1
|
Wang Y, Zhu S, He W, Marchuk H, Richard E, Desviat LR, Young SP, Koeberl D, Kasumov T, Chen X, Zhang GF. The attenuated hepatic clearance of propionate increases cardiac oxidative stress in propionic acidemia. Basic Res Cardiol 2024:10.1007/s00395-024-01066-w. [PMID: 38992300 DOI: 10.1007/s00395-024-01066-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Revised: 06/29/2024] [Accepted: 06/30/2024] [Indexed: 07/13/2024]
Abstract
Propionic acidemia (PA), arising from PCCA or PCCB variants, manifests as life-threatening cardiomyopathy and arrhythmias, with unclear pathophysiology. In this work, propionyl-CoA metabolism in rodent hearts and human pluripotent stem cell-derived cardiomyocytes was investigated with stable isotope tracing analysis. Surprisingly, gut microbiome-derived propionate rather than the propiogenic amino acids (valine, isoleucine, threonine, and methionine) or odd-chain fatty acids was found to be the primary cardiac propionyl-CoA source. In a Pcca-/-(A138T) mouse model and PA patients, accumulated propionyl-CoA and diminished acyl-CoA synthetase short-chain family member 3 impede hepatic propionate disposal, elevating circulating propionate. Prolonged propionate exposure induced significant oxidative stress in PCCA knockdown HL-1 cells and the hearts of Pcca-/-(A138T) mice. Additionally, Pcca-/-(A138T) mice exhibited mild diastolic dysfunction after the propionate challenge. These findings suggest that elevated circulating propionate may cause oxidative damage and functional impairment in the hearts of patients with PA.
Collapse
Affiliation(s)
- You Wang
- School of Basic Medicine, Jining Medical University, Shandong, 272067, China
- Duke Molecular Physiology Institute and Sarah W. Stedman Nutrition and Metabolism Center, Duke University Medical Center, Carmichael Building 48-203, 300 North Duke Street, Durham, NC, 27701, USA
| | - Suhong Zhu
- School of Basic Medicine, Jining Medical University, Shandong, 272067, China
- Duke Molecular Physiology Institute and Sarah W. Stedman Nutrition and Metabolism Center, Duke University Medical Center, Carmichael Building 48-203, 300 North Duke Street, Durham, NC, 27701, USA
| | - Wentao He
- Duke Molecular Physiology Institute and Sarah W. Stedman Nutrition and Metabolism Center, Duke University Medical Center, Carmichael Building 48-203, 300 North Duke Street, Durham, NC, 27701, USA
| | - Hannah Marchuk
- Duke Molecular Physiology Institute and Sarah W. Stedman Nutrition and Metabolism Center, Duke University Medical Center, Carmichael Building 48-203, 300 North Duke Street, Durham, NC, 27701, USA
| | - Eva Richard
- Centro de Biología Molecular Severo Ochoa UAM-CSIC, CIBERER, IdiPaz, IUBM, Universidad Autónoma de Madrid, Madrid, Spain
| | - Lourdes R Desviat
- Centro de Biología Molecular Severo Ochoa UAM-CSIC, CIBERER, IdiPaz, IUBM, Universidad Autónoma de Madrid, Madrid, Spain
| | - Sarah P Young
- Biochemical Genetics Laboratory, Duke University Health System, Durham, NC, USA
- Division of Medical Genetics, Department of Pediatrics, Duke University School of Medicine, Duke University Medical Center, Durham, NC, 27710, USA
| | - Dwight Koeberl
- Division of Medical Genetics, Department of Pediatrics, Duke University School of Medicine, Duke University Medical Center, Durham, NC, 27710, USA
| | - Takhar Kasumov
- Department of Pharmaceutical Sciences, College of Pharmacy, Northeast Ohio Medical University, Rootstown, OH, USA
| | - Xiaoxin Chen
- Surgical Research Lab, Department of Surgery, Cooper University Hospital and Cooper Medical School of Rowan University, Camden, NJ, 08103, USA
- Coriell Institute for Medical Research, Camden, NJ, 08103, USA
- MD Anderson Cancer Center at Cooper, Camden, NJ, 08103, USA
| | - Guo-Fang Zhang
- Duke Molecular Physiology Institute and Sarah W. Stedman Nutrition and Metabolism Center, Duke University Medical Center, Carmichael Building 48-203, 300 North Duke Street, Durham, NC, 27701, USA.
- Department of Medicine, Division of Endocrinology, Metabolism and Nutrition, Duke University Medical Center, Durham, NC, 27701, USA.
| |
Collapse
|
2
|
He W, Marchuk H, Koeberl D, Kasumov T, Chen X, Zhang GF. Fasting alleviates metabolic alterations in mice with propionyl-CoA carboxylase deficiency due to Pcca mutation. Commun Biol 2024; 7:659. [PMID: 38811689 PMCID: PMC11137003 DOI: 10.1038/s42003-024-06362-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 05/20/2024] [Indexed: 05/31/2024] Open
Abstract
Propionic acidemia (PA), resulting from Pcca or Pccb gene mutations, impairs propionyl-CoA metabolism and induces metabolic alterations. While speculation exists that fasting might exacerbate metabolic crises in PA patients by accelerating the breakdown of odd-chain fatty acids and amino acids into propionyl-CoA, direct evidence is lacking. Our investigation into the metabolic effects of fasting in Pcca-/-(A138T) mice, a PA model, reveals surprising outcomes. Propionylcarnitine, a PA biomarker, decreases during fasting, along with the C3/C2 (propionylcarnitine/acetylcarnitine) ratio, ammonia, and methylcitrate. Although moderate amino acid catabolism to propionyl-CoA occurs with a 23-h fasting, a significant reduction in microbiome-produced propionate and increased fatty acid oxidation mitigate metabolic alterations by decreasing propionyl-CoA synthesis and enhancing acetyl-CoA synthesis. Fasting-induced gluconeogenesis further facilitates propionyl-CoA catabolism without changing propionyl-CoA carboxylase activity. These findings suggest that fasting may alleviate metabolic alterations in Pcca-/-(A138T) mice, prompting the need for clinical evaluation of its potential impact on PA patients.
Collapse
Affiliation(s)
- Wentao He
- Sarah W. Stedman Nutrition and Metabolism Center and Duke Molecular Physiology Institute, Duke University, Durham, NC, 27701, USA
| | - Hannah Marchuk
- Sarah W. Stedman Nutrition and Metabolism Center and Duke Molecular Physiology Institute, Duke University, Durham, NC, 27701, USA
| | - Dwight Koeberl
- Division of Medical Genetics, Department of Pediatrics, Duke University School of Medicine, Duke University Medical Center, Durham, NC, 27710, USA
| | - Takhar Kasumov
- Northeast Ohio Medical University, Rootstown, OH, 44272, USA
| | - Xiaoxin Chen
- Department of Surgery, Surgical Research Lab, Cooper University Hospital and Cooper Medical School of Rowan University, Camden, NJ, 08103, USA
- Coriell Institute for Medical Research, Camden, NJ, 08103, USA
- MD Anderson Cancer Center at Cooper, Camden, NJ, 08103, USA
| | - Guo-Fang Zhang
- Sarah W. Stedman Nutrition and Metabolism Center and Duke Molecular Physiology Institute, Duke University, Durham, NC, 27701, USA.
- Division of Endocrinology, Department of Medicine, Metabolism and Nutrition, Duke University Medical Center, Durham, NC, 27701, USA.
| |
Collapse
|
3
|
Meeusen H, Romagnolo A, Holsink SAC, van den Broek TJM, van Helvoort A, Gorter JA, van Vliet EA, Verkuyl JM, Silva JP, Aronica E. A novel hepatocyte ketone production assay to help the selection of nutrients for the ketogenic diet treatment of epilepsy. Sci Rep 2024; 14:11940. [PMID: 38789658 PMCID: PMC11126716 DOI: 10.1038/s41598-024-62723-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Accepted: 05/21/2024] [Indexed: 05/26/2024] Open
Abstract
The classic ketogenic diet is an effective treatment option for drug-resistant epilepsy, but its high fat content challenges patient compliance. Optimizing liver ketone production guided by a method comparing substrates for their ketogenic potential may help to reduce the fat content of the diet without loss in ketosis induction. Here, we present a liver cell assay measuring the β-hydroxybutyrate (βHB) yield from fatty acid substrates. Even chain albumin-conjugated fatty acids comprising between 4 and 18 carbon atoms showed a sigmoidal concentration-βHB response curve (CRC) whereas acetate and omega-3 PUFAs produced no CRC. While CRCs were not distinguished by their half-maximal effective concentration (EC50), they differed by maximum response, which related inversely to the carbon chain length and was highest for butyrate. The assay also suitably assessed the βHB yield from fatty acid blends detecting shifts in maximum response from exchanging medium chain fatty acids for long chain fatty acids. The assay further detected a dual role for butyrate and hexanoic acid as ketogenic substrate at high concentration and ketogenic enhancer at low concentration, augmenting the βHB yield from oleic acid and a fatty acid blend. The assay also found propionate to inhibit ketogenesis from oleic acid and a fatty acid blend at low physiological concentration. Although the in vitro assay shows promise as a tool to optimize the ketogenic yield of a fat blend, its predictive value requires human validation.
Collapse
Affiliation(s)
- Hester Meeusen
- Department of (Neuro)Pathology, Amsterdam UMC, Amsterdam Neuroscience, University of Amsterdam, Amsterdam, The Netherlands
- Department of Nutritional Physiology and Functional Nutrients, Medical & Nutrition Science, Danone Nutricia Research, Uppsalalaan 12, 3584CT, Utrecht, The Netherlands
| | - Alessia Romagnolo
- Department of (Neuro)Pathology, Amsterdam UMC, Amsterdam Neuroscience, University of Amsterdam, Amsterdam, The Netherlands
- Department of Nutritional Physiology and Functional Nutrients, Medical & Nutrition Science, Danone Nutricia Research, Uppsalalaan 12, 3584CT, Utrecht, The Netherlands
| | - Sophie A C Holsink
- Department of Nutritional Physiology and Functional Nutrients, Medical & Nutrition Science, Danone Nutricia Research, Uppsalalaan 12, 3584CT, Utrecht, The Netherlands
| | - Thijs J M van den Broek
- Department of Nutritional Physiology and Functional Nutrients, Medical & Nutrition Science, Danone Nutricia Research, Uppsalalaan 12, 3584CT, Utrecht, The Netherlands
| | - Ardy van Helvoort
- Department of Nutritional Physiology and Functional Nutrients, Medical & Nutrition Science, Danone Nutricia Research, Uppsalalaan 12, 3584CT, Utrecht, The Netherlands
- Department of Respiratory Medicine, NUTRIM - Research Institute of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, Maastricht University, Maastricht, The Netherlands
| | - Jan A Gorter
- Swammerdam Institute for Life Sciences, Center for Neuroscience, University of Amsterdam, Amsterdam, The Netherlands
| | - Erwin A van Vliet
- Department of (Neuro)Pathology, Amsterdam UMC, Amsterdam Neuroscience, University of Amsterdam, Amsterdam, The Netherlands
- Swammerdam Institute for Life Sciences, Center for Neuroscience, University of Amsterdam, Amsterdam, The Netherlands
| | - J Martin Verkuyl
- Department of Nutritional Physiology and Functional Nutrients, Medical & Nutrition Science, Danone Nutricia Research, Uppsalalaan 12, 3584CT, Utrecht, The Netherlands
| | - Jose P Silva
- Department of Nutritional Physiology and Functional Nutrients, Medical & Nutrition Science, Danone Nutricia Research, Uppsalalaan 12, 3584CT, Utrecht, The Netherlands.
| | - Eleonora Aronica
- Department of (Neuro)Pathology, Amsterdam UMC, Amsterdam Neuroscience, University of Amsterdam, Amsterdam, The Netherlands
- Stichting Epilepsie Instellingen Nederland (SEIN), Heemstede, The Netherlands
| |
Collapse
|
4
|
Thaiwatcharamas K, Loilome W, Ho PN, Chusilp S, Tanming P, Klanrit P, Phetcharaburanin J. Children with Hirschsprung disease exhibited alterations in host-microbial co-metabolism after pull-through operation. Pediatr Surg Int 2024; 40:87. [PMID: 38512700 DOI: 10.1007/s00383-024-05667-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/23/2024] [Indexed: 03/23/2024]
Abstract
PURPOSE This study aims to compare the fecal metabolome in post pull-through HD with and without HAEC patients and healthy young children using nuclear magnetic resonance (NMR) spectroscopy. METHODS Fresh fecal samples were collected from children under 5 years of age in both post-pull-through HD patients and healthy Thai children. A total of 20 fecal samples were then analyzed using NMR spectroscopy. RESULTS Thirty-four metabolites identified among HD and healthy children younger than 5 years were compared. HD samples demonstrated a significant decrease in acetoin, phenylacetylglutamine, and N-acetylornithine (corrected p value = 0.01, 0.04, and 0.004, respectively). Succinate and xylose significantly decreased in HD with HAEC group compared to HD without HAEC group (corrected p value = 0.04 and 0.02, respectively). Moreover, glutamine and glutamate metabolism, and alanine, aspartate, and glutamate metabolism were the significant pathways involved, with pathway impact 0.42 and 0.50, respectively (corrected p value = 0.02 and 0.04, respectively). CONCLUSION Differences in class, quantity, and metabolism of protein and other metabolites in young children with HD after pull-through operation were identified. Most of the associated metabolic pathways were correlated with the amino acids metabolism, which is required to maintain intestinal integrity and function.
Collapse
Affiliation(s)
| | - Watcharin Loilome
- Department of Systems Biosciences and Computational Medicine, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
- Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, Thailand
- Khon Kaen University Phenome Centre, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Phuc N Ho
- Department of Systems Biosciences and Computational Medicine, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Sinobol Chusilp
- Department of Surgery, Division of Pediatric Surgery, Khon Kaen University, Khon Kaen, Thailand
| | - Patchareeporn Tanming
- Department of Surgery, Division of Pediatric Surgery, Khon Kaen University, Khon Kaen, Thailand
| | - Poramate Klanrit
- Department of Systems Biosciences and Computational Medicine, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
- Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, Thailand
- Khon Kaen University Phenome Centre, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Jutarop Phetcharaburanin
- Department of Systems Biosciences and Computational Medicine, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand.
- Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, Thailand.
- Khon Kaen University Phenome Centre, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand.
| |
Collapse
|
5
|
Williams AS, Crown SB, Lyons SP, Koves TR, Wilson RJ, Johnson JM, Slentz DH, Kelly DP, Grimsrud PA, Zhang GF, Muoio DM. Ketone flux through BDH1 supports metabolic remodeling of skeletal and cardiac muscles in response to intermittent time-restricted feeding. Cell Metab 2024; 36:422-437.e8. [PMID: 38325337 PMCID: PMC10961007 DOI: 10.1016/j.cmet.2024.01.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 09/20/2023] [Accepted: 01/10/2024] [Indexed: 02/09/2024]
Abstract
Time-restricted feeding (TRF) has gained attention as a dietary regimen that promotes metabolic health. This study questioned if the health benefits of an intermittent TRF (iTRF) schedule require ketone flux specifically in skeletal and cardiac muscles. Notably, we found that the ketolytic enzyme beta-hydroxybutyrate dehydrogenase 1 (BDH1) is uniquely enriched in isolated mitochondria derived from heart and red/oxidative skeletal muscles, which also have high capacity for fatty acid oxidation (FAO). Using mice with BDH1 deficiency in striated muscles, we discover that this enzyme optimizes FAO efficiency and exercise tolerance during acute fasting. Additionally, iTRF leads to robust molecular remodeling of muscle tissues, and muscle BDH1 flux does indeed play an essential role in conferring the full adaptive benefits of this regimen, including increased lean mass, mitochondrial hormesis, and metabolic rerouting of pyruvate. In sum, ketone flux enhances mitochondrial bioenergetics and supports iTRF-induced remodeling of skeletal muscle and heart.
Collapse
Affiliation(s)
- Ashley S Williams
- Duke Molecular Physiology Institute and Sarah W. Stedman Nutrition and Metabolism Center, Duke University Medical Center, Durham, NC 27701, USA
| | - Scott B Crown
- Duke Molecular Physiology Institute and Sarah W. Stedman Nutrition and Metabolism Center, Duke University Medical Center, Durham, NC 27701, USA
| | - Scott P Lyons
- Duke Molecular Physiology Institute and Sarah W. Stedman Nutrition and Metabolism Center, Duke University Medical Center, Durham, NC 27701, USA
| | - Timothy R Koves
- Duke Molecular Physiology Institute and Sarah W. Stedman Nutrition and Metabolism Center, Duke University Medical Center, Durham, NC 27701, USA; Department of Medicine, Division of Geriatrics, Duke University Medical Center, Durham, NC 27710, USA
| | - Rebecca J Wilson
- Duke Molecular Physiology Institute and Sarah W. Stedman Nutrition and Metabolism Center, Duke University Medical Center, Durham, NC 27701, USA
| | - Jordan M Johnson
- Duke Molecular Physiology Institute and Sarah W. Stedman Nutrition and Metabolism Center, Duke University Medical Center, Durham, NC 27701, USA
| | - Dorothy H Slentz
- Duke Molecular Physiology Institute and Sarah W. Stedman Nutrition and Metabolism Center, Duke University Medical Center, Durham, NC 27701, USA
| | - Daniel P Kelly
- Cardiovascular Institute and Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Paul A Grimsrud
- Duke Molecular Physiology Institute and Sarah W. Stedman Nutrition and Metabolism Center, Duke University Medical Center, Durham, NC 27701, USA; Department of Medicine, Division of Endocrinology, Metabolism, and Nutrition, Duke University Medical Center, Durham, NC 27710, USA
| | - Guo-Fang Zhang
- Duke Molecular Physiology Institute and Sarah W. Stedman Nutrition and Metabolism Center, Duke University Medical Center, Durham, NC 27701, USA; Department of Medicine, Division of Endocrinology, Metabolism, and Nutrition, Duke University Medical Center, Durham, NC 27710, USA
| | - Deborah M Muoio
- Duke Molecular Physiology Institute and Sarah W. Stedman Nutrition and Metabolism Center, Duke University Medical Center, Durham, NC 27701, USA; Department of Medicine, Division of Endocrinology, Metabolism, and Nutrition, Duke University Medical Center, Durham, NC 27710, USA; Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC 27710, USA.
| |
Collapse
|
6
|
Das SS, Kar A, Rajkumar S, Lee SHT, Alvarez M, Pietiläinen KH, Pajukanta P. Cross-Tissue Single-Nucleus RNA Sequencing Discovers Tissue-Resident Adipocytes Involved in Propanoate Metabolism in the Human Heart. Arterioscler Thromb Vasc Biol 2023; 43:1788-1804. [PMID: 37409528 PMCID: PMC10538422 DOI: 10.1161/atvbaha.123.319358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 06/26/2023] [Indexed: 07/07/2023]
Abstract
BACKGROUND Adipocytes are crucial regulators of cardiovascular health. However, not much is known about gene expression profiles of adipocytes residing in nonfat cardiovascular tissues, their genetic regulation, and contribution to coronary artery disease. Here, we investigated whether and how the gene expression profiles of adipocytes in the subcutaneous adipose tissue differ from adipocytes residing in the heart. METHODS We used single-nucleus RNA-sequencing data sets of subcutaneous adipose tissue and heart and performed in-depth analysis of tissue-resident adipocytes and their cell-cell interactions. RESULTS We first discovered tissue-specific features of tissue-resident adipocytes, identified functional pathways involved in their tissue specificity, and found genes with cell type-specific expression enrichment in tissue-resident adipocytes. By following up these results, we discovered the propanoate metabolism pathway as a novel distinct characteristic of the heart-resident adipocytes and found a significant enrichment of coronary artery disease genome-wide association study risk variants among the right atrium-specific adipocyte marker genes. Our cell-cell communication analysis identified 22 specific heart adipocyte-associated ligand-receptor pairs and signaling pathways, including THBS (thrombospondin) and EPHA (ephrin type-A), further supporting the distinct tissue-resident role of heart adipocytes. Our results also suggest chamber-level coordination of heart adipocyte expression profiles as we observed a consistently larger number of adipocyte-associated ligand-receptor interactions and functional pathways in the atriums than ventricles. CONCLUSIONS Overall, we introduce a new function and genetic link to coronary artery disease for the previously unexplored heart-resident adipocytes.
Collapse
Affiliation(s)
- Sankha Subhra Das
- Department of Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, USA
| | - Asha Kar
- Department of Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, USA
| | - Sandhya Rajkumar
- Department of Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, USA
| | - Seung Hyuk T. Lee
- Department of Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, USA
| | - Marcus Alvarez
- Department of Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, USA
| | - Kirsi H Pietiläinen
- Obesity Research Unit, Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- HealthyWeightHub, Abdominal Center, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Päivi Pajukanta
- Department of Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, USA
- Bioinformatics Interdepartmental Program, UCLA, Los Angeles, USA
- Institute for Precision Health, David Geffen School of Medicine at UCLA, Los Angeles, USA
| |
Collapse
|
7
|
Marchuk H, Wang Y, Ladd ZA, Chen X, Zhang GF. Pathophysiological mechanisms of complications associated with propionic acidemia. Pharmacol Ther 2023; 249:108501. [PMID: 37482098 PMCID: PMC10529999 DOI: 10.1016/j.pharmthera.2023.108501] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 07/06/2023] [Accepted: 07/18/2023] [Indexed: 07/25/2023]
Abstract
Propionic acidemia (PA) is a genetic metabolic disorder caused by mutations in the mitochondrial enzyme, propionyl-CoA carboxylase (PCC), which is responsible for converting propionyl-CoA to methylmalonyl-CoA for further metabolism in the tricarboxylic acid cycle. When this process is disrupted, propionyl-CoA and its metabolites accumulate, leading to a variety of complications including life-threatening cardiac diseases and other metabolic strokes. While the clinical symptoms and diagnosis of PA are well established, the underlying pathophysiological mechanisms of PA-induced diseases are not fully understood. As a result, there are currently few effective therapies for PA beyond dietary restriction. This review focuses on the pathophysiological mechanisms of the various complications associated with PA, drawing on extensive research and clinical reports. Most research suggests that propionyl-CoA and its metabolites can impair mitochondrial energy metabolism and cause cellular damage by inducing oxidative stress. However, direct evidence from in vivo studies is still lacking. Additionally, elevated levels of ammonia can be toxic, although not all PA patients develop hyperammonemia. The discovery of pathophysiological mechanisms underlying various complications associated with PA can aid in the development of more effective therapeutic treatments. The consequences of elevated odd-chain fatty acids in lipid metabolism and potential gene expression changes mediated by histone propionylation also warrant further investigation.
Collapse
Affiliation(s)
- Hannah Marchuk
- Sarah W. Stedman Nutrition and Metabolism Center & Duke Molecular Physiology Institute, Duke University, Durham, NC 27701, USA
| | - You Wang
- Jining Key Laboratory of Pharmacology, Jining Medical University, Shandong 272067, China.; School of Basic Medicine, Jining Medical University, Shandong 272067, China
| | - Zachary Alec Ladd
- Surgical Research Lab, Department of Surgery, Cooper University Healthcare and Cooper Medical School of Rowan University, Camden, NJ 08103, USA
| | - Xiaoxin Chen
- Surgical Research Lab, Department of Surgery, Cooper University Healthcare and Cooper Medical School of Rowan University, Camden, NJ 08103, USA; Coriell Institute for Medical Research, Camden, NJ 08103, USA; MD Anderson Cancer Center at Cooper, Camden, NJ 08103, USA.
| | - Guo-Fang Zhang
- Sarah W. Stedman Nutrition and Metabolism Center & Duke Molecular Physiology Institute, Duke University, Durham, NC 27701, USA; Department of Medicine, Division of Endocrinology, and Metabolism Nutrition, Duke University Medical Center, Durham, NC 27710, USA.
| |
Collapse
|
8
|
Chen W, Yang Q, Hu L, Wang M, Yang Z, Zeng X, Sun Y. Shared diagnostic genes and potential mechanism between PCOS and recurrent implantation failure revealed by integrated transcriptomic analysis and machine learning. Front Immunol 2023; 14:1175384. [PMID: 37261354 PMCID: PMC10228695 DOI: 10.3389/fimmu.2023.1175384] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 05/04/2023] [Indexed: 06/02/2023] Open
Abstract
Polycystic ovary syndrome (PCOS) is a complex endocrine metabolic disorder that affects 5-10% of women of reproductive age. The endometrium of women with PCOS has altered immune cells resulting in chronic low-grade inflammation, which attribute to recurrent implantation failure (RIF). In this study, we obtained three PCOS and RIF datasets respectively from the Gene Expression Omnibus (GEO) database. By analyzing differentially expressed genes (DEGs) and module genes using weighted gene co-expression networks (WGCNA), functional enrichment analysis, and three machine learning algorithms, we identified twelve diseases shared genes, and two diagnostic genes, including GLIPR1 and MAMLD1. PCOS and RIF validation datasets were assessed using the receiver operating characteristic (ROC) curve, and ideal area under the curve (AUC) values were obtained for each disease. Besides, we collected granulosa cells from healthy and PCOS infertile women, and endometrial tissues of healthy and RIF patients. RT-PCR was used to validate the reliability of GLIPR1 and MAMLD1. Furthermore, we performed gene set enrichment analysis (GSEA) and immune infiltration to explore the underlying mechanism of PCOS and RIF cooccurrence. Through the functional enrichment of twelve shared genes and two diagnostic genes, we found that both PCOS and RIF patients had disturbances in metabolites related to the TCA cycle, which eventually led to the massive activation of immune cells.
Collapse
Affiliation(s)
- Wenhui Chen
- Center for Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Provincial Obstetrical and Gynecological Diseases (Reproductive Medicine) Clinical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Qingling Yang
- Center for Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Provincial Obstetrical and Gynecological Diseases (Reproductive Medicine) Clinical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Linli Hu
- Center for Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Provincial Obstetrical and Gynecological Diseases (Reproductive Medicine) Clinical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Mengchen Wang
- Center for Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Provincial Obstetrical and Gynecological Diseases (Reproductive Medicine) Clinical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Ziyao Yang
- Center for Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Provincial Obstetrical and Gynecological Diseases (Reproductive Medicine) Clinical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xinxin Zeng
- Center for Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Provincial Obstetrical and Gynecological Diseases (Reproductive Medicine) Clinical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yingpu Sun
- Center for Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Provincial Obstetrical and Gynecological Diseases (Reproductive Medicine) Clinical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
9
|
Maines E, Moretti M, Vitturi N, Gugelmo G, Fasan I, Lenzini L, Piccoli G, Gragnaniello V, Maiorana A, Soffiati M, Burlina A, Franceschi R. Understanding the Pathogenesis of Cardiac Complications in Patients with Propionic Acidemia and Exploring Therapeutic Alternatives for Those Who Are Not Eligible or Are Waiting for Liver Transplantation. Metabolites 2023; 13:563. [PMID: 37110221 PMCID: PMC10143878 DOI: 10.3390/metabo13040563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 04/11/2023] [Accepted: 04/12/2023] [Indexed: 04/29/2023] Open
Abstract
The guidelines for the management of patients affected by propionic acidemia (PA) recommend standard cardiac therapy in the presence of cardiac complications. A recent revision questioned the impact of high doses of coenzyme Q10 on cardiac function in patients with cardiomyopathy (CM). Liver transplantation is a therapeutic option for several patients since it may stabilize or reverse CM. Both the patients waiting for liver transplantation and, even more, the ones not eligible for transplant programs urgently need therapies to improve cardiac function. To this aim, the identification of the pathogenetic mechanisms represents a key point. Aims: This review summarizes: (1) the current knowledge of the pathogenetic mechanisms underlying cardiac complications in PA and (2) the available and potential pharmacological options for the prevention or the treatment of cardiac complications in PA. To select articles, we searched the electronic database PubMed using the Mesh terms "propionic acidemia" OR "propionate" AND "cardiomyopathy" OR "Long QT syndrome". We selected 77 studies, enlightening 12 potential disease-specific or non-disease-specific pathogenetic mechanisms, namely: impaired substrate delivery to TCA cycle and TCA dysfunction, secondary mitochondrial electron transport chain dysfunction and oxidative stress, coenzyme Q10 deficiency, metabolic reprogramming, carnitine deficiency, cardiac excitation-contraction coupling alteration, genetics, epigenetics, microRNAs, micronutrients deficiencies, renin-angiotensin-aldosterone system activation, and increased sympathetic activation. We provide a critical discussion of the related therapeutic options. Current literature supports the involvement of multiple cellular pathways in cardiac complications of PA, indicating the growing complexity of their pathophysiology. Elucidating the mechanisms responsible for such abnormalities is essential to identify therapeutic strategies going beyond the correction of the enzymatic defect rather than engaging the dysregulated mechanisms. Although these approaches are not expected to be resolutive, they may improve the quality of life and slow the disease progression. Available pharmacological options are limited and tested in small cohorts. Indeed, a multicenter approach is mandatory to strengthen the efficacy of therapeutic options.
