1
|
Babazade H, Zubarioglu T, Uygur E, Cansever MŞ, Kiykim E, Aktuğlu Zeybek Ç. Carglumic acid as a treatment for persistent hyperammonemia in carnitine-acylcarnitine translocase deficiency: A case study. Mol Genet Metab Rep 2025; 42:101199. [PMID: 40092581 PMCID: PMC11910245 DOI: 10.1016/j.ymgmr.2025.101199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 02/03/2025] [Accepted: 02/03/2025] [Indexed: 03/19/2025] Open
Abstract
Carnitine-acylcarnitine translocase deficiency (CACTD) is a rare autosomal recessive fatty acid oxidation disorder resulting in energy deficiency due to impaired mitochondrial long-chain fatty acid transport. Hyperammonemia is a critical complication, often resistant to conventional treatment. Here, we report the case of a 7-month-old patient with CACTD, initially diagnosed at 10 days old, who presented with persistent hyperammonemia despite optimized medical nutrition therapy and conventional nitrogen scavenging with sodium benzoate. When hyperammonemia persisted, carglumic acid was introduced, leading to a sustained decrease in ammonia levels and effective long-term control. Carglumic acid, typically indicated for organic acidemias, proved beneficial in this CACTD case. The administration of carglumic acid not only provided acute resolution but also stabilized ammonia levels over prolonged follow-up. This case highlights carglumic acid as a potential therapeutic option for managing hyperammonemia in CACTD, underscoring the need for further studies to confirm its efficacy in long-term management of hyperammonemia in fatty acid oxidation disorders.
Collapse
Affiliation(s)
- Hanım Babazade
- İstanbul University-Cerrahpaşa, Cerrahpaşa Medical Faculty, Department of Pediatrics, Division of Nutrition and Metabolism, İstanbul, Turkey
| | - Tanyel Zubarioglu
- İstanbul University-Cerrahpaşa, Cerrahpaşa Medical Faculty, Department of Pediatrics, Division of Nutrition and Metabolism, İstanbul, Turkey
| | - Esma Uygur
- İstanbul University-Cerrahpaşa, Cerrahpaşa Medical Faculty, Department of Pediatrics, Division of Nutrition and Metabolism, İstanbul, Turkey
| | - Mehmet Şerif Cansever
- İstanbul University-Cerrahpaşa, Cerrahpaşa Medical Faculty, Research Laboratory of Metabolism, İstanbul, Turkey
- İstanbul University-Cerrahpaşa, The Vocational School of Health Services, Department of Medical Documentation and Techniques, Division of Medical Laboratory Techniques, İstanbul,Turkey
| | - Ertuğrul Kiykim
- İstanbul University-Cerrahpaşa, Cerrahpaşa Medical Faculty, Department of Pediatrics, Division of Nutrition and Metabolism, İstanbul, Turkey
| | - Çiğdem Aktuğlu Zeybek
- İstanbul University-Cerrahpaşa, Cerrahpaşa Medical Faculty, Department of Pediatrics, Division of Nutrition and Metabolism, İstanbul, Turkey
- İstanbul University-Cerrahpaşa, Cerrahpaşa Medical Faculty, Research Laboratory of Metabolism, İstanbul, Turkey
| |
Collapse
|
2
|
Verberk SGS, Hahn N, Heister D, Haverkamp J, Snelder KS, de Goede KE, Gorki FS, Hendriks JJA, Houtkooper RH, Visser G, Sjouke B, Langeveld M, Van den Bossche J. Monocyte and macrophage profiles in patients with inherited long-chain fatty acid oxidation disorders. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167524. [PMID: 39307292 DOI: 10.1016/j.bbadis.2024.167524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 09/17/2024] [Accepted: 09/17/2024] [Indexed: 09/28/2024]
Abstract
Patients with inherited disorders of the long-chain fatty acid oxidation (lcFAO) machinery present with a heterogeneous profile of disease manifestations and aggravation of symptoms is often triggered by inflammatory activation. Monocytes and macrophages are innate immune cells that play a major role in the onset and resolution of inflammation. These cells undergo metabolic rewiring upon activation including the regulation of the FAO rate. The rewiring of FAO and the effect of lcFAO disorders (lcFAOD) on human monocyte and macrophage phenotype and function remain largely unknown. Here, we performed extensive phenotyping of circulating monocytes and analyzed plasma cytokine levels in 11 lcFAOD patients and 11 matched control subjects. In patients with lcFAOD, we observed induced plasma levels of the inflammatory cytokines IL-1β and IL-6, and enhanced CD206 and CD62L surface marker expression in circulating monocyte subsets. To mimic the most common lcFAOD very-long-chain acyl-CoA dehydrogenase disorder (VLCADD), we used siRNA-mediated knockdown of the ACADVL gene (encoding VLCAD) in macrophages derived from healthy volunteers. Hereby, we found that siVLCAD affected IL-4-induced alternative macrophage activation while leaving LPS responses and cellular metabolism intact. In the same line, monocyte-derived macrophages from lcFAOD patients had elevated levels of the IL-4-induced alternative macrophage markers CD206 and CD200R. Still, they did not show major metabolic defects or changes in the LPS-induced inflammatory response. Our results indicate that monocytes and macrophages from lcFAOD patients present no major inflammatory or metabolic differences and show that IL-4-induced surface markers are intertwined with lcFAO in human macrophages.
Collapse
Affiliation(s)
- Sanne G S Verberk
- Department of Molecular Cell Biology and Immunology, Amsterdam Cardiovascular Sciences, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam Institute for Infection and Immunity, Cancer Centre Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands; Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
| | - Nico Hahn
- Department of Molecular Cell Biology and Immunology, Amsterdam Cardiovascular Sciences, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam Institute for Infection and Immunity, Cancer Centre Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Daan Heister
- Department of Molecular Cell Biology and Immunology, Amsterdam Cardiovascular Sciences, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam Institute for Infection and Immunity, Cancer Centre Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Jorien Haverkamp
- Department Endocrinology and Metabolism, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Khya S Snelder
- Department Endocrinology and Metabolism, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Kyra E de Goede
- Department of Molecular Cell Biology and Immunology, Amsterdam Cardiovascular Sciences, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam Institute for Infection and Immunity, Cancer Centre Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Friederieke S Gorki
- Department of Molecular Cell Biology and Immunology, Amsterdam Cardiovascular Sciences, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam Institute for Infection and Immunity, Cancer Centre Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands; Institute of Innate Immunity, University Hospital Bonn, University of Bonn, Bonn 53127, Germany
| | - Jerome J A Hendriks
- Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
| | - Riekelt H Houtkooper
- Laboratory Genetic Metabolic Diseases, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands; Emma Center for Personalized Medicine, Amsterdam UMC, Amsterdam, the Netherlands
| | - Gepke Visser
- Emma Children's Hospital, Department of Pediatrics, Division of Metabolic Diseases, Amsterdam Gastroenterology Endocrinology and Metabolism, Amsterdam UMC, location University of Amsterdam, Amsterdam, the Netherlands
| | - Barbara Sjouke
- Department Endocrinology and Metabolism, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands; Department of Internal Medicine, Radboud University Medical Centre, Nijmegen, the Netherlands
| | - Mirjam Langeveld
- Department Endocrinology and Metabolism, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Jan Van den Bossche
- Department of Molecular Cell Biology and Immunology, Amsterdam Cardiovascular Sciences, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam Institute for Infection and Immunity, Cancer Centre Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands.
| |
Collapse
|
3
|
Babcock SJ, Houten SM, Gillingham MB. A review of fatty acid oxidation disorder mouse models. Mol Genet Metab 2024; 142:108351. [PMID: 38430613 PMCID: PMC11073919 DOI: 10.1016/j.ymgme.2024.108351] [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: 11/22/2023] [Revised: 02/20/2024] [Accepted: 02/21/2024] [Indexed: 03/05/2024]
Abstract
Fatty acid oxidation disorders (FAODs) are a family of rare, genetic disorders that affect any part of the fatty acid oxidation pathway. Patients present with severe phenotypes, such as hypoketotic hypoglycemia, cardiomyopathy, and rhabdomyolysis, and currently manage these symptoms by the avoidance of fasting and maintaining a low-fat, high-carbohydrate diet. Because knowledge about FAODs is limited due to the small number of patients, rodent models have been crucial in learning more about these disorders, particularly in studying the molecular mechanisms involved in different phenotypes and in evaluating treatments for patients. The purpose of this review is to present the different FAOD mouse models and highlight the benefits and limitations of using these models. Specifically, we discuss the phenotypes of the available FAOD mouse models, the potential molecular causes of prominent FAOD phenotypes that have been studied using FAOD mouse models, and how FAOD mouse models have been used to evaluate treatments for patients.
