Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 54
Filter
1.
Article in English | MEDLINE | ID: mdl-38783717

ABSTRACT

CONTEXT: Small cohorts of youth with congenital adrenal hyperplasia (CAH) demonstrate increased risk of obesity and poor cardiometabolic health. OBJECTIVE: To determine the odds of cardiometabolic-related diagnoses in youth with CAH compared to matched controls in a cross-sectional analysis in a large, multisite database (PEDSnet). DESIGN: Electronic health record data (2009-2019) were used to determine odds of cardiometabolic-related outcomes based on diagnosis, anthropometric and laboratory data using logistic regression among youth with CAH vs. controls. SETTING: Six PEDSnet sites. PATIENTS OR OTHER PARTICIPANTS: Youth with CAH and >1 outpatient visit in PEDSnet (n=1,647) were propensity-score matched on 8 variables to controls (n=6,588). A subset of youth with classic CAH (n=547, with glucocorticoid and mineralocorticoid prescriptions) were matched to controls (n=2,188). INTERVENTION(S): N/A. MAIN OUTCOME MEASURE(S): Odds of having cardiometabolic-related diagnoses among youth over 2 years with CAH compared to matched controls. RESULTS: Outcomes were calculated for all individuals with CAH (median age at last visit 12.9 years [7.3, 17.6]) and a subset with classic CAH (median age at last visit 11.6 years [4.7, 17.5]) compared to their matched controls. All patients with CAH had higher odds of overweight/obesity (odds ratio [95% confidence interval] 3.63 [3.24,4.07]), hypertension (3.07 [2.60,3.64]), dysglycemia (1.95 [1.35,2.82], dyslipidemia (2.28 [1.79,2.91]) and liver dysfunction (2.30 [1.91,2.76]) compared to matched controls. Patients with classic CAH had higher odds of overweight/obesity (3.21 [2.61,3.93]), hypertension (8.22 [6.71,10.08]), and liver dysfunction (2.11 [1.55,2.89]) compared to matched controls. CONCLUSIONS: Overall, youth with CAH are at increased risk of diagnoses related to worse cardiometabolic health.

2.
Am J Physiol Heart Circ Physiol ; 327(2): H315-H330, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38819385

ABSTRACT

Cardiovascular dysfunctions complicate 10-20% of pregnancies, increasing the risk for postpartum mortality. Various gestational insults, including preeclampsia are reported to be associated with adverse maternal cardiovascular outcomes. One such insult, gestational hyperandrogenism increases the risk for preeclampsia and other gestational morbidities but its impact on postpartum maternal health is not well known. We hypothesize that gestational hyperandrogenism such as testosterone (T) excess will adversely impact the maternal heart in the postpartum period. Pregnant ewes were injected with T propionate from day 30 to day 90 of gestation (term 147 days). Three months postpartum, echocardiograms, plasma cytokine profiles, cardiac morphometric, and molecular analysis were conducted [control (C) n = 6, T-treated (T) n = 7 number of animals]. Data were analyzed by two-tailed Student's t test and Cohen's effect size (d) analysis. There was a nonsignificant large magnitude decrease in cardiac output (7.64 ± 1.27 L/min vs. 10.19 ± 1.40, P = 0.22, d = 0.81) and fractional shortening in the T ewes compared with C (35.83 ± 2.33% vs. 41.50 ± 2.84, P = 0.15, d = 0.89). T treatment significantly increased 1) left ventricle (LV) weight-to-body weight ratio (2.82 ± 0.14 g/kg vs. 2.46 ± 0.08) and LV thickness (14.56 ± 0.52 mm vs. 12.50 ± 0.75), 2) proinflammatory marker [tumor necrosis factor-alpha (TNF-α)] in LV (1.66 ± 0.35 vs. 1.06 ± 0.18), 3) LV collagen (Masson's Trichrome stain: 3.38 ± 0.35 vs. 1.49 ± 0.15 and Picrosirius red stain: 5.50 ± 0.32 vs. 3.01 ± 0.23), 4) markers of LV apoptosis, including TUNEL (8.3 ± 1.1 vs. 0.9 ± 0.18), bcl-2-associated X protein (Bax)+-to-b-cell lymphoma 2 (Bcl2)+ ratio (0.68 ± 0.30 vs. 0.13 ± 0.02), and cleaved caspase 3 (15.4 ± 1.7 vs. 4.4 ± 0.38). These findings suggest that gestational testosterone excess adversely programs the maternal LV, leading to adverse structural and functional consequences in the postpartum period.NEW & NOTEWORTHY Using a sheep model of human translational relevance, this study provides evidence that excess gestational testosterone exposure such as that seen in hyperandrogenic disorders adversely impacts postpartum maternal hearts.