Collapse
Affiliation(s)
- Evelina Maines
- Division of Pediatrics, Santa Chiara General Hospital, APSS, 38122 Trento, Italy
| | - Michele Moretti
- Division of Cardiology, Santa Chiara General Hospital, APSS, 38122 Trento, Italy
| | - Nicola Vitturi
- Division of Metabolic Diseases, Department of Medicine-DIMED, University Hospital, 35128 Padova, Italy
| | - Giorgia Gugelmo
- Division of Clinical Nutrition, Department of Medicine-DIMED, University Hospital, 35128 Padova, Italy
| | - Ilaria Fasan
- Division of Clinical Nutrition, Department of Medicine-DIMED, University Hospital, 35128 Padova, Italy
| | - Livia Lenzini
- Emergency Medicine Unit, Department of Medicine-DIMED, University Hospital, 35128 Padova, Italy
| | - Giovanni Piccoli
- CIBIO, Department of Cellular, Computational and Integrative Biology, Italy & Dulbecco Telethon Institute, Università degli Studi di Trento, 38123 Trento, Italy
| | - Vincenza Gragnaniello
- Division of Inherited Metabolic Diseases, Reference Centre Expanded Newborn Screening, Department of Women’s and Children’s Health, University Hospital, 35128 Padova, Italy
| | - Arianna Maiorana
- Division of Metabolism and Research Unit of Metabolic Biochemistry, Bambino Gesù Children’s Hospital-IRCCS, 00165 Rome, Italy
| | - Massimo Soffiati
- Division of Pediatrics, Santa Chiara General Hospital, APSS, 38122 Trento, Italy
| | - Alberto Burlina
- Division of Inherited Metabolic Diseases, Reference Centre Expanded Newborn Screening, Department of Women’s and Children’s Health, University Hospital, 35128 Padova, Italy
| | - Roberto Franceschi
- Division of Pediatrics, Santa Chiara General Hospital, APSS, 38122 Trento, Italy
| |
Collapse
|
10
|
Koves TR, Zhang GF, Davidson MT, Chaves AB, Crown SB, Johnson JM, Slentz DH, Grimsrud PA, Muoio DM. Pyruvate-supported flux through medium-chain ketothiolase promotes mitochondrial lipid tolerance in cardiac and skeletal muscles. Cell Metab 2023:S1550-4131(23)00094-3. [PMID: 37060901 DOI: 10.1016/j.cmet.2023.03.016] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 02/07/2023] [Accepted: 03/24/2023] [Indexed: 04/17/2023]
Abstract
Even-chain acylcarnitine (AC) metabolites, most of which are generated as byproducts of incomplete fatty acid oxidation (FAO), are viewed as biomarkers of mitochondrial lipid stress attributable to one or more metabolic bottlenecks in the β-oxidation pathway. The origins and functional implications of FAO bottlenecks remain poorly understood. Here, we combined a sophisticated mitochondrial phenotyping platform with state-of-the-art molecular profiling tools and multiple two-state mouse models of respiratory function to uncover a mechanism that connects AC accumulation to lipid intolerance, metabolic inflexibility, and respiratory inefficiency in skeletal muscle mitochondria. These studies also identified a short-chain carbon circuit at the C4 node of FAO wherein reverse flux of glucose-derived acetyl CoA through medium-chain ketothiolase enhances lipid tolerance and redox stability in heart mitochondria by regenerating free CoA and NAD+. The findings help to explain why diminished FAO capacity, AC accumulation, and metabolic inflexibility are tightly linked to poor health outcomes.
Collapse
Affiliation(s)
- Timothy R Koves
- Duke Molecular Physiology Institute and Sarah W. Stedman Nutrition and Metabolism Center, Duke University Medical Center, Durham, NC 27701, USA; Department of Medicine, Division of Geriatrics, Duke University Medical Center, Durham, NC 27710, USA
| | - Guo-Fang Zhang
- Duke Molecular Physiology Institute and Sarah W. Stedman Nutrition and Metabolism Center, Duke University Medical Center, Durham, NC 27701, USA; Department of Medicine, Division of Endocrinology, Metabolism, and Nutrition, Duke University Medical Center, Durham, NC 27710, USA
| | - Michael T Davidson
- Duke Molecular Physiology Institute and Sarah W. Stedman Nutrition and Metabolism Center, Duke University Medical Center, Durham, NC 27701, USA
| | - Alec B Chaves
- Duke Molecular Physiology Institute and Sarah W. Stedman Nutrition and Metabolism Center, Duke University Medical Center, Durham, NC 27701, USA
| | - Scott B Crown
- Duke Molecular Physiology Institute and Sarah W. Stedman Nutrition and Metabolism Center, Duke University Medical Center, Durham, NC 27701, USA
| | - Jordan M Johnson
- Duke Molecular Physiology Institute and Sarah W. Stedman Nutrition and Metabolism Center, Duke University Medical Center, Durham, NC 27701, USA
| | - Dorothy H Slentz
- Duke Molecular Physiology Institute and Sarah W. Stedman Nutrition and Metabolism Center, Duke University Medical Center, Durham, NC 27701, USA
| | - Paul A Grimsrud
- Duke Molecular Physiology Institute and Sarah W. Stedman Nutrition and Metabolism Center, Duke University Medical Center, Durham, NC 27701, USA; Department of Medicine, Division of Endocrinology, Metabolism, and Nutrition, Duke University Medical Center, Durham, NC 27710, USA
| | - Deborah M Muoio
- Duke Molecular Physiology Institute and Sarah W. Stedman Nutrition and Metabolism Center, Duke University Medical Center, Durham, NC 27701, USA; Department of Medicine, Division of Endocrinology, Metabolism, and Nutrition, Duke University Medical Center, Durham, NC 27710, USA; Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC, USA.
| |
Collapse
|
11
|
He W, Berthiaume JM, Previs S, Kasumov T, Zhang GF. Ischemia promotes acyl-CoAs dephosphorylation and propionyl-CoA accumulation. Metabolomics 2023; 19:12. [PMID: 36750484 PMCID: PMC11238255 DOI: 10.1007/s11306-023-01975-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Accepted: 01/23/2023] [Indexed: 02/09/2023]
Abstract
INTRODUCTION Our untargeted metabolic data unveiled that Acyl-CoAs undergo dephosphorylation, however little is known about these novel metabolites and their physiology/pathology relevance. OBJECTIVES To understand the relationship between acyl-CoAs dephosphorylation and energy status as implied in our previous work, we seek to investigate how ischemia (energy depletion) triggers metabolic changes, specifically acyl-CoAs dephosphorylation in this work. METHODS Rat hearts were isolated and perfused in Langendorff mode for 15 min followed by 0, 5, 15, and 30 minutes of global ischemia. The heart tissues were harvested for metabolic analysis. RESULTS As expected, ATP and phosphocreatine were significantly decreased during ischemia. Most short- and medium-chain acyl-CoAs progressively increased with ischemic time from 0 to 15 min, whereas a 30-minute ischemia did not lead to further change. Unlike other acyl-CoAs, propionyl-CoA accumulated progressively in the hearts that underwent ischemia from 0 to 30 min. Progressive dephosphorylation occurred to all assayed acyl-CoAs and free CoA regardless their level changes during the ischemia. CONCLUSION The present work further confirms that dephosphorylation of acyl-CoAs is an energy-dependent process and how this dephosphorylation is mediated warrants further investigations. It is plausible that dephosphorylation of acyl-CoAs and limited anaplerosis are involved in ischemic injuries to heart. Further investigations are warranted to examine the mechanisms of acyl-CoA dephosphorylation and how the dephosphorylation is possibly involved in ischemic injuries.
Collapse
Affiliation(s)
- Wentao He
- Sarah W. Stedman Nutrition and Metabolism Center & Duke Molecular Physiology Institute, Duke University, Durham, NC, 27701, USA
| | - Jessica M Berthiaume
- Department of Physiology & Biophysics, Case Western Reserve University, Cleveland, OH, 44104, USA
- Inotiv Westminster, 7581 W 103rd Ave, Westminster, CO, 80021, USA
| | - Stephen Previs
- Merck & Co., Inc, 2000 Galloping Hill Rd, Kenilworth, NJ, 07033, USA
| | - Takhar Kasumov
- Northeast Ohio Medical University, Rootstown, OH, 44272, USA
| | - Guo-Fang Zhang
- Sarah W. Stedman Nutrition and Metabolism Center & Duke Molecular Physiology Institute, Duke University, Durham, NC, 27701, USA.
- Department of Medicine, Division of Endocrinology, Metabolism Nutrition, Duke University Medical Center, Durham, NC, 27701, USA.
| |
Collapse
|
12
|
Li Y, Yang S, Jin X, Li D, Lu J, Wang X, Wu M. Mitochondria as novel mediators linking gut microbiota to atherosclerosis that is ameliorated by herbal medicine: A review. Front Pharmacol 2023; 14:1082817. [PMID: 36733506 PMCID: PMC9886688 DOI: 10.3389/fphar.2023.1082817] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Accepted: 01/06/2023] [Indexed: 01/18/2023] Open
Abstract
Atherosclerosis (AS) is the main cause of cardiovascular disease (CVD) and is characterized by endothelial damage, lipid deposition, and chronic inflammation. Gut microbiota plays an important role in the occurrence and development of AS by regulating host metabolism and immunity. As human mitochondria evolved from primordial bacteria have homologous characteristics, they are attacked by microbial pathogens as target organelles, thus contributing to energy metabolism disorders, oxidative stress, and apoptosis. Therefore, mitochondria may be a key mediator of intestinal microbiota disorders and AS aggravation. Microbial metabolites, such as short-chain fatty acids, trimethylamine, hydrogen sulfide, and bile acids, also affect mitochondrial function, including mtDNA mutation, oxidative stress, and mitophagy, promoting low-grade inflammation. This further damages cellular homeostasis and the balance of innate immunity, aggravating AS. Herbal medicines and their monomers can effectively ameliorate the intestinal flora and their metabolites, improve mitochondrial function, and inhibit atherosclerotic plaques. This review focuses on the interaction between gut microbiota and mitochondria in AS and explores a therapeutic strategy for restoring mitochondrial function and intestinal microbiota disorders using herbal medicines, aiming to provide new insights for the prevention and treatment of AS.
Collapse
Affiliation(s)
- Yujuan Li
- Guang’an Men Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Shengjie Yang
- Guang’an Men Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xiao Jin
- Guang’an Men Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Dan Li
- Guang’an Men Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jing Lu
- Guang’an Men Hospital, China Academy of Chinese Medical Sciences, Beijing, China,Beijing University of Chinese Medicine, Beijing, China
| | - Xinyue Wang
- Guang’an Men Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Min Wu
- Guang’an Men Hospital, China Academy of Chinese Medical Sciences, Beijing, China,*Correspondence: Min Wu,
| |
Collapse
|
13
|
Subramanian C, Frank MW, Tangallapally R, Yun MK, White SW, Lee RE, Rock CO, Jackowski S. Relief of CoA sequestration and restoration of mitochondrial function in a mouse model of propionic acidemia. J Inherit Metab Dis 2023; 46:28-42. [PMID: 36251252 PMCID: PMC10092110 DOI: 10.1002/jimd.12570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 10/06/2022] [Accepted: 10/10/2022] [Indexed: 01/19/2023]
Abstract
Propionic acidemia (PA, OMIM 606054) is a devastating inborn error of metabolism arising from mutations that reduce the activity of the mitochondrial enzyme propionyl-CoA carboxylase (PCC). The defects in PCC reduce the concentrations of nonesterified coenzyme A (CoASH), thus compromising mitochondrial function and disrupting intermediary metabolism. Here, we use a hypomorphic PA mouse model to test the effectiveness of BBP-671 in correcting the metabolic imbalances in PA. BBP-671 is a high-affinity allosteric pantothenate kinase activator that counteracts feedback inhibition of the enzyme to increase the intracellular concentration of CoA. Liver CoASH and acetyl-CoA are depressed in PA mice and BBP-671 treatment normalizes the cellular concentrations of these two key cofactors. Hepatic propionyl-CoA is also reduced by BBP-671 leading to an improved intracellular C3:C2-CoA ratio. Elevated plasma C3:C2-carnitine ratio and methylcitrate, hallmark biomarkers of PA, are significantly reduced by BBP-671. The large elevations of malate and α-ketoglutarate in the urine of PA mice are biomarkers for compromised tricarboxylic acid cycle activity and BBP-671 therapy reduces the amounts of both metabolites. Furthermore, the low survival of PA mice is restored to normal by BBP-671. These data show that BBP-671 relieves CoA sequestration, improves mitochondrial function, reduces plasma PA biomarkers, and extends the lifespan of PA mice, providing the preclinical foundation for the therapeutic potential of BBP-671.