Collapse
Affiliation(s)
- Shannon J Babcock
- Department of Molecular and Medical Genetics, Oregon Health and Science University, Portland, OR, USA.
| | - Sander M Houten
- Deparment of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Melanie B Gillingham
- Department of Molecular and Medical Genetics, Oregon Health and Science University, Portland, OR, USA
| |
Collapse
|
4
|
Nurjanah S, Gerding A, Vieira-Lara MA, Evers B, Langelaar-Makkinje M, Spiekerkoetter U, Bakker BM, Tucci S. Heptanoate Improves Compensatory Mechanism of Glucose Homeostasis in Mitochondrial Long-Chain Fatty Acid Oxidation Defect. Nutrients 2023; 15:4689. [PMID: 37960342 PMCID: PMC10649308 DOI: 10.3390/nu15214689] [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: 09/22/2023] [Revised: 10/31/2023] [Accepted: 11/01/2023] [Indexed: 11/15/2023] Open
Abstract
Defects in mitochondrial fatty acid β-oxidation (FAO) impair metabolic flexibility, which is an essential process for energy homeostasis. Very-long-chain acyl-CoA dehydrogenase (VLCADD; OMIM 609575) deficiency is the most common long-chain mitochondrial FAO disorder presenting with hypoglycemia as a common clinical manifestation. To prevent hypoglycemia, triheptanoin-a triglyceride composed of three heptanoates (C7) esterified with a glycerol backbone-can be used as a dietary treatment, since it is metabolized into precursors for gluconeogenesis. However, studies investigating the effect of triheptanoin on glucose homeostasis are limited. To understand the role of gluconeogenesis in the pathophysiology of long-chain mitochondrial FAO defects, we injected VLCAD-deficient (VLCAD-/-) mice with 13C3-glycerol in the presence and absence of heptanoate (C7). The incorporation of 13C3-glycerol into blood glucose was higher in VLCAD-/- mice than in WT mice, whereas the difference disappeared in the presence of C7. The result correlates with 13C enrichment of liver metabolites in VLCAD-/- mice. In contrast, the C7 bolus significantly decreased the 13C enrichment. These data suggest that the increased contribution of gluconeogenesis to the overall glucose production in VLCAD-/- mice increases the need for gluconeogenesis substrate, thereby avoiding hypoglycemia. Heptanoate is a suitable substrate to induce glucose production in mitochondrial FAO defect.
Collapse
Affiliation(s)
- Siti Nurjanah
- Department of General Pediatrics, Adolescent Medicine and Neonatology, Medical Centre, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany (U.S.)
- Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany
| | - Albert Gerding
- Laboratory of Pediatrics, Systems Medicine of Metabolism and Signaling, University Medical Center Groningen, University of Groningen, 9700 RB Groningen, The Netherlands (M.L.-M.)
- Laboratory of Metabolic Diseases, Department of Laboratory Medicine, University Medical Center Groningen, University of Groningen, 9700 RB Groningen, The Netherlands
| | - Marcel A. Vieira-Lara
- Laboratory of Pediatrics, Systems Medicine of Metabolism and Signaling, University Medical Center Groningen, University of Groningen, 9700 RB Groningen, The Netherlands (M.L.-M.)
| | - Bernard Evers
- Laboratory of Pediatrics, Systems Medicine of Metabolism and Signaling, University Medical Center Groningen, University of Groningen, 9700 RB Groningen, The Netherlands (M.L.-M.)
| | - Miriam Langelaar-Makkinje
- Laboratory of Pediatrics, Systems Medicine of Metabolism and Signaling, University Medical Center Groningen, University of Groningen, 9700 RB Groningen, The Netherlands (M.L.-M.)
| | - Ute Spiekerkoetter
- Department of General Pediatrics, Adolescent Medicine and Neonatology, Medical Centre, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany (U.S.)
| | - Barbara M. Bakker
- Laboratory of Pediatrics, Systems Medicine of Metabolism and Signaling, University Medical Center Groningen, University of Groningen, 9700 RB Groningen, The Netherlands (M.L.-M.)
| | - Sara Tucci
- Pharmacy, Medical Center, University of Freiburg, 79106 Freiburg, Germany
- G.E.R.N. Research Center for Tissue Replacement, Regeneration & Neogenesis, Department of Prosthetic Dentistry, Medical Center, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| |
Collapse
|
5
|
Rossi A, Assunto A, Rosano C, Tucci S, Ruoppolo M, Caterino M, Pirozzi F, Strisciuglio P, Parenti G, Melis D. Mitochondrial reprogramming in peripheral blood mononuclear cells of patients with glycogen storage disease type Ia. GENES & NUTRITION 2023; 18:10. [PMID: 37280548 DOI: 10.1186/s12263-023-00729-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Accepted: 05/05/2023] [Indexed: 06/08/2023]
Abstract
BACKGROUND Glycogen storage disease type Ia (GSDIa) is an inborn metabolic disorder caused by the deficiency of glucose-6-phospatase-α (G6Pase-α) leading to mitochondrial dysfunction. It remains unclear whether mitochondrial dysfunction is present in patients' peripheral blood mononuclear cells (PBMC) and whether dietary treatment can play a role. The aim of this study was to investigate mitochondrial function in PBMC of GSDIa patients. METHODS Ten GSDIa patients and 10 age-, sex- and fasting-time matched controls were enrolled. Expression of genes involved in mitochondrial function and activity of key fatty acid oxidation (FAO) and Krebs cycle proteins were assessed in PBMC. Targeted metabolomics and assessment of metabolic control markers were also performed. RESULTS Adult GSDIa patients showed increased CPT1A, SDHB, TFAM, mTOR expression (p < 0.05) and increased VLCAD, CPT2 and citrate synthase activity in PBMC (p < 0.05). VLCAD activity directly correlated with WC (p < 0.01), BMI (p < 0.05), serum malonycarnitine levels (p < 0.05). CPT2 activity directly correlated with BMI (p < 0.05). CONCLUSION Mitochondrial reprogramming is detectable in PBMC of GSDIa patients. This feature may develop as an adaptation to the liver enzyme defect and may be triggered by dietary (over)treatment in the frame of G6Pase-α deficiency. PBMC can represent an adequate mean to assess (diet-induced) metabolic disturbances in GSDIa.
Collapse
Affiliation(s)
- Alessandro Rossi
- Department of Translational Medicine, Section of Pediatrics, University of Naples Federico II, Naples, Italy
| | - Antonia Assunto
- Department of Translational Medicine, Section of Pediatrics, University of Naples Federico II, Naples, Italy
| | - Carmen Rosano
- Department of Translational Medicine, Section of Pediatrics, University of Naples Federico II, Naples, Italy
| | - Sara Tucci
- Pharmacy, Medical Center - University of Freiburg, Hugstetterstr. 55, D-79106, Freiburg, Germany
| | - Margherita Ruoppolo
- Department of Molecular Medicine and Medical Biotechnology, University of Naples "Federico II", Naples, Italy
- CEINGE Advanced Biotechnologies, Naples, Italy
| | - Marianna Caterino
- Department of Molecular Medicine and Medical Biotechnology, University of Naples "Federico II", Naples, Italy
- CEINGE Advanced Biotechnologies, Naples, Italy
| | - Francesca Pirozzi
- Department of Molecular Medicine and Medical Biotechnology, University of Naples "Federico II", Naples, Italy
- CEINGE Advanced Biotechnologies, Naples, Italy
| | - Pietro Strisciuglio
- Department of Translational Medicine, Section of Pediatrics, University of Naples Federico II, Naples, Italy
| | - Giancarlo Parenti
- Department of Translational Medicine, Section of Pediatrics, University of Naples Federico II, Naples, Italy
- Telethon Institute of Genetics and Medicine, Pozzuoli, Italy
| | - Daniela Melis
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", Section of Pediatrics, University of Salerno, Via Salvador Allende, 43 84081, Baronissi (Salerno), Italy.
