Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
1.
Nat Cardiovasc Res ; 3(7): 869-882, 2024 Jul.
Article in English | MEDLINE | ID: mdl-39196175

ABSTRACT

Differentiation of cardiac fibroblasts to myofibroblasts is necessary for matrix remodeling and fibrosis in heart failure. We previously reported that mitochondrial calcium signaling drives α-ketoglutarate-dependent histone demethylation, promoting myofibroblast formation. Here we investigate the role of ATP-citrate lyase (ACLY), a key enzyme for acetyl-CoA biosynthesis, in histone acetylation regulating myofibroblast fate and persistence in cardiac fibrosis. We show that inactivation of ACLY prevents myofibroblast differentiation and reverses myofibroblasts towards quiescence. Genetic deletion of Acly in post-activated myofibroblasts prevents fibrosis and preserves cardiac function in pressure-overload heart failure. TGFß stimulation enhances ACLY nuclear localization and ACLY-SMAD2/3 interaction, and increases H3K27ac at fibrotic gene loci. Pharmacological inhibition of ACLY or forced nuclear expression of a dominant-negative ACLY mutant prevents myofibroblast formation and H3K27ac. Our data indicate that nuclear ACLY activity is necessary for myofibroblast differentiation and persistence by maintaining histone acetylation at TGFß-induced myofibroblast genes. These findings provide targets to prevent and reverse pathological fibrosis.


Subject(s)
ATP Citrate (pro-S)-Lyase , Cell Differentiation , Fibrosis , Histones , Myofibroblasts , Smad2 Protein , Myofibroblasts/metabolism , Myofibroblasts/drug effects , ATP Citrate (pro-S)-Lyase/metabolism , ATP Citrate (pro-S)-Lyase/genetics , Animals , Fibrosis/metabolism , Cell Differentiation/drug effects , Histones/metabolism , Smad2 Protein/metabolism , Smad2 Protein/genetics , Acetylation/drug effects , Cell Nucleus/metabolism , Cell Nucleus/drug effects , Smad3 Protein/metabolism , Smad3 Protein/genetics , Cells, Cultured , Chromatin/metabolism , Mice, Knockout , Transforming Growth Factor beta/metabolism , Disease Models, Animal , Signal Transduction , Mice, Inbred C57BL , Male , Mice , Gene Expression Regulation/drug effects
2.
Microbiol Spectr ; 11(6): e0215723, 2023 Dec 12.
Article in English | MEDLINE | ID: mdl-37929974

ABSTRACT

IMPORTANCE: The yeast C. albicans exhibits metabolic flexibility for adaptability to host niches with varying availability of nutrients including essential metals like iron. For example, blood is iron deplete, while the oral cavity and the intestinal lumen are considered iron replete. We show here that C. albicans can tolerate very high levels of environmental iron, despite an increase in high iron-induced reactive oxygen species (ROS) that it mitigates with the help of a unique oxidase, known as alternative oxidase (AOX). High iron induces AOX1/2 that limits mitochondrial accumulation of ROS. Genetic elimination of AOX1/2 resulted in diminished virulence during oropharyngeal candidiasis in high iron mice. Since human mitochondria lack AOX protein, it represents a unique target for treatment of fungal infections.


Subject(s)
Candida albicans , Oxidoreductases , Humans , Animals , Mice , Candida albicans/genetics , Candida albicans/metabolism , Reactive Oxygen Species/metabolism , Oxidoreductases/genetics , Oxidoreductases/metabolism , Iron/metabolism , Fungal Proteins/genetics , Fungal Proteins/metabolism
3.
Redox Biol ; 63: 102740, 2023 07.
Article in English | MEDLINE | ID: mdl-37210780

