Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
Am J Respir Cell Mol Biol ; 64(3): 368-378, 2021 03.
Article in English | MEDLINE | ID: mdl-33290178

ABSTRACT

Human genome-wide association studies (GWASs) have identified more than 270 loci associated with pulmonary function; however, follow-up studies to determine causal genes at these loci are few. SNPs in low-density lipoprotein receptor-related protein 1 (LRP1) are associated with human pulmonary function in GWASs. Using murine models, we investigated the effect of genetic disruption of the Lrp1 gene in smooth muscle cells on pulmonary function in naive animals and after exposure to bacterial LPS or house dust mite extract. Disruption of Lrp1 in smooth muscle cells leads to an increase in tissue resistance, elastance, and tissue elastance at baseline. Furthermore, disruption of Lrp1 in smooth muscle increases airway responsiveness as measured by increased total lung resistance and airway resistance after methacholine. Immune cell counts in BAL fluid were increased in animals with Lrp1 disruption. The difference in airway responsiveness by genotype observed in naive animals was not observed after LPS or house dust mite extract exposure. To further explore the mechanisms contributing to changes in pulmonary function, we identified several ligands dysregulated with Lrp1 disruption in smooth muscle cells. These data suggest that dysregulation of LRP1 in smooth muscle cells affects baseline pulmonary function and airway responsiveness and helps establish LRP1 as the causal gene at this GWAS locus.


Subject(s)
Genome-Wide Association Study , Low Density Lipoprotein Receptor-Related Protein-1/metabolism , Lung/physiology , Animals , Bronchoalveolar Lavage Fluid , Humans , Lipopolysaccharides/pharmacology , Mice , Muscle, Smooth/drug effects , Muscle, Smooth/metabolism , Polymorphism, Single Nucleotide/genetics , Proteome/metabolism , Pyroglyphidae/physiology , Quantitative Trait Loci/genetics
2.
Environ Epidemiol ; 3(4): e055, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31538136

ABSTRACT

BACKGROUND: Numerous health effects of smoking are well-known; associations with semen quality are uncertain. Most previous studies did not adjust for potential confounders and had limited information on age at smoking initiation or smoking cessation. METHODS: We investigated 1,631 healthy fertile men in the Nanjing Medical University Longitudinal Investigation of Fertility and the Environment (NMU-LIFE) study. Relationships were examined using multivariable linear regression controlling for potential covariates. RESULTS: We found a significant decrease in semen volume (ß = -0.10, P = 0.001) and total sperm count (ß = -0.42, P = 0.037), and significant increase in total motility (ß = 6.02, P = 0.037) and progressive motility (ß = 5.52, P = 0.037) in ever smokers of pack-years ≥10 compared with never smokers. We observed an inverse dose-dependent relation between smoking pack-years and semen volume (P < 0.001) and total sperm count (P = 0.010) and a positive dose-dependent relation between smoking pack-years and both total motility and progressive motility (P = 0.042 and 0.048, respectively). No significant differences in semen quality were detected among ever smokers with different ages at smoking initiation nor in former smokers compared with never smokers. CONCLUSIONS: Cigarette smoking was associated with lower semen volume and total sperm count and higher sperm motility. Smoking cessation might have a restorative effect on semen quality. This finding has important implications for public health research and for understanding the development of abnormal semen quality.

3.
Clin Epigenetics ; 11(1): 37, 2019 02 28.
Article in English | MEDLINE | ID: mdl-30819252

ABSTRACT

BACKGROUND: Ambient air pollution is associated with numerous adverse health outcomes, but the underlying mechanisms are not well understood; epigenetic effects including altered DNA methylation could play a role. To evaluate associations of long-term air pollution exposure with DNA methylation in blood, we conducted an epigenome-wide association study in a Korean chronic obstructive pulmonary disease cohort (N = 100 including 60 cases) using Illumina's Infinium HumanMethylation450K Beadchip. Annual average concentrations of particulate matter ≤ 10 µm in diameter (PM10) and nitrogen dioxide (NO2) were estimated at participants' residential addresses using exposure prediction models. We used robust linear regression to identify differentially methylated probes (DMPs) and two different approaches, DMRcate and comb-p, to identify differentially methylated regions (DMRs). RESULTS: After multiple testing correction (false discovery rate < 0.05), there were 12 DMPs and 27 DMRs associated with PM10 and 45 DMPs and 57 DMRs related to NO2. DMP cg06992688 (OTUB2) and several DMRs were associated with both exposures. Eleven DMPs in relation to NO2 confirmed previous findings in Europeans; the remainder were novel. Methylation levels of 39 DMPs were associated with expression levels of nearby genes in a separate dataset of 3075 individuals. Enriched networks were related to outcomes associated with air pollution including cardiovascular and respiratory diseases as well as inflammatory and immune responses. CONCLUSIONS: This study provides evidence that long-term ambient air pollution exposure impacts DNA methylation. The differential methylation signals can serve as potential air pollution biomarkers. These results may help better understand the influences of ambient air pollution on human health.