Subject(s)
Postpartum Period , Animals , Female , Pregnancy , Sheep , Testosterone/blood , Ventricular Function, Left , Testosterone Propionate/toxicity , Cytokines/blood , Cytokines/metabolism , Cardiac Output , Gestational Age
3.
Mol Cell Endocrinol ; 588: 112234, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38588858

ABSTRACT

Hyperandrogenic disorders, such as polycystic ovary syndrome, are often associated with metabolic disruptions such as insulin resistance and hyperinsulinemia. Studies in sheep, a precocial model of translational relevance, provide evidence that in utero exposure to excess testosterone during days 30-90 of gestation (the sexually dimorphic window where males naturally experience elevated androgens) programs insulin resistance and hyperinsulinemia in female offspring. Extending earlier findings that adverse effects of testosterone excess are evident in fetal day 90 pancreas, the end of testosterone treatment, the present study provides evidence that transcriptomic and phenotypic effects of in utero testosterone excess on female pancreas persist after cessation of treatment, suggesting lasting organizational changes, and induce a male-like phenotype in female pancreas. These findings demonstrate that the female pancreas is susceptible to programmed masculinization during the sexually dimorphic window of fetal development and shed light on underlying connections between hyperandrogenism and metabolic homeostasis.


Subject(s)
Pancreas , Testosterone , Transcriptome , Animals , Female , Sheep , Transcriptome/drug effects , Transcriptome/genetics , Pregnancy , Pancreas/metabolism , Pancreas/drug effects , Male , Prenatal Exposure Delayed Effects/metabolism , Insulin Resistance , Hyperandrogenism/metabolism , Hyperandrogenism/genetics , Fetal Development/drug effects , Sex Characteristics
4.
Sci Rep ; 14(1): 6230, 2024 03 14.
Article in English | MEDLINE | ID: mdl-38486090

ABSTRACT

Gestational hyperandrogenism is a risk factor for adverse maternal and offspring outcomes with effects likely mediated in part via disruptions in maternal lipid homeostasis. Using a translationally relevant sheep model of gestational testosterone (T) excess that manifests maternal hyperinsulinemia, intrauterine growth restriction (IUGR), and adverse offspring cardiometabolic outcomes, we tested if gestational T excess disrupts maternal lipidome. Dimensionality reduction models following shotgun lipidomics of gestational day 127.1 ± 5.3 (term 147 days) plasma revealed clear differences between control and T-treated sheep. Lipid signatures of gestational T-treated sheep included higher phosphoinositides (PI 36:2, 39:4) and lower acylcarnitines (CAR 16:0, 18:0, 18:1), phosphatidylcholines (PC 38:4, 40:5) and fatty acids (linoleic, arachidonic, Oleic). Gestational T excess activated phosphatidylethanolamines (PE) and PI biosynthesis. The reduction in key fatty acids may underlie IUGR and activated PI for the maternal hyperinsulinemia evidenced in this model. Maternal circulatory lipids contributing to adverse cardiometabolic outcomes are modifiable by dietary interventions.


Subject(s)
Cardiovascular Diseases , Hyperandrogenism , Hyperinsulinism , Pregnancy , Female , Sheep , Animals , Phosphatidylethanolamines , Phosphatidylinositols , Testosterone , Fatty Acids , Homeostasis
5.
Sci Rep ; 14(1): 7401, 2024 03 28.
Article in English | MEDLINE | ID: mdl-38548847