ABSTRACT

Mitochondrial supercomplexes are observed in mammalian tissues with high energy demand and may influence metabolism and redox signaling. Nevertheless, the mechanisms that regulate supercomplex abundance remain unclear. In this study, we examined the composition of supercomplexes derived from murine cardiac mitochondria and determined how their abundance changes with substrate provision or by genetically induced changes to the cardiac glucose-fatty acid cycle. Protein complexes from digitonin-solubilized cardiac mitochondria were resolved by blue-native polyacrylamide gel electrophoresis and were identified by mass spectrometry and immunoblotting to contain constituents of Complexes I, III, IV, and V as well as accessory proteins involved in supercomplex assembly and stability, cristae architecture, carbohydrate and fat oxidation, and oxidant detoxification. Respiratory analysis of high molecular mass supercomplexes confirmed the presence of intact respirasomes, capable of transferring electrons from NADH to O2. Provision of respiratory substrates to isolated mitochondria augmented supercomplex abundance, with fatty acyl substrate (octanoylcarnitine) promoting higher supercomplex abundance than carbohydrate-derived substrate (pyruvate). Mitochondria isolated from transgenic hearts that express kinase-deficient 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase (GlycoLo), which decreases glucose utilization and increases reliance on fatty acid oxidation for energy, had higher mitochondrial supercomplex abundance and activity compared with mitochondria from wild-type or phosphatase-deficient 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase-expressing hearts (GlycoHi), the latter of which encourages reliance on glucose catabolism for energy. These findings indicate that high energetic reliance on fatty acid catabolism bolsters levels of mitochondrial supercomplexes, supporting the idea that the energetic state of the heart is regulatory factor in supercomplex assembly or stability.


Subject(s)
Heart , Phosphofructokinase-2 , Mice , Animals , Phosphofructokinase-2/metabolism , Mitochondria, Heart/metabolism , Glucose/metabolism , Fatty Acids/metabolism , Mammals/metabolism
4.
Nat Cardiovasc Res ; 2(12): 1109-1111, 2023 Dec.
Article in English | MEDLINE | ID: mdl-39188987

ABSTRACT

The molecular mechanisms that link propionyl-CoA metabolism and epigenetic regulation of gene expression are unclear, as are the implications for heart function. Now, new insights into the modulation of chromatin acylation and transcription by aberrant oxidation of propionyl-CoA are revealed in the dysfunctional hearts of mice with propionic acidemia.


Subject(s)
Acyl Coenzyme A , Chromatin , Transcription, Genetic , Animals , Mice , Acyl Coenzyme A/metabolism , Acylation , Chromatin/metabolism , Chromatin/genetics , Epigenesis, Genetic , Myocardium/metabolism , Oxidation-Reduction , Propionic Acidemia/metabolism , Propionic Acidemia/genetics
5.
J Mol Cell Cardiol ; 172: 78-89, 2022 11.
Article in English | MEDLINE | ID: mdl-35988357

ABSTRACT

BACKGROUND: Fibrosis and extracellular matrix remodeling are mediated by resident cardiac fibroblasts (CFs). In response to injury, fibroblasts activate, differentiating into specialized synthetic and contractile myofibroblasts producing copious extracellular matrix proteins (e.g., collagens). Myofibroblast persistence in chronic diseases, such as HF, leads to progressive cardiac dysfunction and maladaptive remodeling. We recently reported that an increase in αKG (alpha-ketoglutarate) bioavailability, which contributes to enhanced αKG-dependent lysine demethylase activity and chromatin remodeling, is required for myofibroblast formation. Therefore, we aimed to determine the substrates and metabolic pathways contributing to αKG biosynthesis and their requirement for myofibroblast formation. METHODS: Stable isotope metabolomics identified glutaminolysis as a key metabolic pathway required for αKG biosynthesis and myofibroblast formation, therefore we tested the effects of pharmacologic inhibition (CB-839) or genetic deletion of glutaminase (Gls1-/-) on myofibroblast formation in both murine and human cardiac fibroblasts. We employed immunofluorescence staining, functional gel contraction, western blotting, and bioenergetic assays to determine the myofibroblast phenotype. RESULTS: Carbon tracing indicated enhanced glutaminolysis mediating increased αKG abundance. Pharmacological and genetic inhibition of glutaminolysis prevented myofibroblast formation indicated by a reduction in αSMA+ cells, collagen gel contraction, collagen abundance, and the bioenergetic response. Inhibition of glutaminolysis also prevented TGFß-mediated histone demethylation and supplementation with cell-permeable αKG rescued the myofibroblast phenotype. Importantly, inhibition of glutaminolysis was sufficient to prevent myofibroblast formation in CFs isolated from the human failing heart. CONCLUSIONS: These results define glutaminolysis as necessary for myofibroblast formation and persistence, providing substantial rationale to evaluate several new therapeutic targets to treat cardiac fibrosis.


