all phthalate metabolites (Σall phth.m) were protective against PCOS in adolescence (P=0.001 andP=0.005 respectively). There were negative associations of MEP with PCO (P=0.022) and of MEP with serum AMH (P=0.031). Consequently, our data suggest that antenatal exposure to environmental phthalates may be associated with oestrogenic and/or anti-androgenic reproductive effects in adolescent girls.
- Published
- 2014
- Full Text
- View/download PDF
47. Perinatal programming of gut microbiota and immunity
- Author
-
Deborah M. Sloboda and A. L. Kozyrskyj
- Subjects
0301 basic medicine ,Pregnancy ,biology ,Gastrointestinal Microbiome ,Medicine (miscellaneous) ,Gut flora ,biology.organism_classification ,medicine.disease ,Infant newborn ,03 medical and health sciences ,030104 developmental biology ,Immunity ,Immunology ,medicine - Published
- 2015
- Full Text
- View/download PDF
48. Fructose, pregnancy and later life impacts
- Author
-
Sheridan Gentili, Deborah M. Sloboda, Carla R Toop, Ousseynou Sarr, Timothy R. H. Regnault, Regnault, Timothy RH, Gentili, Sheridan, Sarr, Ousseynou, Toop, Carla R, and Sloboda, Deborah M
- Subjects
Male ,medicine.medical_specialty ,Sucrose ,placenta ,Physiology ,Offspring ,Fructose ,Biology ,liver ,Diet, High-Fat ,Models, Biological ,fructose ,Fetal Development ,chemistry.chemical_compound ,Child Development ,Dietary Sucrose ,Non-alcoholic Fatty Liver Disease ,Pregnancy ,Physiology (medical) ,Internal medicine ,Diabetes mellitus ,Dietary Carbohydrates ,medicine ,Animals ,Humans ,Obesity ,Child ,Pharmacology ,offspring ,Perinatal Exposure ,Infant ,sucrose ,Maternal Nutritional Physiological Phenomena ,medicine.disease ,Placentation ,Fatty Liver ,Gestational diabetes ,fetus ,Endocrinology ,Diabetes Mellitus, Type 2 ,Liver ,chemistry ,Female ,pregnancy ,Child Nutritional Physiological Phenomena - Abstract
Fructose is an increasingly common constituent of the Westernized diet due to cost and production efficiencies. Although an integral component of our pre-industrial revolution diet, over the past two decades human and animal studies have highlighted that excessive fructose intake appears to be associated with adverse metabolic effects. Excessive intake of fructose is the combined result of increased total energy consumption and increased portion sizes of foods, which often incorporate the fructose-containing sugars sucrose and high-fructose corn-syrup (HFCS). The adverse metabolic effects following excessive fructose consumption have become a hot topic in mainstream media and there is now rigorous scientific debate regarding periods of exposure, dosage levels, interactive effects with other sugars and fats and mechanisms underlying the actions of fructose. There is still a degree of controversy regarding the extent to which sugars such as sucrose and HFCS have contributed to the current epidemic of obesity and diabetes. Furthermore, an increasing number of infants are being exposed to sugar-sweetened food and beverages before birth and during early postnatal life, highlighting the importance of determining the long-term effects of this perinatal exposure on the developing offspring. There are limited human observational and controlled studies identifying associations of excessive sweetened food and beverage consumption with poor pregnancy outcomes. Animal research has demonstrated an increased incidence of gestational diabetes as well as altered maternal, fetal and offspring metabolic function, although the long-term effects and the mechanism underlying these perturbations are ill defined. This review aims to understand the role of early life fructose exposure in modifying postnatal risk of disease in the offspring, focusing on fructose intake during pregnancy and in early postnatal life.