Subject(s)
Air Pollution/analysis , DNA Methylation , Pulmonary Disease, Chronic Obstructive/genetics , Whole Genome Sequencing/methods , Aged , Aged, 80 and over , Air Pollution/adverse effects , Cohort Studies , DNA Methylation/drug effects , Epigenesis, Genetic , Female , Gene Expression Regulation , Humans , Male , Nitrogen Dioxide/adverse effects , Nitrogen Dioxide/analysis , Particulate Matter/analysis , Pulmonary Disease, Chronic Obstructive/chemically induced , Republic of Korea
4.
Nanotoxicology ; 13(5): 644-663, 2019 06.
Article in English | MEDLINE | ID: mdl-30704319

ABSTRACT

Nano-titanium dioxide (nano-TiO2), though one of the most utilized and produced engineered nanomaterials (ENMs), diminishes cardiovascular function through dysregulation of metabolism and mitochondrial bioenergetics following inhalation exposure. The molecular mechanisms governing this cardiac dysfunction remain largely unknown. The purpose of this study was to elucidate molecular mediators that connect nano-TiO2 exposure with impaired cardiac function. Specifically, we were interested in the role of microRNA (miRNA) expression in the resulting dysfunction. Not only are miRNA global regulators of gene expression, but also miRNA-based therapeutics provide a realistic treatment modality. Wild type and MiRNA-378a knockout mice were exposed to nano-TiO2 with an aerodynamic diameter of 182 ± 1.70 nm and a mass concentration of 11.09 mg/m3 for 4 h. Cardiac function, utilizing the Vevo 2100 Imaging System, electron transport chain complex activities, and mitochondrial respiration assessed cardiac and mitochondrial function. Immunoblotting and qPCR examined molecular targets of miRNA-378a. MiRNA-378a-3p expression was increased 48 h post inhalation exposure to nano-TiO2. Knockout of miRNA-378a preserved cardiac function following exposure as revealed by preserved E/A ratio and E/SR ratio. In knockout animals, complex I, III, and IV activities (∼2- to 6-fold) and fatty acid respiration (∼5-fold) were significantly increased. MiRNA-378a regulated proteins involved in mitochondrial fusion, transcription, and fatty acid metabolism. MiRNA-378a-3p acts as a negative regulator of mitochondrial metabolic and biogenesis pathways. MiRNA-378a knockout animals provide a protective effect against nano-TiO2 inhalation exposure by altering mitochondrial structure and function. This is the first study to manipulate a miRNA to attenuate the effects of ENM exposure.


Subject(s)
Cardiovascular Physiological Phenomena/drug effects , Heart/drug effects , Inhalation Exposure/adverse effects , MicroRNAs/genetics , Nanoparticles/toxicity , Titanium/toxicity , Animals , Cardiovascular Physiological Phenomena/genetics , Echocardiography , Gene Expression/drug effects , Heart/diagnostic imaging , Mice , Mice, Knockout , Mitochondria/drug effects , Mitochondria/metabolism , Nanoparticles/chemistry , Titanium/chemistry
5.
J Mol Cell Cardiol ; 119: 104-115, 2018 06.
Article in English | MEDLINE | ID: mdl-29733819

ABSTRACT

>99% of the mitochondrial proteome is nuclear-encoded. The mitochondrion relies on a coordinated multi-complex process for nuclear genome-encoded mitochondrial protein import. Mitochondrial heat shock protein 70 (mtHsp70) is a key component of this process and a central constituent of the protein import motor. Type 2 diabetes mellitus (T2DM) disrupts mitochondrial proteomic signature which is associated with decreased protein import efficiency. The goal of this study was to manipulate the mitochondrial protein import process through targeted restoration of mtHsp70, in an effort to restore proteomic signature and mitochondrial function in the T2DM heart. A novel line of cardiac-specific mtHsp70 transgenic mice on the db/db background were generated and cardiac mitochondrial subpopulations were isolated with proteomic evaluation and mitochondrial function assessed. MicroRNA and epigenetic regulation of the mtHsp70 gene during T2DM were also evaluated. MtHsp70 overexpression restored cardiac function and nuclear-encoded mitochondrial protein import, contributing to a beneficial impact on proteome signature and enhanced mitochondrial function during T2DM. Further, transcriptional repression at the mtHsp70 genomic locus through increased localization of H3K27me3 during T2DM insult was observed. Our results suggest that restoration of a key protein import constituent, mtHsp70, provides therapeutic benefit through attenuation of mitochondrial and contractile dysfunction in T2DM.


