Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 24(4)2023 Feb 12.
Article in English | MEDLINE | ID: mdl-36835096

ABSTRACT

Infants exposed to diabetic pregnancy are at higher risk of cardiomyopathy at birth and early onset cardiovascular disease (CVD) as adults. Using a rat model, we showed how fetal exposure to maternal diabetes causes cardiac disease through fuel-mediated mitochondrial dysfunction, and that a maternal high-fat diet (HFD) exaggerates the risk. Diabetic pregnancy increases circulating maternal ketones which can have a cardioprotective effect, but whether diabetes-mediated complex I dysfunction impairs myocardial metabolism of ketones postnatally remains unknown. The objective of this study was to determine whether neonatal rat cardiomyocytes (NRCM) from diabetes- and HFD-exposed offspring oxidize ketones as an alternative fuel source. To test our hypothesis, we developed a novel ketone stress test (KST) using extracellular flux analyses to compare real-time ß-hydroxybutyrate (ßHOB) metabolism in NRCM. We also compared myocardial expression of genes responsible for ketone and lipid metabolism. NRCM had a dose-dependent increase in respiration with increasing concentrations of ßHOB, demonstrating that both control and combination exposed NRCM can metabolize ketones postnatally. Ketone treatment also enhanced the glycolytic capacity of combination exposed NRCM with a dose-dependent increase in the glucose-mediated proton efflux rate (PER) from CO2 (aerobic glycolysis) alongside a decreased reliance on PER from lactate (anaerobic glycolysis). Expression of genes responsible for ketone body metabolism was higher in combination exposed males. Findings demonstrate that myocardial ketone body metabolism is preserved and improves fuel flexibility in NRCM from diabetes- and HFD-exposed offspring, which suggests that ketones might serve a protective role in neonatal cardiomyopathy due to maternal diabetes.


Subject(s)
Diabetes, Gestational , Pregnancy in Diabetics , Prenatal Exposure Delayed Effects , Pregnancy , Male , Humans , Female , Rats , Animals , Diet, High-Fat , Prenatal Exposure Delayed Effects/metabolism , Myocytes, Cardiac/metabolism , Ketones
2.
J Nutr Biochem ; 97: 108797, 2021 11.
Article in English | MEDLINE | ID: mdl-34126202

ABSTRACT

Agricultural workers, especially those who work in swine confinement facilities, are at increased risk for developing pulmonary diseases including asthma, chronic obstructive pulmonary disease, and chronic bronchitis due to exposures to fumes, vapors, and organic dust. Repetitive exposure to agricultural dust leads to unresolved inflammation, a common underlying mechanism that worsens lung disease. Besides occupational exposure to dusts, diet also significantly contributes to inflammation and disease progression. Since DHA (docosahexaenoic acid), a polyunsaturated omega-3 fatty acid and its bioactive metabolites have key roles in inflammation resolution, we rationalized that individuals chronically exposed to organic dusts can benefit from dietary modifications. Here, we evaluated the role of DHA in modifying airway inflammation in a murine model of repetitive exposure to an aqueous extract of agricultural dust (three-week exposure to swine confinement dust extract, HDE) and after a one-week resolution/recovery period. We found that mice fed a high DHA diet had significantly increased bronchoalveolar lavage fluid (BALF) levels of DHA-derived resolvins and lower TNFα along with altered plasma levels of endocannabinoids and related lipid mediators. Following the one-week recovery we identified significantly reduced BALF cellularity and cytokine/chemokine release along with increased BALF amphiregulin and resolvins in DHA diet-fed versus control diet-fed mice challenged with HDE. We further report observations on the effects of repetitive HDE exposure on lung Ym1+ and Arg-1+ macrophages. Overall, our findings support a protective role for DHA and identify DHA-derived resolvins and endocannabinoids among the potential mediators of DHA in altering airway inflammation in chronic agricultural dust exposure.


