Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Nat Commun ; 11(1): 698, 2020 02 04.
Article in English | MEDLINE | ID: mdl-32019928

ABSTRACT

Profound metabolic changes are characteristic of macrophages during classical activation and have been implicated in this phenotype. Here we demonstrate that nitric oxide (NO) produced by murine macrophages is responsible for TCA cycle alterations and citrate accumulation associated with polarization. 13C tracing and mitochondrial respiration experiments map NO-mediated suppression of metabolism to mitochondrial aconitase (ACO2). Moreover, we find that inflammatory macrophages reroute pyruvate away from pyruvate dehydrogenase (PDH) in an NO-dependent and hypoxia-inducible factor 1α (Hif1α)-independent manner, thereby promoting glutamine-based anaplerosis. Ultimately, NO accumulation leads to suppression and loss of mitochondrial electron transport chain (ETC) complexes. Our data reveal that macrophages metabolic rewiring, in vitro and in vivo, is dependent on NO targeting specific pathways, resulting in reduced production of inflammatory mediators. Our findings require modification to current models of macrophage biology and demonstrate that reprogramming of metabolism should be considered a result rather than a mediator of inflammatory polarization.


Subject(s)
Aconitate Hydratase/metabolism , Macrophages/enzymology , Nitric Oxide/metabolism , Pyruvate Dehydrogenase Acetyl-Transferring Kinase/metabolism , Aconitate Hydratase/genetics , Animals , Citric Acid/metabolism , Citric Acid Cycle , Electron Transport Chain Complex Proteins/genetics , Electron Transport Chain Complex Proteins/metabolism , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Inflammation/genetics , Inflammation/metabolism , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/enzymology , Mitochondria/metabolism , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/metabolism , Pyruvate Dehydrogenase Acetyl-Transferring Kinase/genetics , Pyruvic Acid/metabolism
2.
Inflamm Bowel Dis ; 23(11): 1983-1995, 2017 11.
Article in English | MEDLINE | ID: mdl-29019857

ABSTRACT

BACKGROUND: If treatment with intravenous steroids fail, inflammatory bowel disease patients with acute severe colitis face systemic anti-tumor necrosis factor biologic rescue therapy or colectomy. Interleukin (IL)-27 is a cytokine with an immunosuppressive role in adaptive immune responses. However, the IL-27 receptor complex is also expressed on innate immune cells, and there is evidence that IL-27 can impact the function of innate cell subsets, although this particular functionality in vivo is not understood. Our aim was to define the efficacy of IL-27 in acute severe colitis and characterize novel IL-27-driven mechanisms of immunosuppression in the colonic mucosa. METHODS: We assessed oral delivery of Lactococcus lactis expressing an IL-27 hyperkine on the innate immune response in vivo in a genetically intact, noninfective, acute murine colitis model induced by intrarectal instillation of 2,4,6-trinitrobenzenesulfonic acid in SJL/J mice. RESULTS: IL-27 attenuates acute severe colitis through the reduction of colonic mucosal neutrophil infiltrate associated with a decreased CXC chemokine gradient. This suppression was T cell independent and IL-10 dependent, initially featuring enhanced mucosal IL-10. IL-27 was associated with a reduction in colonic proinflammatory cytokines and induced a multifocal, strong, positive nuclear expression of phosphorylated STAT-1 in mucosal epithelial cells. CONCLUSION: We have defined novel mechanisms of IL-27 immunosuppression toward colonic innate immune responses in vivo. Mucosal delivery of IL-27 has translational potential as a novel therapeutic for inflammatory bowel disease, and it is a future mucosal directed rescue therapy in acute severe inflammatory bowel disease.


Subject(s)
Colitis/drug therapy , Colon/immunology , Immunity, Innate , Interleukin-10/metabolism , Interleukin-27/pharmacology , Intestinal Mucosa/metabolism , Acute Disease , Animals , Colitis/chemically induced , Colon/physiopathology , Disease Models, Animal , Inflammation/pathology , Inflammatory Bowel Diseases/drug therapy , Inflammatory Bowel Diseases/pathology , Interleukin-27/immunology , Intestinal Mucosa/drug effects , Male , Mice , Mice, Knockout , T-Lymphocytes/metabolism , Trinitrobenzenesulfonic Acid/administration & dosage
3.
Redox Biol ; 10: 12-23, 2016 12.
Article in English | MEDLINE | ID: mdl-27676159