Subject(s)
Diabetes Mellitus, Type 2/genetics , HSP70 Heat-Shock Proteins/genetics , Mitochondrial Proteins/genetics , Myocardium/metabolism , Animals , Diabetes Mellitus, Type 2/pathology , Epigenesis, Genetic , Humans , Lipid Peroxidation/genetics , Mice , Mice, Transgenic , Mitochondria, Heart/genetics , Myocardium/pathology , Oxidative Stress/genetics , Protein Transport/genetics , Proteome/genetics
6.
Nanotoxicology ; 12(1): 32-48, 2018 02.
Article in English | MEDLINE | ID: mdl-29243970

ABSTRACT

Nanotechnology offers innovation in products from cosmetics to drug delivery, leading to increased engineered nanomaterial (ENM) exposure. Unfortunately, health impacts of ENM are not fully realized. Titanium dioxide (TiO2) is among the most widely produced ENM due to its use in numerous applications. Extrapulmonary effects following pulmonary exposure have been identified and may involve reactive oxygen species (ROS). The goal of this study was to determine the extent of ROS involvement on cardiac function and the mitochondrion following nano-TiO2 exposure. To address this question, we utilized a transgenic mouse model with overexpression of a novel mitochondrially-targeted antioxidant enzyme (phospholipid hydroperoxide glutathione peroxidase; mPHGPx) which provides protection against oxidative stress to lipid membranes. MPHGPx mice and littermate controls were exposed to nano-TiO2 aerosols (Evonik, P25) to provide a calculated pulmonary deposition of 11 µg/mouse. Twenty-four hours following exposure, we observed diastolic dysfunction as evidenced by E/A ratios greater than 2 and increased radial strain during diastole in wild-type mice (p < 0.05 for both), indicative of restrictive filling. Overexpression of mPHGPx mitigated the contractile deficits resulting from nano-TiO2 exposure. To investigate the cellular mechanisms associated with the observed cardiac dysfunction, we focused our attention on the mitochondrion. We observed a significant increase in ROS production (p < 0.05) and decreased mitochondrial respiratory function (p < 0.05) following nano-TiO2 exposure which were attenuated in mPHGPx transgenic mice. In summary, nano-TiO2 inhalation exposure is associated with cardiac diastolic dysfunction and mitochondrial functional alterations, which can be mitigated by the overexpression of mPHGPx, suggesting ROS contribution in the development of contractile and bioenergetic dysfunction.


Subject(s)
Heart/drug effects , Mitochondria/drug effects , Mitochondria/pathology , Nanostructures/toxicity , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism , Titanium/administration & dosage , Titanium/toxicity , Administration, Inhalation , Animals , Antioxidants/pharmacology , Glutathione Peroxidase/genetics , Heart/physiopathology , Male , Mice , Mice, Transgenic , Mitochondria/metabolism , Nanostructures/administration & dosage , Phospholipid Hydroperoxide Glutathione Peroxidase
7.
J Mol Cell Cardiol ; 110: 15-25, 2017 09.
Article in English | MEDLINE | ID: mdl-28709769

ABSTRACT

Cardiovascular disease is the primary cause of mortality for individuals with type 2 diabetes mellitus. During the diabetic condition, cardiovascular dysfunction can be partially attributed to molecular changes in the tissue, including alterations in microRNA (miRNA) interactions. MiRNAs have been reported in the mitochondrion and their presence may influence cellular bioenergetics, creating decrements in functional capacity. In this study, we examined the roles of Argonaute 2 (Ago2), a protein associated with cytosolic and mitochondrial miRNAs, and Polynucleotide Phosphorylase (PNPase), a protein found in the inner membrane space of the mitochondrion, to determine their role in mitochondrial miRNA import. In cardiac tissue from human and mouse models of type 2 diabetes mellitus, Ago2 protein levels were unchanged while PNPase protein expression levels were increased; also, there was an increase in the association between both proteins in the diabetic state. MiRNA-378 was found to be significantly increased in db/db mice, leading to decrements in ATP6 levels and ATP synthase activity, which was also exhibited when overexpressing PNPase in HL-1 cardiomyocytes and in HL-1 cells with stable miRNA-378 overexpression (HL-1-378). To assess potential therapeutic interventions, flow cytometry evaluated the capacity for targeting miRNA-378 species in mitochondria through antimiR treatment, revealing miRNA-378 level-dependent inhibition. Our study establishes PNPase as a contributor to mitochondrial miRNA import through the transport of miRNA-378, which may regulate bioenergetics during type 2 diabetes mellitus. Further, our data provide evidence that manipulation of PNPase levels may enhance the delivery of antimiR therapeutics to mitochondria in physiological and pathological conditions.


Subject(s)
MicroRNAs/metabolism , Mitochondria/metabolism , Polyribonucleotide Nucleotidyltransferase/metabolism , RNA Transport , Animals , Antagomirs , Argonaute Proteins/metabolism , Cell Line , Diabetes Mellitus, Type 2/enzymology , Diabetes Mellitus, Type 2/metabolism , Disease Models, Animal , Energy Metabolism , Fluorescence , Gene Expression Regulation , Gene Knockdown Techniques , Humans , Mice , Protein Binding
8.
Am J Physiol Lung Cell Mol Physiol ; 312(4): L520-L530, 2017 Apr 01.
Article in English | MEDLINE | ID: mdl-28130264