ABSTRACT

Lipids play a critical role in neonate development and breastmilk is the newborn's major source of lipids. Milk lipids directly influence the neonate plasma lipid profile. The milk lipidome is dynamic, influenced by maternal factors and related to the maternal plasma lipidome. The close inter-relationship between the maternal plasma, milk and neonate plasma lipidomes is critical to understanding maternal-child health and nutrition. In this exploratory study, lipidomes of blood and breast milk from Suffolk sheep and matched lamb blood (n = 13), were profiled on day 34 post birth by untargeted mass spectrometry. Comparative multivariate analysis of the three matrices identified distinct differences in lipids and class of lipids amongst them. Paired analysis identified 346 differential lipids (DL) and 31 correlated lipids (CL) in maternal plasma and milk, 340 DL and 32 CL in lamb plasma and milk and 295 DL and 16 CL in maternal plasma and lamb plasma. Conversion of phosphatidic acid to phosphatidyl inositol was the most active pathway in lamb plasma compared to maternal plasma. This exploratory study illustrates the partitioning of lipids across maternal plasma, milk and lamb plasma and the dynamic relationship between them, reiterating the need to study these three matrices as one biological system.


Subject(s)
Lipidomics , Milk , Female , Animals , Sheep , Humans , Milk/metabolism , Milk, Human/metabolism , Nutritional Status , Plasma , Lipids
6.
Mol Cell Endocrinol ; 588: 112202, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38552943

ABSTRACT

Developmental exposure to endocrine disruptors like bisphenol A (BPA) are implicated in later-life metabolic dysfunction. Leveraging a unique sheep model of developmental programming, we conducted an exploratory analysis of the programming effects of BPA on the endocrine pancreas. Pregnant ewes were administered environmentally relevant doses of BPA during gestational days (GD) 30-90, and pancreata from female fetuses and adult offspring were analyzed. Prenatal BPA exposure induced a trend toward decreased islet insulin staining and ß-cell count, increased glucagon staining and α-cell count, and increased α-cell/ß-cell ratio. Findings were most consistent in fetal pancreata assessed at GD90 and in adult offspring exposed to the lowest BPA dose. While not assessed in fetuses, adult islet fibrosis was increased. Collectively, these data provide further evidence that early-life BPA exposure is a likely threat to human metabolic health. Future studies should corroborate these findings and decipher the molecular mechanisms of BPA's developmental endocrine toxicity.


Subject(s)
Benzhydryl Compounds , Islets of Langerhans , Phenols , Prenatal Exposure Delayed Effects , Animals , Benzhydryl Compounds/toxicity , Female , Phenols/toxicity , Pregnancy , Sheep , Prenatal Exposure Delayed Effects/chemically induced , Prenatal Exposure Delayed Effects/pathology , Islets of Langerhans/drug effects , Islets of Langerhans/metabolism , Islets of Langerhans/pathology , Endocrine Disruptors/toxicity , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/pathology , Maternal Exposure/adverse effects , Insulin/metabolism , Fetus/drug effects , Glucagon-Secreting Cells/drug effects , Glucagon-Secreting Cells/metabolism , Glucagon-Secreting Cells/pathology
7.
Endocrinology ; 165(2)2023 Dec 23.
Article in English | MEDLINE | ID: mdl-38060679

ABSTRACT

Gestational hyperandrogenism adversely impacts offspring health. Using an ovine model, we found that prenatal testosterone (T) excess adversely affects growth and cardiometabolic outcomes in female offspring and produces sex-specific effects on fetal myocardium. Since lipids are essential to cardiometabolic function, we hypothesized that prenatal T excess leads to sex-specific disruptions in lipid metabolism at birth. Shotgun lipidomics was performed on the plasma samples collected 48 hours after birth from female (F) and male (M) lambs of control (C) and (T) sheep (CF = 4, TF = 7, CM = 5, TM = 10) and data were analyzed by univariate analysis, multivariate dimensionality reduction modeling followed by functional enrichment, and pathway analyses. Biosynthesis of phosphatidylserine was the major pathway responsible for sex differences in controls. Unsupervised and supervised models showed separation between C and T in both sexes with glycerophospholipids and glycerolipids classes being responsible for the sex differences between C and T. T excess increased cholesterol in females while decreasing phosphatidylcholine levels in male lambs. Specifically, T excess: 1) suppressed the phosphatidylethanolamine N-methyltransferase (PEMT) phosphatidylcholine synthesis pathway overall and in TM lambs as opposed to suppression of carnitine levels overall and TF lambs; and 2) activated biosynthesis of ether-linked (O-)phosphatidylethanolamine and O-phosphatidylcholine from O-diacylglycerol overall and in TF lambs. Higher cholesterol levels could underlie adverse cardiometabolic outcomes in TF lambs, whereas suppressed PEMT pathway in TM lambs could lead to endoplasmic reticulum stress and defective lipid transport. These novel findings point to sex-specific effects of prenatal T excess on lipid metabolism in newborn lambs, a precocial ovine model of translational relevance.