Collapse
Affiliation(s)
- Chitra Subramanian
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, Tennessee, United States
| | - Matthew W Frank
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, Tennessee, United States
| | - Rajendra Tangallapally
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, Tennessee, United States
| | - Mi-Kyung Yun
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, Tennessee, United States
| | - Stephen W White
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, Tennessee, United States
- St. Jude Graduate School of Biomedical Sciences, St. Jude Children's Research Hospital, Memphis, Tennessee, United States
- Department of Microbiology, Immunology and Biochemistry, University of Tennessee Health Science Center, Memphis, Tennessee, United States
| | - Richard E Lee
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, Tennessee, United States
| | - Charles O Rock
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, Tennessee, United States
- Department of Microbiology, Immunology and Biochemistry, University of Tennessee Health Science Center, Memphis, Tennessee, United States
| | - Suzanne Jackowski
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, Tennessee, United States
| |
Collapse
|
14
|
Hareng L, Schuster P, Haake V, Walk T, Herold M, Laue H, Natsch A. Towards the mechanism of spermatotoxicity of p-tert-butyl-alpha-methylhydrocinnamic aldehyde: inhibition of late stage ex-vivo spermatogenesis in rat seminiferous tubule cultures by para-tert-butyl- benzoic acid. Arch Toxicol 2023; 97:279-294. [PMID: 36173422 DOI: 10.1007/s00204-022-03379-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Accepted: 09/07/2022] [Indexed: 01/19/2023]
Abstract
Molecules metabolized to para-tert-butyl-benzoic acid (p-TBBA) affect male reproduction in rats through effects on spermatogenesis. This toxicity is specific to p-TBBA and not observed in meta-substituted analogues. The underlying mode of action was evaluated by comparing effects of p-TBBA and the position isomer m-TBBA (2-50 µM) in an ex vivo 3D primary seminiferous tubule cell culture system from juvenile Sprague Dawley rats (Bio-AlteR®). Treated cultures were evaluated for CoA-conjugate formation, cytotoxicity, blood-testis barrier functionality and different germ cell populations to assess effects on spermatogenesis. In addition, an evaluation of the metabolome of treated cultures was performed by using MxP® Broad Profiling via a LC-MS/MS and GC-MS platform. Para-TBBA decreased germ cell populations of late stages of spermatogenesis and led to the formation of CoA-conjugates in the ex vivo tissue. In addition, p-TBBA had a pronounced effect on the metabolome by affecting lipid balance and other CoA-dependent pathways contributing to energy production and the redox system. Meta-TBBA did not affect germ cell populations and no m-TBBA related CoA-conjugates were detectable. The metabolic profile of m-TBBA treated cells was comparable to vehicle control treated cultures, indicating that formation of CoA-conjugates, inhibition of spermatogenesis, and effects on the metabolome are mechanistically linked events. Thus, for this specific chemical group an adverse outcome pathway can be postulated, including the formation of benzoic acid metabolites, accumulation of CoA-conjugates to a certain threshold and CoA depletion, which affects the metabolic and lipid profile and leads to tissue specific effects with impaired functionalities such as spermatogenesis.
Collapse
Affiliation(s)
- Lars Hareng
- Product Safety, Regulations, Toxicology and Ecology, BASF SE, Z470, 67056, Ludwigshafen, Germany.
| | | | | | - Tilman Walk
- BASF Metabolome Solutions GmbH, Berlin, Germany
| | | | - Heike Laue
- Fragrances S&T, Ingredients Research, Givaudan Schweiz AG, 8310, Kemptthal, Switzerland
| | - Andreas Natsch
- Fragrances S&T, Ingredients Research, Givaudan Schweiz AG, 8310, Kemptthal, Switzerland
| |
Collapse
|
15
|
Tirzepatide induces a thermogenic-like amino acid signature in brown adipose tissue. Mol Metab 2022; 64:101550. [PMID: 35921984 PMCID: PMC9396640 DOI: 10.1016/j.molmet.2022.101550] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 07/12/2022] [Accepted: 07/13/2022] [Indexed: 11/30/2022] Open
Abstract
Objectives Tirzepatide, a dual GIP and GLP-1 receptor agonist, delivered superior glycemic control and weight loss compared to selective GLP-1 receptor (GLP-1R) agonism in patients with type 2 diabetes (T2D). These results have fueled mechanistic studies focused on understanding how tirzepatide achieves its therapeutic efficacy. Recently, we found that treatment with tirzepatide improves insulin sensitivity in humans with T2D and obese mice in concert with a reduction in circulating levels of branched-chain amino (BCAAs) and keto (BCKAs) acids, metabolites associated with development of systemic insulin resistance (IR) and T2D. Importantly, these systemic effects were found to be coupled to increased expression of BCAA catabolic genes in thermogenic brown adipose tissue (BAT) in mice. These findings led us to hypothesize that tirzepatide may lower circulating BCAAs/BCKAs by promoting their catabolism in BAT. Methods To address this question, we utilized a murine model of diet-induced obesity and employed stable-isotope tracer studies in combination with metabolomic analyses in BAT and other tissues. Results Treatment with tirzepatide stimulated catabolism of BCAAs/BCKAs in BAT, as demonstrated by increased labeling of BCKA-derived metabolites, and increases in levels of byproducts of BCAA breakdown, including glutamate, alanine, and 3-hydroxyisobutyric acid (3-HIB). Further, chronic administration of tirzepatide increased levels of multiple amino acids in BAT that have previously been shown to be elevated in response to cold exposure. Finally, chronic treatment with tirzepatide led to a substantial increase in several TCA cycle intermediates (α-ketoglutarate, fumarate, and malate) in BAT. Conclusions These findings suggest that tirzepatide induces a thermogenic-like amino acid profile in BAT, an effect that may account for reduced systemic levels of BCAAs in obese IR mice. Tirzepatide augments the catabolism of BCAA in brown adipose tissue (BAT) of obese mice. Tirzepatide promotes BCAA catabolism in BAT, despite its lower potency to activate the mouse GIPR relative to mouse GIP. Tirzepatide increases amino acids and TCA cycle intermediates in BAT, as also observed in BAT thermogenesis.
Collapse
|
16
|
The Regulation and Characterization of Mitochondrial-Derived Methylmalonic Acid in Mitochondrial Dysfunction and Oxidative Stress: From Basic Research to Clinical Practice. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:7043883. [PMID: 35656023 PMCID: PMC9155905 DOI: 10.1155/2022/7043883] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 04/16/2022] [Accepted: 04/23/2022] [Indexed: 01/11/2023]
Abstract
Methylmalonic acid (MMA) can act as a diagnosis of hereditary methylmalonic acidemia and assess the status of vitamin B12. Moreover, as a new potential biomarker, it has been widely reported to be associated with the progression and prognosis of chronic diseases such as cardiovascular events, renal insufficiency, cognitive impairment, and cancer. MMA accumulation may cause oxidative stress and impair mitochondrial function, disrupt cellular energy metabolism, and trigger cell death. This review primarily focuses on the mechanisms and epidemiology or progression in the clinical study on MMA.
Collapse
|
17
|
Shen L, Yu Y, Zhou Y, Pruett-Miller SM, Zhang GF, Karner CM. SLC38A2 provides proline to fulfil unique synthetic demands arising during osteoblast differentiation and bone formation. eLife 2022; 11:76963. [PMID: 35261338 PMCID: PMC9007586 DOI: 10.7554/elife.76963] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 03/08/2022] [Indexed: 11/13/2022] Open
Abstract
Cellular differentiation is associated with the acquisition of a unique protein signature which is essential to attain the ultimate cellular function and activity of the differentiated cell. This is predicted to result in unique biosynthetic demands that arise during differentiation. Using a bioinformatic approach, we discovered osteoblast differentiation is associated with increased demand for the amino acid proline. When compared to other differentiated cells, osteoblast-associated proteins including RUNX2, OSX, OCN and COL1A1 are significantly enriched in proline. Using a genetic and metabolomic approach, we demonstrate that the neutral amino acid transporter SLC38A2 acts cell autonomously to provide proline to facilitate the efficient synthesis of proline-rich osteoblast proteins. Genetic ablation of SLC38A2 in osteoblasts limits both osteoblast differentiation and bone formation in mice. Mechanistically, proline is primarily incorporated into nascent protein with little metabolism observed. Collectively, these data highlight a requirement for proline in fulfilling the unique biosynthetic requirements that arise during osteoblast differentiation and bone formation.
Collapse
Affiliation(s)
- Leyao Shen
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, United States
| | - Yilin Yu
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, United States
| | - Yunji Zhou
- Department of Biostatistics and Bioinformatics, Duke University, Durham, United States
| | - Shondra M Pruett-Miller
- Department of Cell and Molecular Biology, St Jude Children's Research Hospital, Memphis, United States
| | - Guo-Fang Zhang
- Sarah W Stedman Nutrition and Metabolism Center, Duke University Medical Center, Durham, United States
| | - Courtney M Karner
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, United States
| |
Collapse
|
18
|
Liu Y, Yang L, Shuai R, Huang S, Zhang B, Han L, Sun K, Wu Y. Different Pattern of Cardiovascular Impairment in Methylmalonic Acidaemia Subtypes. Front Pediatr 2022; 10:810495. [PMID: 35281223 PMCID: PMC8904414 DOI: 10.3389/fped.2022.810495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Accepted: 01/06/2022] [Indexed: 11/26/2022] Open
Abstract
Methylmalonic acidaemia (MMA) has been reported to be associated with cardiovascular involvement, especially for the combined type with homocystinuria. We have screened 80 control subjects and 99 MMA patients (23 isolated type and 76 combined type) using electrocardiograph and echocardiography. 32 cases (34%) of ECG changes were found including sinus tachycardia (n = 11), prolonged QTc interval (n = 1), I-degree atrioventricular block (n = 1), left axis deviation (n = 5) and T wave change (n = 14). By echocardiography, 8 cases of congenital heart disease were found in 4 combined MMA patients (5.3%) including ventricular septal defect (n = 2), atrial septal defect (n = 3), patent ductus arteriosus (n = 1) and coronary artery-pulmonary artery fistula (n =2). Pulmonary hypertension (n = 2) and hypertrophic cardiomyopathy (n = 1) in combined subtype were also noted. Moreover, echocardiographic parameters were analyzed by multiple regression to clarify the influence of different subtypes on cardiac function. It was found that the left ventricular mass index (LVMI) was significantly reduced only in combined subtype [R = -3.0, 95%CI (-5.4, -0.5), P = 0.017]. For left ventricle, the mitral E' velocity was significantly reduced [isolated type: R = -1.8, 95%CI (-3.3, -0.4), P = 0.016; combined type: R = -2.5, 95%CI (-3.5, -1.5), P < 0.001], the global longitudinal strain (GLS) was the same [isolated type: R = -1.4, 95%CI (-2.3, -0.4), P = 0.007; Combined type: R = -1.1, 95%CI (-1.8, -0.4), P = 0.001], suggesting weakened left ventricular diastolic and systolic functions in both subtypes. For right ventricle, only in combined subtype, the tricuspid E' velocity was significantly reduced [R = -1.4, 95%CI (-2.6, -0.2), P = 0.021], and the tricuspid annular plane systolic excursion (TAPSE) was the same [R = -1.3, 95%CI (-2.3, -0.3), P=0.013], suggesting impaired right ventricular systolic and diastolic function. In conclusion, isolated and combined types showed different pattern of cardiac dysfunction, specifically the former only affected the left ventricle while the latter affected both ventricles. And it is necessary to perform echocardiographic screening and follow up in both MMA subtypes.
Collapse
Affiliation(s)
- Ying Liu
- Department of Pediatric Cardiology, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Ling Yang
- Department of Pediatric Cardiology, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Ruixue Shuai
- Department of Pediatrics, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Suqiu Huang
- Department of Pediatric Cardiology, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Bingyao Zhang
- Department of Pediatric Cardiology, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Lianshu Han
- Department of Pediatric Genetic Endocrinology, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Kun Sun
- Department of Pediatric Cardiology, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yurong Wu
- Department of Pediatric Cardiology, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
19
|
Karlstaedt A. Stable Isotopes for Tracing Cardiac Metabolism in Diseases. Front Cardiovasc Med 2021; 8:734364. [PMID: 34859064 PMCID: PMC8631909 DOI: 10.3389/fcvm.2021.734364] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 10/18/2021] [Indexed: 12/28/2022] Open
Abstract
Although metabolic remodeling during cardiovascular diseases has been well-recognized for decades, the recent development of analytical platforms and mathematical tools has driven the emergence of assessing cardiac metabolism using tracers. Metabolism is a critical component of cellular functions and adaptation to stress. The pathogenesis of cardiovascular disease involves metabolic adaptation to maintain cardiac contractile function even in advanced disease stages. Stable-isotope tracer measurements are a powerful tool for measuring flux distributions at the whole organism level and assessing metabolic changes at a systems level in vivo. The goal of this review is to summarize techniques and concepts for in vivo or ex vivo stable isotope labeling in cardiovascular research, to highlight mathematical concepts and their limitations, to describe analytical methods at the tissue and single-cell level, and to discuss opportunities to leverage metabolic models to address important mechanistic questions relevant to all patients with cardiovascular disease.
Collapse
Affiliation(s)
- Anja Karlstaedt
- Department of Cardiology, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States.,Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| |
Collapse
|
20
|
Jankauskas SS, Kansakar U, Varzideh F, Wilson S, Mone P, Lombardi A, Gambardella J, Santulli G. Heart failure in diabetes. Metabolism 2021; 125:154910. [PMID: 34627874 PMCID: PMC8941799 DOI: 10.1016/j.metabol.2021.154910] [Citation(s) in RCA: 97] [Impact Index Per Article: 32.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Revised: 10/02/2021] [Accepted: 10/04/2021] [Indexed: 12/16/2022]
Abstract
Heart failure and cardiovascular disorders represent the leading cause of death in diabetic patients. Here we present a systematic review of the main mechanisms underlying the development of diabetic cardiomyopathy. We also provide an excursus on the relative contribution of cardiomyocytes, fibroblasts, endothelial and smooth muscle cells to the pathophysiology of heart failure in diabetes. After having described the preclinical tools currently available to dissect the mechanisms of this complex disease, we conclude with a section on the most recent updates of the literature on clinical management.