ABSTRACT

Human genome-wide association studies have identified over 50 loci associated with pulmonary function and related phenotypes, yet follow-up studies to determine causal genes or variants are rare. Single nucleotide polymorphisms in serotonin receptor 4 (HTR4) are associated with human pulmonary function in genome-wide association studies and follow-up animal work has demonstrated that Htr4 is causally associated with pulmonary function in mice, although the precise mechanisms were not identified. We sought to elucidate the role of neural innervation and pulmonary architecture in the lung phenotype of Htr4-/- animals. We report here that the Htr4-/- phenotype in mouse is dependent on vagal innervation to the lung. Both ex vivo tracheal ring reactivity and in vivo flexiVent pulmonary functional analyses demonstrate that vagotomy abrogates the Htr4-/- airway hyperresponsiveness phenotype. Hyperpolarized 3He gas magnetic resonance imaging and stereological assessment of wild-type and Htr4-/- mice reveal no observable differences in lung volume, inflation characteristics, or pulmonary microarchitecture. Finally, control of breathing experiments reveal substantive differences in baseline breathing characteristics between mice with/without functional HTR4 in breathing frequency, relaxation time, flow rate, minute volume, time of inspiration and expiration and breathing pauses. These results suggest that HTR4's role in pulmonary function likely relates to neural innervation and control of breathing.


Subject(s)
Lung/innervation , Lung/physiology , Receptors, Serotonin, 5-HT4/deficiency , Vagus Nerve/physiology , Animals , Genotype , In Vitro Techniques , Mice, Inbred C57BL , Muscle Contraction/physiology , Phenotype , Pulmonary Ventilation/physiology , Receptors, Serotonin, 5-HT4/metabolism , Respiration , Respiratory Function Tests , Respiratory Hypersensitivity/physiopathology , Trachea/physiology , Vagotomy , Vagus Nerve/surgery
9.
Am J Physiol Heart Circ Physiol ; 312(3): H446-H458, 2017 Mar 01.
Article in English | MEDLINE | ID: mdl-28011589

ABSTRACT

Nanomaterial production is expanding as new industrial and consumer applications are introduced. Nevertheless, the impacts of exposure to these compounds are not fully realized. The present study was designed to determine whether gestational nano-sized titanium dioxide exposure impacts cardiac and metabolic function of developing progeny. Pregnant Sprague-Dawley rats were exposed to nano-aerosols (~10 mg/m3, 130- to 150-nm count median aerodynamic diameter) for 7-8 nonconsecutive days, beginning at gestational day 5-6 Physiological and bioenergetic effects on heart function and cardiomyocytes across three time points, fetal (gestational day 20), neonatal (4-10 days), and young adult (6-12 wk), were evaluated. Functional analysis utilizing echocardiography, speckle-tracking based strain, and cardiomyocyte contractility, coupled with mitochondrial energetics, revealed effects of nano-exposure. Maternal exposed progeny demonstrated a decrease in E- and A-wave velocities, with a 15% higher E-to-A ratio than controls. Myocytes isolated from exposed animals exhibited ~30% decrease in total contractility, departure velocity, and area of contraction. Bioenergetic analysis revealed a significant increase in proton leak across all ages, accompanied by decreases in metabolic function, including basal respiration, maximal respiration, and spare capacity. Finally, electron transport chain complex I and IV activities were negatively impacted in the exposed group, which may be linked to a metabolic shift. Molecular data suggest that an increase in fatty acid metabolism, uncoupling, and cellular stress proteins may be associated with functional deficits of the heart. In conclusion, gestational nano-exposure significantly impairs the functional capabilities of the heart through cardiomyocyte impairment, which is associated with mitochondrial dysfunction.NEW & NOTEWORTHY Cardiac function is evaluated, for the first time, in progeny following maternal nanomaterial inhalation. The findings indicate that exposure to nano-sized titanium dioxide (nano-TiO2) during gestation negatively impacts cardiac function and mitochondrial respiration and bioenergetics. We conclude that maternal nano-TiO2 inhalation contributes to adverse cardiovascular health effects, lasting into adulthood.


Subject(s)
Energy Metabolism/drug effects , Heart/diagnostic imaging , Myocardium/pathology , Nanostructures/toxicity , Prenatal Exposure Delayed Effects/pathology , Aging , Animals , Echocardiography , Electron Transport Complex I/drug effects , Electron Transport Complex I/metabolism , Electron Transport Complex IV/drug effects , Electron Transport Complex IV/metabolism , Female , Heart Diseases/chemically induced , Heart Diseases/diagnostic imaging , Heart Diseases/pathology , Heart Function Tests , Myocardial Contraction/drug effects , Myocytes, Cardiac/drug effects , Pregnancy , Rats , Rats, Sprague-Dawley , Titanium/toxicity
10.
J Mol Cell Cardiol ; 90: 74-83, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26654913

ABSTRACT

Enhanced sensitivity in echocardiographic analyses may allow for early detection of changes in cardiac function beyond the detection limits of conventional echocardiographic analyses, particularly in a small animal model. The goal of this study was to compare conventional echocardiographic measurements and speckle-tracking based strain imaging analyses in a small animal model of type 1 diabetes mellitus. Conventional analyses revealed differences in ejection fraction, fractional shortening, cardiac output, and stroke volume in diabetic animals relative to controls at 6-weeks post-diabetic onset. In contrast, when assessing short- and long-axis speckle-tracking based strain analyses, diabetic mice showed changes in average systolic radial strain, radial strain rate, radial displacement, and radial velocity, as well as decreased circumferential and longitudinal strain rate, as early as 1-week post-diabetic onset and persisting throughout the diabetic study. Further, we performed regional analyses for the LV and found that the free wall region was affected in both the short- and long-axis when assessing radial dimension parameters. These changes began 1-week post-diabetic onset and remained throughout the progression of the disease. These findings demonstrate the use of speckle-tracking based strain as an approach to elucidate cardiac dysfunction from a global perspective, identifying left ventricular cardiac regions affected during the progression of type 1 diabetes mellitus earlier than contractile changes detected by conventional echocardiographic measurements.