ABSTRACT

Inflammatory maturation of M1 macrophages by proinflammatory stimuli such as toll like receptor ligands results in profound metabolic reprogramming resulting in commitment to aerobic glycolysis as evidenced by repression of mitochondrial oxidative phosphorylation (OXPHOS) and enhanced glucose utilization. In contrast, "alternatively activated" macrophages adopt a metabolic program dominated by fatty acid-fueled OXPHOS. Despite the known importance of these developmental stages on the qualitative aspects of an inflammatory response, relatively little is know regarding the regulation of these metabolic adjustments. Here we provide evidence that the immunosuppressive cytokine IL-10 defines a metabolic regulatory loop. Our data show for the first time that lipopolysaccharide (LPS)-induced glycolytic flux controls IL-10-production via regulation of mammalian target of rapamycin (mTOR) and that autocrine IL-10 in turn regulates macrophage nitric oxide (NO) production. Genetic and pharmacological manipulation of IL-10 and nitric oxide (NO) establish that metabolically regulated autocrine IL-10 controls glycolytic commitment by limiting NO-mediated suppression of OXPHOS. Together these data support a model where autocine IL-10 production is controlled by glycolytic flux in turn regulating glycolytic commitment by preserving OXPHOS via suppression of NO. We propose that this IL-10-driven metabolic rheostat maintains metabolic equilibrium during M1 macrophage differentiation and that perturbation of this regulatory loop, either directly by exogenous cellular sources of IL-10 or indirectly via limitations in glucose availability, skews the cellular metabolic program altering the balance between inflammatory and immunosuppressive phenotypes.


Subject(s)
Interleukin-10/metabolism , Lipopolysaccharides/pharmacology , Macrophages/drug effects , Nitric Oxide/metabolism , TOR Serine-Threonine Kinases/metabolism , Animals , Autocrine Communication , Cell Differentiation , Cells, Cultured , Glucose/metabolism , Glycolysis , Humans , Macrophage Activation/drug effects , Macrophages/cytology , Macrophages/metabolism , Mice , Mitochondria/drug effects , Mitochondria/metabolism , Oxidative Phosphorylation/drug effects
4.
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
5.
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
6.
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
7.
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
8.
Am J Physiol Cell Physiol ; 303(12): C1244-51, 2012 Dec 15.
Article in English | MEDLINE | ID: mdl-23034391

ABSTRACT

Dysfunctional mitochondria are central in the pathogenesis of diabetic cardiomyopathy. Mitochondrial proteomic alterations resulting from diabetes mellitus have been reported although the mechanisms driving changes in proteomic signatures are unknown. microRNAs (miRNAs) have been considered as potential regulators of proteins. The goal of this study was to determine whether miRNAs play a role in diabetes-induced mitochondrial proteomic alterations. Quanitative RT-PCR miRNA screening in diabetic mice, 5 wk following multiple low-dose streptozotocin treatment was associated with alteration in the expression of 29 miRNAs in the diabetic heart compared with control. Among those miRNAs upregulated in the diabetic heart was miR-141 (P < 0.002). miRNA target prediction analyses identified miR-141 as a potential regulator of the inner mitochondrial membrane phosphate transporter, solute carrier family 25 member 3 (Slc25a3), which provides inorganic phosphate to the mitochondrial matrix and is essential for ATP production. With the use of a luciferase reporter construct with a Slc25a3 3'-untranslated region (UTR) target sequence, overexpression of miR-141 downregulated luciferase activity levels confirming miR-141/Slc25a3 3'-UTR binding. miR-141 overexpression in HL-1 cells elicited a decrease in Slc25a3 protein content, ATP production and a decrease in ATP synthase activity, similar to the diabetic phenotype (P < 0.05, for both). Diabetic interfibrillar mitochondria (IFM) displayed decreased Slc25a3 protein content, which was inversely correlated with increased miR-141 expression. Further, diabetic IFM ATP synthase activity was also decreased (P < 0.05). Together these results indicate that miR-141 can regulate Slc25a3 protein expression in the diabetic heart. Further, diabetes-induced miRNA changes may influence mitochondrial proteomes and functional processes such as mitochondrial ATP production.


Subject(s)
Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Type 1/metabolism , Diabetic Cardiomyopathies/metabolism , MicroRNAs/metabolism , Mitochondrial Proteins/biosynthesis , Phosphate Transport Proteins/biosynthesis , Animals , Male , Mice , Mitochondria, Heart/metabolism , Mitochondrial Proton-Translocating ATPases/metabolism
9.
Congest Heart Fail ; 17(6): 262-8, 2011.
Article in English | MEDLINE | ID: mdl-22103917

ABSTRACT

Heart failure (HF) is a common disease that has been attributed, in part, to deprivation of cardiac energy. As a result, the interplay between metabolism and adenosine triphosphate production is fundamental in determining the mechanisms driving the disease progression. Due to its central role in energy production, metabolism, calcium homeostasis, and oxidative stress, the mitochondrion has been suggested to play a pivotal role in the progression of the heart to failure. Nevertheless, the mitochondrion's specific role(s) and the proteins contributing to the development and progression of HF are not entirely clear. Thus, changes in mitochondrial proteomic make-up during HF have garnered great interest. With the continued development of advanced tools for assessing proteomic make-up, characterization of mitochondrial proteomic changes during disease states such as HF are being realized. These studies have begun to identify potential biomarkers of disease progression as well as protein targets that may provide an avenue for therapeutic intervention. The goal of this review is to highlight some of the changes in mitochondrial proteomic make-up that are associated with the development of HF in an effort to identify target axes and candidate proteins contributing to disease development. Results from a number of different HF models will be evaluated to gain insight into some of the similarities and differences in mitochondrial proteomic alterations associated with morphological and functional changes that result from the disease. Congest Heart Fail.