Subject(s)
Cardiovascular Diseases , Hyperandrogenism , Pregnancy , Animals , Sheep , Female , Male , Animals, Newborn , Lipidomics , Testosterone/pharmacology , Phosphatidylcholines , Cholesterol
8.
Placenta ; 142: 119-127, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37699274

ABSTRACT

INTRODUCTION: This study was designed to test the efficacy of an ultrasound flow measurement method to evaluate placental function in a hyperandrogenic sheep model that produces placental morphologic changes and an intrauterine growth restriction (IUGR) phenotype. MATERIALS AND METHODS: Pregnant ewes were assigned randomly between control (n = 12) and testosterone-treatment (T-treated, n = 22) groups. The T-treated group was injected twice weekly intramuscularly (IM) with 100 mg testosterone propionate. Control sheep were injected with corn oil vehicle. Lambs were delivered at 119.5 ± 0.48 days gestation. At the time of delivery of each lamb, flow spectra were generated from one fetal artery and two fetal veins, and the spectral envelopes examined using fast Fourier transform analysis. Base 10 logarithms of the ratio of the amplitudes of the maternal and fetal spectral peaks (LRSP) in the venous power spectrum were compared in the T-treated and control populations. In addition, we calculated the resistive index (RI) for the artery defined as ((peak systole - min diastole)/peak systole). Two-tailed T-tests were used for comparisons. RESULTS: LRSPs, after removal of significant outliers, were -0.158 ± 0.238 for T-treated and 0.057 ± 0.213 for control (p = 0.015) animals. RIs for the T-treated sheep fetuses were 0.506 ± 0.137 and 0.497 ± 0.086 for controls (p = 0.792) DISCUSSION: LRSP analysis distinguishes between T-treated and control sheep, whereas RIs do not. LRSP has the potential to identify compromised pregnancies.


Subject(s)
Fetus , Placenta , Sheep , Pregnancy , Animals , Female , Humans , Placenta/blood supply , Fetus/blood supply , Umbilical Veins , Arteries , Umbilical Arteries , Fetal Growth Retardation/veterinary
9.
Endocrinology ; 164(6)2023 04 17.
Article in English | MEDLINE | ID: mdl-37042476

ABSTRACT

Cardiovascular disease affects 1% to 4% of the nearly 4 million pregnancies in the United States each year and is the primary cause of pregnancy-related mortality. Adverse pregnancy outcomes are associated with cardiovascular complications during pregnancy persisting into the postpartum period. Recently, investigations have identified an altered sex hormone milieu, such as in the case of hyperandrogenism, as a causative factor in the development of gestational cardiovascular dysfunction. The mechanisms involved in the development of cardiovascular disease in postpartum women are largely unknown. Animal studies have attempted to recapitulate adverse pregnancy outcomes to investigate causal relationships and molecular underpinnings of adverse gestational cardiac events and progression to the development of cardiovascular disease postpartum. This review will focus on summarizing clinical and animal studies detailing the impact of adverse pregnancy outcomes, including preeclampsia, gestational diabetes mellitus, and maternal obesity, on gestational cardiometabolic dysfunction and postpartum cardiovascular disease. Specifically, we will highlight the adverse impact of gestational hyperandrogenism and its potential to serve as a biomarker for maternal gestational and postpartum cardiovascular dysfunctions.


Subject(s)
Cardiovascular Diseases , Diabetes, Gestational , Hyperandrogenism , Pregnancy , Female , Humans , Cardiovascular Diseases/etiology , Hyperandrogenism/complications , Pregnancy Outcome , Postpartum Period
10.
Sci Rep ; 13(1): 2682, 2023 02 15.
Article in English | MEDLINE | ID: mdl-36792653