Subject(s)
Diabetes Mellitus, Experimental/physiopathology , Diabetic Cardiomyopathies/diagnosis , Echocardiography/methods , Heart Ventricles/physiopathology , Ventricular Dysfunction, Left/diagnosis , Animals , Cardiac Output , Diabetic Cardiomyopathies/physiopathology , Diastole , Male , Mice , Stroke Volume , Systole , Ventricular Dysfunction, Left/physiopathology
11.
Am J Physiol Heart Circ Physiol ; 309(12): H2017-30, 2015 Dec 15.
Article in English | MEDLINE | ID: mdl-26497962

ABSTRACT

Throughout the United States, air pollution correlates with adverse health outcomes, and cardiovascular disease incidence is commonly increased following environmental exposure. In areas surrounding active mountaintop removal mines (MTM), a further increase in cardiovascular morbidity is observed and may be attributed in part to particulate matter (PM) released from the mine. The mitochondrion has been shown to be central in the etiology of many cardiovascular diseases, yet its roles in PM-related cardiovascular effects are not realized. In this study, we sought to elucidate the cardiac processes that are disrupted following exposure to mountaintop removal mining particulate matter (PM MTM). To address this question, we exposed male Sprague-Dawley rats to PM MTM, collected within one mile of an active MTM site, using intratracheal instillation. Twenty-four hours following exposure, we evaluated cardiac function, apoptotic indices, and mitochondrial function. PM MTM exposure elicited a significant decrease in ejection fraction and fractional shortening compared with controls. Investigation into the cellular impacts of PM MTM exposure identified a significant increase in mitochondrial-induced apoptotic signaling, as reflected by an increase in TUNEL-positive nuclei and increased caspase-3 and -9 activities. Finally, a significant increase in mitochondrial transition pore opening leading to decreased mitochondrial function was identified following exposure. In conclusion, our data suggest that pulmonary exposure to PM MTM increases cardiac mitochondrial-associated apoptotic signaling and decreases mitochondrial function concomitant with decreased cardiac function. These results suggest that increased cardiovascular disease incidence in populations surrounding MTM mines may be associated with increased cardiac cell apoptotic signaling and decreased mitochondrial function.


Subject(s)
Air Pollutants, Occupational/toxicity , Air Pollution/adverse effects , Heart Diseases/chemically induced , Mitochondrial Diseases/chemically induced , Particulate Matter/toxicity , Animals , Apoptosis/drug effects , Caspases/metabolism , Echocardiography , Environmental Exposure , Environmental Monitoring , Heart Diseases/diagnostic imaging , In Situ Nick-End Labeling , Injections, Spinal , Male , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondria/ultrastructure , Mitochondrial Diseases/diagnostic imaging , Myocardial Contraction/drug effects , Oxygen Consumption/drug effects , Rats , Rats, Sprague-Dawley
12.
Circ Cardiovasc Genet ; 8(6): 785-802, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26377859

ABSTRACT

BACKGROUND: Cardiomyocytes are rich in mitochondria which are situated in spatially distinct subcellular regions, including those under the plasma membrane, subsarcolemmal mitochondria, and those between the myofibrils, interfibrillar mitochondria. We previously observed subpopulation-specific differences in mitochondrial proteomes following diabetic insult. The objective of this study was to determine whether mitochondrial genome-encoded proteins are regulated by microRNAs inside the mitochondrion and whether subcellular spatial location or diabetes mellitus influences the dynamics. METHODS AND RESULTS: Using microarray technology coupled with cross-linking immunoprecipitation and next generation sequencing, we identified a pool of mitochondrial microRNAs, termed mitomiRs, that are redistributed in spatially distinct mitochondrial subpopulations in an inverse manner following diabetic insult. Redistributed mitomiRs displayed distinct interactions with the mitochondrial genome requiring specific stoichiometric associations with RNA-induced silencing complex constituents argonaute-2 (Ago2) and fragile X mental retardation-related protein 1 (FXR1) for translational regulation. In the presence of Ago2 and FXR1, redistribution of mitomiR-378 to the interfibrillar mitochondria following diabetic insult led to downregulation of mitochondrially encoded F0 component ATP6. Next generation sequencing analyses identified specific transcriptome and mitomiR sequences associated with ATP6 regulation. Overexpression of mitomiR-378 in HL-1 cells resulted in its accumulation in the mitochondrion and downregulation of functional ATP6 protein, whereas antagomir blockade restored functional ATP6 protein and cardiac pump function. CONCLUSIONS: We propose mitomiRs can translationally regulate mitochondrially encoded proteins in spatially distinct mitochondrial subpopulations during diabetes mellitus. The results reveal the requirement of RNA-induced silencing complex constituents in the mitochondrion for functional mitomiR translational regulation and provide a connecting link between diabetic insult and ATP synthase function.