| |
Collapse
|
6
|
Lund M, Heaton R, Hargreaves IP, Gregersen N, Olsen RKJ. Odd- and even-numbered medium-chained fatty acids protect against glutathione depletion in very long-chain acyl-CoA dehydrogenase deficiency. Biochim Biophys Acta Mol Cell Biol Lipids 2023; 1868:159248. [PMID: 36356723 DOI: 10.1016/j.bbalip.2022.159248] [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: 01/11/2022] [Revised: 10/09/2022] [Accepted: 10/17/2022] [Indexed: 11/09/2022]
Abstract
Recent trials have reported the ability of triheptanoin to improve clinical outcomes for the severe symptoms associated with long-chain fatty acid oxidation disorders, including very long-chain acyl-CoA dehydrogenase (VLCAD) deficiency. However, the milder myopathic symptoms are still challenging to treat satisfactorily. Myopathic pathogenesis is multifactorial, but oxidative stress is an important component. We have previously shown that metabolic stress increases the oxidative burden in VLCAD-deficient cell lines and can deplete the antioxidant glutathione (GSH). We investigated whether medium-chain fatty acids provide protection against GSH depletion during metabolic stress in VLCAD-deficient fibroblasts. To investigate the effect of differences in anaplerotic capacity, we included both even-(octanoate) and odd-numbered (heptanoate) medium-chain fatty acids. Overall, we show that modulation of the concentration of medium-chain fatty acids in culture media affects levels of GSH retained during metabolic stress in VLCAD-deficient cell lines but not in controls. Lowered glutamine concentration in the culture media during metabolic stress led to GSH depletion and decreased viability in VLCAD deficient cells, which could be rescued by both heptanoate and octanoate in a dose-dependent manner. Unlike GSH levels, the levels of total thiols increased after metabolic stress exposure, the size of this increase was not affected by differences in cell culture medium concentrations of glutamine, heptanoate or octanoate. Addition of a PPAR agonist further exacerbated stress-related GSH-depletion and viability loss, requiring higher concentrations of fatty acids to restore GSH levels and cell viability. Both odd- and even-numbered medium-chain fatty acids efficiently protect VLCADdeficient cells against metabolic stress-induced antioxidant depletion.
Collapse
Affiliation(s)
- Martin Lund
- Research Unit for Molecular Medicine, Department of Clinical Medicine, Aarhus University and Aarhus University Hospital, Palle Juel-Jensens Boulevard 99, 8200 Aarhus, Denmark.
| | - Robert Heaton
- School of Pharmacy, Liverpool John Moore University, Byrom Street, Liverpool L3 3AF, United Kingdom
| | - Iain P Hargreaves
- School of Pharmacy, Liverpool John Moore University, Byrom Street, Liverpool L3 3AF, United Kingdom
| | - Niels Gregersen
- Research Unit for Molecular Medicine, Department of Clinical Medicine, Aarhus University and Aarhus University Hospital, Palle Juel-Jensens Boulevard 99, 8200 Aarhus, Denmark
| | - Rikke K J Olsen
- Research Unit for Molecular Medicine, Department of Clinical Medicine, Aarhus University and Aarhus University Hospital, Palle Juel-Jensens Boulevard 99, 8200 Aarhus, Denmark.
| |
Collapse
|
7
|
Dong Q, Yan S, Li D, Zhou C, Tian S, Wang Y, Miao P, Zhu W, Zhu S, Pan C. Feeding foliar nano-selenium biofortified panax notoginseng could reduce the occurrence of glycolipid metabolism disorder in mice caused by high-fat diets. Front Nutr 2022; 9:973027. [PMID: 36091251 PMCID: PMC9450130 DOI: 10.3389/fnut.2022.973027] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2022] [Accepted: 08/02/2022] [Indexed: 11/30/2022] Open
Abstract
Nano-selenium (nano-Se) has been extensively explored as a biostimulant for improving the quality of grain crops. However, there are few reports about the effect on the medicinal components of Chinese herbal medicine cultured with nano-Se. Here, we sprayed nano-Se during the cultivation of Panax notoginseng (SePN), and measured the changes of medicinal components compared with conventional Panax notoginseng (PN). Furthermore, we identified a more pronounced effect of SePN on reducing obesity in animals compared with PN. By measuring antioxidant capacity, histopathology, gene expression related to glycolipid metabolism, and gut microbiota composition, we propose a potential mechanism for SePN to improve animal health. Compared with the control groups, foliar spraying of nano-Se increased saponins contents (Rb2, Rb3, Rc, F2, Rb2, and Rf) in the roots of Panax notoginseng, the content of Rb2 increased by 3.9 times particularly. Interestingly, animal studies indicated that taking selenium-rich Panax notoginseng (SePN) can further ameliorate liver antioxidation (SOD, MDA, and GSH) and enzyme activities involved in glycolipid metabolism (ATGL and PFK). It also relieved inflammation and regulated the expression of genes (MCAD, PPAR-α, and PCSK9) related to fatty acid oxidation. The abundance ratio of Firmicutes/Bacteroides and beneficial bacteria abundance (Bifidobacterium, Butyricimonas, and Parasutterella) in gut microbiota were improved relative to the control. In summary, the application of nano-Se on PN may effectively raise the content of Panax notoginseng saponins (PNS) and immensely lower the risk of metabolic disorders of glycolipids.
Collapse
Affiliation(s)
- Qinyong Dong
- Department of Applied Chemistry, College of Science, Innovation Center of Pesticide Research, China Agricultural University, Beijing, China
| | - Sen Yan
- Department of Applied Chemistry, College of Science, Innovation Center of Pesticide Research, China Agricultural University, Beijing, China
| | - Dong Li
- Department of Applied Chemistry, College of Science, Innovation Center of Pesticide Research, China Agricultural University, Beijing, China
| | - Chunran Zhou
- Department of Applied Chemistry, College of Science, Innovation Center of Pesticide Research, China Agricultural University, Beijing, China
| | - Sinuo Tian
- Department of Applied Chemistry, College of Science, Innovation Center of Pesticide Research, China Agricultural University, Beijing, China
| | - Yu Wang
- Department of Applied Chemistry, College of Science, Innovation Center of Pesticide Research, China Agricultural University, Beijing, China
| | - Peijuan Miao
- Department of Applied Chemistry, College of Science, Innovation Center of Pesticide Research, China Agricultural University, Beijing, China
| | - Wentao Zhu
- Department of Applied Chemistry, College of Science, Innovation Center of Pesticide Research, China Agricultural University, Beijing, China
| | - Shusheng Zhu
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, College of Plant Protection, National Engineering Research Center for Applied Technology of Agricultural Biodiversity, Yunnan Agricultural University, Kunming, China
| | - Canping Pan
- Department of Applied Chemistry, College of Science, Innovation Center of Pesticide Research, China Agricultural University, Beijing, China
- *Correspondence: Canping Pan
| |
Collapse
|
8
|
Ryder B, Inbar-Feigenberg M, Glamuzina E, Halligan R, Vara R, Elliot A, Coman D, Minto T, Lewis K, Schiff M, Vijay S, Akroyd R, Thompson S, MacDonald A, Woodward AJM, Gribben JEL, Grunewald S, Belaramani K, Hall M, van der Haak N, Devanapalli B, Tolun AA, Wilson C, Bhattacharya K. New insights into carnitine-acylcarnitine translocase deficiency from 23 cases: Management challenges and potential therapeutic approaches. J Inherit Metab Dis 2021; 44:903-915. [PMID: 33634872 DOI: 10.1002/jimd.12371] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 02/19/2021] [Accepted: 02/23/2021] [Indexed: 12/22/2022]
Abstract
Carnitine acyl-carnitine translocase deficiency (CACTD) is a rare autosomal recessive disorder of mitochondrial long-chain fatty-acid transport. Most patients present in the first 2 days of life, with hypoketotic hypoglycaemia, hyperammonaemia, cardiomyopathy or arrhythmia, hepatomegaly and elevated liver enzymes. Multi-centre international retrospective chart review of clinical presentation, biochemistry, treatment modalities including diet, subsequent complications, and mode of death of all patients. Twenty-three patients from nine tertiary metabolic units were identified. Seven attenuated patients of Pakistani heritage, six of these homozygous c.82G>T, had later onset manifestations and long-term survival without chronic hyperammonemia. Of the 16 classical cases, 15 had cardiac involvement at presentation comprising cardiac arrhythmias (9/15), cardiac arrest (7/15), and cardiac hypertrophy (9/15). Where recorded, ammonia levels were elevated in all but one severe case (13/14 measured) and 14/16 had hypoglycaemia. Nine classical patients survived longer-term-most with feeding difficulties and cognitive delay. Hyperammonaemia appears refractory to ammonia scavenger treatment and carglumic acid, but responds well to high glucose delivery during acute metabolic crises. High-energy intake seems necessary to prevent decompensation. Anaplerosis utilising therapeutic d,l-3-hydroxybutyrate, Triheptanoin and increased protein intake, appeared to improve chronic hyperammonemia and metabolic stability where trialled in individual cases. CACTD is a rare disorder of fatty acid oxidation with a preponderance to severe cardiac dysfunction. Long-term survival is possible in classical early-onset cases with long-chain fat restriction, judicious use of glucose infusions, and medium chain triglyceride supplementation. Adjunctive therapies supporting anaplerosis may improve longer-term outcomes.