ABSTRACT

Adverse in-utero insults during fetal life alters offspring's developmental trajectory, including that of the cardiovascular system. Gestational hyperandrogenism is once such adverse in-utero insult. Gestational testosterone (T)-treatment, an environment of gestational hyperandrogenism, manifests as hypertension and pathological left ventricular (LV) remodeling in adult ovine offspring. Furthermore, sexual dimorphism is noted in cardiomyocyte number and morphology in fetal life and at birth. This study investigated transcriptional changes and potential biomarkers of prenatal T excess-induced adverse cardiac programming. Genome-wide coding and non-coding (nc) RNA expression were compared between prenatal T-treated (T propionate 100 mg intramuscular twice weekly from days 30 to 90 of gestation; Term: 147 days) and control ovine LV at day 90 fetus in both sexes. Prenatal T induced differential expression of mRNAs in the LV of female (2 down, 5 up) and male (3 down, 1 up) (FDR < 0.05, absolute log2 fold change > 0.5); pathways analysis demonstrated 205 pathways unique to the female, 382 unique to the male and 23 common pathways. In the male, analysis of ncRNA showed differential regulation of 15 lncRNAs (14 down, 1 up) and 27 snoRNAs (26 down and 1 up). These findings suggest sexual dimorphic modulation of cardiac coding and ncRNA with gestational T excess.


Subject(s)
Hyperandrogenism , Prenatal Exposure Delayed Effects , Pregnancy , Humans , Sheep , Animals , Female , Male , Heart Ventricles/metabolism , Hyperandrogenism/chemically induced , Testosterone/pharmacology , Fetus/metabolism , Myocytes, Cardiac/metabolism , Prenatal Exposure Delayed Effects/chemically induced
11.
J Perinatol ; 42(12): 1576-1586, 2022 12.
Article in English | MEDLINE | ID: mdl-36376450

ABSTRACT

Thyroid hormones play an important role in the development and function of the cardiac myocyte. Dysregulation of the thyroid hormone milieu affects the fetal cardiac cells via complex molecular mechanisms, either by altering gene expression or directly by affecting post-translational processes. This review offers a comprehensive summary of the effects of thyroid hormones on the developing cardiovascular system and its adaptation. Furthermore, we will highlight the gaps in knowledge and provide suggestions for future research.


Subject(s)
Thyroid Gland , Thyroid Hormones , Humans , Infant, Newborn , Fetus , Heart
12.
iScience ; 25(8): 104704, 2022 Aug 19.
Article in English | MEDLINE | ID: mdl-35854813

ABSTRACT

The impact of the recent pandemic on healthcare workers highlights the need to improve the working environment in hospitals. This is especially true in procedural rooms such as the operating and delivery rooms, which inherently require extended exposure to the virus, allows no social distancing, and generates aerosolized virus into the room through the use of the equipment. While reviewing the history of the development of the current Heating, Ventilation, and Air Conditioning Systems (HVAC), we identified inadequacies in the architecture and regulations of the system that resulted in insufficient protection during the current pandemic. Thus, we worked with building/facilities management, the operating room and nursing staff, and learned from research on airplane cabin air circulation to modify HVAC systems to address this issue. The modification includes calculating and implementing appropriate air changes per hour of the HVAC system. Modifying the existing system allows sufficient exchange of air within the procedure room to reduce the amount of exposure to viruses which results in safer working environments for healthcare workers. In the future, there will continue to be more pandemics, thus it is important to start creating safer working environments now, such as revisiting the hospital architecture and HVAC system, so that they can be improved upon and so that we are more prepared for the future.

13.
Toxicol Appl Pharmacol ; 450: 116164, 2022 09 01.
Article in English | MEDLINE | ID: mdl-35842136

ABSTRACT

Substance use (SU) during pregnancy is on the rise, posing significant risks to the developing fetus. The adverse impact of maternal alcohol and nicotine use during the perinatal period on offspring health has been well established, including their associations with adverse cardiovascular health in offspring. However, limited studies examine the impact of other well-known SU utilized during pregnancy on offspring's cardiovascular health. This review summarizes the proposed mechanism of action of four commonly utilized substances: cocaine, marijuana, methamphetamine, and opioids, and their cardiovascular impact. Furthermore, we will review the current understanding of the adverse impact of substance use during pregnancy on offspring's cardiovascular system based on existing studies. This review will also highlight possible molecular mechanisms underlying the in-utero adverse programming of offspring's cardiovascular system secondary to SU in pregnancy and address the gaps in current understanding of how SU adversely impacts the developing cardiovascular system of offspring in utero.