Subject(s)
Diet , Docosahexaenoic Acids/administration & dosage , Dust , Inhalation Exposure/adverse effects , Respiratory Tract Diseases/diet therapy , Agricultural Workers' Diseases/diet therapy , Agricultural Workers' Diseases/pathology , Animals , Bronchoalveolar Lavage Fluid/chemistry , Disease Models, Animal , Eicosapentaenoic Acid/analogs & derivatives , Eicosapentaenoic Acid/metabolism , Endocannabinoids/blood , Fatty Acids, Unsaturated/blood , Inflammation/diet therapy , Inflammation/pathology , Lung/pathology , Macrophages, Alveolar/physiology , Male , Mice , Mice, Inbred C57BL , Respiratory Tract Diseases/pathology , Swine , Tumor Necrosis Factor-alpha/metabolism
3.
Int J Mol Sci ; 22(5)2021 Feb 27.
Article in English | MEDLINE | ID: mdl-33673574

ABSTRACT

Offspring born to diabetic or obese mothers have a higher lifetime risk of heart disease. Previously, we found that rat offspring exposed to late-gestational diabetes mellitus (LGDM) and maternal high-fat (HF) diet develop mitochondrial dysfunction, impaired cardiomyocyte bioenergetics, and cardiac dysfunction at birth and again during aging. Here, we compared echocardiography, cardiomyocyte bioenergetics, oxidative damage, and mitochondria-mediated cell death among control, pregestational diabetes mellitus (PGDM)-exposed, HF-diet-exposed, and combination-exposed newborn offspring. We hypothesized that PGDM exposure, similar to LGDM, causes mitochondrial dysfunction to play a central, pathogenic role in neonatal cardiomyopathy. We found that PGDM-exposed offspring, similar to LGDM-exposed offspring, have cardiac dysfunction at birth, but their isolated cardiomyocytes have seemingly less bioenergetics impairment. This finding was due to confounding by impaired viability related to poorer ATP generation, more lipid peroxidation, and faster apoptosis under metabolic stress. To mechanistically isolate and test the role of mitochondria, we transferred mitochondria from normal rat myocardium to control and exposed neonatal rat cardiomyocytes. As expected, transfer provides a respiratory boost to cardiomyocytes from all groups. They also reduce apoptosis in PGDM-exposed males, but not in females. Findings highlight sex-specific differences in mitochondria-mediated mechanisms of developmentally programmed heart disease and underscore potential caveats of therapeutic mitochondrial transfer.


Subject(s)
Diabetes Mellitus, Experimental/complications , Diabetes, Gestational/physiopathology , Energy Metabolism , Heart Diseases/prevention & control , Mitochondria/transplantation , Myocytes, Cardiac/cytology , Myocytes, Cardiac/physiology , Animals , Diet, High-Fat/adverse effects , Female , Heart Diseases/etiology , Heart Diseases/pathology , Male , Maternal Nutritional Physiological Phenomena , Pregnancy , Prenatal Exposure Delayed Effects/etiology , Prenatal Exposure Delayed Effects/pathology , Prenatal Exposure Delayed Effects/prevention & control , Rats , Rats, Sprague-Dawley , Sex Factors
4.
Reprod Sci ; 28(5): 1476-1488, 2021 05.
Article in English | MEDLINE | ID: mdl-33398850

ABSTRACT

The placenta represents a critical node in fetal lipid acquisition, yet the mechanisms by which the placenta handles lipids under normal and pathologic conditions are incompletely understood. A key player in placental lipid handling is peroxisome proliferator-activated receptor gamma (PPARγ). PPARγ influences global gene expression via its regulation of the epigenetic modifier lysine methyltransferase 5A (KMT5A), which places a methyl group on histone 4 lysine 20 (H4K20me) of target genes. Here we test the hypothesis that KMT5A is present in both the human and rat placentas and is affected by uteroplacental insufficiency (UPI) in the rat in association with increased placental lipid accumulation. We assessed levels and localization of KMT5A, as well as lipid droplet accumulation, in human placental tissue collected from maternal donors after delivery by planned cesarean section. Using a rat model of UPI, we also evaluated the effects of UPI on lipid accumulation, PPARγ, KMT5A, and H4K20me in the rat placenta. In this study, we show for the first time the presence and activity of KMT5A, in human and in rat placentas. We also demonstrate that in the rat placenta, UPI increases hypoxia, KMT5a expression, and activity in association with increased lipid accumulation in placenta supporting male fetuses. Placental PPARγ-KMT5A axis may be an important mediator of placental lipid handling.