Collapse
Affiliation(s)
- Bryony Ryder
- Division of Clinical and Metabolic Genetics, The Hospital for Sick Children, Toronto, Ontario, Canada
- National Metabolic Service, Starship Children's Hospital, Auckland, New Zealand
| | - Michal Inbar-Feigenberg
- Division of Clinical and Metabolic Genetics, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Emma Glamuzina
- National Metabolic Service, Starship Children's Hospital, Auckland, New Zealand
| | - Rebecca Halligan
- Department of Inherited Metabolic Disorders, Birmingham Women's and Children's Hospital Foundation Trust, Birmingham, UK
- Department of Metabolic Medicine, Evelina Children's Hospital, London, UK
| | - Roshni Vara
- Department of Metabolic Medicine, Evelina Children's Hospital, London, UK
| | - Aoife Elliot
- Queensland Lifespan Metabolic Medicine Service, Queensland Children's Hospital, Brisbane, QLD, Australia
| | - David Coman
- Queensland Lifespan Metabolic Medicine Service, Queensland Children's Hospital, Brisbane, QLD, Australia
- School of Medicine University of Queensland and Griffith University, Brisbane, Queensland, Australia
| | - Tahlee Minto
- Queensland Lifespan Metabolic Medicine Service, Queensland Children's Hospital, Brisbane, QLD, Australia
| | - Katherine Lewis
- Queensland Lifespan Metabolic Medicine Service, Queensland Children's Hospital, Brisbane, QLD, Australia
| | - Manuel Schiff
- Reference Centre for Inherited Metabolic Diseases, AP-HP, Necker University Hospital, University of Paris, Paris, France
- INSERM U1163, Institut Imagine, Paris, France
| | - Suresh Vijay
- Department of Inherited Metabolic Disorders, Birmingham Women's and Children's Hospital Foundation Trust, Birmingham, UK
| | - Rhonda Akroyd
- National Metabolic Service, Starship Children's Hospital, Auckland, New Zealand
| | - Sue Thompson
- Department of Metabolic Genetics, Sydney Children's Hospitals' Network NSW, Sydney, New South Wales, Australia
- Faculty of Health and Medical Science, University of Sydney, Sydney, New South Wales, Australia
| | - Anita MacDonald
- Department of Inherited Metabolic Disorders, Birmingham Women's and Children's Hospital Foundation Trust, Birmingham, UK
| | - Abigail J M Woodward
- Department of Nutrition & Dietetics, Evelina London Children's Hospital, London, UK
| | - Joanne E L Gribben
- Department of Nutrition & Dietetics, Evelina London Children's Hospital, London, UK
| | - Stephanie Grunewald
- Metabolic Medicine Department, Great Ormond Street Hospital, Institute of Child Health University College London, NIHR Biomedical Research Centre, London, UK
| | - Kiran Belaramani
- Department of Metabolic Medicine, Hong Kong Children's Hospital, Ngau Tau Kok, Hong Kong
| | - Madeleine Hall
- Departments of Metabolic Medicine & Nutrition, Women's and Children's Hospital, North Adelaide, South Australia, Australia
| | - Natalie van der Haak
- Departments of Metabolic Medicine & Nutrition, Women's and Children's Hospital, North Adelaide, South Australia, Australia
| | - Beena Devanapalli
- Department of Metabolic Genetics, Sydney Children's Hospitals' Network NSW, Sydney, New South Wales, Australia
| | - Adviye Ayper Tolun
- Department of Metabolic Genetics, Sydney Children's Hospitals' Network NSW, Sydney, New South Wales, Australia
| | - Callum Wilson
- National Metabolic Service, Starship Children's Hospital, Auckland, New Zealand
| | - Kaustuv Bhattacharya
- Department of Metabolic Genetics, Sydney Children's Hospitals' Network NSW, Sydney, New South Wales, Australia
- Faculty of Health and Medical Science, University of Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
9
|
Adu-Amankwaah J, Adzika GK, Adekunle AO, Ndzie Noah ML, Mprah R, Bushi A, Akhter N, Xu Y, Huang F, Chatambarara B, Sun H. The Synergy of ADAM17-Induced Myocardial Inflammation and Metabolic Lipids Dysregulation During Acute Stress: New Pathophysiologic Insights Into Takotsubo Cardiomyopathy. Front Cardiovasc Med 2021; 8:696413. [PMID: 34150874 PMCID: PMC8212952 DOI: 10.3389/fcvm.2021.696413] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Accepted: 05/11/2021] [Indexed: 12/11/2022] Open
Abstract
Due to its reversible nature, Takotsubo cardiomyopathy (TTC) is considered an intriguing and fascinating cardiovascular disease characterized by a transient wall motion abnormality of the left ventricle, affecting more than one coronary artery territory, often in a circumferential apical distribution. Takotsubo cardiomyopathy was discovered by a Japanese cardiovascular expert and classified as acquired primary cardiomyopathy by the American Heart Association (AHA) in 1990 and 2006, respectively. Regardless of the extensive research efforts, its pathophysiology is still unclear; therefore, there are no well-established guidelines specifically for treating and managing TTC patients. Increasing evidence suggests that sympatho-adrenergic stimulation is strongly associated with the pathogenesis of this disease. Under acute stressful conditions, the hyperstimulation of beta-adrenergic receptors (β-ARs) resulting from excessive release of catecholamines induces intracellular kinases capable of phosphorylating and activating “A Disintegrin and Metalloprotease 17” (ADAM17), a type-I transmembrane protease that plays a central role in acute myocardial inflammation and metabolic lipids dysregulation which are the main hallmarks of TTC. However, our understanding of this is limited; hence this concise review provides a comprehensive insight into the key role of ADAM17 in acute myocardial inflammation and metabolic lipids dysregulation during acute stress. Also, how the synergy of ADAM17-induced acute inflammation and lipids dysregulation causes TTC is explained. Finally, potential therapeutic targets for TTC are also discussed.
Collapse
Affiliation(s)
| | | | | | | | - Richard Mprah
- Department of Physiology, Xuzhou Medical University, Xuzhou, China
| | - Aisha Bushi
- Department of Medicine, Xuzhou Medical University, Xuzhou, China
| | - Nazma Akhter
- Department of Physiology, Xuzhou Medical University, Xuzhou, China
| | - Yaxin Xu
- Department of Physiology, Xuzhou Medical University, Xuzhou, China
| | - Fei Huang
- Department of Physiology, Xuzhou Medical University, Xuzhou, China
| | | | - Hong Sun
- Department of Physiology, Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
10
|
Altered Metabolic Flexibility in Inherited Metabolic Diseases of Mitochondrial Fatty Acid Metabolism. Int J Mol Sci 2021; 22:ijms22073799. [PMID: 33917608 PMCID: PMC8038842 DOI: 10.3390/ijms22073799] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 04/03/2021] [Accepted: 04/05/2021] [Indexed: 12/14/2022] Open
Abstract
In general, metabolic flexibility refers to an organism's capacity to adapt to metabolic changes due to differing energy demands. The aim of this work is to summarize and discuss recent findings regarding variables that modulate energy regulation in two different pathways of mitochondrial fatty metabolism: β-oxidation and fatty acid biosynthesis. We focus specifically on two diseases: very long-chain acyl-CoA dehydrogenase deficiency (VLCADD) and malonyl-CoA synthetase deficiency (acyl-CoA synthetase family member 3 (ACSF3)) deficiency, which are both characterized by alterations in metabolic flexibility. On the one hand, in a mouse model of VLCAD-deficient (VLCAD-/-) mice, the white skeletal muscle undergoes metabolic and morphologic transdifferentiation towards glycolytic muscle fiber types via the up-regulation of mitochondrial fatty acid biosynthesis (mtFAS). On the other hand, in ACSF3-deficient patients, fibroblasts show impaired mitochondrial respiration, reduced lipoylation, and reduced glycolytic flux, which are compensated for by an increased β-oxidation rate and the use of anaplerotic amino acids to address the energy needs. Here, we discuss a possible co-regulation by mtFAS and β-oxidation in the maintenance of energy homeostasis.