Subject(s)
Myofibroblasts , Humans , Mice , Animals , Myofibroblasts/metabolism , Glutamine/metabolism , Fibroblasts/metabolism , Collagen/metabolism , Cells, Cultured
7.
Ann Am Thorac Soc ; 19(7): 1102-1111, 2022 07.
Article in English | MEDLINE | ID: mdl-35007497

ABSTRACT

Rationale: Cardiovascular disease accounts for one-third of deaths in patients with chronic obstructive pulmonary disease (COPD). Better control of cardiovascular risk factors in primary care could improve outcomes. Objectives: To define the prevalence, monitoring, treatment, and control of risk factors in patients with COPD. Methods: Repeated cross-sectional analysis of primary care electronic medical records for all patients with COPD in the Canadian Primary Care Sentinel Surveillance Network from 2013 to 2018 (n = 32,695 in 2018). A control group was matched 1:1 for age, sex, and rural residence (n = 32,638 in 2018). Five risk factors were defined using validated definitions including laboratory results: hypertension, dyslipidemia, diabetes, obesity, and smoking. Results: All risk factors were more common in patients with COPD compared with matched control subjects, including hypertension (52.3% vs. 44.9%), dyslipidemia (62.0% vs. 57.8%), diabetes (25.0% vs. 20.2%), obesity (40.8% vs. 36.8%), and smoking (40.9% vs. 11.4%), respectively. The mean Framingham risk score was 20.6% versus 18.6%, with 53.8% of patients with COPD being high risk (⩾20%). Monitoring of risk factors within the last year in patients with COPD in 2018 was suboptimal: 71.8% hypertension, 39.4% dyslipidemia, 74.5% diabetes, 52.3% obesity. Smoking status was infrequently recorded in the electronic record. In those monitored, guideline recommended targets were achieved in 60.8%, 46.6%, 57.4%, 10.6% and 12.0% for each risk factor. Cardiovascular therapies including angiotensin-converting enzyme inhibitors (69%), statins (69%), and smoking cessation therapies (27%) were underused. Conclusions: In patients with COPD, major cardiovascular risk factors are common, yet inadequately monitored, undertreated, and poorly controlled. Strategies are needed to improve comprehensive risk factor management proven to reduce cardiovascular morbidity and mortality.


Subject(s)
Cardiovascular Diseases , Dyslipidemias , Hypertension , Pulmonary Disease, Chronic Obstructive , Canada/epidemiology , Cardiovascular Diseases/epidemiology , Cardiovascular Diseases/prevention & control , Cross-Sectional Studies , Heart Disease Risk Factors , Humans , Hypertension/epidemiology , Obesity/complications , Obesity/epidemiology , Pulmonary Disease, Chronic Obstructive/epidemiology , Risk Factors
8.
JACC Basic Transl Sci ; 6(8): 650-672, 2021 Aug.
Article in English | MEDLINE | ID: mdl-34466752

ABSTRACT

In this study the authors used systems biology to define progressive changes in metabolism and transcription in a large animal model of heart failure with preserved ejection fraction (HFpEF). Transcriptomic analysis of cardiac tissue, 1-month post-banding, revealed loss of electron transport chain components, and this was supported by changes in metabolism and mitochondrial function, altogether signifying alterations in oxidative metabolism. Established HFpEF, 4 months post-banding, resulted in changes in intermediary metabolism with normalized mitochondrial function. Mitochondrial dysfunction and energetic deficiencies were noted in skeletal muscle at early and late phases of disease, suggesting cardiac-derived signaling contributes to peripheral tissue maladaptation in HFpEF. Collectively, these results provide insights into the cellular biology underlying HFpEF progression.