Subject(s)
Hypoxia/metabolism , Methyltransferases/metabolism , PPAR gamma/metabolism , Placenta Diseases/metabolism , Uterine Diseases/metabolism , Animals , Disease Models, Animal , Female , Humans , Lipid Accumulation Product , Pregnancy , Rats, Sprague-Dawley , Signal Transduction , Up-Regulation
5.
iScience ; 23(11): 101746, 2020 Nov 20.
Article in English | MEDLINE | ID: mdl-33225249

ABSTRACT

Infants of diabetic mothers are at risk of cardiomyopathy at birth and myocardial infarction in adulthood, but prevention is hindered because mechanisms remain unknown. We previously showed that maternal glucolipotoxicity increases the risk of cardiomyopathy and mortality in newborn rats through fuel-mediated mitochondrial dysfunction. Here we demonstrate ongoing cardiometabolic consequences by cross-fostering and following echocardiography, cardiomyocyte bioenergetics, mitochondria-mediated turnover, and cell death following metabolic stress in aged adults. Like humans, cardiac function improves by weaning with no apparent differences in early adulthood but declines again in aged diabetes-exposed offspring. This is preceded by impaired oxidative phosphorylation, exaggerated age-related increase in mitochondrial number, and higher oxygen consumption. Prenatally exposed male cardiomyocytes have more mitolysosomes indicating high baseline turnover; when exposed to metabolic stress, mitophagy cannot increase and cardiomyocytes have faster mitochondrial membrane potential loss and mitochondria-mediated cell death. Details highlight age- and sex-specific roles of mitochondria in developmentally programmed adult heart disease.

6.
Front Endocrinol (Lausanne) ; 11: 570846, 2020.
Article in English | MEDLINE | ID: mdl-33042024

ABSTRACT

Background: Children born to diabetic or obese mothers have a higher risk of heart disease at birth and later in life. Using chromatin immunoprecipitation sequencing, we previously demonstrated that late-gestation diabetes, maternal high fat (HF) diet, and the combination causes distinct fuel-mediated epigenetic reprogramming of rat cardiac tissue during fetal cardiogenesis. The objective of the present study was to investigate the overall transcriptional signature of newborn offspring exposed to maternal diabetes and maternal H diet. Methods: Microarray gene expression profiling of hearts from diabetes exposed, HF diet exposed, and combination exposed newborn rats was compared to controls. Functional annotation, pathway and network analysis of differentially expressed genes were performed in combination exposed and control newborn rat hearts. Further downstream metabolic assessments included measurement of total and phosphorylated AKT2 and GSK3ß, as well as quantification of glycolytic capacity by extracellular flux analysis and glycogen staining. Results: Transcriptional analysis identified significant fuel-mediated changes in offspring cardiac gene expression. Specifically, functional pathways analysis identified two key signaling cascades that were functionally prioritized in combination exposed offspring hearts: (1) downregulation of fibroblast growth factor (FGF) activated PI3K/AKT pathway and (2) upregulation of peroxisome proliferator-activated receptor gamma coactivator alpha (PGC1α) mitochondrial biogenesis signaling. Functional metabolic and histochemical assays supported these transcriptome changes, corroborating diabetes- and diet-induced cardiac transcriptome remodeling and cardiac metabolism in offspring. Conclusion: This study provides the first data accounting for the compounding effects of maternal hyperglycemia and hyperlipidemia on the developmental cardiac transcriptome, and elucidates nuanced and novel features of maternal diabetes and diet on regulation of heart health.


Subject(s)
Diabetes, Gestational/metabolism , Diet, High-Fat/adverse effects , Gene Regulatory Networks/physiology , Maternal Nutritional Physiological Phenomena/physiology , Myocardium/metabolism , Transcriptome/physiology , Animals , Animals, Newborn , Diabetes, Gestational/genetics , Diabetes, Gestational/pathology , Diet, High-Fat/trends , Female , Gene Expression Profiling/methods , Male , Myocardium/pathology , Organelle Biogenesis , Pregnancy , Rats
7.
Nutrients ; 12(8)2020 Aug 04.
Article in English | MEDLINE | ID: mdl-32759853