Collapse
|
11
|
Chen M, Zheng J, Zou X, Ye C, Xia H, Yang M, Gao Q, Yang Q, Liu H. Ligustrum robustum (Roxb.) blume extract modulates gut microbiota and prevents metabolic syndrome in high-fat diet-fed mice. JOURNAL OF ETHNOPHARMACOLOGY 2021; 268:113695. [PMID: 33316365 DOI: 10.1016/j.jep.2020.113695] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 12/07/2020] [Accepted: 12/10/2020] [Indexed: 06/12/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE In Chinese folk medicine, Ligustrum robustum (Roxb.) Blume has been widely used as a healthy tea beverage for improvement in obesity and lipidemic metabolic disorders. AIM OF THE STUDY We aimed to investigate the effect of L. robustum extract (LRE) on metabolic syndrome in high-fat diet (HFD)-fed mice and to explore the underlying role of gut microbiota during the treatment. MATERIALS AND METHODS The ground dried leaves of L. robustum (Roxb.) Blume were extracted with ethanol and then purified by a resin column. The composition of L. robustum extract (LRE) was analyzed by high-performance liquid chromatography-tandem mass spectrometry (HPLC-MS/MS). C57BL/6 J mice fed with HFD were treated with LRE for 16 weeks. RT-qPCR and morphological staining were utilized to reveal the impact of LRE on hepatic glucolipid metabolism and gut integrity. The next-generation sequencing of 16 S rDNA was applied for analyzing the gut microbial community of fecal samples. RESULTS LRE, mainly composed of ligupurpuroside A and aceteoside, alleviated insulin resistance, improved hepatic metabolism, enhanced intestinal integrity, and suppressed inflammatory responses in HFD-fed mice. Moreover, LRE treatment reshaped the gut microbiota structure by increasing the levels of genera Streptococcus, Lactobacillus, and Mucispirillum and decreasing the populations of Alistipes and Lachnospiraceae NK4A136 group in HFD-fed mice. The alteration of gut microbiota was associated with several metabolic pathways of gut bacteria. Spearman's correlation analysis further confirmed the links between the changed intestinal bacteria and multiple disease indices. CONCLUSIONS LRE prevented gut microbiota dysbiosis and metabolic disorder in HFD-fed mice, which helps to promote the application in LRE-mediated prevention from metabolic syndrome as a gut microbial regulator.
Collapse
Affiliation(s)
- Man Chen
- College of Basic Medicine, Hubei University of Chinese Medicine, Wuhan, Hubei, 430065, PR China
| | - Junping Zheng
- College of Basic Medicine, Hubei University of Chinese Medicine, Wuhan, Hubei, 430065, PR China
| | - Xiaojuan Zou
- College of Basic Medicine, Hubei University of Chinese Medicine, Wuhan, Hubei, 430065, PR China
| | - Cheng Ye
- Wuhan Customs Technology Center, Qintai Avenue 588, Wuhan, 430050, PR China
| | - Hui Xia
- College of Basic Medicine, Hubei University of Chinese Medicine, Wuhan, Hubei, 430065, PR China
| | - Ming Yang
- State Engineering Technology Institute for Karst Desertification Control, School of Karst Science, Guizhou Normal University, Guiyang, 550001, PR China
| | - Qinghua Gao
- College of Basic Medicine, Hubei University of Chinese Medicine, Wuhan, Hubei, 430065, PR China
| | - Qingxiong Yang
- State Engineering Technology Institute for Karst Desertification Control, School of Karst Science, Guizhou Normal University, Guiyang, 550001, PR China.
| | - Hongtao Liu
- College of Basic Medicine, Hubei University of Chinese Medicine, Wuhan, Hubei, 430065, PR China; Chongqing Academy of Chinese Materia Medica, Nanshan Road 34, Chongqing, 400065, PR China.
| |
Collapse
|
12
|
Manzo T, Prentice BM, Anderson KG, Raman A, Schalck A, Codreanu GS, Nava Lauson CB, Tiberti S, Raimondi A, Jones MA, Reyzer M, Bates BM, Spraggins JM, Patterson NH, McLean JA, Rai K, Tacchetti C, Tucci S, Wargo JA, Rodighiero S, Clise-Dwyer K, Sherrod SD, Kim M, Navin NE, Caprioli RM, Greenberg PD, Draetta G, Nezi L. Accumulation of long-chain fatty acids in the tumor microenvironment drives dysfunction in intrapancreatic CD8+ T cells. J Exp Med 2021; 217:151833. [PMID: 32491160 PMCID: PMC7398173 DOI: 10.1084/jem.20191920] [Citation(s) in RCA: 195] [Impact Index Per Article: 48.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 01/14/2020] [Accepted: 03/19/2020] [Indexed: 12/13/2022] Open
Abstract
CD8+ T cells are master effectors of antitumor immunity, and their presence at tumor sites correlates with favorable outcomes. However, metabolic constraints imposed by the tumor microenvironment (TME) can dampen their ability to control tumor progression. We describe lipid accumulation in the TME areas of pancreatic ductal adenocarcinoma (PDA) populated by CD8+ T cells infiltrating both murine and human tumors. In this lipid-rich but otherwise nutrient-poor TME, access to using lipid metabolism becomes particularly valuable for sustaining cell functions. Here, we found that intrapancreatic CD8+ T cells progressively accumulate specific long-chain fatty acids (LCFAs), which, rather than provide a fuel source, impair their mitochondrial function and trigger major transcriptional reprogramming of pathways involved in lipid metabolism, with the subsequent reduction of fatty acid catabolism. In particular, intrapancreatic CD8+ T cells specifically exhibit down-regulation of the very-long-chain acyl-CoA dehydrogenase (VLCAD) enzyme, which exacerbates accumulation of LCFAs and very-long-chain fatty acids (VLCFAs) that mediate lipotoxicity. Metabolic reprogramming of tumor-specific T cells through enforced expression of ACADVL enabled enhanced intratumoral T cell survival and persistence in an engineered mouse model of PDA, overcoming one of the major hurdles to immunotherapy for PDA.