Subject(s)
Prenatal Exposure Delayed Effects , Substance-Related Disorders , Female , Heart , Humans , Pregnancy , Substance-Related Disorders/epidemiology
14.
Am J Physiol Heart Circ Physiol ; 322(4): H607-H621, 2022 04 01.
Article in English | MEDLINE | ID: mdl-35119334

ABSTRACT

Perturbed in utero hormone milieu leads to intrauterine growth retardation (IUGR), a known risk factor for left ventricular (LV) dysfunction later in life. Gestational testosterone (T) excess predisposes offspring to IUGR and leads to LV myocardial disarray and hypertension in adult females. However, the early impact of T excess on LV programming and if it is female specific is unknown. LV tissues were obtained at day 90 gestation from days 30-90 T-treated or control fetuses (n = 6/group/sex) and morphometric and molecular analyses were conducted. Gestational T treatment increased cardiomyocyte number only in female fetuses. T excess upregulated receptor expression of insulin and insulin-like growth factor. Furthermore, in a sex-specific manner, T increased expression of phosphatidylinositol 3-kinase (PI3K) while downregulating phosphorylated mammalian target of rapamycin (pmTOR)-to-mTOR ratio suggestive of compensatory response. T excess 1) upregulated atrial natriuretic peptide (ANP) and brain natriuretic peptide (BNP), markers of stress and cardiac hypertrophy and 2) upregulated estrogen receptors1 (ESR1) and 2 (ESR2), but not in androgen receptor (AR). Thus, gestational T excess upregulated markers of cardiac stress and hypertrophy in both sexes while inducing cardiomyocyte hyperplasia only in females, likely mediated via insulin and estrogenic programming.NEW & NOTEWORTHY The present study demonstrates sex-specific effects of gestational T excess between days 30 and 90 of gestation on the cardiac phenotype. Furthermore, the sex-specific programming is likely secondary to perturbation in both estrogen and insulin signaling pathways collectively. These findings are supportive of the role of androgen excess to serve as early biomarkers of CVD and could be critical in identifying therapeutic targets for LV hypertrophy and predict long-term CVD.


Subject(s)
Sex Characteristics , Testosterone , Animals , Atrial Natriuretic Factor/genetics , Female , Insulin/metabolism , Male , Mammals/metabolism , Myocytes, Cardiac/metabolism , Natriuretic Peptide, Brain/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Sheep , Testosterone/metabolism
15.
Reproduction ; 162(6): R85-R98, 2021 10 28.
Article in English | MEDLINE | ID: mdl-34715674

ABSTRACT

A growing body of research suggests that alterations to the human microbiome are associated with disease states, including obesity and diabetes. During pregnancy, these disease states are associated with maternal microbial dysbiosis. This review discusses the current literature regarding the typical maternal and offspring microbiome as well as alterations to the microbiome in the context of obesity, type 2 diabetes mellitus, and gestational diabetes mellitus. Furthermore, this review outlines the proposed mechanisms linking associations between the maternal microbiome in the aforementioned disease states and offspring microbiome. Additionally, this review highlights associations between alterations in offspring microbiome and postnatal health outcomes.


Subject(s)
Diabetes Mellitus, Type 2 , Diabetes, Gestational , Microbiota , Prenatal Exposure Delayed Effects , Diabetes, Gestational/metabolism , Female , Humans , Obesity/metabolism , Pregnancy , Prenatal Exposure Delayed Effects/metabolism
16.
J Clin Med ; 10(17)2021 Aug 26.
Article in English | MEDLINE | ID: mdl-34501285

ABSTRACT

Cardiovascular diseases (CVD) remain the leading cause of death in men and women. Biological sex plays a major role in cardiovascular physiology and pathological cardiovascular remodeling. Traditionally, pathological remodeling of cardiovascular system refers to the molecular, cellular, and morphological changes that result from insults, such as myocardial infarction or hypertension. Regular exercise training is known to induce physiological cardiovascular remodeling and beneficial functional adaptation of the cardiovascular apparatus. However, impact of exercise-induced cardiovascular remodeling and functional adaptation varies between males and females. This review aims to compare and contrast sex-specific manifestations of exercise-induced cardiovascular remodeling and functional adaptation. Specifically, we review (1) sex disparities in cardiovascular function, (2) influence of biological sex on exercise-induced cardiovascular remodeling and functional adaptation, and (3) sex-specific impacts of various types, intensities, and durations of exercise training on cardiovascular apparatus. The review highlights both animal and human studies in order to give an all-encompassing view of the exercise-induced sex differences in cardiovascular system and addresses the gaps in knowledge in the field.