9.
CMAJ Open ; 9(2): E376-E383, 2021.
Article in English | MEDLINE | ID: mdl-33863795

ABSTRACT

BACKGROUND: Heart failure (HF) poses a substantial global health burden, particularly in patients with chronic obstructive pulmonary disease (COPD). The objective of this study was to validate an electronic medical record-based definition of HF in patients with COPD in primary care practices in the province of British Columbia, Canada. METHODS: We conducted a cross-sectional retrospective chart review from Sept. 1, 2018, to Dec. 31, 2018, for a cohort of patients from primary care practices in BC whose physicians were recruited through the BC node of the Canadian Primary Care Sentinel Surveillance Network. Heart failure case definitions were developed by combining diagnostic codes, medication information and laboratory values available in primary care electronic medical records. These were compared with HF diagnoses identified through detailed chart review as the gold standard. Sensitivity, specificity, negative (NPV) and positive predictive values (PPV) were calculated for each definition. RESULTS: Charts of 311 patients with COPD were reviewed, of whom 72 (23.2%) had HF. Five categories of definitions were constructed, all of which had appropriate sensitivity, specificity and NPV. The optimal case definition consisted of 1 HF billing code or a specific combination of medications for HF. This definition had an excellent specificity (93.3%, 95% confidence interval [CI] 89.4%-96.1%), sensitivity (90.3%, 95% CI 81.0%-96.0%), PPV (80.2%, 95% CI 69.9%-88.3%) and NPV (97.0%, 95% CI 93.8%-98.8%). INTERPRETATION: This comprehensive case definition improves upon previous primary care HF definitions to include medication codes and laboratory data, along with previously used billing codes. A case definition for HF was derived and validated and can be used with data from electronic medical records to identify HF in patients with COPD in primary care accurately.


Subject(s)
Heart Failure , Primary Health Care , Pulmonary Disease, Chronic Obstructive , British Columbia/epidemiology , Clinical Laboratory Information Systems/statistics & numerical data , Cross-Sectional Studies , Databases, Pharmaceutical/statistics & numerical data , Electronic Health Records/statistics & numerical data , Female , Health Information Systems/organization & administration , Heart Failure/diagnosis , Heart Failure/epidemiology , Heart Failure/therapy , Humans , International Classification of Diseases , Male , Middle Aged , Predictive Value of Tests , Primary Health Care/methods , Primary Health Care/organization & administration , Primary Health Care/standards , Pulmonary Disease, Chronic Obstructive/diagnosis , Pulmonary Disease, Chronic Obstructive/epidemiology , Pulmonary Disease, Chronic Obstructive/therapy , Quality Improvement , Retrospective Studies , Sensitivity and Specificity , Sentinel Surveillance
10.
Circ Res ; 128(1): 92-114, 2021 01 08.
Article in English | MEDLINE | ID: mdl-33092464

ABSTRACT

RATIONALE: Ca2+-induced Ca2+ release (CICR) in normal hearts requires close approximation of L-type calcium channels (LTCCs) within the transverse tubules (T-tubules) and RyR (ryanodine receptors) within the junctional sarcoplasmic reticulum. CICR is disrupted in cardiac hypertrophy and heart failure, which is associated with loss of T-tubules and disruption of cardiac dyads. In these conditions, LTCCs are redistributed from the T-tubules to disrupt CICR. The molecular mechanism responsible for LTCCs recruitment to and from the T-tubules is not well known. JPH (junctophilin) 2 enables close association between T-tubules and the junctional sarcoplasmic reticulum to ensure efficient CICR. JPH2 has a so-called joining region that is located near domains that interact with T-tubular plasma membrane, where LTCCs are housed. The idea that this joining region directly interacts with LTCCs and contributes to LTCC recruitment to T-tubules is unknown. OBJECTIVE: To determine if the joining region in JPH2 recruits LTCCs to T-tubules through direct molecular interaction in cardiomyocytes to enable efficient CICR. METHODS AND RESULTS: Modified abundance of JPH2 and redistribution of LTCC were studied in left ventricular hypertrophy in vivo and in cultured adult feline and rat ventricular myocytes. Protein-protein interaction studies showed that the joining region in JPH2 interacts with LTCC-α1C subunit and causes LTCCs distribution to the dyads, where they colocalize with RyRs. A JPH2 with induced mutations in the joining region (mutPG1JPH2) caused T-tubule remodeling and dyad loss, showing that an interaction between LTCC and JPH2 is crucial for T-tubule stabilization. mutPG1JPH2 caused asynchronous Ca2+-release with impaired excitation-contraction coupling after ß-adrenergic stimulation. The disturbed Ca2+ regulation in mutPG1JPH2 overexpressing myocytes caused calcium/calmodulin-dependent kinase II activation and altered myocyte bioenergetics. CONCLUSIONS: The interaction between LTCC and the joining region in JPH2 facilitates dyad assembly and maintains normal CICR in cardiomyocytes.