Collapse
Affiliation(s)
- Teresa Manzo
- Department of Experimental Oncology, IRCCS European Institute of Oncology, Milano, Italy.,Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Boone M Prentice
- Department of Biochemistry, Mass Spectrometry Research Center, Department of Chemistry, Department of Pharmacology and Medicine, Vanderbilt University, Nashville, TN
| | - Kristin G Anderson
- Clinical Research Division and Program in Immunology, Fred Hutchinson Cancer Research Center, Seattle, WA.,Departments of Medicine/Oncology and Immunology, University of Washington School of Medicine, Seattle, WA
| | - Ayush Raman
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Aislyn Schalck
- Department of Genetics and Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | | | - Carina B Nava Lauson
- Department of Experimental Oncology, IRCCS European Institute of Oncology, Milano, Italy
| | - Silvia Tiberti
- Department of Experimental Oncology, IRCCS European Institute of Oncology, Milano, Italy
| | - Andrea Raimondi
- Experimental Imaging Center, IRCCS San Raffaele Scientific Institute, San Raffaele Vita-Salute University, Milano, Italy
| | - Marissa A Jones
- Department of Biochemistry, Mass Spectrometry Research Center, Department of Chemistry, Department of Pharmacology and Medicine, Vanderbilt University, Nashville, TN
| | - Michelle Reyzer
- Department of Biochemistry, Mass Spectrometry Research Center, Department of Chemistry, Department of Pharmacology and Medicine, Vanderbilt University, Nashville, TN
| | - Breanna M Bates
- Clinical Research Division and Program in Immunology, Fred Hutchinson Cancer Research Center, Seattle, WA.,Departments of Medicine/Oncology and Immunology, University of Washington School of Medicine, Seattle, WA
| | - Jeffrey M Spraggins
- Department of Biochemistry, Mass Spectrometry Research Center, Department of Chemistry, Department of Pharmacology and Medicine, Vanderbilt University, Nashville, TN
| | - Nathan H Patterson
- Department of Biochemistry, Mass Spectrometry Research Center, Department of Chemistry, Department of Pharmacology and Medicine, Vanderbilt University, Nashville, TN
| | - John A McLean
- Center for Innovative Technology, Vanderbilt University, Nashville, TN
| | - Kunal Rai
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Carlo Tacchetti
- Experimental Imaging Center, IRCCS San Raffaele Scientific Institute, San Raffaele Vita-Salute University, Milano, Italy
| | - Sara Tucci
- Laboratory of Clinical Biochemistry and Metabolism Center for Pediatrics and Adolescent Medicine, University of Freiburg, Freiburg, Germany
| | - Jennifer A Wargo
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX.,Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Simona Rodighiero
- Department of Experimental Oncology, IRCCS European Institute of Oncology, Milano, Italy
| | - Karen Clise-Dwyer
- Department of Stem Cell Transplantation, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Stacy D Sherrod
- Center for Innovative Technology, Vanderbilt University, Nashville, TN
| | - Michael Kim
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Nicholas E Navin
- Department of Genetics and Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Richard M Caprioli
- Department of Biochemistry, Mass Spectrometry Research Center, Department of Chemistry, Department of Pharmacology and Medicine, Vanderbilt University, Nashville, TN
| | - Philip D Greenberg
- Clinical Research Division and Program in Immunology, Fred Hutchinson Cancer Research Center, Seattle, WA.,Departments of Medicine/Oncology and Immunology, University of Washington School of Medicine, Seattle, WA
| | - Giulio Draetta
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Luigi Nezi
- Department of Experimental Oncology, IRCCS European Institute of Oncology, Milano, Italy.,Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX
| |
Collapse
|
13
|
Lund M, Andersen KG, Heaton R, Hargreaves IP, Gregersen N, Olsen RKJ. Bezafibrate activation of PPAR drives disturbances in mitochondrial redox bioenergetics and decreases the viability of cells from patients with VLCAD deficiency. Biochim Biophys Acta Mol Basis Dis 2021; 1867:166100. [PMID: 33549744 DOI: 10.1016/j.bbadis.2021.166100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 01/11/2021] [Accepted: 02/01/2021] [Indexed: 10/22/2022]
Abstract
Very long-chain acyl-CoA dehydrogenase (VLCAD) deficiency is the most common inborn long-chain fatty acid oxidation (FAO) disorder. VLCAD deficiency is characterized by distinct phenotypes. The severe phenotypes are potentially life-threatening and affect the heart or liver, with a comparatively milder phenotype characterized by myopathic symptoms. There is an unmet clinical need for effective treatment options for the myopathic phenotype. The molecular mechanisms driving the gradual decrease in mitochondrial function and associated alterations of muscle fibers are unclear. The peroxisome proliferator-activated receptor (PPAR) pan-agonist bezafibrate is a potent modulator of FAO and multiple other mitochondrial functions and has been proposed as a potential medication for myopathic cases of long-chain FAO disorders. In vitro experiments have demonstrated the ability of bezafibrate to increase VLCAD expression and activity. However, the outcome of small-scale clinical trials has been controversial. We found VLCAD deficient patient fibroblasts to have an increased oxidative stress burden and deranged mitochondrial bioenergetic capacity, compared to controls. Applying heat stress under fasting conditions to bezafibrate pretreated patient cells, caused a marked further increase of mitochondrial superoxide levels. Patient cells failed to maintain levels of the essential thiol peptide antioxidant glutathione and experienced a decrease in cellular viability. Our findings indicate that chronic PPAR activation is a plausible initiator of long-term pathogenesis in VLCAD deficiency. Our findings further implicate disruption of redox homeostasis as a key pathogenic mechanism in VLCAD deficiency and support the notion that a deranged thiol metabolism might be an important pathogenic factor in VLCAD deficiency.
Collapse
Affiliation(s)
- Martin Lund
- Research Unit for Molecular Medicine, Department of Clinical Medicine, Aarhus University and Aarhus University Hospital, Palle Juel-Jensens Boulevard 99, 8200 Aarhus, Denmark
| | - Kathrine G Andersen
- Research Unit for Molecular Medicine, Department of Clinical Medicine, Aarhus University and Aarhus University Hospital, Palle Juel-Jensens Boulevard 99, 8200 Aarhus, Denmark
| | - Robert Heaton
- School of Pharmacy, Liverpool John Moore University, Byrom Street, Liverpool L3 3AF, United Kingdom
| | - Iain P Hargreaves
- School of Pharmacy, Liverpool John Moore University, Byrom Street, Liverpool L3 3AF, United Kingdom
| | - Niels Gregersen
- Research Unit for Molecular Medicine, Department of Clinical Medicine, Aarhus University and Aarhus University Hospital, Palle Juel-Jensens Boulevard 99, 8200 Aarhus, Denmark
| | - Rikke K J Olsen
- Research Unit for Molecular Medicine, Department of Clinical Medicine, Aarhus University and Aarhus University Hospital, Palle Juel-Jensens Boulevard 99, 8200 Aarhus, Denmark.
| |
Collapse
|
14
|
Gaston G, Gangoiti JA, Winn S, Chan B, Barshop BA, Harding CO, Gillingham MB. Cardiac tissue citric acid cycle intermediates in exercised very long-chain acyl-CoA dehydrogenase-deficient mice fed triheptanoin or medium-chain triglyceride. J Inherit Metab Dis 2020; 43:1232-1242. [PMID: 33448436 DOI: 10.1002/jimd.12284] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Revised: 07/01/2020] [Accepted: 07/07/2020] [Indexed: 11/11/2022]
Abstract
Anaplerotic odd-chain fatty acid supplementation has been suggested as an approach to replenish citric acid cycle intermediate (CACi) pools and facilitate adenosine triphosphate (ATP) production in subjects with long-chain fatty acid oxidation disorders, but the evidence that cellular CACi depletion exists and that repletion occurs following anaplerotic substrate supplementation is limited. We exercised very long-chain acyl-CoA dehydrogenase-deficient (VLCAD-/-) and wild-type (WT) mice to exhaustion and collected cardiac tissue for measurement of CACi by targeted metabolomics. In a second experimental group, VLCAD-/- and WT mice that had been fed chow prepared with either medium-chain triglyceride (MCT) oil or triheptanoin for 4 weeks were exercised for 60 minutes. VLCAD-/- mice exhibited lower succinate in cardiac muscle at exhaustion than WT mice suggesting lower CACi in VLCAD-/- with prolonged exercise. In mice fed either MCT or triheptanoin, succinate and malate were greater in VLCAD-/- mice fed triheptanoin compared to VLCAD-/- animals fed MCT but lower than WT mice fed triheptanoin. Long-chain odd acylcarnitines such as C19 were elevated in VLCAD-/- and WT mice fed triheptanoin suggesting some elongation of the heptanoate, but it is unknown what proportion of heptanoate was oxidized vs elongated. Prolonged exercise was associated with decreased cardiac muscle succinate in VLCAD-/- mice in comparison to WT mice. VLCAD-/- fed triheptanoin had increased succinate compared to VLCAD-/- mice fed MCT but lower than WT mice fed triheptanoin. Cardiac CACi were higher following dietary ingestion of an anaplerotic substrate, triheptanoin, in comparison to MCT.
Collapse
Affiliation(s)
- Garen Gaston
- Department of Molecular and Medical Genetics, Oregon Health & Science University, Portland, Oregon, USA
| | - Jon A Gangoiti
- Department of Pediatrics, Genetics Division, Biochemical Genetics Program, University of California San Diego, La Jolla, California, USA
| | - Shelley Winn
- Department of Molecular and Medical Genetics, Oregon Health & Science University, Portland, Oregon, USA
| | - Benjamin Chan
- Biostatistics and Design Program, School of Public Health, Oregon Health & Science University, Portland, Oregon, USA
| | - Bruce A Barshop
- Department of Pediatrics, Genetics Division, Biochemical Genetics Program, University of California San Diego, La Jolla, California, USA
| | - Cary O Harding
- Department of Molecular and Medical Genetics, Oregon Health & Science University, Portland, Oregon, USA
| | - Melanie B Gillingham
- Department of Molecular and Medical Genetics, Oregon Health & Science University, Portland, Oregon, USA
| |
Collapse
|
15
|
Tsuchiya Y, Ozai R, Sugino T, Kawashima K, Kushibiki S, Kim YH, Sato S. Changes in peripheral blood oxidative stress markers and hepatic gene expression related to oxidative stress in Holstein cows with and without subacute ruminal acidosis during the periparturient period. J Vet Med Sci 2020; 82:1529-1536. [PMID: 32893200 PMCID: PMC7653322 DOI: 10.1292/jvms.20-0426] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
We investigated changes in peripheral blood metabolites, oxidative stress markers
(malondialdehyde, potential antioxidant capacity, and glutathione peroxidase [GPX]), and
hepatic gene expression related to oxidative stress in Holstein cows with and without
subacute ruminal acidosis (SARA) during the periparturient period. Eighteen multiparous
Holstein cows were categorized into SARA (n=9) or non-SARA (n=9) groups depending on
whether they developed SARA; reticulo-ruminal pH was <5.6 for more than 3 hr per day,
during the 2 weeks after parturition. Blood and liver tissue samples were collected 3
weeks prepartum and 2 and 6 weeks postpartum, with an additional blood sample collected 0
and 4 weeks postpartum. Blood aspartate transaminase (AST) and nonesterified fatty acid
(NEFA) increased significantly (P<0.05) after parturition in both
groups. GPX activity decreased gradually after parturition in the SARA group. In the SARA
group, gene expression of GPX 1 and microsomal glutathione S-transferase
3 (MGST3) decreased significantly (P<0.05), and
expression of metallothionein 2A increased significantly (P<0.05)
after parturition in the SARA group. Superoxide dismutase 1 and MGST3
decreased significantly (P<0.05) 2 weeks postpartum in the non-SARA
group. Gene expression related to oxidative stress was negatively correlated with AST,
NEFA and total ketone body levels. Therefore, the hepatic gene expression related to
oxidative stress might change associated with a negative energy balance, and might relate
the high oxidative stress in the SARA group during periparturient period.