Subject(s)
Adenosine Triphosphate/biosynthesis , Energy Metabolism , Heart Failure/metabolism , Mitochondria, Heart/metabolism , Oxidative Stress/physiology , Proteome/physiology , Animals , Apoptosis , Heart Failure/pathology , Humans , Reactive Oxygen Species/metabolism
10.
Am J Physiol Regul Integr Comp Physiol ; 300(2): R186-200, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21048079

ABSTRACT

Diabetic cardiomyopathy is associated with increased risk of heart failure in type 1 diabetic patients. Mitochondrial dysfunction is suggested as an underlying contributor to diabetic cardiomyopathy. Cardiac mitochondria are characterized by subcellular spatial locale, including mitochondria located beneath the sarcolemma, subsarcolemmal mitochondria (SSM), and mitochondria situated between the myofibrils, interfibrillar mitochondria (IFM). The goal of this study was to determine whether type 1 diabetic insult in the heart influences proteomic make-up of spatially distinct mitochondrial subpopulations and to evaluate the role of nuclear encoded mitochondrial protein import. Utilizing multiple proteomic approaches (iTRAQ and two-dimensional-differential in-gel electrophoresis), IFM proteomic make-up was impacted by type 1 diabetes mellitus to a greater extent than SSM, as evidenced by decreased abundance of fatty acid oxidation and electron transport chain proteins. Mitochondrial phosphate carrier and adenine nucleotide translocator, as well as inner membrane translocases, were decreased in the diabetic IFM (P < 0.05 for both). Mitofilin, a protein involved in cristae morphology, was diminished in the diabetic IFM (P < 0.05). Posttranslational modifications, including oxidations and deamidations, were most prevalent in the diabetic IFM. Mitochondrial heat shock protein 70 (mtHsp70) was significantly decreased in diabetic IFM (P < 0.05). Mitochondrial protein import was decreased in the diabetic IFM with no change in the diabetic SSM (P < 0.05). Taken together, these results indicate that mitochondrial proteomic alterations in the type 1 diabetic heart are more pronounced in the IFM. Further, proteomic alterations are associated with nuclear encoded mitochondrial protein import dysfunction and loss of an essential mitochondrial protein import constituent, mtHsp70, implicating this process in the pathogenesis of the diabetic heart.


Subject(s)
Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Type 1/metabolism , Diabetic Cardiomyopathies/metabolism , Mitochondria, Heart/metabolism , Myocardium/metabolism , Protein Transport/physiology , Proteome/metabolism , 3-Hydroxyacyl CoA Dehydrogenases/metabolism , Acetyl-CoA C-Acyltransferase/metabolism , Animals , Blood Glucose/metabolism , Carbon-Carbon Double Bond Isomerases/metabolism , Carrier Proteins/genetics , Carrier Proteins/metabolism , Citric Acid Cycle/physiology , Diabetes Mellitus, Experimental/blood , Diabetes Mellitus, Experimental/physiopathology , Diabetes Mellitus, Type 1/blood , Diabetes Mellitus, Type 1/physiopathology , Diabetic Cardiomyopathies/physiopathology , Down-Regulation/physiology , Electron Transport Chain Complex Proteins/metabolism , Enoyl-CoA Hydratase/metabolism , Gene Expression/physiology , HSP70 Heat-Shock Proteins/metabolism , Heart/physiopathology , Insulin/blood , Male , Membrane Potential, Mitochondrial/physiology , Mice , Mice, Inbred Strains , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Muscle Proteins/metabolism , Protein Processing, Post-Translational/physiology , Proteome/genetics , Proteomics , Racemases and Epimerases/metabolism , Recombinant Fusion Proteins/metabolism , Up-Regulation/physiology
11.
Am J Physiol Heart Circ Physiol ; 299(2): H529-40, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20543078