- Published
- 2013
- Full Text
- View/download PDF
49. Effects of glucocorticoid treatment given in early or late gestation on growth and development in sheep
- Author
-
Thorsten Braun, Timothy J. M. Moss, Dorota A. Doherty, Graeme R. Polglase, Ilias Nitsos, Shaofu Li, John P. Newnham, Deborah M. Sloboda, and John R. G. Challis
- Subjects
Fetus ,medicine.medical_specialty ,Pregnancy ,business.industry ,Medicine (miscellaneous) ,medicine.disease ,Postnatal age ,Endocrinology ,Internal medicine ,embryonic structures ,medicine ,Betamethasone ,Gestation ,business ,hormones, hormone substitutes, and hormone antagonists ,Dexamethasone ,Glucocorticoid ,medicine.drug ,Hormone - Abstract
Antenatal corticosteroids are used to augment fetal lung maturity in human pregnancy. Dexamethasone (DEX) is also used to treat congenital adrenal hyperplasia of the fetus in early pregnancy. We previously reported effects of synthetic corticosteroids given to sheep in early or late gestation on pregnancy length and fetal cortisol levels and glucocorticoids alter plasma insulin-like growth factor (IGF) and insulin-like growth factor binding protein (IGFBP) concentrations in late pregnancy and reduce fetal weight. The effects of administering DEX in early pregnancy on fetal organ weights and betamethasone (BET) given in late gestation on weights of fetal brain regions or organ development have not been reported. We hypothesized that BET or DEX administration at either stage of pregnancy would have deleterious effects on fetal development and associated hormones. In early pregnancy, DEX was administered as four injections at 12-hourly intervals over 48 h commencing at 40–42 days of gestation (dG). There was no consistent effect on fetal weight, or individual fetal organ weights, except in females at 7 months postnatal age. When BET was administered at 104, 111 and 118 dG, the previously reported reduction in total fetal weight was associated with significant reductions in weights of fetal brain, cerebellum, heart, kidney and liver. Fetal plasma insulin, leptin and triiodothyronine were also reduced at different times in fetal and postnatal life. We conclude that at the amounts given, the sheep fetus is sensitive to maternal administration of synthetic glucocorticoid in late gestation, with effects on growth and metabolic hormones that may persist into postnatal life.
- Published
- 2013
- Full Text
- View/download PDF
50. Maternal High-Fat Diet-Induced Loss of Fetal Oocytes Is Associated with Compromised Follicle Growth in Adult Rat Offspring1
- Author
-
Jim Petrik, Caroline J. Moore, Wajiha Gohir, Pauline E. Chang, Michael W. Tsoulis, Kaitlyn A. Chan, Deborah M. Sloboda, Mark H. Vickers, and Kristin L. Connor
- Subjects
0301 basic medicine ,endocrine system ,medicine.medical_specialty ,Pregnancy ,Offspring ,Follicular atresia ,Ovary ,Cell Biology ,General Medicine ,Biology ,medicine.disease ,Antral follicle ,03 medical and health sciences ,030104 developmental biology ,medicine.anatomical_structure ,Endocrinology ,Reproductive Medicine ,Internal medicine ,Lactation ,medicine ,Folliculogenesis ,Ovarian follicle - Abstract
Maternal obesity predisposes offspring to metabolic and reproductive dysfunction. We have shown previously that female rat offspring born to mothers fed a high-fat (HF) diet throughout pregnancy and lactation enter puberty early and display aberrant reproductive cyclicity. The mechanisms driving this reproductive phenotype are currently unknown thus we investigated whether changes in ovarian function were involved. Wistar rats were mated and randomized to: dams fed a control diet (CON) or dams fed a HF diet from conception until the end of lactation (HF). Ovaries were collected from fetuses at Embryonic Day (E) 20, and neonatal ovaries at Day 4 (P4), prepubertal ovaries at P27 and adult ovaries at P120. In a subset of offspring, the effects of a HF diet fed postweaning were evaluated. The present study shows that fetuses of mothers fed a HF diet had significantly fewer oocytes at E20, and in neonates, have reduced AMH signaling that may facilitate an increased number of assembled primordial follicles. Both prepubertally and in adulthood, ovaries show increased follicular atresia. As adults, offspring have reduced FSH responsiveness, low expression levels of estrogen receptor alpha (Eralpha), the oocyte-secreted factor, Gdf9, oocyte-specific RNA binding protein, Dazl, and high expression levels of the granulosa-cell derived factor, AMH, in antral follicles. Together, these data suggest that ovarian compromise in offspring born to HF-fed mothers may arise from changes already observable in the fetus and neonate and in the long term, associated with increased follicular atresia through adulthood.
- Published
- 2016
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.