Collapse
Affiliation(s)
- Yoshiyuki Tsuchiya
- Graduate School of Veterinary Sciences, Iwate University, Morioka, Iwate 020-8550, Japan.,Yamagata Prefectural Agricultural Mutual Aid Association, Tendo, Yamagata 994-8511, Japan
| | - Reiko Ozai
- Cooperative Department of Veterinary Medicine, Faculty of Agriculture, Iwate University, Morioka, Iwate 020-8550, Japan
| | - Toshihisa Sugino
- The Research Center for Animal Science, Graduate School of Integrated Science for Life, Hiroshima University, Higashi-Hiroshima 739-8528, Japan
| | - Kenji Kawashima
- Chiba Prefectural Livestock Research Center, Yachimata, Chiba 289-1113, Japan
| | - Shiro Kushibiki
- Institute of Livestock and Grassland Science, NARO, Tsukuba, Ibaraki 305-0901, Japan
| | - Yo-Han Kim
- Cooperative Department of Veterinary Medicine, Faculty of Agriculture, Iwate University, Morioka, Iwate 020-8550, Japan
| | - Shigeru Sato
- Graduate School of Veterinary Sciences, Iwate University, Morioka, Iwate 020-8550, Japan.,Cooperative Department of Veterinary Medicine, Faculty of Agriculture, Iwate University, Morioka, Iwate 020-8550, Japan
| |
Collapse
|
16
|
Bleeker JC, Visser G, Clarke K, Ferdinandusse S, de Haan FH, Houtkooper RH, IJlst L, Kok IL, Langeveld M, van der Pol WL, de Sain‐van der Velden MGM, Sibeijn‐Kuiper A, Takken T, Wanders RJA, van Weeghel M, Wijburg FA, van der Woude LH, Wüst RCI, Cox PJ, Jeneson JAL. Nutritional ketosis improves exercise metabolism in patients with very long-chain acyl-CoA dehydrogenase deficiency. J Inherit Metab Dis 2020; 43:787-799. [PMID: 31955429 PMCID: PMC7384182 DOI: 10.1002/jimd.12217] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 12/23/2019] [Accepted: 01/14/2020] [Indexed: 12/11/2022]
Abstract
A maladaptive shift from fat to carbohydrate (CHO) oxidation during exercise is thought to underlie myopathy and exercise-induced rhabdomyolysis in patients with fatty acid oxidation (FAO) disorders. We hypothesised that ingestion of a ketone ester (KE) drink prior to exercise could serve as an alternative oxidative substrate supply to boost muscular ATP homeostasis. To establish a rational basis for therapeutic use of KE supplementation in FAO, we tested this hypothesis in patients deficient in Very Long-Chain acyl-CoA Dehydrogenase (VLCAD). Five patients (range 17-45 y; 4 M/1F) patients were included in an investigator-initiated, randomised, blinded, placebo-controlled, 2-way cross-over study. Patients drank either a KE + CHO mix or an isocaloric CHO equivalent and performed 35 minutes upright cycling followed by 10 minutes supine cycling inside a Magnetic Resonance scanner at individual maximal FAO work rate (fatmax; approximately 40% VO2 max). The protocol was repeated after a 1-week interval with the alternate drink. Primary outcome measures were quadriceps phosphocreatine (PCr), Pi and pH dynamics during exercise and recovery assayed by in vivo 31 P-MR spectroscopy. Secondary outcomes included plasma and muscle metabolites and respiratory gas exchange recordings. Ingestion of KE rapidly induced mild ketosis and increased muscle BHB content. During exercise at FATMAX, VLCADD-specific plasma acylcarnitine levels, quadriceps glycolytic intermediate levels and in vivo Pi/PCr ratio were all lower in KE + CHO than CHO. These results provide a rational basis for future clinical trials of synthetic ketone ester supplementation therapy in patients with FAO disorders. Trial registration: ClinicalTrials.gov. Protocol ID: NCT03531554; METC2014.492; ABR51222.042.14.
Collapse
Affiliation(s)
- Jeannette C. Bleeker
- Department of Metabolic Diseases, Wilhelmina Children's HospitalUniversity Medical Center UtrechtUtrechtThe Netherlands
- Department of Metabolic Diseases, Emma Children's Hospital, Amsterdam UMCUniversity of AmsterdamAmsterdamThe Netherlands
| | - Gepke Visser
- Department of Metabolic Diseases, Wilhelmina Children's HospitalUniversity Medical Center UtrechtUtrechtThe Netherlands
- Department of Metabolic Diseases, Emma Children's Hospital, Amsterdam UMCUniversity of AmsterdamAmsterdamThe Netherlands
| | - Kieran Clarke
- Department of Physiology, Anatomy and GeneticsUniversity of OxfordOxfordUK
| | - Sacha Ferdinandusse
- Laboratory Genetic Metabolic Diseases, Amsterdam Gastroenterology and Metabolism, Amsterdam UMCUniversity of AmsterdamAmsterdamThe Netherlands
| | - Ferdinand H. de Haan
- ACHIEVE, Center for Applied Research, Faculty of HealthUniversity of Applied Sciences AmsterdamAmsterdamThe Netherlands
| | - Riekelt H. Houtkooper
- Laboratory Genetic Metabolic Diseases, Amsterdam Gastroenterology and Metabolism, Amsterdam UMCUniversity of AmsterdamAmsterdamThe Netherlands
| | - Lodewijk IJlst
- Laboratory Genetic Metabolic Diseases, Amsterdam Gastroenterology and Metabolism, Amsterdam UMCUniversity of AmsterdamAmsterdamThe Netherlands
| | - Irene L. Kok
- Department of Metabolic Diseases, Wilhelmina Children's HospitalUniversity Medical Center UtrechtUtrechtThe Netherlands
| | - Mirjam Langeveld
- Department of Endocrinology and Metabolism, Amsterdam UMCUniversity of AmsterdamAmsterdamThe Netherlands
| | - W. Ludo van der Pol
- Department of Neurology and Neurosurgery, Rudolf Magnus Institute of Neuroscience, Spieren voor Spieren KindercentrumUniversity Medical Center UtrechtUtrechtThe Netherlands
| | | | - Anita Sibeijn‐Kuiper
- Neuroimaging Center, Department of Biomedical Sciences of Cells and SystemsUniversity Medical Center GroningenGroningenThe Netherlands
| | - Tim Takken
- Center for Child Development & Exercise, Department of Medical PhysiologyUniversity Medical Center UtrechtUtrechtThe Netherlands
| | - Ronald J. A. Wanders
- Department of Metabolic Diseases, Emma Children's Hospital, Amsterdam UMCUniversity of AmsterdamAmsterdamThe Netherlands
- Laboratory Genetic Metabolic Diseases, Amsterdam Gastroenterology and Metabolism, Amsterdam UMCUniversity of AmsterdamAmsterdamThe Netherlands
| | - Michel van Weeghel
- Laboratory Genetic Metabolic Diseases, Amsterdam Gastroenterology and Metabolism, Amsterdam UMCUniversity of AmsterdamAmsterdamThe Netherlands
- Laboratory Genetic Metabolic Diseases, Amsterdam UMCUniversity of Amsterdam, Amsterdam Cardiovascular SciencesAmsterdamThe Netherlands
- Core Facility Metabolomics, Amsterdam UMCUniversity of AmsterdamAmsterdamThe Netherlands
| | - Frits A. Wijburg
- Department of Metabolic Diseases, Emma Children's Hospital, Amsterdam UMCUniversity of AmsterdamAmsterdamThe Netherlands
| | - Luc H. van der Woude
- Human Movement SciencesUniversity Medical Center GroningenGroningenThe Netherlands
| | - Rob C. I. Wüst
- Laboratory Genetic Metabolic Diseases, Amsterdam Gastroenterology and Metabolism, Amsterdam UMCUniversity of AmsterdamAmsterdamThe Netherlands
| | - Pete J. Cox
- Department of Physiology, Anatomy and GeneticsUniversity of OxfordOxfordUK
| | - Jeroen A. L. Jeneson
- Neuroimaging Center, Department of Biomedical Sciences of Cells and SystemsUniversity Medical Center GroningenGroningenThe Netherlands