17.
AACE Clin Case Rep ; 7(1): 65-68, 2021.
Article in English | MEDLINE | ID: mdl-33851023

ABSTRACT

OBJECTIVE: Liddle syndrome (LS) is a rare autosomal dominant condition secondary to a gain-of-function mutation affecting the epithelial sodium channels (ENaCs) in the distal nephron. It presents with early-onset hypertension, hypokalemia, and metabolic alkalosis in the face of hyporeninemia and hypoaldosteronism. We report a novel mutation affecting the ENaCs in a normotensive adolescent with LS. METHODS: We describe a pediatric case of LS with a novel mutation and review the condition's presentation and management. To date, 31 different mutations in the ß- or γ-subunit of ENaCs have been reported as associated with LS. RESULTS: We describe a 16-year-old girl presenting with muscle cramps with a strong family history of hypertension and hypokalemia. Initial investigations revealed hypokalemia together with hypoaldosteronism and hyporeninemia. Subsequent genetic testing revealed a novel mutation in SCNN1B (deletion: c.1713delC), leading to the premature termination of the sodium channel epithelial 1 subunit-ß protein and the LS phenotype. Treatment with triamterene (50 mg, twice daily) and potassium chloride (20 mEq, once daily) normalized the serum potassium and led to resolution of her muscle cramps. CONCLUSION: It is essential to consider investigating the presence of rare genetic syndromes, like LS, when a patient presents with hypokalemia. Further studies are needed to understand the variable presentation of this condition.

18.
PLoS One ; 16(1): e0245012, 2021.
Article in English | MEDLINE | ID: mdl-33411822

ABSTRACT

OBJECTIVES: To examine the temporal patterns of hospitalizations with diabetic ketoacidosis (DKA) in the pediatric population and their associated fiscal impact. METHODS: The Texas Inpatient Public Use Data File was used to identify hospitalizations of state residents aged 1month-19 years with a primary diagnosis of DKA during 2005-2014. Temporal changes of population-adjusted hospitalization rates and hospitalization volumes were examined for the whole cohort and on stratified analyses of sociodemographic attributes. Changes in the aggregate and per-hospitalization charges were assessed overall and on stratified analyses. RESULTS: There were 24,072 DKA hospitalizations during the study period. The population-adjusted hospitalization rate for the whole cohort increased from 31.3 to 35.9 per 100,000 between 2005-2006 and 2013-2014. Hospitalization volume increased by 30.2% over the same period, driven mainly by males, ethnic minorities, those with Medicaid insurance and uninsured patients. The aggregate hospital charges increased from approximately $69 million to $130 million between 2005-2006 and 2013-2014, with 66% of the rise being due to increased per-hospitalization charges. CONCLUSIONS: There was progressive rise in pediatric DKA hospitalizations over the last decade, with concurrent near-doubling of the associated fiscal footprint. Marked disparities were noted in the increasing hospitalization burden of DKA, born predominantly by racial and ethnic minorities, as well as by the underinsured and the uninsured. Further studies are needed to identify scalable preventive measures to achieve an equitable reduction of pediatric DKA events.


Subject(s)
Diabetic Ketoacidosis/therapy , Hospital Charges , Hospitalization/trends , Adolescent , Child , Child, Preschool , Databases, Factual , Diabetic Ketoacidosis/economics , Female , Hospitalization/economics , Humans , Infant , Male , Retrospective Studies , Sex Factors , Texas
19.
BMC Microbiol ; 20(1): 157, 2020 06 11.
Article in English | MEDLINE | ID: mdl-32527226