Collapse
Affiliation(s)
- Stanislovas S Jankauskas
- Department of Medicine, Fleischer Institute for Diabetes and Metabolism (FIDAM), Einstein-Mount Sinai Diabetes Research Center (ES-DRC), Albert Einstein College of Medicine, New York, NY 10461, USA; Department of Molecular Pharmacology, Einstein Institute for Neuroimmunology and Inflammation, Wilf Family Cardiovascular Research Institute, Einstein Institute for Aging Research, Albert Einstein College of Medicine, New York, NY 10461, USA
| | - Urna Kansakar
- Department of Medicine, Fleischer Institute for Diabetes and Metabolism (FIDAM), Einstein-Mount Sinai Diabetes Research Center (ES-DRC), Albert Einstein College of Medicine, New York, NY 10461, USA; Department of Molecular Pharmacology, Einstein Institute for Neuroimmunology and Inflammation, Wilf Family Cardiovascular Research Institute, Einstein Institute for Aging Research, Albert Einstein College of Medicine, New York, NY 10461, USA
| | - Fahimeh Varzideh
- Department of Medicine, Fleischer Institute for Diabetes and Metabolism (FIDAM), Einstein-Mount Sinai Diabetes Research Center (ES-DRC), Albert Einstein College of Medicine, New York, NY 10461, USA; Department of Molecular Pharmacology, Einstein Institute for Neuroimmunology and Inflammation, Wilf Family Cardiovascular Research Institute, Einstein Institute for Aging Research, Albert Einstein College of Medicine, New York, NY 10461, USA
| | - Scott Wilson
- Department of Medicine, Fleischer Institute for Diabetes and Metabolism (FIDAM), Einstein-Mount Sinai Diabetes Research Center (ES-DRC), Albert Einstein College of Medicine, New York, NY 10461, USA
| | - Pasquale Mone
- Department of Medicine, Fleischer Institute for Diabetes and Metabolism (FIDAM), Einstein-Mount Sinai Diabetes Research Center (ES-DRC), Albert Einstein College of Medicine, New York, NY 10461, USA
| | - Angela Lombardi
- Department of Medicine, Fleischer Institute for Diabetes and Metabolism (FIDAM), Einstein-Mount Sinai Diabetes Research Center (ES-DRC), Albert Einstein College of Medicine, New York, NY 10461, USA
| | - Jessica Gambardella
- Department of Medicine, Fleischer Institute for Diabetes and Metabolism (FIDAM), Einstein-Mount Sinai Diabetes Research Center (ES-DRC), Albert Einstein College of Medicine, New York, NY 10461, USA; Department of Molecular Pharmacology, Einstein Institute for Neuroimmunology and Inflammation, Wilf Family Cardiovascular Research Institute, Einstein Institute for Aging Research, Albert Einstein College of Medicine, New York, NY 10461, USA; International Translational Research and Medical Education (ITME), Department of Advanced Biomedical Science, "Federico II" University, 80131 Naples, Italy
| | - Gaetano Santulli
- Department of Medicine, Fleischer Institute for Diabetes and Metabolism (FIDAM), Einstein-Mount Sinai Diabetes Research Center (ES-DRC), Albert Einstein College of Medicine, New York, NY 10461, USA; Department of Molecular Pharmacology, Einstein Institute for Neuroimmunology and Inflammation, Wilf Family Cardiovascular Research Institute, Einstein Institute for Aging Research, Albert Einstein College of Medicine, New York, NY 10461, USA; International Translational Research and Medical Education (ITME), Department of Advanced Biomedical Science, "Federico II" University, 80131 Naples, Italy.
| |
Collapse
|
21
|
He W, Wang Y, Xie EJ, Barry MA, Zhang GF. Metabolic perturbations mediated by propionyl-CoA accumulation in organs of mouse model of propionic acidemia. Mol Genet Metab 2021; 134:257-266. [PMID: 34635437 DOI: 10.1016/j.ymgme.2021.09.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2021] [Revised: 09/28/2021] [Accepted: 09/29/2021] [Indexed: 12/18/2022]
Abstract
Propionic acidemia (PA) is an autosomal recessive metabolic disorder after gene encoding propionyl-CoA carboxylase, Pcca or Pccb, is mutated. This genetic disorder could develop various complications which are ascribed to dysregulated propionyl-CoA metabolism in organs. However, the effect of attenuated PCC on propionyl-CoA metabolism in different organs remains to be fully understood. We investigated metabolic perturbations in organs of Pcca-/-(A138T) mice (a mouse model of PA) under chow diet and acute administration of [13C3]propionate to gain insight into pathological mechanisms of PA. With chow diet, the metabolic alteration is organ dependent. l-Carnitine reduction induced by propionylcarnitine accumulation only occurs in lung and liver of Pcca-/- (A138T) mice. [13C3]Propionate tracing data demonstrated that PCC activity was dramatically reduced in Pcca-/-(A138T) brain, lung, liver, kidney, and adipose tissues, but not significantly changed in Pcca-/-(A138T) muscles (heart and skeletal muscles) and pancreas, which was largely supported by PCCA expression data. The largest expansion of propionylcarnitine in Pcca-/-(A138T) heart after acute administration of propionate indicated the vulnerability of heart to high circulating propionate. The overwhelming propionate in blood also stimulated ketone production from the increased fatty acid oxidation in Pcca-/-(A138T) liver by lowering malonyl-CoA, which has been observed in cases where metabolic decompensation occurs in PA patients. This work shed light on organ-specific metabolic alternations under varying severities of PA.
Collapse
Affiliation(s)
- Wentao He
- Duke Molecular Physiology Institute and Sarah W. Stedman Nutrition and Metabolism Center, Duke University Medical Center, Durham, NC 27701, USA
| | - You Wang
- School of Basic Medicine, Jining Medical University, Shandong 272067, China
| | - Erik J Xie
- Duke Molecular Physiology Institute and Sarah W. Stedman Nutrition and Metabolism Center, Duke University Medical Center, Durham, NC 27701, USA
| | - Michael A Barry
- Department of Medicine, Division of Infectious Diseases, Mayo Clinic, Rochester, MN 55905, USA
| | - Guo-Fang Zhang
- Duke Molecular Physiology Institute and Sarah W. Stedman Nutrition and Metabolism Center, Duke University Medical Center, Durham, NC 27701, USA; Department of Medicine, Division of Endocrinology, Metabolism Nutrition, Duke University Medical Center, Durham, NC 27701, USA.
| |
Collapse
|
22
|
Subramanian C, Frank MW, Tangallapally R, Yun MK, Edwards A, White SW, Lee RE, Rock CO, Jackowski S. Pantothenate kinase activation relieves coenzyme A sequestration and improves mitochondrial function in mice with propionic acidemia. Sci Transl Med 2021; 13:eabf5965. [PMID: 34524863 DOI: 10.1126/scitranslmed.abf5965] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Chitra Subramanian
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Matthew W Frank
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Rajendra Tangallapally
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Mi-Kyung Yun
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis TN, 38105, USA
| | - Anne Edwards
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Stephen W White
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis TN, 38105, USA.,St. Jude Graduate School of Biomedical Sciences, St. Jude Children's Research Hospital, Memphis, TN 38105, USA.,Department of Microbiology, Immunology and Biochemistry, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Richard E Lee
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, TN 38105, USA.,Center for Pediatric Experimental Therapeutics, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Charles O Rock
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, TN 38105, USA.,Department of Microbiology, Immunology and Biochemistry, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Suzanne Jackowski
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| |
Collapse
|
23
|
Zhang Z, Liu S, Ma H, Xiang X, Nie D, Hu P, Tang G. Propionic Acid-Based PET Imaging of Prostate Cancer. Mol Imaging Biol 2021; 23:836-845. [PMID: 33876336 DOI: 10.1007/s11307-021-01608-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 04/08/2021] [Accepted: 04/12/2021] [Indexed: 10/21/2022]
Abstract
PURPOSE This study aimed to evaluate the potential value of 2-[18F]fluoropropionic acid ([18F]FPA) for PET imaging of prostate cancer (PCa) and to explore the relationship between [18F]FPA accumulation and fatty acid synthase (FASN) levels in PCa models. The results of the first [18F]FPA PET study of a PCa patient are reported. PROCEDURES The LNCaP, PC-3 cell lines with high FASN expression, and DU145 cell lines with low FASN expression were selected for cell culture. A PET imaging comparison of [18F]FDG and [18F]FPA was performed in LNCaP, PC-3, and DU145 tumors. Additionally, in vivo inhibition experiments in those models were conducted with orlistat. In a human PET study, a patient with PCa before surgery was examined with [18F]FPA PET and [18F]FDG PET. RESULTS The uptake of [18F]FPA in the LNCaP and PC-3 tumors was higher than that of [18F]FDG (P<0.05 and P<0.05), but was lower in DU145 tumors (P<0.05). The accumulation (% ID/g) of [18F]FPA in the LNCaP, PC-3, and DU145 tumors decreased by 27.6, 40.5, and 11.7 %, respectively, after treatment with orlistat. The [18F]FPA showed higher radioactive uptake than [18F]FDG in the first PCa patient. CONCLUSIONS The [18F]FPA uptake in PCa models may be varies with fatty acid synthase activity and could be reduced after administration of a single FASN inhibitor, albeit the activity that is not measured directly. The [18F]FPA seems to be a potential broad-spectrum PET imaging agent and may serve as a valuable tool in the diagnosis of PCa in humans.
Collapse
Affiliation(s)
- Zhanwen Zhang
- Department of Nuclear Medicine, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, China.,Department of Nuclear Medicine and Medical Imaging, Guangdong Engineering Research Center for Translational Application of Medical Radiopharmaceuticals, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
| | - Shaoyu Liu
- Department of Nuclear Medicine and Medical Imaging, Guangdong Engineering Research Center for Translational Application of Medical Radiopharmaceuticals, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China.,Department of Nuclear Medicine, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China
| | - Hui Ma
- Department of Nuclear Medicine and Medical Imaging, Guangdong Engineering Research Center for Translational Application of Medical Radiopharmaceuticals, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China.,Department of Radiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
| | - Xianhong Xiang
- Department of Nuclear Medicine and Medical Imaging, Guangdong Engineering Research Center for Translational Application of Medical Radiopharmaceuticals, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
| | - Dahong Nie
- Department of Nuclear Medicine and Medical Imaging, Guangdong Engineering Research Center for Translational Application of Medical Radiopharmaceuticals, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
| | - Ping Hu
- Department of Nuclear Medicine, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, China.
| | - Ganghua Tang
- Department of Nuclear Medicine and Medical Imaging, Guangdong Engineering Research Center for Translational Application of Medical Radiopharmaceuticals, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China. .,Nanfang PET Center and Department of Nuclear Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
| |
Collapse
|
24
|
Zhang GF, Jensen MV, Gray SM, El K, Wang Y, Lu D, Becker TC, Campbell JE, Newgard CB. Reductive TCA cycle metabolism fuels glutamine- and glucose-stimulated insulin secretion. Cell Metab 2021; 33:804-817.e5. [PMID: 33321098 PMCID: PMC8115731 DOI: 10.1016/j.cmet.2020.11.020] [Citation(s) in RCA: 82] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Revised: 11/06/2020] [Accepted: 11/25/2020] [Indexed: 12/11/2022]
Abstract
Metabolic fuels regulate insulin secretion by generating second messengers that drive insulin granule exocytosis, but the biochemical pathways involved are incompletely understood. Here we demonstrate that stimulation of rat insulinoma cells or primary rat islets with glucose or glutamine + 2-aminobicyclo-(2,2,1)-heptane-2-carboxylic acid (Gln + BCH) induces reductive, "counter-clockwise" tricarboxylic acid (TCA) cycle flux of glutamine to citrate. Molecular or pharmacologic suppression of isocitrate dehydrogenase-2 (IDH2), which catalyzes reductive carboxylation of 2-ketoglutarate to isocitrate, results in impairment of glucose- and Gln + BCH-stimulated reductive TCA cycle flux, lowering of NADPH levels, and inhibition of insulin secretion. Pharmacologic suppression of IDH2 also inhibits insulin secretion in living mice. Reductive TCA cycle flux has been proposed as a mechanism for generation of biomass in cancer cells. Here we demonstrate that reductive TCA cycle flux also produces stimulus-secretion coupling factors that regulate insulin secretion, including in non-dividing cells.