Subject(s)
Diabetes Mellitus, Experimental/metabolism , Diabetic Cardiomyopathies/metabolism , Genome, Mitochondrial , MicroRNAs/metabolism , Mitochondria, Heart/metabolism , Protein Biosynthesis , RNA/metabolism , Animals , Diabetes Mellitus, Experimental/pathology , Diabetic Cardiomyopathies/pathology , Mice , Mitochondria, Heart/pathology , Mitochondrial Proteins , RNA, Mitochondrial
13.
J Mol Cell Cardiol ; 79: 212-23, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25463274

ABSTRACT

Mitofilin, also known as heart muscle protein, is an inner mitochondrial membrane structural protein that plays a central role in maintaining cristae morphology and structure. It is a critical component of the mitochondrial contact site and cristae organizing system (MICOS) complex which is important for mitochondrial architecture and cristae morphology. Our laboratory has previously reported alterations in mitochondrial morphology and proteomic make-up during type 1 diabetes mellitus, with mitofilin being significantly down-regulated in interfibrillar mitochondria (IFM). The goal of this study was to investigate whether overexpression of mitofilin can limit mitochondrial disruption associated with the diabetic heart through restoration of mitochondrial morphology and function. A transgenic mouse line overexpressing mitofilin was generated and mice injected intraperitoneally with streptozotocin using a multi low-dose approach. Five weeks following diabetes mellitus onset, cardiac contractile function was assessed. Restoration of ejection fraction and fractional shortening was observed in mitofilin diabetic mice as compared to wild-type controls (P<0.05 for both). Decrements observed in electron transport chain (ETC) complex I, III, IV and V activities, state 3 respiration, lipid peroxidation as well as mitochondria membrane potential in type 1 diabetic IFM were restored in mitofilin diabetic mice (P<0.05 for all). Qualitative analyses of electron micrographs revealed restoration of mitochondrial cristae structure in mitofilin diabetic mice as compared to wild-type controls. Furthermore, measurement of mitochondrial internal complexity using flow cytometry displayed significant reduction in internal complexity in diabetic IFM which was restored in mitofilin diabetic IFM (P<0.05). Taken together these results suggest that transgenic overexpression of mitofilin preserves mitochondrial structure, leading to restoration of mitochondrial function and attenuation of cardiac contractile dysfunction in the diabetic heart.


Subject(s)
Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Experimental/physiopathology , Heart/physiopathology , Mitochondria, Heart/metabolism , Mitochondrial Proteins/metabolism , Muscle Proteins/metabolism , Animals , Blotting, Western , Body Weight , Diabetes Mellitus, Experimental/metabolism , Electron Transport , Humans , Lipid Peroxidation , Male , Membrane Potential, Mitochondrial , Mice, Transgenic , Mitochondria, Heart/ultrastructure , Mitochondrial Dynamics , Myocardial Contraction , Native Polyacrylamide Gel Electrophoresis , Organ Size , Oxidative Stress
14.
Nanotoxicology ; 9(8): 941-51, 2015.
Article in English | MEDLINE | ID: mdl-25475392

ABSTRACT

Due to the ongoing evolution of nanotechnology, there is a growing need to assess the toxicological outcomes in under-studied populations in order to properly consider the potential of engineered nanomaterials (ENM) and fully enhance their safety. Recently, we and others have explored the vascular consequences associated with gestational nanomaterial exposure, reporting microvascular dysfunction within the uterine circulation of pregnant dams and the tail artery of fetal pups. It has been proposed (via work derived by the Barker Hypothesis) that mitochondrial dysfunction and subsequent oxidative stress mechanisms as a possible link between a hostile gestational environment and adult disease. Therefore, in this study, we exposed pregnant Sprague-Dawley rats to nanosized titanium dioxide aerosols after implantation (gestational day 6). Pups were delivered, and the progeny grew into adulthood. Microvascular reactivity, mitochondrial respiration and hydrogen peroxide production of the coronary and uterine circulations of the female offspring were evaluated. While there were no significant differences within the maternal or litter characteristics, endothelium-dependent dilation and active mechanotransduction in both coronary and uterine arterioles were significantly impaired. In addition, there was a significant reduction in maximal mitochondrial respiration (state 3) in the left ventricle and uterus. These studies demonstrate microvascular dysfunction and coincide with mitochondrial inefficiencies in both the cardiac and uterine tissues, which may represent initial evidence that prenatal ENM exposure produces microvascular impairments that persist throughout multiple developmental stages.