Subject(s)
Calcium Channels, L-Type/metabolism , Calcium Signaling , Calcium/metabolism , Hypertrophy, Left Ventricular/metabolism , Membrane Proteins/metabolism , Muscle Proteins/metabolism , Myocytes, Cardiac/metabolism , Animals , Calcium Channels, L-Type/genetics , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Cats , Cells, Cultured , Disease Models, Animal , Excitation Contraction Coupling , Humans , Hypertrophy, Left Ventricular/pathology , Hypertrophy, Left Ventricular/physiopathology , Kinetics , Male , Membrane Proteins/genetics , Mitochondria, Heart/metabolism , Mitochondria, Heart/pathology , Muscle Proteins/genetics , Mutation , Myocytes, Cardiac/pathology , Organelle Biogenesis , Protein Binding , Protein Interaction Domains and Motifs , Rats, Sprague-Dawley , Ryanodine Receptor Calcium Release Channel
11.
Circ Res ; 127(3): 427-447, 2020 07 17.
Article in English | MEDLINE | ID: mdl-32673537

ABSTRACT

Cardiac fibrosis is mediated by the activation of resident cardiac fibroblasts, which differentiate into myofibroblasts in response to injury or stress. Although myofibroblast formation is a physiological response to acute injury, such as myocardial infarction, myofibroblast persistence, as occurs in heart failure, contributes to maladaptive remodeling and progressive functional decline. Although traditional pathways of activation, such as TGFß (transforming growth factor ß) and AngII (angiotensin II), have been well characterized, less understood are the alterations in mitochondrial function and cellular metabolism that are necessary to initiate and sustain myofibroblast formation and function. In this review, we highlight recent reports detailing the mitochondrial and metabolic mechanisms that contribute to myofibroblast differentiation, persistence, and function with the hope of identifying novel therapeutic targets to treat, and potentially reverse, tissue organ fibrosis.


Subject(s)
Cell Differentiation , Energy Metabolism , Heart Diseases/metabolism , Mitochondria, Heart/metabolism , Myofibroblasts/metabolism , Animals , Calcium Signaling , Fibrosis , Heart Diseases/pathology , Humans , Mitochondria, Heart/pathology , Myofibroblasts/pathology
12.
J Clin Invest ; 130(4): 2081-2096, 2020 04 01.
Article in English | MEDLINE | ID: mdl-31945018

ABSTRACT

Macrophages have been linked to tumor initiation, progression, metastasis, and treatment resistance. However, the transcriptional regulation of macrophages driving the protumor function remains elusive. Here, we demonstrate that the transcription factor c-Maf is a critical controller for immunosuppressive macrophage polarization and function in cancer. c-Maf controls many M2-related genes and has direct binding sites within a conserved noncoding sequence of the Csf-1r gene and promotes M2-like macrophage-mediated T cell suppression and tumor progression. c-Maf also serves as a metabolic checkpoint regulating the TCA cycle and UDP-GlcNAc biosynthesis, thus promoting M2-like macrophage polarization and activation. Additionally, c-Maf is highly expressed in tumor-associated macrophages (TAMs) and regulates TAM immunosuppressive function. Deletion of c-Maf specifically in myeloid cells results in reduced tumor burden with enhanced antitumor T cell immunity. Inhibition of c-Maf partly overcomes resistance to anti-PD-1 therapy in a subcutaneous LLC tumor model. Similarly, c-Maf is expressed in human M2 and tumor-infiltrating macrophages/monocytes as well as circulating monocytes of human non-small cell lung carcinoma (NSCLC) patients and critically regulates their immunosuppressive activity. The natural compound ß-glucan downregulates c-Maf expression on macrophages, leading to enhanced antitumor immunity in mice. These findings establish a paradigm for immunosuppressive macrophage polarization and transcriptional regulation by c-Maf and suggest that c-Maf is a potential target for effective tumor immunotherapy.