ABSTRACT

Agricultural workers are at risk for the development of acute and chronic lung diseases due to their exposure to organic agricultural dusts. A diet intervention using the omega-3 fatty acid docosahexaenoic acid (DHA) has been shown to be an effective therapeutic approach for alleviating a dust-induced inflammatory response. We thus hypothesized a high-DHA diet would alter the dust-induced inflammatory response through the increased production of specialized pro-resolving mediators (SPMs). Mice were pre-treated with a DHA-rich diet 4 weeks before being intranasally challenged with a single dose of an extract made from dust collected from a concentrated swine feeding operation (HDE). This omega-3-fatty-acid-rich diet led to reduced arachidonic acid levels in the blood, enhanced macrophage recruitment, and increased the production of the DHA-derived SPM Resolvin D1 (RvD1) in the lung following HDE exposure. An assessment of transcript-level changes in the immune response demonstrated significant differences in immune pathway activation and alterations of numerous macrophage-associated genes among HDE-challenged mice fed a high DHA diet. Our data indicate that consuming a DHA-rich diet leads to the enhanced production of SPMs during an acute inflammatory challenge to dust, supporting a role for dietary DHA supplementation as a potential therapeutic strategy for reducing dust-induced lung inflammation.


Subject(s)
Diet, High-Fat/methods , Docosahexaenoic Acids/administration & dosage , Dust , Inhalation Exposure/adverse effects , Pneumonia/diet therapy , Animal Feed/adverse effects , Animals , Arachidonic Acid/blood , Dietary Supplements , Disease Models, Animal , Docosahexaenoic Acids/biosynthesis , Lung/physiopathology , Male , Mice , Mice, Inbred C57BL , Pneumonia/etiology , Swine
8.
Int J Mol Sci ; 20(12)2019 Jun 25.
Article in English | MEDLINE | ID: mdl-31242551

ABSTRACT

Infants born to diabetic or obese mothers are at greater risk of heart disease at birth and throughout life, but prevention is hindered because underlying mechanisms remain poorly understood. Using a rat model, we showed that prenatal exposure to maternal diabetes and a high-fat diet caused diastolic and systolic dysfunction, myocardial lipid accumulation, decreased respiratory capacity, and oxidative stress in newborn offspring hearts. This study aimed to determine whether mitochondrial dynamism played a role. Using confocal live-cell imaging, we examined mitochondrial dynamics in neonatal rat cardiomyocytes (NRCM) from four prenatally exposed groups: controls, diabetes, high-fat diet, and combination exposed. Cardiac expression of dynamism-related genes and proteins were compared, and gender-specific differences were evaluated. Findings show that normal NRCM have highly dynamic mitochondria with a well-balanced number of fusion and fission events. Prenatal exposure to diabetes or a high-fat diet impaired dynamism resulting in shorter, wider mitochondria. Mechanisms of impaired dynamism were gender-specific and protein regulated. Females had higher expression of fusion proteins which may confer a cardioprotective effect. Prenatally exposed male hearts had post-translational modifications known to impair dynamism and influence mitophagy-mediated cell death. This study identifies mitochondrial fusion and fission proteins as targetable, pathogenic regulators of heart health in offspring exposed to excess circulating maternal fuels.


Subject(s)
Diet, High-Fat/adverse effects , Fetal Development , Heart/embryology , Mitochondrial Dynamics , Organogenesis , Pregnancy in Diabetics , Animals , Animals, Newborn , Biomarkers , Female , Fetal Development/genetics , Fluorescent Antibody Technique , Gene Expression Regulation , Male , Mitochondria, Heart/genetics , Mitochondria, Heart/metabolism , Mitochondria, Heart/ultrastructure , Mitochondrial Dynamics/genetics , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Myocytes, Cardiac/metabolism , Organogenesis/genetics , Pregnancy , Protein Processing, Post-Translational , Rats , Sex Factors
9.
Pediatr Res ; 83(3): 712-722, 2018 03.
Article in English | MEDLINE | ID: mdl-29166372

ABSTRACT

BackgroundDiabetes and obesity during pregnancy have an impact on the health of both mothers and developing babies. Prevention focuses on glycemic control, but increasing evidence implicates a role for lipids. Using a rat model, we showed that a maternal high-fat (HF) diet increased perinatal morbidity and mortality, but lipid processing across the maternal-placental-fetal triad remained unstudied. We hypothesized that HF diet would disrupt placental lipid processing to exaggerate fuel-mediated consequences of diabetic pregnancy.MethodsWe compared circulating lipid profiles, hormones, and inflammatory markers in dams and rat offspring from normal, diabetes-exposed, HF-diet-exposed, and combination-exposed pregnancies. Placentae were examined for lipid accumulation and expression of fuel transporters.ResultsMaternal HF diet exaggerated hyperlipidemia of pregnancy, with diabetes marked dyslipidemia developed in dams but not in offspring. Placentae demonstrated lipid accumulation and lower expression of fatty acid (FA) transporters. Diet-exposed offspring had a lower fraction of circulating essential FAs. Pregnancy loss was significantly higher in diet-exposed but not in diabetes-exposed pregnancies, which could not be explained by differences in hormone production. Although not confirmed, inflammation may play a role.ConclusionMaternal hyperlipidemia contributes to placental lipid droplet accumulation, perinatal mortality, and aberrant FA profiles that may influence the health of the developing offspring.