Subject(s)
Microvessels/drug effects , Mitochondria/drug effects , Mitochondria/pathology , Nanoparticles/toxicity , Prenatal Exposure Delayed Effects/metabolism , Titanium/toxicity , Animals , Cell Respiration/drug effects , Coronary Vessels/drug effects , Female , Hydrogen Peroxide/metabolism , Mechanotransduction, Cellular , Mitochondria/metabolism , Nanoparticles/administration & dosage , Nanoparticles/chemistry , Oxidative Stress/drug effects , Pregnancy , Rats , Titanium/administration & dosage , Titanium/chemistry , Uterine Artery/drug effects
15.
Am J Physiol Heart Circ Physiol ; 307(1): H54-65, 2014 Jul 01.
Article in English | MEDLINE | ID: mdl-24778174

ABSTRACT

The mitochondrion has been implicated in the development of diabetic cardiomyopathy. Examination of cardiac mitochondria is complicated by the existence of spatially distinct subpopulations including subsarcolemmal (SSM) and interfibrillar (IFM). Dysfunction to cardiac SSM has been reported in murine models of type 2 diabetes mellitus; however, subpopulation-based mitochondrial analyses have not been explored in type 2 diabetic human heart. The goal of this study was to determine the impact of type 2 diabetes mellitus on cardiac mitochondrial function in the human patient. Mitochondrial subpopulations from atrial appendages of patients with and without type 2 diabetes were examined. Complex I- and fatty acid-mediated mitochondrial respiration rates were decreased in diabetic SSM compared with nondiabetic (P ≤ 0.05 for both), with no change in IFM. Electron transport chain (ETC) complexes I and IV activities were decreased in diabetic SSM compared with nondiabetic (P ≤ 0.05 for both), with a concomitant decline in their levels (P ≤ 0.05 for both). Regression analyses comparing comorbidities determined that diabetes mellitus was the primary factor accounting for mitochondrial dysfunction. Linear spline models examining correlative risk for mitochondrial dysfunction indicated that patients with diabetes display the same degree of state 3 and electron transport chain complex I dysfunction in SSM regardless of the extent of glycated hemoglobin (HbA1c) and hyperglycemia. Overall, the results suggest that independent of other pathologies, mitochondrial dysfunction is present in cardiac SSM of patients with type 2 diabetes and the degree of dysfunction is consistent regardless of the extent of elevated HbA1c or blood glucose levels.


Subject(s)
Diabetes Mellitus, Type 2/physiopathology , Diabetic Cardiomyopathies/physiopathology , Membrane Potential, Mitochondrial , Mitochondria, Heart/metabolism , Mitochondria, Heart/pathology , Sarcolemma/metabolism , Cell Respiration , Cells, Cultured , Diabetes Mellitus, Type 2/pathology , Diabetic Cardiomyopathies/pathology , Female , Humans , Male , Middle Aged , Sarcolemma/pathology
16.
Life Sci ; 93(8): 313-22, 2013 Sep 03.
Article in English | MEDLINE | ID: mdl-23872101

ABSTRACT

AIMS: We have previously reported alterations in cardiolipin content and inner mitochondrial membrane (IMM) proteomic make-up specifically in interfibrillar mitochondria (IFM) in the type 1 diabetic heart; however, the mechanism underlying this alteration is unknown. The goal of this study was to determine how the cardiolipin biosynthetic pathway and cardiolipin-IMM protein interactions are impacted by type 1 diabetes mellitus. MAIN METHODS: Male FVB mice were made diabetic by multiple low-dose streptozotocin injections and sacrificed five weeks post-diabetic onset. Messenger RNA was measured and cardiac mitochondrial subpopulations were isolated. Further mitochondrial functional experimentation included evaluating the protein expression of the enzymes directly responsible for cardiolipin biosynthesis, as well as ATP synthase activity. Interactions between cardiolipin and ATP synthase subunits were also examined. KEY FINDINGS: Western blot analysis revealed a significant decrease in cardiolipin synthase (CRLS) protein content in diabetic IFM, with a concomitant decrease in its activity. ATP synthase activity was also significantly decreased. We identified two novel direct interactions between two subunits of the ATP synthase F0 complex (ATP5F1 and ATP5H), both of which were significantly decreased in diabetic IFM. SIGNIFICANCE: Overall, these results indicate that type 1 diabetes mellitus negatively impacts the cardiolipin biosynthetic pathway specifically at CRLS, contributing to decreased cardiolipin content and loss of interactions with key ATP synthase F0 complex constituents in the IFM.


Subject(s)
Cardiolipins/biosynthesis , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Type 1/metabolism , Mitochondria, Heart/metabolism , Mitochondrial Membranes/metabolism , Animals , Blotting, Western , Male , Membrane Proteins/metabolism , Mice , Mitochondrial Proton-Translocating ATPases/metabolism , RNA, Messenger/metabolism , Streptozocin , Transferases (Other Substituted Phosphate Groups)/metabolism
17.
J Appl Physiol (1985) ; 115(4): 514-24, 2013 Aug 15.
Article in English | MEDLINE | ID: mdl-23722706