Subject(s)
Carcinoma, Non-Small-Cell Lung/immunology , Immunity, Cellular , Lung Neoplasms/immunology , Macrophage Activation , Macrophages/immunology , Neoplasms, Experimental/immunology , Proto-Oncogene Proteins c-maf/immunology , Animals , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Non-Small-Cell Lung/therapy , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic/immunology , Humans , Lung Neoplasms/pathology , Lung Neoplasms/therapy , Macrophages/pathology , Male , Mice , Mice, Knockout , Monocytes/immunology , Monocytes/pathology , Neoplasms, Experimental/genetics , Neoplasms, Experimental/pathology , Neoplasms, Experimental/therapy , Proto-Oncogene Proteins c-maf/genetics , T-Lymphocytes/immunology , T-Lymphocytes/pathology
13.
Nat Commun ; 10(1): 4509, 2019 10 04.
Article in English | MEDLINE | ID: mdl-31586055

ABSTRACT

Fibroblast to myofibroblast differentiation is crucial for the initial healing response but excessive myofibroblast activation leads to pathological fibrosis. Therefore, it is imperative to understand the mechanisms underlying myofibroblast formation. Here we report that mitochondrial calcium (mCa2+) signaling is a regulatory mechanism in myofibroblast differentiation and fibrosis. We demonstrate that fibrotic signaling alters gating of the mitochondrial calcium uniporter (mtCU) in a MICU1-dependent fashion to reduce mCa2+ uptake and induce coordinated changes in metabolism, i.e., increased glycolysis feeding anabolic pathways and glutaminolysis yielding increased α-ketoglutarate (αKG) bioavailability. mCa2+-dependent metabolic reprogramming leads to the activation of αKG-dependent histone demethylases, enhancing chromatin accessibility in loci specific to the myofibroblast gene program, resulting in differentiation. Our results uncover an important role for the mtCU beyond metabolic regulation and cell death and demonstrate that mCa2+ signaling regulates the epigenome to influence cellular differentiation.


Subject(s)
Calcium Signaling/physiology , Cell Differentiation/genetics , Epigenesis, Genetic/physiology , Myocardial Infarction/pathology , Myofibroblasts/physiology , Animals , Calcium Channels/genetics , Calcium Channels/metabolism , Calcium-Binding Proteins/metabolism , DNA Methylation/physiology , Disease Models, Animal , Embryo, Mammalian , Epigenome , Female , Fibrosis , Glycolysis/physiology , Humans , Ketoglutaric Acids/metabolism , Mice , Mice, Knockout , Mitochondria/metabolism , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Myocardial Infarction/diagnostic imaging , Myocardial Infarction/etiology , Myocardium/cytology , Myocardium/pathology , Primary Cell Culture
14.
Sci Rep ; 9(1): 13705, 2019 09 23.
Article in English | MEDLINE | ID: mdl-31548575

ABSTRACT

Stable isotope-resolved metabolomics (SIRM) provides information regarding the relative activity of numerous metabolic pathways and the contribution of nutrients to specific metabolite pools; however, SIRM experiments can be difficult to execute, and data interpretation is challenging. Furthermore, standardization of analytical procedures and workflows remain significant obstacles for widespread reproducibility. Here, we demonstrate the workflow of a typical SIRM experiment and suggest experimental controls and measures of cross-validation that improve data interpretation. Inhibitors of glycolysis and oxidative phosphorylation as well as mitochondrial uncouplers serve as pharmacological controls, which help define metabolic flux configurations that occur under well-controlled metabolic states. We demonstrate how such controls and time course labeling experiments improve confidence in metabolite assignments as well as delineate metabolic pathway relationships. Moreover, we demonstrate how radiolabeled tracers and extracellular flux analyses integrate with SIRM to improve data interpretation. Collectively, these results show how integration of flux methodologies and use of pharmacological controls increase confidence in SIRM data and provide new biological insights.