Subject(s)
Diet, High-Fat , Lipid Metabolism , Maternal Nutritional Physiological Phenomena , Placenta/metabolism , Animals , Diabetes Mellitus, Experimental/metabolism , Diabetes, Gestational/metabolism , Female , Inflammation , Lipids/blood , Obesity/physiopathology , Pregnancy , Prenatal Exposure Delayed Effects/metabolism , Rats , Rats, Sprague-Dawley , Triglycerides/chemistry
10.
Nutrients ; 9(4)2017 Apr 20.
Article in English | MEDLINE | ID: mdl-28425976

ABSTRACT

Infants born to women with diabetes or obesity are exposed to excess circulating fuels during fetal heart development and are at higher risk of cardiac diseases. We have previously shown that late-gestation diabetes, especially in conjunction with a maternal high-fat (HF) diet, impairs cardiac functions in rat-offspring. This study investigated changes in genome-wide histone modifications in newborn hearts from rat-pups exposed to maternal diabetes and HF-diet. Chromatin-immunoprecipitation-sequencing revealed a differential peak distribution on gene promoters in exposed pups with respect to acetylation of lysines 9 and 14 and to trimethylation of lysines 4 and 27 in histone H3 (all, false discovery rate, FDR < 0.1). In the HF-diet exposed offspring, 54% of the annotated genes showed the gene-activating mark trimethylated lysine 4. Many of these genes (1) are associated with the "metabolic process" in general and particularly with "positive regulation of cholesterol biosynthesis" (FDR = 0.03); (2) overlap with 455 quantitative trait loci for blood pressure, body weight, serum cholesterol (all, FDR < 0.1); and (3) are linked to cardiac disease susceptibility/progression, based on disease ontology analyses and scientific literature. These results indicate that maternal HF-diet changes the cardiac histone signature in offspring suggesting a fuel-mediated epigenetic reprogramming of cardiac tissue in utero.


Subject(s)
Cardiovascular Diseases/genetics , Diet, High-Fat/adverse effects , Histone Code , Metabolic Syndrome/genetics , Prenatal Exposure Delayed Effects/genetics , Animals , Animals, Newborn , Blood Pressure , Body Weight , Cardiovascular Diseases/etiology , Cholesterol/blood , Diabetes Mellitus, Experimental , Epigenesis, Genetic , Female , Fetal Development , Gene Expression Regulation, Developmental , Genetic Association Studies , Genetic Predisposition to Disease , Maternal Nutritional Physiological Phenomena , Metabolic Syndrome/etiology , Pregnancy , Promoter Regions, Genetic , Quantitative Trait Loci , Rats , Rats, Sprague-Dawley , Sequence Analysis, DNA
11.
PLoS One ; 11(8): e0160818, 2016.
Article in English | MEDLINE | ID: mdl-27518105