Collapse
Affiliation(s)
- Guo-Fang Zhang
- Sarah W. Stedman Nutrition and Metabolism Center and Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC 27701, USA; Department of Medicine, Endocrinology and Metabolism Division, Duke University Medical Center, Durham, NC 27701, USA
| | - Mette V Jensen
- Sarah W. Stedman Nutrition and Metabolism Center and Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC 27701, USA
| | - Sarah M Gray
- Sarah W. Stedman Nutrition and Metabolism Center and Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC 27701, USA
| | - Kimberley El
- Sarah W. Stedman Nutrition and Metabolism Center and Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC 27701, USA
| | - You Wang
- Sarah W. Stedman Nutrition and Metabolism Center and Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC 27701, USA
| | - Danhong Lu
- Sarah W. Stedman Nutrition and Metabolism Center and Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC 27701, USA
| | - Thomas C Becker
- Sarah W. Stedman Nutrition and Metabolism Center and Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC 27701, USA; Department of Medicine, Endocrinology and Metabolism Division, Duke University Medical Center, Durham, NC 27701, USA
| | - Jonathan E Campbell
- Sarah W. Stedman Nutrition and Metabolism Center and Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC 27701, USA; Department of Medicine, Endocrinology and Metabolism Division, Duke University Medical Center, Durham, NC 27701, USA; Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC 27701, USA
| | - Christopher B Newgard
- Sarah W. Stedman Nutrition and Metabolism Center and Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC 27701, USA; Department of Medicine, Endocrinology and Metabolism Division, Duke University Medical Center, Durham, NC 27701, USA; Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC 27701, USA.
| |
Collapse
|
25
|
Pathmanapan S, Ilkayeva O, Martin JT, Loe AKH, Zhang H, Zhang GF, Newgard CB, Wunder JS, Alman BA. Mutant IDH and non-mutant chondrosarcomas display distinct cellular metabolomes. Cancer Metab 2021; 9:13. [PMID: 33762012 PMCID: PMC7992867 DOI: 10.1186/s40170-021-00247-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 03/03/2021] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND Majority of chondrosarcomas are associated with a number of genetic alterations, including somatic mutations in isocitrate dehydrogenase 1 (IDH1) and IDH2 genes, but the downstream effects of these mutated enzymes on cellular metabolism and tumor energetics are unknown. As IDH mutations are likely to be involved in malignant transformation of chondrosarcomas, we aimed to exploit metabolomic changes in IDH mutant and non-mutant chondrosarcomas. METHODS Here, we profiled over 69 metabolites in 17 patient-derived xenografts by targeted mass spectrometry to determine if metabolomic differences exist in mutant IDH1, mutant IDH2, and non-mutant chondrosarcomas. UMAP (Uniform Manifold Approximation and Projection) analysis was performed on our dataset to examine potential similarities that may exist between each chondrosarcoma based on genotype. RESULTS UMAP revealed that mutant IDH chondrosarcomas possess a distinct metabolic profile compared with non-mutant chondrosarcomas. More specifically, our targeted metabolomics study revealed large-scale differences in organic acid intermediates of the tricarboxylic acid (TCA) cycle, amino acids, and specific acylcarnitines in chondrosarcomas. Lactate and late TCA cycle intermediates were elevated in mutant IDH chondrosarcomas, suggestive of increased glycolytic metabolism and possible anaplerotic influx to the TCA cycle. A broad elevation of amino acids was found in mutant IDH chondrosarcomas. A few acylcarnitines of varying carbon chain lengths were also elevated in mutant IDH chondrosarcomas, but with minimal clustering in accordance with tumor genotype. Analysis of previously published gene expression profiling revealed increased expression of several metabolism genes in mutant IDH chondrosarcomas, which also correlated to patient survival. CONCLUSIONS Overall, our findings suggest that IDH mutations induce global metabolic changes in chondrosarcomas and shed light on deranged metabolic pathways.
Collapse
Affiliation(s)
- Sinthu Pathmanapan
- Developmental and Stem Cell Biology, Hospital for Sick Children, Toronto, ON, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| | - Olga Ilkayeva
- Department of Pharmacology & Cancer Biology, Duke University, Durham, NC, USA
- Sarah W. Stedman Nutrition and Metabolism Center and Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC, USA
| | - John T Martin
- Department of Orthopaedic Surgery, Duke University, 311 Trent, Durham, NC, 27710, USA
| | - Adrian Kwan Ho Loe
- Developmental and Stem Cell Biology, Hospital for Sick Children, Toronto, ON, Canada
| | - Hongyuan Zhang
- Department of Orthopaedic Surgery, Duke University, 311 Trent, Durham, NC, 27710, USA
| | - Guo-Fang Zhang
- Sarah W. Stedman Nutrition and Metabolism Center and Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC, USA
| | - Christopher B Newgard
- Department of Pharmacology & Cancer Biology, Duke University, Durham, NC, USA
- Sarah W. Stedman Nutrition and Metabolism Center and Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC, USA
| | - Jay S Wunder
- Lunenfeld-Tanenbaum Research Institute, and the University Musculoskeletal Oncology Unit, Mount Sinai Hospital, Toronto, ON, Canada
| | - Benjamin A Alman
- Department of Orthopaedic Surgery, Duke University, 311 Trent, Durham, NC, 27710, USA.
| |
Collapse
|
26
|
Walejko JM, Christopher BA, Crown SB, Zhang GF, Pickar-Oliver A, Yoneshiro T, Foster MW, Page S, van Vliet S, Ilkayeva O, Muehlbauer MJ, Carson MW, Brozinick JT, Hammond CD, Gimeno RE, Moseley MA, Kajimura S, Gersbach CA, Newgard CB, White PJ, McGarrah RW. Branched-chain α-ketoacids are preferentially reaminated and activate protein synthesis in the heart. Nat Commun 2021; 12:1680. [PMID: 33723250 PMCID: PMC7960706 DOI: 10.1038/s41467-021-21962-2] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 02/18/2021] [Indexed: 12/20/2022] Open
Abstract
Branched-chain amino acids (BCAA) and their cognate α-ketoacids (BCKA) are elevated in an array of cardiometabolic diseases. Here we demonstrate that the major metabolic fate of uniformly-13C-labeled α-ketoisovalerate ([U-13C]KIV) in the heart is reamination to valine. Activation of cardiac branched-chain α-ketoacid dehydrogenase (BCKDH) by treatment with the BCKDH kinase inhibitor, BT2, does not impede the strong flux of [U-13C]KIV to valine. Sequestration of BCAA and BCKA away from mitochondrial oxidation is likely due to low levels of expression of the mitochondrial BCAA transporter SLC25A44 in the heart, as its overexpression significantly lowers accumulation of [13C]-labeled valine from [U-13C]KIV. Finally, exposure of perfused hearts to levels of BCKA found in obese rats increases phosphorylation of the translational repressor 4E-BP1 as well as multiple proteins in the MEK-ERK pathway, leading to a doubling of total protein synthesis. These data suggest that elevated BCKA levels found in obesity may contribute to pathologic cardiac hypertrophy via chronic activation of protein synthesis. Systemic modulation of branched-chain keto acid (BCKA) metabolism alters cardiac health. Here, the authors define the major fates of BCKA in the heart and demonstrate that acute exposure to BCKA levels found in obesity activates cardiac protein synthesis and markedly alters the heart phosphoproteome.
Collapse
Affiliation(s)
- Jacquelyn M Walejko
- Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, NC, USA
| | - Bridgette A Christopher
- Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, NC, USA.,Department of Medicine, Division of Cardiology, Duke University School of Medicine, Durham, NC, USA
| | - Scott B Crown
- Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, NC, USA
| | - Guo-Fang Zhang
- Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, NC, USA.,Department of Medicine, Division of Endocrinology, Metabolism and Nutrition, Duke University School of Medicine, Durham, NC, USA.,Sarah W. Stedman Nutrition and Metabolism Center, Duke University School of Medicine, Durham, NC, USA
| | - Adrian Pickar-Oliver
- Department of Biomedical Engineering, Duke University, Durham, NC, USA.,Center for Advanced Genomic Technologies, Duke University, Durham, NC, USA
| | | | - Matthew W Foster
- Duke Proteomics and Metabolomics Shared Resource, Duke University School of Medicine, Durham, NC, USA
| | - Stephani Page
- Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, NC, USA
| | - Stephan van Vliet
- Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, NC, USA
| | - Olga Ilkayeva
- Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, NC, USA.,Department of Medicine, Division of Endocrinology, Metabolism and Nutrition, Duke University School of Medicine, Durham, NC, USA.,Sarah W. Stedman Nutrition and Metabolism Center, Duke University School of Medicine, Durham, NC, USA
| | - Michael J Muehlbauer
- Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, NC, USA.,Sarah W. Stedman Nutrition and Metabolism Center, Duke University School of Medicine, Durham, NC, USA
| | | | | | | | | | - M Arthur Moseley
- Duke Proteomics and Metabolomics Shared Resource, Duke University School of Medicine, Durham, NC, USA
| | | | - Charles A Gersbach
- Department of Biomedical Engineering, Duke University, Durham, NC, USA.,Center for Advanced Genomic Technologies, Duke University, Durham, NC, USA.,Department of Surgery, Duke University School of Medicine, Durham, NC, USA
| | - Christopher B Newgard
- Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, NC, USA.,Department of Medicine, Division of Endocrinology, Metabolism and Nutrition, Duke University School of Medicine, Durham, NC, USA.,Sarah W. Stedman Nutrition and Metabolism Center, Duke University School of Medicine, Durham, NC, USA.,Department of Pharmacology and Cancer Biology, Duke University, Durham, NC, USA
| | - Phillip J White
- Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, NC, USA. .,Department of Medicine, Division of Endocrinology, Metabolism and Nutrition, Duke University School of Medicine, Durham, NC, USA. .,Sarah W. Stedman Nutrition and Metabolism Center, Duke University School of Medicine, Durham, NC, USA. .,Department of Pharmacology and Cancer Biology, Duke University, Durham, NC, USA.
| | - Robert W McGarrah
- Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, NC, USA. .,Department of Medicine, Division of Cardiology, Duke University School of Medicine, Durham, NC, USA. .,Sarah W. Stedman Nutrition and Metabolism Center, Duke University School of Medicine, Durham, NC, USA.
| |
Collapse
|
27
|
Lagerwaard B, Pougovkina O, Bekebrede AF, te Brinke H, Wanders RJ, Nieuwenhuizen AG, Keijer J, de Boer VCJ. Increased protein propionylation contributes to mitochondrial dysfunction in liver cells and fibroblasts, but not in myotubes. J Inherit Metab Dis 2021; 44:438-449. [PMID: 32740932 PMCID: PMC8049071 DOI: 10.1002/jimd.12296] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 07/14/2020] [Accepted: 07/30/2020] [Indexed: 12/22/2022]
Abstract
Post-translational protein modifications derived from metabolic intermediates, such as acyl-CoAs, have been shown to regulate mitochondrial function. Patients with a genetic defect in the propionyl-CoA carboxylase (PCC) gene clinically present symptoms related to mitochondrial disorders and are characterised by decreased mitochondrial respiration. Since propionyl-CoA accumulates in PCC deficient patients and protein propionylation can be driven by the level of propionyl-CoA, we hypothesised that protein propionylation could play a role in the pathology of the disease. Indeed, we identified increased protein propionylation due to pathologic propionyl-CoA accumulation in patient-derived fibroblasts and this was accompanied by defective mitochondrial respiration, as was shown by a decrease in complex I-driven respiration. To mimic pathological protein propionylation levels, we exposed cultured fibroblasts, Fao liver cells and C2C12 muscle myotubes to propionate levels that are typically found in these patients. This induced a global increase in protein propionylation and histone protein propionylation and was also accompanied by a decrease in mitochondrial respiration in liver and fibroblasts. However, in C2C12 myotubes propionate exposure did not decrease mitochondrial respiration, possibly due to differences in propionyl-CoA metabolism as compared to the liver. Therefore, protein propionylation could contribute to the pathology in these patients, especially in the liver, and could therefore be an interesting target to pursue in the treatment of this metabolic disease.
Collapse
Affiliation(s)
- Bart Lagerwaard
- Human and Animal Physiology, Wageningen University and ResearchWageningenNetherlands
- TI Food and NutritionWageningenNetherlands
| | - Olga Pougovkina
- Laboratory Genetic Metabolic Diseases, Department of Clinical ChemistryAcademic Medical Center, University of AmsterdamAmsterdamNetherlands
| | - Anna F. Bekebrede
- Human and Animal Physiology, Wageningen University and ResearchWageningenNetherlands
| | - Heleen te Brinke
- Laboratory Genetic Metabolic Diseases, Department of Clinical ChemistryAcademic Medical Center, University of AmsterdamAmsterdamNetherlands
| | - Ronald J.A. Wanders
- Laboratory Genetic Metabolic Diseases, Department of Clinical ChemistryAcademic Medical Center, University of AmsterdamAmsterdamNetherlands
- Department of PediatricsEmma Children's Hospital, Academic Medical Center, University of AmsterdamAmsterdamNetherlands
| | - Arie G. Nieuwenhuizen
- Human and Animal Physiology, Wageningen University and ResearchWageningenNetherlands
| | - Jaap Keijer
- Human and Animal Physiology, Wageningen University and ResearchWageningenNetherlands
| | - Vincent C. J. de Boer
- Human and Animal Physiology, Wageningen University and ResearchWageningenNetherlands
- Laboratory Genetic Metabolic Diseases, Department of Clinical ChemistryAcademic Medical Center, University of AmsterdamAmsterdamNetherlands
- Department of PediatricsEmma Children's Hospital, Academic Medical Center, University of AmsterdamAmsterdamNetherlands
| |
Collapse
|
28
|
Park KC, Krywawych S, Richard E, Desviat LR, Swietach P. Cardiac Complications of Propionic and Other Inherited Organic Acidemias. Front Cardiovasc Med 2020; 7:617451. [PMID: 33415129 PMCID: PMC7782273 DOI: 10.3389/fcvm.2020.617451] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Accepted: 11/24/2020] [Indexed: 12/12/2022] Open
Abstract
Clinical observations and experimental studies have determined that systemic acid-base disturbances can profoundly affect the heart. A wealth of information is available on the effects of altered pH on cardiac function but, by comparison, much less is known about the actions of the organic anions that accumulate alongside H+ ions in acidosis. In the blood and other body fluids, these organic chemical species can collectively reach concentrations of several millimolar in severe metabolic acidoses, as in the case of inherited organic acidemias, and exert powerful biological actions on the heart that are not intuitive to predict. Indeed, cardiac pathologies, such as cardiomyopathy and arrhythmia, are frequently reported in organic acidemia patients, but the underlying pathophysiological mechanisms are not well established. Research efforts in the area of organic anion physiology have increased dramatically in recent years, particularly for propionate, which accumulates in propionic acidemia, one of the commonest organic acidemias characterized by a high incidence of cardiac disease. This Review provides a comprehensive historical overview of all known organic acidemias that feature cardiac complications and a state-of-the-art overview of the cardiac sequelae observed in propionic acidemia. The article identifies the most promising candidates for molecular mechanisms that become aberrantly engaged by propionate anions (and its metabolites), and discusses how these may result in cardiac derangements in propionic acidemia. Key clinical and experimental findings are considered in the context of potential therapies in the near future.