ABSTRACT

Compelling clinical reports reveal that behavioral stress alone is sufficient to cause reversible myocardial dysfunction in selected individuals. We developed a rodent stress cardiomyopathy model by a combination of prenatal and postnatal behavioral stresses (Stress). We previously reported a decrease in percent fractional shortening by echo, both systolic and diastolic dysfunction by catheter-based hemodynamics, as well as attenuated hemodynamic and inotropic responses to the ß-adrenergic agonist, isoproterenol (ISO) in Stress rats compared with matched controls (Kan H, Birkle D, Jain AC, Failinger C, Xie S, Finkel MS. J Appl Physiol 98: 77-82, 2005). We now report enhanced catecholamine responses to behavioral stress, as evidenced by increased circulating plasma levels of norepinephrine (P < 0.01) and epinephrine (P < 0.01) in Stress rats vs. controls. Cardiac myocytes isolated from Stress rats also reveal evidence of oxidative stress, as indicated by decreased ATP, increased GSSG, and decreased GSH-to-GSSG ratio in the presence of increased GSH peroxidase and catalase activities (P < 0.01, for each). We also report blunted inotropic and intracellular Ca(2+) concentration responses to extracellular Ca(2+) (P < 0.05), as well as altered inotropic responses to the intracellular calcium regulator, caffeine (20 mM; P < 0.01). Treatment of cardiac myocytes with N-acetylcysteine (NAC) (10(-3) M) normalized calcium handling in response to ISO and extracellular Ca(2+) concentration and inotropic response to caffeine (P < 0.01, for each). NAC also attenuated the blunted inotropic response to ISO and Ca(2+) (P < 0.01, for each). Surprisingly, NAC did not reverse the changes in GSH, GSSG, or GSH-to-GSSG ratio. These data support a GSH-independent salutary effect of NAC on intracellular calcium signaling in this rodent model of stress-induced cardiomyopathy.


Subject(s)
Acetylcysteine/pharmacology , Cardiomyopathies/drug therapy , Myocytes, Cardiac/drug effects , Oxidative Stress/drug effects , Adenosine Triphosphate/metabolism , Animals , Calcium/metabolism , Calcium Signaling/drug effects , Cardiomyopathies/blood , Cardiomyopathies/metabolism , Catalase/metabolism , Disease Models, Animal , Epinephrine/blood , Female , Glutathione/metabolism , Glutathione Disulfide/metabolism , Glutathione Peroxidase/metabolism , Isoproterenol/pharmacology , Male , Myocardium/metabolism , Myocytes, Cardiac/metabolism , Norepinephrine/blood , Rats , Rats, Sprague-Dawley
18.
Am J Physiol Regul Integr Comp Physiol ; 304(7): R553-65, 2013 Apr 01.
Article in English | MEDLINE | ID: mdl-23408027

ABSTRACT

Mitochondrial dysfunction is a contributor to diabetic cardiomyopathy. Previously, we observed proteomic decrements within the inner mitochondrial membrane (IMM) and matrix of diabetic cardiac interfibrillar mitochondria (IFM) correlating with dysfunctional mitochondrial protein import. The goal of this study was to determine whether overexpression of mitochondria phospholipid hydroperoxide glutathione peroxidase 4 (mPHGPx), an antioxidant enzyme capable of scavenging membrane-associated lipid peroxides in the IMM, could reverse proteomic alterations, dysfunctional protein import, and ultimately, mitochondrial dysfunction associated with the diabetic heart. MPHGPx transgenic mice and controls were made diabetic by multiple low-dose streptozotocin injections and examined after 5 wk of hyperglycemia. Five weeks after hyperglycemia onset, in vivo analysis of cardiac contractile function revealed decreased ejection fraction and fractional shortening in diabetic hearts that was reversed with mPHGPx overexpression. MPHGPx overexpression increased electron transport chain function while attenuating hydrogen peroxide production and lipid peroxidation in diabetic mPHGPx IFM. MPHGPx overexpression lessened proteomic loss observed in diabetic IFM. Posttranslational modifications, including oxidations and deamidations, were attenuated in diabetic IFM with mPHGPx overexpression. Mitochondrial protein import dysfunction in diabetic IFM was reversed with mPHGPx overexpression correlating with protein import constituent preservation. Ingenuity Pathway Analyses indicated that oxidative phosphorylation, tricarboxylic acid cycle, and fatty acid oxidation processes most influenced in diabetic IFM were preserved by mPHGPx overexpression. Specific mitochondrial networks preserved included complex I and II, mitochondrial ultrastructure, and mitochondrial protein import. These results indicate that mPHGPx overexpression can preserve the mitochondrial proteome and provide cardioprotective benefits to the diabetic heart.


Subject(s)
Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Type 1/metabolism , Diabetic Cardiomyopathies/metabolism , Gene Expression Regulation, Enzymologic/physiology , Glutathione Peroxidase/metabolism , Mitochondria, Heart/metabolism , Animals , Biological Transport , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Type 1/complications , Diabetic Cardiomyopathies/complications , Female , Glutathione Peroxidase/genetics , Lipid Peroxidation , Male , Mass Spectrometry , Mice , Mice, Transgenic , Mitochondria, Heart/enzymology , Mitochondria, Heart/genetics , Oxidative Stress , Phospholipid Hydroperoxide Glutathione Peroxidase , Proteomics , Reactive Oxygen Species/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...