ABSTRACT

RATIONALE: Infants born to diabetic or obese mothers are at risk of respiratory distress and persistent pulmonary hypertension of the newborn (PPHN), conceivably through fuel-mediated pathogenic mechanisms. Prior research and preventative measures focus on controlling maternal hyperglycemia, but growing evidence suggests a role for additional circulating fuels including lipids. Little is known about the individual or additive effects of a maternal high-fat diet on fetal lung development. OBJECTIVE: The objective of this study was to determine the effects of a maternal high-fat diet, alone and alongside late-gestation diabetes, on lung alveologenesis and vasculogenesis, as well as to ascertain if consequences persist beyond the perinatal period. METHODS: A rat model was used to study lung development in offspring from control, diabetes-exposed, high-fat diet-exposed and combination-exposed pregnancies via morphometric, histologic (alveolarization and vasculogenesis) and physiologic (echocardiography, pulmonary function) analyses at birth and 3 weeks of age. Outcomes were interrogated for diet, diabetes and interaction effect using ANOVA with significance set at p≤0.05. Findings prompted additional mechanistic inquiry of key molecular pathways. RESULTS: Offspring exposed to maternal diabetes or high-fat diet, alone and in combination, had smaller lungs and larger hearts at birth. High-fat diet-exposed, but not diabetes-exposed offspring, had a higher perinatal death rate and echocardiographic evidence of PPHN at birth. Alveolar mean linear intercept, septal thickness, and airspace area (D2) were not significantly different between the groups; however, markers of lung maturity were. Both diabetes-exposed and diet-exposed offspring expressed more T1α protein, a marker of type I cells. Diet-exposed newborn pups expressed less surfactant protein B and had fewer pulmonary vessels enumerated. Mechanistic inquiry revealed alterations in AKT activation, higher endothelin-1 expression, and an impaired Txnip/VEGF pathway that are important for vessel growth and migration. After 3 weeks, mortality remained highest and static lung compliance and hysteresis were lowest in combination-exposed offspring. CONCLUSION: This study emphasizes the effects of a maternal high-fat diet, especially alongside late-gestation diabetes, on pulmonary vasculogenesis, demonstrates adverse consequences beyond the perinatal period and directs attention to mechanistic pathways of interest. Findings provide a foundation for additional investigation of preventative and therapeutic strategies aimed at decreasing pulmonary morbidity in at-risk infants.


Subject(s)
Diabetes, Gestational , Diet, High-Fat/adverse effects , Lung/growth & development , Maternal Nutritional Physiological Phenomena , Prenatal Exposure Delayed Effects/pathology , Prenatal Exposure Delayed Effects/physiopathology , Animals , Animals, Newborn , Female , Hemodynamics , Lung/blood supply , Lung/pathology , Lung/physiopathology , Pregnancy , Prenatal Exposure Delayed Effects/mortality , Pulmonary Alveoli/pathology , Pulmonary Artery/pathology , Pulmonary Veins/pathology , Rats , Rats, Sprague-Dawley
12.
PLoS One ; 11(2): e0149002, 2016.
Article in English | MEDLINE | ID: mdl-26872351

ABSTRACT

Mitochondrial dysfunction is increasingly recognized and studied as a mediator of heart disease. Extracellular flux analysis (XF) has emerged as a powerful tool to investigate cellular bioenergetics in the context of cardiac health and disease, however its use and interpretation requires improved understanding of the normal metabolic differences in cardiomyocytes (CM) at various stages of maturation. This study standardized XF analyses methods (mitochondrial stress test, glycolytic stress test and palmitate oxidation test) and established age related differences in bioenergetics profiles of healthy CMs at newborn (NB1), weaning (3WK), adult (10WK) and aged (12-18MO) time points. Findings show that immature CMs demonstrate a more robust and sustained glycolytic capacity and a relative inability to oxidize fatty acids when compared to older CMs. The study also highlights the need to recognize the contribution of CO2 from the Krebs cycle as well as lactate from anaerobic glycolysis to the proton production rate before interpreting glycolytic capacity in CMs. Overall, this study demonstrates that caution should be taken to assure that translatable developmental time points are used to investigate mitochondrial dysfunction as a cause of cardiac disease. Specifically, XF analysis of newborn CMs should be reserved to study fetal/neonatal disease and older CMs (≥10 weeks) should be used to investigate adult disease pathogenesis. Knowledge gained will aid in improved investigation of developmentally programmed heart disease and stress the importance of discerning maturational differences in bioenergetics when developing mitochondrial targeted preventative and therapeutic strategies for cardiac disease.


Subject(s)
Metabolic Flux Analysis , Myocytes, Cardiac/metabolism , Oxidative Phosphorylation , Aging/metabolism , Animals , Animals, Newborn , Carbon Dioxide/metabolism , Fatty Acids/metabolism , Female , Glycolysis/physiology , Lactic Acid/metabolism , Mitochondria/metabolism , Primary Cell Culture , Rats , Rats, Sprague-Dawley , Weaning
13.
Am J Physiol Heart Circ Physiol ; 310(6): H681-92, 2016 Mar 15.
Article in English | MEDLINE | ID: mdl-26801311