Subject(s)
Mass Spectrometry/methods , Metabolomics/methods , Workflow , Data Interpretation, Statistical , Isotope Labeling/methods , Mass Spectrometry/standards , Metabolic Networks and Pathways , Metabolomics/standards , Reproducibility of Results
15.
J Clin Invest ; 129(1): 63-65, 2019 01 02.
Article in English | MEDLINE | ID: mdl-30507608

ABSTRACT

Individuals with the rs671 SNP in the gene encoding aldehyde dehydrogenase 2 (ALDH2) are at increased risk of cardiovascular disease (CVD); however, it has been unclear if this mutation contributes to CVD development. In this issue of the JCI, Zhong et al. perform an elegant set of experiments that reveal a pathway wherein the ALDH2 rs671 mutant is phosphorylated by AMPK and translocates to the nucleus where it represses the transcription of a lysosomal H+ pump subunit that is critical for lipid degradation and foam cell formation, as occurs in atherosclerosis. The discovery of this pathway may explain how subjects harboring ALDH2 rs671 are at a greater risk for numerous other disease states and thereby provide new targets for therapeutic intervention.


Subject(s)
Atherosclerosis , Polymorphism, Single Nucleotide , AMP-Activated Protein Kinases , Aldehyde Dehydrogenase, Mitochondrial , Aldehyde Oxidoreductases , Foam Cells , Humans
16.
Redox Biol ; 17: 440-449, 2018 07.
Article in English | MEDLINE | ID: mdl-29885625

ABSTRACT

Pathological cardiac remodeling during heart failure is associated with higher levels of lipid peroxidation products and lower abundance of several aldehyde detoxification enzymes, including aldehyde dehydrogenase 2 (ALDH2). An emerging idea that could explain these findings concerns the role of electrophilic species in redox signaling, which may be important for adaptive responses to stress or injury. The purpose of this study was to determine whether genetically increasing ALDH2 activity affects pressure overload-induced cardiac dysfunction. Mice subjected to transverse aortic constriction (TAC) for 12 weeks developed myocardial hypertrophy and cardiac dysfunction, which were associated with diminished ALDH2 expression and activity. Cardiac-specific expression of the human ALDH2 gene in mice augmented myocardial ALDH2 activity but did not improve cardiac function in response to pressure overload. After 12 weeks of TAC, ALDH2 transgenic mice had larger hearts than their wild-type littermates and lower capillary density. These findings show that overexpression of ALDH2 augments the hypertrophic response to pressure overload and imply that downregulation of ALDH2 may be an adaptive response to certain forms of cardiac pathology.


Subject(s)
Aldehyde Dehydrogenase, Mitochondrial/genetics , Heart Failure/genetics , Oxidative Stress/genetics , Ventricular Remodeling/genetics , Animals , Aorta/metabolism , Gene Expression Regulation , Heart Failure/metabolism , Heart Failure/pathology , Humans , Mice , Mice, Transgenic , Mitochondria/metabolism , Myocardium/metabolism , Myocardium/pathology , Oxidation-Reduction , Pressure , Signal Transduction/genetics
17.
Circ Res ; 123(1): 107-128, 2018 06 22.
Article in English | MEDLINE | ID: mdl-29929976

ABSTRACT

Metabolic pathways integrate to support tissue homeostasis and to prompt changes in cell phenotype. In particular, the heart consumes relatively large amounts of substrate not only to regenerate ATP for contraction but also to sustain biosynthetic reactions for replacement of cellular building blocks. Metabolic pathways also control intracellular redox state, and metabolic intermediates and end products provide signals that prompt changes in enzymatic activity and gene expression. Mounting evidence suggests that the changes in cardiac metabolism that occur during development, exercise, and pregnancy as well as with pathological stress (eg, myocardial infarction, pressure overload) are causative in cardiac remodeling. Metabolism-mediated changes in gene expression, metabolite signaling, and the channeling of glucose-derived carbon toward anabolic pathways seem critical for physiological growth of the heart, and metabolic inefficiency and loss of coordinated anabolic activity are emerging as proximal causes of pathological remodeling. This review integrates knowledge of different forms of cardiac remodeling to develop general models of how relationships between catabolic and anabolic glucose metabolism may fortify cardiac health or promote (mal)adaptive myocardial remodeling. Adoption of conceptual frameworks based in relational biology may enable further understanding of how metabolism regulates cardiac structure and function.