ABSTRACT

BACKGROUND: A placental microbiome, which may be altered in gestational diabetes mellitus (GDM), has been described. However, publications raising doubts about the existence of a placental microbiome that is different than contaminants in DNA extraction kits and reagents ("kitomes") have emerged. The aims of this study were to confirm the existence of a placental microbiome distinct from contaminants and determine if it is altered in GDM mothers. RESULTS: We first enrolled normal weight, obese and GDM mothers (N = 17) at term elective cesarean section delivery in a pilot case control study. Bacterial DNA was extracted from placental parenchyma, maternal and cord blood, maternal vaginal-rectal swabs, and positive and negative controls with the standard Qiagen/MoBio Power Soil kit. Placentas had significantly higher copies of bacterial 16S rRNA genes than negative controls, but the placental microbiome was similar in all three groups and could not be distinguished from contaminants in blank controls. To determine the source and composition of the putative placental bacterial community identified in the pilot study, we expanded the study to 10 subjects per group (N = 30) and increased the number and variety of negative controls (N = 53). We modified our protocol to use an ultraclean DNA extraction kit (Qiagen QIAamp UCP with Pathogen Lysis Tube S), which reduced the "kitome" contamination, but we were still unable to distinguish a placental microbiome from contaminants in negative controls. We noted microbial DNA from the high biomass vaginal-rectal swabs and positive controls in placental and negative control samples and determined that this resulted from close proximity well-to-well cross contamination or "splashome". We eliminated this source of contamination by repeating the sequencing run with a minimum of four wells separating high biomass from low biomass samples. This reduced the reads of bacterial 16S rRNA genes in placental samples to insignificant numbers. CONCLUSIONS: We identified the problem of well-to-well contamination ("splashome") as an additional source of error in microbiome studies of low biomass samples and found a method of eliminating it. Once "kitome" and "splashome" contaminants were eliminated, we were unable to identify a unique placental microbiome.


Subject(s)
Bacteria/classification , Diabetes, Gestational/microbiology , Obesity/microbiology , Placenta/microbiology , Sequence Analysis, DNA/methods , Adult , Bacteria/genetics , Bacteria/isolation & purification , Case-Control Studies , Cesarean Section , Female , Fetal Blood/microbiology , Humans , Infant , Organ Specificity , Pilot Projects , Pregnancy , RNA, Ribosomal, 16S/genetics , Rectum/microbiology , Vagina/microbiology , Young Adult
20.
J Intensive Care Med ; 35(11): 1314-1322, 2020 Nov.
Article in English | MEDLINE | ID: mdl-31409186

ABSTRACT

OBJECTIVE: To examine the hospital-level variation in intensive care unit (ICU) utilization and quantify the relative contribution of patient and hospital characteristics versus individual hospital factors to the variation in ICU admission rates among pediatric hospitalizations with diabetic ketoacidosis (DKA). METHODS: The Texas Inpatient Public Use Data File was used to identify hospitalizations of state residents aged 1 month to 19 years with a primary diagnosis of DKA between 2005 and 2014. Multilevel, mixed-effects logistic regression modeling was performed to examine the association of patient- and hospital-level factors with ICU admission. Risk and reliability adjustment was then performed to assess hospital-level variation in ICU utilization. Intraclass correlation coefficient was used to quantify variation in use of ICU attributable to individual hospitals. The association between adjusted rates of ICU admission and total hospital charges and length of stay was examined using linear regression. RESULTS: Of the 23 585 DKA hospitalizations, 14 638 (62.1%) were admitted to ICU. On multilevel analysis, the odds of ICU admission progressively decreased with rising volume of DKA hospitalizations (adjusted odds ratio: 0.08 [highest vs lowest quartile]; 95% confidence interval [CI]: 0.03-0.24). The crude median (interquartile range [IQR]; range) of ICU admissions across hospitals was 82.6% (73%-90%; 11.1%-100%). The median (IQR) risk- and reliability-adjusted ICU admission rate was 81.0% (73.0%-86.9%), ranging from 11.2% to 94%. Following risk and reliability adjustment, the intraclass correlation coefficient was 0.005 (95% CI: 0.004-0.006). For each 10% increase in adjusted ICU admission rate, total hospital charges rose by 7% (95% CI: 3%-11%). There was no association between ICU admission rates and hospital length of stay. CONCLUSION: Although high variation in ICU utilization was noted across hospitals among pediatric DKA hospitalizations, the proportion of variation attributable to individual hospitals was negligible, once adjusted for patient mix and hospital characteristics.


Subject(s)
Diabetic Ketoacidosis , Child , Critical Care , Diabetic Ketoacidosis/epidemiology , Diabetic Ketoacidosis/therapy , Humans , Intensive Care Units , Length of Stay , Reproducibility of Results , Retrospective Studies
SELECTION OF CITATIONS
SEARCH DETAIL
...