ABSTRACT

Offspring of diabetic pregnancies are at risk of cardiovascular disease at birth and throughout life, purportedly through fuel-mediated influences on the developing heart. Preventative measures focus on glycemic control, but the contribution of additional offenders, including lipids, is not understood. Cellular bioenergetics can be influenced by both diabetes and hyperlipidemia and play a pivotal role in the pathophysiology of adult cardiovascular disease. This study investigated whether a maternal high-fat diet, independently or additively with diabetes, could impair fuel metabolism, mitochondrial function, and cardiac physiology in the developing offspring's heart. Sprague-Dawley rats fed a control or high-fat diet were administered placebo or streptozotocin to induce diabetes during pregnancy and then delivered offspring from four groups: control, diabetes exposed, diet exposed, and combination exposed. Cardiac function, cellular bioenergetics (mitochondrial stress test, glycolytic stress test, and palmitate oxidation assay), lipid peroxidation, mitochondrial histology, and copy number were determined. Diabetes-exposed offspring had impaired glycolytic and respiratory capacity and a reduced proton leak. High-fat diet-exposed offspring had increased mitochondrial copy number, increased lipid peroxidation, and evidence of mitochondrial dysfunction. Combination-exposed pups were most severely affected and demonstrated cardiac lipid droplet accumulation and diastolic/systolic cardiac dysfunction that mimics that of adult diabetic cardiomyopathy. This study is the first to demonstrate that a maternal high-fat diet impairs cardiac function in offspring of diabetic pregnancies through metabolic stress and serves as a critical step in understanding the role of cellular bioenergetics in developmentally programmed cardiac disease.


Subject(s)
Diabetes Mellitus, Experimental/metabolism , Diabetes, Gestational/metabolism , Diet, High-Fat , Heart/physiopathology , Mitochondria, Heart/metabolism , Myocytes, Cardiac/pathology , Stress, Physiological , Animals , Animals, Newborn , Cell Respiration , DNA, Mitochondrial/metabolism , Diabetes Mellitus, Experimental/pathology , Diabetes, Gestational/pathology , Echocardiography , Female , Glycolysis , Lipid Peroxidation , Mitochondria, Heart/pathology , Pregnancy , Rats , Rats, Sprague-Dawley
14.
J Biol Chem ; 286(46): 40296-306, 2011 Nov 18.
Article in English | MEDLINE | ID: mdl-21949239

ABSTRACT

Myotonic dystrophy 1 (DM1) is a multisystemic disease caused by a triplet nucleotide repeat expansion in the 3' untranslated region of the gene coding for myotonic dystrophy protein kinase (DMPK). DMPK is a nuclear envelope (NE) protein that promotes myogenic gene expression in skeletal myoblasts. Muscular dystrophy research has revealed the NE to be a key determinant of nuclear structure, gene regulation, and muscle function. To investigate the role of DMPK in NE stability, we analyzed DMPK expression in epithelial and myoblast cells. We found that DMPK localizes to the NE and coimmunoprecipitates with Lamin-A/C. Overexpression of DMPK in HeLa cells or C2C12 myoblasts disrupts Lamin-A/C and Lamin-B1 localization and causes nuclear fragmentation. Depletion of DMPK also disrupts NE lamina, showing that DMPK is required for NE stability. Our data demonstrate for the first time that DMPK is a critical component of the NE. These novel findings suggest that reduced DMPK may contribute to NE instability, a common mechanism of skeletal muscle wasting in muscular dystrophies.


Subject(s)
Epithelial Cells/enzymology , Muscle Proteins/metabolism , Myoblasts, Skeletal/enzymology , Myotonic Dystrophy/enzymology , Nuclear Envelope/enzymology , Protein Serine-Threonine Kinases/metabolism , Epithelial Cells/pathology , Gene Expression Regulation/genetics , HeLa Cells , Humans , Lamins/genetics , Lamins/metabolism , Muscle Proteins/genetics , Myoblasts, Skeletal/pathology , Myotonic Dystrophy/genetics , Myotonic Dystrophy/pathology , Myotonin-Protein Kinase , Nuclear Envelope/genetics , Nuclear Envelope/pathology , Protein Serine-Threonine Kinases/genetics
15.
Dev Dyn ; 237(9): 2353-66, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18729234

ABSTRACT

Myotonic dystrophy (DM1) is a multi-systemic disease caused by a triplet nucleotide repeat expansion in the 3' untranslated region of the gene coding for myotonic dystrophy protein kinase (DMPK). The primary pathophysiology of DM1 is thought to result from RNA transport and processing defects. The function of DMPK in development or any potential role in DM1 remains unknown. Here we report a novel role for DMPK in myogenesis. We have discovered a specific expression pattern of DMPK in mouse and chick embryonic development. DMPK is expressed in postmitotic cardiac and skeletal myocytes and developmental signaling centers. During cardiac myocyte maturation, DMPK migrates from perinuclear to cellular membrane localization. Manipulating DMPK levels in cultured cardiac and skeletal myocytes has revealed a key role for DMPK in myocyte differentiation. Overexpression of DMPK induces cell rounding and apoptosis in myocytes. In addition, DMPK is necessary for myogenin expression in differentiating C2C12 myoblasts.