Subject(s)
Cardiomegaly/metabolism , Exercise/physiology , Heart/growth & development , Myocardium/metabolism , Ventricular Remodeling/physiology , Adaptation, Physiological , Adenosine Triphosphate/biosynthesis , Cardiomegaly/etiology , Cardiomegaly, Exercise-Induced/physiology , Diabetes Mellitus/metabolism , Energy Metabolism , Female , Fetal Heart/metabolism , Gene Expression , Glucose/metabolism , Heart Failure/metabolism , Humans , Pregnancy , Signal Transduction
18.
Regeneration (Oxf) ; 5(1): 78-86, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29721328

ABSTRACT

Planarians are outstanding models for studying mechanisms of regeneration; however, there are few methods to measure changes in their metabolism. Examining metabolism in planarians is important because the regenerative process is dependent on numerous integrated metabolic pathways, which provide the energy required for tissue repair as well as the ability to synthesize the cellular building blocks needed to form new tissue. Therefore, we standardized an extracellular flux analysis method to measure mitochondrial and glycolytic activity in live planarians during normal growth as well as during regeneration. Small, uninjured planarians showed higher rates of oxygen consumption compared with large planarians, with no difference in glycolytic activity; however, glycolysis increased during planarian regeneration. Exposure of planarians to koningic acid, a specific inhibitor of glyceraldehyde-3-phosphate dehydrogenase, completely abolished extracellular acidification with little effect on oxygen consumption, which suggests that the majority of glucose catabolized in planarians is fated for aerobic glycolysis. These studies describe a useful method for measuring respiration and glycolysis in planarians and provide data implicating changes in glucose metabolism in the regenerative response.

19.
JCI Insight ; 3(3)2018 02 08.
Article in English | MEDLINE | ID: mdl-29415881

ABSTRACT

Skeletal muscle mass is regulated by a complex array of signaling pathways. TGF-ß-activated kinase 1 (TAK1) is an important signaling protein, which regulates context-dependent activation of multiple intracellular pathways. However, the role of TAK1 in the regulation of skeletal muscle mass remains unknown. Here, we report that inducible inactivation of TAK1 causes severe muscle wasting, leading to kyphosis, in both young and adult mice.. Inactivation of TAK1 inhibits protein synthesis and induces proteolysis, potentially through upregulating the activity of the ubiquitin-proteasome system and autophagy. Phosphorylation and enzymatic activity of AMPK are increased, whereas levels of phosphorylated mTOR and p38 MAPK are diminished upon inducible inactivation of TAK1 in skeletal muscle. In addition, targeted inactivation of TAK1 leads to the accumulation of dysfunctional mitochondria and oxidative stress in skeletal muscle of adult mice. Inhibition of TAK1 does not attenuate denervation-induced muscle wasting in adult mice. Finally, TAK1 activity is highly upregulated during overload-induced skeletal muscle growth, and inactivation of TAK1 prevents myofiber hypertrophy in response to functional overload. Overall, our study demonstrates that TAK1 is a key regulator of skeletal muscle mass and oxidative metabolism.


Subject(s)
MAP Kinase Kinase Kinases/metabolism , Mitochondria/metabolism , Muscle Weakness/pathology , Muscle, Skeletal/pathology , Animals , Autophagy/physiology , Disease Models, Animal , Female , Humans , Hypertrophy , Kyphosis/etiology , MAP Kinase Kinase Kinases/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/pathology , Muscle Weakness/complications , Muscle Weakness/genetics , Muscle, Skeletal/cytology , Muscle, Skeletal/metabolism , Oxidative Stress/physiology , Proteasome Endopeptidase Complex/metabolism , Proteolysis , Signal Transduction/physiology
SELECTION OF CITATIONS
SEARCH DETAIL