ABSTRACT

Cardiac complications and heart failure are the leading cause of death in type 2 diabetic patients. Mitochondrial dysfunction is central in the pathogenesis of the type 2 diabetic heart. However, it is unclear whether this dysfunction is specific for a particular subcellular region. The purpose of this study was to determine whether mitochondrial dysfunction in the type 2 diabetic heart is specific to a spatially distinct subset of mitochondria. We investigated mitochondrial morphology, function, and proteomic composition of subsarcolemmal mitochondria (SSM) and interfibrillar mitochondria (IFM) in 18-wk-old db/db mice. Oxidative damage was assessed in subpopulations through the measurement of lipid peroxidation byproducts and nitrotyrosine residues. Proteomic profiles and posttranslational modifications were assessed in mitochondrial subpopulations using iTRAQ and multi-dimensional protein identification technologies, respectively. SSM from db/db hearts had altered morphology, including a decrease in size and internal complexity, whereas db/db IFM were increased in internal complexity. Db/db SSM displayed decreased state 3 respiration rates, electron transport chain activities, ATP synthase activities, and mitochondrial membrane potential and increased oxidative damage, with no change in IFM. Proteomic assessment revealed a greater impact on db/db SSM compared with db/db IFM. Inner mitochondrial membrane proteins, including electron transport chain, ATP synthesis, and mitochondrial protein import machinery, were predominantly decreased. We provide evidence that mitochondrial dysfunction in the type 2 diabetic heart is associated with a specific subcellular locale. Furthermore, mitochondrial morphological and functional indexes are impacted differently during type 2 diabetic insult and may result from the modulation of spatially distinct mitochondrial proteomes.


Subject(s)
Diabetes Mellitus, Type 2/metabolism , Mitochondria, Heart/metabolism , Mitochondrial Proteins/metabolism , Proteome , Animals , Antioxidants/metabolism , Cell Respiration , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/pathology , Disease Models, Animal , Electron Transport , Electron Transport Chain Complex Proteins/metabolism , Ion Channels/metabolism , Lipid Peroxidation , Male , Membrane Potential, Mitochondrial , Mice , Mitochondria, Heart/pathology , Mitochondrial ADP, ATP Translocases/metabolism , Mitochondrial Proton-Translocating ATPases/metabolism , Mitochondrial Size , Oxidative Stress , Protein Processing, Post-Translational , Proteomics/methods , Tyrosine/analogs & derivatives , Tyrosine/metabolism , Uncoupling Protein 3
12.
Am J Physiol Heart Circ Physiol ; 298(2): H633-42, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19966057

ABSTRACT

Cardiovascular complications, such as diabetic cardiomyopathy, account for the majority of deaths associated with diabetes mellitus. Mitochondria are particularly susceptible to the damaging effects of diabetes mellitus and have been implicated in the pathogenesis of diabetic cardiomyopathy. Cardiac mitochondria consist of two spatially distinct subpopulations, termed subsarcolemmal mitochondria (SSM) and interfibrillar mitochondria (IFM). The goal of this study was to determine whether subcellular spatial location is associated with apoptotic propensity of cardiac mitochondrial subpopulations during diabetic insult. Swiss Webster mice were subjected to intraperitoneal injection of streptozotocin or citrate saline vehicle. Ten weeks following injection, diabetic hearts displayed increased caspase-3 and caspase-9 activities, indicating enhanced apoptotic signaling (P < 0.05, for both). Mitochondrial size (forward scatter) and internal complexity (side scatter) were decreased in diabetic IFM (P < 0.05, for both) but not in diabetic SSM. Mitochondrial membrane potential (Delta(Psim)) was lower in diabetic IFM (P < 0.01) but not in diabetic SSM. Mitochondrial permeability transition pore (mPTP) opening was increased in diabetic compared with control IFM (P < 0.05), whereas no differences were observed in diabetic compared with control SSM. Examination of mPTP constituents revealed increases in cyclophilin D in diabetic IFM. Furthermore, diabetic IFM possessed lower cytochrome c and BcL-2 levels and increased Bax levels (P < 0.05, for all 3). No significant changes in these proteins were observed in diabetic SSM compared with control. These results indicate that diabetes mellitus is associated with an enhanced apoptotic propensity in IFM, suggesting a differential apoptotic susceptibility of distinct mitochondrial subpopulations based upon subcellular location.


Subject(s)
Apoptosis/physiology , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Experimental/physiopathology , Mitochondria, Heart/pathology , Mitochondria, Heart/physiology , Myocardium/pathology , Sarcolemma/pathology , Animals , Caspase 3/metabolism , Caspase 9/metabolism , Peptidyl-Prolyl Isomerase F , Cyclophilins/metabolism , Cytochromes c/metabolism , Diabetes Mellitus, Experimental/metabolism , Disease Models, Animal , Female , Membrane Potential, Mitochondrial/physiology , Mice , Mitochondria, Heart/ultrastructure , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Permeability Transition Pore , Myocardium/metabolism , Myocardium/ultrastructure , Sarcolemma/metabolism , Sarcolemma/ultrastructure , Streptozocin , bcl-2-Associated X Protein/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...