Collapse
Affiliation(s)
- Kyung Chan Park
- Department of Anatomy, Physiology and Genetics, Burdon Sanderson Cardiac Science Centre, British Heart Foundation Centre of Research Excellence, University of Oxford, Oxford, United Kingdom
| | - Steve Krywawych
- Department of Chemical Pathology, Great Ormond Street Hospital, London, United Kingdom
| | - Eva Richard
- Centro de Biología Molecular Severo Ochoa, Universidad Autonoma de Madrid-Consejo Superior de Investigaciones Cientificas (UAM-CSIC), Centro de Investigacion Biomedica en Red de Enfermedades Raras (CIBERER), IdiPaz, Universidad Autónoma de Madrid, Madrid, Spain
| | - Lourdes R Desviat
- Centro de Biología Molecular Severo Ochoa, Universidad Autonoma de Madrid-Consejo Superior de Investigaciones Cientificas (UAM-CSIC), Centro de Investigacion Biomedica en Red de Enfermedades Raras (CIBERER), IdiPaz, Universidad Autónoma de Madrid, Madrid, Spain
| | - Pawel Swietach
- Department of Anatomy, Physiology and Genetics, Burdon Sanderson Cardiac Science Centre, British Heart Foundation Centre of Research Excellence, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
29
|
Fulgencio-Covián A, Alonso-Barroso E, Guenzel AJ, Rivera-Barahona A, Ugarte M, Pérez B, Barry MA, Pérez-Cerdá C, Richard E, Desviat LR. Pathogenic implications of dysregulated miRNAs in propionic acidemia related cardiomyopathy. Transl Res 2020; 218:43-56. [PMID: 31951825 DOI: 10.1016/j.trsl.2019.12.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Revised: 12/03/2019] [Accepted: 12/23/2019] [Indexed: 12/25/2022]
Abstract
Cardiac alterations (hypertrophic/dilated cardiomyopathy, and rhythm alterations) are one of the major causes of mortality and morbidity in propionic acidemia (PA), caused by the deficiency of the mitochondrial enzyme propionyl-CoA carboxylase (PCC), involved in the catabolism of branched-chain amino acids, cholesterol, and odd-chain fatty acids. Impaired mitochondrial oxidative phosphorylation has been documented in heart biopsies of PA patients, as well as in the hypomorphic Pcca-/-(A138T) mouse model, in the latter correlating with increased oxidative damage and elevated expression of cardiac dysfunction biomarkers atrial and brain natriuretic peptides (ANP and BNP) and beta-myosin heavy chain (β-MHC). Here we characterize the cardiac phenotype in the PA mouse model by histological and echocardiography studies and identify a series of upregulated cardiac-enriched microRNAs (miRNAs) in the PA mouse heart, some of them also altered as circulating miRNAs in PA patients' plasma samples. In PA mice hearts, we show alterations in signaling pathways regulated by the identified miRNAs, which could be contributing to cardiac remodeling and dysfunction; notably, an activation of the mammalian target of rapamycin (mTOR) pathway and a decrease in autophagy, which are reverted by rapamycin treatment. In vitro studies in HL-1 cardiomyocytes indicate that propionate, the major toxic metabolite accumulating in the disease, triggers the increase in expression levels of miRNAs, BNP, and β-MHC, concomitant with an increase in reactive oxygen species. Our results highlight miRNAs and signaling alterations in the PCC-deficient heart which may contribute to the development of PA-associated cardiomyopathy and provide a basis to identify new targets for therapeutic intervention.
Collapse
Affiliation(s)
- Alejandro Fulgencio-Covián
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa CSIC-UAM, Universidad Autónoma de Madrid, Madrid, Spain; Centro de Diagnóstico de Enfermedades Moleculares (CEDEM), Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), ISCIII, Madrid, Spain; Instituto de Investigación Sanitaria Hospital La Paz (IdiPaz), ISCIII, Madrid, Spain
| | - Esmeralda Alonso-Barroso
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa CSIC-UAM, Universidad Autónoma de Madrid, Madrid, Spain; Centro de Diagnóstico de Enfermedades Moleculares (CEDEM), Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), ISCIII, Madrid, Spain; Instituto de Investigación Sanitaria Hospital La Paz (IdiPaz), ISCIII, Madrid, Spain
| | | | - Ana Rivera-Barahona
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa CSIC-UAM, Universidad Autónoma de Madrid, Madrid, Spain; Centro de Diagnóstico de Enfermedades Moleculares (CEDEM), Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), ISCIII, Madrid, Spain; Instituto de Investigación Sanitaria Hospital La Paz (IdiPaz), ISCIII, Madrid, Spain
| | - Magdalena Ugarte
- Centro de Diagnóstico de Enfermedades Moleculares (CEDEM), Madrid, Spain
| | - Belén Pérez
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa CSIC-UAM, Universidad Autónoma de Madrid, Madrid, Spain; Centro de Diagnóstico de Enfermedades Moleculares (CEDEM), Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), ISCIII, Madrid, Spain; Instituto de Investigación Sanitaria Hospital La Paz (IdiPaz), ISCIII, Madrid, Spain
| | | | - Celia Pérez-Cerdá
- Centro de Diagnóstico de Enfermedades Moleculares (CEDEM), Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), ISCIII, Madrid, Spain; Instituto de Investigación Sanitaria Hospital La Paz (IdiPaz), ISCIII, Madrid, Spain
| | - Eva Richard
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa CSIC-UAM, Universidad Autónoma de Madrid, Madrid, Spain; Centro de Diagnóstico de Enfermedades Moleculares (CEDEM), Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), ISCIII, Madrid, Spain; Instituto de Investigación Sanitaria Hospital La Paz (IdiPaz), ISCIII, Madrid, Spain
| | - Lourdes R Desviat
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa CSIC-UAM, Universidad Autónoma de Madrid, Madrid, Spain; Centro de Diagnóstico de Enfermedades Moleculares (CEDEM), Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), ISCIII, Madrid, Spain; Instituto de Investigación Sanitaria Hospital La Paz (IdiPaz), ISCIII, Madrid, Spain.
| |
Collapse
|
30
|
McGarrah RW, Zhang GF, Christopher BA, Deleye Y, Walejko JM, Page S, Ilkayeva O, White PJ, Newgard CB. Dietary branched-chain amino acid restriction alters fuel selection and reduces triglyceride stores in hearts of Zucker fatty rats. Am J Physiol Endocrinol Metab 2020; 318:E216-E223. [PMID: 31794262 PMCID: PMC7052576 DOI: 10.1152/ajpendo.00334.2019] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Elevations in circulating levels of branched-chain amino acids (BCAAs) are associated with a variety of cardiometabolic diseases and conditions. Restriction of dietary BCAAs in rodent models of obesity lowers circulating BCAA levels and improves whole-animal and skeletal-muscle insulin sensitivity and lipid homeostasis, but the impact of BCAA supply on heart metabolism has not been studied. Here, we report that feeding a BCAA-restricted chow diet to Zucker fatty rats (ZFRs) causes a shift in cardiac fuel metabolism that favors fatty acid relative to glucose catabolism. This is illustrated by an increase in labeling of acetyl-CoA from [1-13C]palmitate and a decrease in labeling of acetyl-CoA and malonyl-CoA from [U-13C]glucose, accompanied by a decrease in cardiac hexokinase II and glucose transporter 4 protein levels. Metabolomic profiling of heart tissue supports these findings by demonstrating an increase in levels of a host of fatty-acid-derived metabolites in hearts from ZFRs and Zucker lean rats (ZLRs) fed the BCAA-restricted diet. In addition, the twofold increase in cardiac triglyceride stores in ZFRs compared with ZLRs fed on chow diet is eliminated in ZFRs fed on the BCAA-restricted diet. Finally, the enzymatic activity of branched-chain ketoacid dehydrogenase (BCKDH) is not influenced by BCAA restriction, and levels of BCAA in the heart instead reflect their levels in circulation. In summary, reducing BCAA supply in obesity improves cardiac metabolic health by a mechanism independent of alterations in BCKDH activity.
Collapse
Affiliation(s)
- Robert W McGarrah
- Sarah W. Stedman Nutrition and Metabolism Center, Duke University Medical Center, Durham, North Carolina
- Duke Molecular Physiology Institute, Duke University Medical Center, Durham, North Carolina
- Cardiology Division, Department of Medicine, Duke University Medical Center, Durham, North Carolina
| | - Guo-Fang Zhang
- Sarah W. Stedman Nutrition and Metabolism Center, Duke University Medical Center, Durham, North Carolina
- Duke Molecular Physiology Institute, Duke University Medical Center, Durham, North Carolina
- Endocrinology Division, Department of Medicine, Duke University Medical Center, Durham, North Carolina
| | - Bridgette A Christopher
- Sarah W. Stedman Nutrition and Metabolism Center, Duke University Medical Center, Durham, North Carolina
- Duke Molecular Physiology Institute, Duke University Medical Center, Durham, North Carolina
- Cardiology Division, Department of Medicine, Duke University Medical Center, Durham, North Carolina
| | - Yann Deleye
- Sarah W. Stedman Nutrition and Metabolism Center, Duke University Medical Center, Durham, North Carolina
- Duke Molecular Physiology Institute, Duke University Medical Center, Durham, North Carolina
| | - Jacquelyn M Walejko
- Sarah W. Stedman Nutrition and Metabolism Center, Duke University Medical Center, Durham, North Carolina
- Duke Molecular Physiology Institute, Duke University Medical Center, Durham, North Carolina
| | - Stephani Page
- Sarah W. Stedman Nutrition and Metabolism Center, Duke University Medical Center, Durham, North Carolina
- Duke Molecular Physiology Institute, Duke University Medical Center, Durham, North Carolina
| | - Olga Ilkayeva
- Sarah W. Stedman Nutrition and Metabolism Center, Duke University Medical Center, Durham, North Carolina
- Duke Molecular Physiology Institute, Duke University Medical Center, Durham, North Carolina
| | - Phillip J White
- Sarah W. Stedman Nutrition and Metabolism Center, Duke University Medical Center, Durham, North Carolina
- Duke Molecular Physiology Institute, Duke University Medical Center, Durham, North Carolina
- Endocrinology Division, Department of Medicine, Duke University Medical Center, Durham, North Carolina
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina
| | - Christopher B Newgard
- Sarah W. Stedman Nutrition and Metabolism Center, Duke University Medical Center, Durham, North Carolina
- Duke Molecular Physiology Institute, Duke University Medical Center, Durham, North Carolina
- Endocrinology Division, Department of Medicine, Duke University Medical Center, Durham, North Carolina
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina
| |
Collapse
|
31
|
Liu Y, Wang R, Zheng K, Xin Y, Jia S, Zhao X. Metabonomics analysis of liver in rats administered with chronic low-dose acrylamide. Xenobiotica 2020; 50:894-905. [PMID: 31928121 DOI: 10.1080/00498254.2020.1714791] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The current study aimed to investigate the hepatotoxicity of rats administered with chronic low-dose acrylamide (AA) by using metabonomics technology on the basis of ultraperformance liquid chromatography-mass spectrometry (UPLC-MS). A total of 40 male Wistar rats were randomly divided into the following four groups: control, low-dose AA (0.2 mg/kg bw, non-carcinogenic end-point based on the induction of morphological nerve changes in rats), middle-dose AA (1 mg/kg bw), and high-dose AA (5 mg/kg bw). The rats continuously received AA by administering it in drinking water daily for 16 weeks. After the treatment, rat livers were collected for metabonomics analysis and histopathology examination. Principal components analysis (PCA) and partial least-squares discriminant analysis (PLS-DA) were used to investigate the metabonomics profile changes in rat liver tissues and screen the potential biomarkers.Fourteen metabolites were identified with significant changes in intensities (increased or decreased compared with the control group) as a result of treatment (p < 0.05 or p < 0.01). These metabolites included tauro-b-muricholic acid, docosapentaenoic acid, sphingosine 1-phosphate, taurodeoxycholic acid, lysoPE(20:5), cervonyl carnitine, linoleyl carnitine, docosahexaenoic acid, lysoPC(20:4), lysoPE(18:3), PA(20:4), stearidonyl carnitine, alpha-linolenic acid, and lysoPA(18:0).Results showed that chronic exposure to AA at NOAEL (0.2 mg/kg bw) exhibited no toxic effect in rat livers at the metabolic level. AA induced oxidative stress to the liver and disrupted lipid metabolism. The results of liver histopathology examination further supported the metabonomic results.
Collapse
Affiliation(s)
- Yanli Liu
- Department of Nutrition and Food Hygiene, Public Health College, Harbin Medical University, Harbin, China
| | - Ruijuan Wang
- Department of Nutrition and Food Hygiene, Public Health College, Harbin Medical University, Harbin, China
| | - Kai Zheng
- Department of Nutrition and Food Hygiene, Public Health College, Harbin Medical University, Harbin, China
| | - Youwei Xin
- Department of Nutrition and Food Hygiene, Public Health College, Harbin Medical University, Harbin, China
| | - Siqi Jia
- Department of Nutrition and Food Hygiene, Public Health College, Harbin Medical University, Harbin, China
| | - Xiujuan Zhao
- Department of Nutrition and Food Hygiene, Public Health College, Harbin Medical University, Harbin, China
| |
Collapse
|
32
|
Roginski AC, Wajner A, Cecatto C, Wajner SM, Castilho RF, Wajner M, Amaral AU. Disturbance of bioenergetics and calcium homeostasis provoked by metabolites accumulating in propionic acidemia in heart mitochondria of developing rats. Biochim Biophys Acta Mol Basis Dis 2020; 1866:165682. [PMID: 31931102 DOI: 10.1016/j.bbadis.2020.165682] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Revised: 01/08/2020] [Accepted: 01/09/2020] [Indexed: 12/14/2022]
Abstract
Propionic acidemia is caused by lack of propionyl-CoA carboxylase activity. It is biochemically characterized by accumulation of propionic (PA) and 3-hydroxypropionic (3OHPA) acids and clinically by severe encephalopathy and cardiomyopathy. High urinary excretion of maleic acid (MA) and 2-methylcitric acid (2MCA) is also found in the affected patients. Considering that the underlying mechanisms of cardiac disease in propionic acidemia are practically unknown, we investigated the effects of PA, 3OHPA, MA and 2MCA (0.05-5 mM) on important mitochondrial functions in isolated rat heart mitochondria, as well as in crude heart homogenates and cultured cardiomyocytes. MA markedly inhibited state 3 (ADP-stimulated), state 4 (non-phosphorylating) and uncoupled (CCCP-stimulated) respiration in mitochondria supported by pyruvate plus malate or α-ketoglutarate associated with reduced ATP production, whereas PA and 3OHPA provoked less intense inhibitory effects and 2MCA no alterations at all. MA-induced impaired respiration was attenuated by coenzyme A supplementation. In addition, MA significantly inhibited α-ketoglutarate dehydrogenase activity. Similar data were obtained in heart crude homogenates and permeabilized cardiomyocytes. MA, and PA to a lesser degree, also decreased mitochondrial membrane potential (ΔΨm), NAD(P)H content and Ca2+ retention capacity, and caused swelling in Ca2+-loaded mitochondria. Noteworthy, ΔΨm collapse and mitochondrial swelling were fully prevented or attenuated by cyclosporin A and ADP, indicating the involvement of mitochondrial permeability transition. It is therefore proposed that disturbance of mitochondrial energy and calcium homeostasis caused by MA, as well as by PA and 3OHPA to a lesser extent, may be involved in the cardiomyopathy commonly affecting propionic acidemic patients.