Subject(s)
Muscle Cells/metabolism , Muscle Fibers, Skeletal/metabolism , Myocytes, Cardiac/metabolism , Protein Serine-Threonine Kinases/metabolism , Animals , Apoptosis/physiology , Blotting, Western , Cell Differentiation/physiology , Cell Line , Cells, Cultured , Chick Embryo , Chickens , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Immunohistochemistry , Mice , Microscopy, Confocal , Muscle Cells/cytology , Muscle Fibers, Skeletal/cytology , Myocytes, Cardiac/cytology , Myotonin-Protein Kinase , Protein Serine-Threonine Kinases/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Transfection
16.
J Mol Recognit ; 19(3): 215-26, 2006.
Article in English | MEDLINE | ID: mdl-16680721

ABSTRACT

Myotonic dystrophy protein kinase (DMPK) was the initial representative of a ubiquitous protein kinase family that regulates cell size and shape. DMPK is highly expressed in heart and skeletal muscle and transgenic over-expression induces cardiac hypertrophy. The characterization of DMPK has been limited by the paucity of immunological reagents with high affinity and well-defined specificity. Amino acid sequence data was used to predict the surface exposure of the coil-coiled domain of DMPK. These exposed amino acids were substituted into an extremely stable coiled-coil template to produce a peptide antigen. Sera from mice immunized with the peptide conjugated to keyhole limpet hemocyanin were screened against recombinant DMPK using Western blots. Murine spleens expressing DMPK antibodies were used to produce hybridoma cell lines. Hybridoma supernatants were further screened against recombinant DMPK and four clonal hybridoma cell lines expressing DMPK antibodies were generated. These four monoclonal antibodies recognized recombinant DMPK in Western blots of COS-1 cell lysates expressing high levels of recombinant DMPK and immunoprecipitated recombinant DMPK from COS-1 cell lysates. The identity of the immunoprecipitated DMPK was confirmed by MALDI-TOF mass spectrometry and peptide mass fingerprinting. DMPK was the only protein detected in the immunoprecipitates, indicating the high specificity of the antibodies. Western blots immunostained with two of the monoclonal antibodies specifically recognized the two isoforms of endogenous DMPK, DMPK-1 and DMPK-2, that are expressed at low levels in the human heart. The recognition of low amounts of DMPK-1 and DMPK-2 indicates the high affinity of these antibodies. A human heart lysate was subjected to ammonium sulfate precipitation and column chromatography to produce a fraction that was enriched in DMPK. One of the monoclonal antibodies immunoprecipitated endogenous DMPK from this fraction. This antibody was used for immuno-localization studies of an adenoviral DMPK construct, expressed in adult mouse cardiac myocytes. This construct was localized to the intercalated disc, the site of endogenous DMPK, indicating that this antibody is applicable to immuno-localization studies. This study demonstrates the utility of the described procedure for generation of specific monoclonal antibodies with high affinity for epitopes in coiled-coiled domains of mammalian proteins expressed at low levels.


Subject(s)
Antibodies, Monoclonal/immunology , Protein Serine-Threonine Kinases/immunology , 3T3 Cells , Adult , Amino Acid Sequence , Animals , Antibodies, Monoclonal/biosynthesis , Antibodies, Monoclonal/isolation & purification , Antibody Specificity/immunology , Blotting, Western , COS Cells , Chlorocebus aethiops , Humans , Immunohistochemistry , Isoenzymes/immunology , Isoenzymes/metabolism , Male , Mice , Molecular Sequence Data , Myocardium/enzymology , Myocardium/metabolism , Myotonin-Protein Kinase , Peptide Fragments/chemistry , Peptide Fragments/immunology , Protein Serine-Threonine Kinases/metabolism , Protein Structure, Secondary , Sequence Homology, Amino Acid , Spleen/enzymology , Spleen/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...