Collapse
Affiliation(s)
- Ana Cristina Roginski
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Alessandro Wajner
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Cristiane Cecatto
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Simone Magagnin Wajner
- Departamento de Medicina Interna, Faculdade de Medicina, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Roger Frigério Castilho
- Departamento de Patologia Clínica, Faculdade de Ciências Médicas, Universidade Estadual de Campinas, Campinas, SP, Brazil
| | - Moacir Wajner
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil; Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil; Serviço de Genética Médica, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil
| | - Alexandre Umpierrez Amaral
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil; Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil; Departamento de Ciências Biológicas, Universidade Regional Integrada do Alto Uruguai e das Missões, Erechim, RS, Brazil.
| |
Collapse
|
33
|
Lu PC, Hsu CN, Lin IC, Lo MH, Yang MY, Tain YL. The Association Between Changes in Plasma Short-Chain Fatty Acid Concentrations and Hypertension in Children With Chronic Kidney Disease. Front Pediatr 2020; 8:613641. [PMID: 33614542 PMCID: PMC7890123 DOI: 10.3389/fped.2020.613641] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Accepted: 12/21/2020] [Indexed: 12/22/2022] Open
Abstract
Background: Some children with chronic kidney disease (CKD) develop hypertension faster than others. This may be attributable to endothelial dysfunction, among other reasons. Short-chain fatty acids (SCFAs), that is, acetate, butyrate, and propionate, are known for reducing cardiovascular risks via preserving endothelial function. This study aimed to investigate the association between changes in plasma SCFA concentrations and in cardiovascular and endothelial parameters in children with CKD. Methods: In total, 105 children and adolescents who met the CKD criteria were enrolled in this study, and 65 patients aged >6 years were divided into two groups based on the ambulatory BP measurements. The parameters of plasma SCFAs, endothelial function and morphology, and echocardiography were examined at the index visit and followed up after 1 year. Results: We observed that 27.69% of 65 patients developed hypertension during the study period. Plasma acetate increased by 22.75 μM in the stable group (P < 0.001), whereas there was no change in the worsened BP group. The index higher plasma butyrate was positively correlated with worsened BP (adjusted odd ratio, 1.381; P = 0.013). At the follow-up, plasma butyrate decreased by 2.12 and 4.41 μM in the stable and worsened BP groups, respectively (P < 0.001). In 105 subjects, higher index plasma propionate was positively correlated with decreasing ejection fraction (adjusted odd ratio, 1.281; P = 0.046). Conclusions: Plasma acetate seemed to play a role in preventing hypertension in children with CKD. However, the index plasma propionate and butyrate concentrations seemed to imply the development of cardiovascular problems in our 1-year study.
Collapse
Affiliation(s)
- Pei-Chen Lu
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital and College of Medicine, Chang Gung University, Kaohsiung, Taiwan.,Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Chien-Ning Hsu
- Department of Pharmacy, Kaohsiung Chang Gung Memorial Hospital and College of Medicine, Chang Gung University, Kaohsiung, Taiwan
| | - I-Chun Lin
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital and College of Medicine, Chang Gung University, Kaohsiung, Taiwan
| | - Mao-Hung Lo
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital and College of Medicine, Chang Gung University, Kaohsiung, Taiwan
| | - Ming-Yu Yang
- Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - You-Lin Tain
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital and College of Medicine, Chang Gung University, Kaohsiung, Taiwan.,Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| |
Collapse
|
34
|
Tamayo M, Fulgencio-Covián A, Navarro-García JA, Val-Blasco A, Ruiz-Hurtado G, Gil-Fernández M, Martín-Nunes L, Lopez JA, Desviat LR, Delgado C, Richard E, Fernández-Velasco M. Intracellular calcium mishandling leads to cardiac dysfunction and ventricular arrhythmias in a mouse model of propionic acidemia. Biochim Biophys Acta Mol Basis Dis 2019; 1866:165586. [PMID: 31678161 DOI: 10.1016/j.bbadis.2019.165586] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Revised: 09/16/2019] [Accepted: 09/17/2019] [Indexed: 12/20/2022]
Abstract
Propionic acidemia (PA) is a rare metabolic disease associated with mutations in genes encoding the α and β subunits of the enzyme propionyl-CoA carboxylase. The accumulation of toxic metabolites results in mitochondrial dysfunction, increased reactive oxygen species production and oxidative damage, which have been associated with the disease pathophysiology. Clinical symptoms are heterogeneous and include cardiac complications, mainly cardiac dysfunction and arrhythmias, which are recognized as one of the major life-threatening manifestations in patients. We aimed to investigate the molecular mechanisms underlying the cardiac phenotype using a hypomorphic mouse model (Pcca-/-(A138T)) that recapitulates some biochemical and clinical characteristics of PA. We demonstrate that Pcca-/-(A138T) mice present with depressed cardiac function along with impaired cell contractility when compared to the wild-type mice. Cardiac dysfunction in Pcca-/-(A138T) mice was associated with lower systolic Ca2+ release ([Ca2+]i transients), impairment in the sarcoplasmic reticulum (SR) Ca2+ load and decreased Ca2+ re-uptake by SR-Ca2+ ATPase (SERCA2a). These functional changes correlated well with the depressed activity of SERCA2a, the elevated ROS levels and SERCA2a oxidation rate in cardiomyocytes isolated from Pcca-/-(A138T) mice. In addition, decreased SR-Ca2+ load in Pcca-/-(A138T) cardiomyocytes was associated with increased diastolic Ca2+ release. The increase in Ca2+ sparks, Ca2+ waves and spontaneous [Ca2+]i transients in Pcca-/-(A138T) cardiomyocytes could be responsible for the induction of ventricular arrhythmias detected in these mice. Overall, our results uncover the role of impaired Ca2+ handling in arrhythmias and cardiac dysfunction in PA, and identify new targets for the development of therapeutic approaches for this devastating metabolic disease.
Collapse
Affiliation(s)
- M Tamayo
- Biomedical Research Institute "Alberto Sols" CSIC-UAM, Madrid, Spain
| | - A Fulgencio-Covián
- Centro de Biología Molecular "Severo Ochoa" UAM-CSIC, Universidad Autónoma de Madrid, Spain
| | - J A Navarro-García
- Cardiorenal Translational Laboratory, Institute of Research i+12, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - A Val-Blasco
- Innate Immune Response Group, IdiPAZ, La Paz University Hospital, Madrid, Spain
| | - G Ruiz-Hurtado
- Cardiorenal Translational Laboratory, Institute of Research i+12, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - M Gil-Fernández
- Innate Immune Response Group, IdiPAZ, La Paz University Hospital, Madrid, Spain
| | - L Martín-Nunes
- Biomedical Research Institute "Alberto Sols" CSIC-UAM, Madrid, Spain
| | - J A Lopez
- Laboratorio de Proteomica Cardiovascular, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - L R Desviat
- Centro de Biología Molecular "Severo Ochoa" UAM-CSIC, Universidad Autónoma de Madrid, Spain
| | - C Delgado
- Biomedical Research Institute "Alberto Sols" CSIC-UAM, Madrid, Spain.
| | - E Richard
- Centro de Biología Molecular "Severo Ochoa" UAM-CSIC, Universidad Autónoma de Madrid, Spain.
| | - M Fernández-Velasco
- Innate Immune Response Group, IdiPAZ, La Paz University Hospital, Madrid, Spain.
| |
Collapse
|
35
|
Wellen KE, Snyder NW. Should we consider subcellular compartmentalization of metabolites, and if so, how do we measure them? Curr Opin Clin Nutr Metab Care 2019; 22:347-354. [PMID: 31365463 PMCID: PMC6824478 DOI: 10.1097/mco.0000000000000580] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
PURPOSE OF REVIEW To examine the consequences of metabolism compartmentalized at the subcellular level, provide prototypical examples of compartmentalized metabolism, and describe methods to examine compartmentalized metabolism. RECENT FINDINGS Progress in metabolomics and isotope tracing has underscored the importance of subcellular compartments of metabolism. The discovery of biological effects of metabolites as bioenergetic intermediates, anabolic building blocks, signaling mediators, and effectors in posttranslation modifications of proteins and nucleic acids have highlighted the role of compartmentalization in determining metabolic fate. Recent advances in both direct and indirect methods to quantify compartmentalized metabolism have improved upon historical approaches. Genetically encoded metabolite sensors, chemical probes, immunoaffinity purification, and compartment-resolved metabolic modeling have all been recently applied to study compartmentalization. SUMMARY Accurate measurement of metabolites in distinct subcellular compartments is important for understanding and pharmacologically targeting metabolic pathways in diverse disease contexts, including cancer, diabetes, heart failure, obesity, and regulation of the immune system. Direct and indirect approaches to quantify compartmentalized metabolism are advancing rapidly. Yet, major challenges remain in the generalizability, rigor, and interpretation of data from the available methods to quantify compartmentalized metabolism.
Collapse
Affiliation(s)
- Kathryn E Wellen
- Department of Cancer Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
- Abramson Family Cancer Research Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | | |
Collapse
|
36
|
Yu Y, Newman H, Shen L, Sharma D, Hu G, Mirando AJ, Zhang H, Knudsen E, Zhang GF, Hilton MJ, Karner CM. Glutamine Metabolism Regulates Proliferation and Lineage Allocation in Skeletal Stem Cells. Cell Metab 2019; 29:966-978.e4. [PMID: 30773468 PMCID: PMC7062112 DOI: 10.1016/j.cmet.2019.01.016] [Citation(s) in RCA: 159] [Impact Index Per Article: 31.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Revised: 11/15/2018] [Accepted: 01/20/2019] [Indexed: 12/27/2022]
Abstract
Skeletal stem cells (SSCs) are postulated to provide a continuous supply of osteoblasts throughout life. However, under certain conditions, the SSC population can become incorrectly specified or is not maintained, resulting in reduced osteoblast formation, decreased bone mass, and in severe cases, osteoporosis. Glutamine metabolism has emerged as a critical regulator of many cellular processes in diverse pathologies. The enzyme glutaminase (GLS) deaminates glutamine to form glutamate-the rate-limiting first step in glutamine metabolism. Using genetic and metabolic approaches, we demonstrate GLS and glutamine metabolism are required in SSCs to regulate osteoblast and adipocyte specification and bone formation. Mechanistically, transaminase-dependent α-ketoglutarate production is critical for the proliferation, specification, and differentiation of SSCs. Collectively, these data suggest stimulating GLS activity may provide a therapeutic approach to expand SSCs in aged individuals and enhance osteoblast differentiation and activity to increase bone mass.
Collapse
Affiliation(s)
- Yilin Yu
- Department of Orthopaedic Surgery, Duke Orthopaedic Cellular, Developmental, and Genome Laboratories, Duke University School of Medicine, Durham, NC 27710, USA
| | - Hunter Newman
- Department of Orthopaedic Surgery, Duke Orthopaedic Cellular, Developmental, and Genome Laboratories, Duke University School of Medicine, Durham, NC 27710, USA
| | - Leyao Shen
- Department of Orthopaedic Surgery, Duke Orthopaedic Cellular, Developmental, and Genome Laboratories, Duke University School of Medicine, Durham, NC 27710, USA
| | - Deepika Sharma
- Department of Orthopaedic Surgery, Duke Orthopaedic Cellular, Developmental, and Genome Laboratories, Duke University School of Medicine, Durham, NC 27710, USA
| | - Guoli Hu
- Department of Orthopaedic Surgery, Duke Orthopaedic Cellular, Developmental, and Genome Laboratories, Duke University School of Medicine, Durham, NC 27710, USA
| | - Anthony J Mirando
- Department of Orthopaedic Surgery, Duke Orthopaedic Cellular, Developmental, and Genome Laboratories, Duke University School of Medicine, Durham, NC 27710, USA
| | - Hongyuan Zhang
- Department of Orthopaedic Surgery, Duke Orthopaedic Cellular, Developmental, and Genome Laboratories, Duke University School of Medicine, Durham, NC 27710, USA
| | - Everett Knudsen
- Department of Orthopaedic Surgery, Duke Orthopaedic Cellular, Developmental, and Genome Laboratories, Duke University School of Medicine, Durham, NC 27710, USA
| | - Guo-Fang Zhang
- Sarah W. Stedman Nutrition and Metabolism Center & Duke Molecular Physiology Institute, Duke University Medical Center, 300 North Duke Street, Durham, NC 27701, USA; Department of Medicine, Duke University School of Medicine, Durham, NC 27701, USA
| | - Matthew J Hilton
- Department of Orthopaedic Surgery, Duke Orthopaedic Cellular, Developmental, and Genome Laboratories, Duke University School of Medicine, Durham, NC 27710, USA; Department of Cell Biology, Duke University, Durham, NC 27710, USA
| | - Courtney M Karner
- Department of Orthopaedic Surgery, Duke Orthopaedic Cellular, Developmental, and Genome Laboratories, Duke University School of Medicine, Durham, NC 27710, USA; Department of Cell Biology, Duke University, Durham, NC 27710, USA.
| |
Collapse
|