- Center for Child Development & Exercise, Department of Medical PhysiologyUniversity Medical Center UtrechtUtrechtThe Netherlands
- Department of Radiology, Amsterdam UMCUniversity of AmsterdamAmsterdamThe Netherlands
| |
Collapse
|
17
|
Wang S, Leng J, Diao C, Wang Y, Zheng R. Genetic characteristics and follow-up of patients with fatty acid β-oxidation disorders through expanded newborn screening in a Northern Chinese population. J Pediatr Endocrinol Metab 2020; 33:683-690. [PMID: 32447334 DOI: 10.1515/jpem-2019-0551] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2019] [Accepted: 03/16/2020] [Indexed: 12/22/2022]
Abstract
Background Fatty acid β-oxidation disorders (FAODs) include more than 15 distinct disorders and have a wide variety of symptoms, usually not evident between episodes of acute decompensation. After the introduction of newborn screening (NBS) using tandem mass spectrometry (MS/MS), early identification of FAODs has become feasible. We analyzed the MS/MS results in Tianjin, China during a six-year period to evaluate the incidence, disease spectrum, and genetic characteristics of FAODs. Methods We analyzed the MS/MS results for screening FAODs from May 2013 to December 2018 in Tianjin, China. Infants with positive screening results were confirmed through next-generation sequencing and validated by Sanger sequencing. Results A total of 220,443 infants were screened and 25 FAODs patients were identified (1:8,817). Primary carnitine deficiency (PCD) with an incidence rate up to 1:20,040 was the most common disorder among all FAODs. Recurrent mutations of relatively common diseases, like PCD and short-chain acyl-CoA dehydrogenase deficiency (SCADD), were identified. During the follow-up, two patients suffered from sudden death due to carnitine palmitoyl transferase-Ⅱ deficiency (CPT Ⅱ) and very-long-chain acyl-CoA dehydrogenase deficiency (VLCAD). Conclusion Our data indicated that FAODs are relatively common in Tianjin and may even cause infant death in certain cases. The elucidated disease spectrum and genetic backgrounds elucidated in this study may contribute to the treatment and prenatal genetic counseling of FAODs.
Collapse
Affiliation(s)
- Shuting Wang
- Pediatric Department, Tianjin Medical University General Hospital, Tianjin, PR China
- Tianjin Women and Children's Health Center, Tianjin, PR China
| | - Junhong Leng
- Tianjin Women and Children's Health Center, Tianjin, PR China
| | - Chengming Diao
- Tianjin Women and Children's Health Center, Tianjin, PR China
| | - Yuan Wang
- Tianjin Medical Laboratory, BGI-Tianjin, BGI-Shenzhen, Tianjin, PR China
- Binhai Genomics Institute, BGI-Tianjin, BGI-Shenzhen, Tianjin, PR China
| | - Rongxiu Zheng
- Pediatric Department, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, 300052, Tianjin, PR China
| |
Collapse
|
18
|
Wehbe Z, Behringer S, Alatibi K, Watkins D, Rosenblatt D, Spiekerkoetter U, Tucci S. The emerging role of the mitochondrial fatty-acid synthase (mtFASII) in the regulation of energy metabolism. Biochim Biophys Acta Mol Cell Biol Lipids 2019; 1864:1629-1643. [PMID: 31376476 DOI: 10.1016/j.bbalip.2019.07.012] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Revised: 07/17/2019] [Accepted: 07/28/2019] [Indexed: 11/30/2022]
Abstract
Malonyl-CoA synthetase (ACSF3) catalyzes the first step of the mitochondrial fatty acid biosynthesis (mtFASII). Mutations in ACSF3 cause CMAMMA a rare inborn error of metabolism. The clinical phenotype is very heterogeneous, with some patients presenting with neurologic manifestations. In some children, presenting symptoms such as coma, ketoacidosis and hypoglycemia are suggestive of an intermediary metabolic disorder. The overall pathophysiological mechanisms are not understood. In order to study the role of mtFASII in the regulation of energy metabolism we performed a comprehensive metabolic phenotyping with Seahorse technology proteomics in fibroblasts from healthy controls and ACSF3 patients. SILAC-based proteomics and lipidomic analysis were performed to investigate the effects of hypofunctional mtFASII on proteome and lipid homeostasis of complex lipids. Our data clearly confirmed an impaired mitochondrial flexibility characterized by reduced mitochondrial respiration and glycolytic flux due to a lower lipoylation degree. These findings were accompanied by the adaptational upregulation of β-oxidation and by the reduction of anaplerotic amino acids as compensatory mechanism to address the required energy need. Finally, lipidomic analysis demonstrated that the content of the bioactive lipids sphingomyelins and cardiolipins was strongly increased. Our data clearly demonstrate the role of mtFASII in metabolic regulation. Moreover, we show that mtFASII acts as mediator in the lipid-mediated signaling processes in the regulation of energy homeostasis and metabolic flexibility.
Collapse
Affiliation(s)
- Zeinab Wehbe
- Department of General Pediatrics, Center for Pediatrics and Adolescent Medicine, Faculty of Medicine and Medical Centre, University of Freiburg, 79106 Freiburg, Germany; University of Freiburg, Faculty of Biology, Schaenzlestrasse 1, D-79104 Freiburg, Germany
| | - Sidney Behringer
- Department of General Pediatrics, Center for Pediatrics and Adolescent Medicine, Faculty of Medicine and Medical Centre, University of Freiburg, 79106 Freiburg, Germany
| | - Khaled Alatibi
- Department of General Pediatrics, Center for Pediatrics and Adolescent Medicine, Faculty of Medicine and Medical Centre, University of Freiburg, 79106 Freiburg, Germany; University of Freiburg, Faculty of Biology, Schaenzlestrasse 1, D-79104 Freiburg, Germany
| | - David Watkins
- Department of Human Genetics, McGill University and Research Institute McGill University Health Centre, H4A 3J1 Montreal, Quebec, Canada
| | - David Rosenblatt
- Department of Human Genetics, McGill University and Research Institute McGill University Health Centre, H4A 3J1 Montreal, Quebec, Canada
| | - Ute Spiekerkoetter
- Department of General Pediatrics, Center for Pediatrics and Adolescent Medicine, Faculty of Medicine and Medical Centre, University of Freiburg, 79106 Freiburg, Germany
| | - Sara Tucci
- Department of General Pediatrics, Center for Pediatrics and Adolescent Medicine, Faculty of Medicine and Medical Centre, University of Freiburg, 79106 Freiburg, Germany.
| |
Collapse
|
19
|
Herrera-Olivares AM, Fernández-Luque JA, Paradas C, Lucia A, Santalla A. Combined HIIT and Resistance Training in Very Long-Chain Acyl-CoA Dehydrogenase Deficiency: A Case Report. Front Physiol 2019; 10:650. [PMID: 31191348 PMCID: PMC6547021 DOI: 10.3389/fphys.2019.00650] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2019] [Accepted: 05/08/2019] [Indexed: 12/13/2022] Open
Abstract
Very long-chain acyl-CoA dehydrogenase deficiency (VLCADD) is a rare disorder of mitochondrial fatty acid β-oxidation characterized by a spectrum of clinical manifestations. Patients with the adult-onset form can present with muscle pain, rhabdomyolysis and myoglobinuria after physiological stress, such as fasting and exercise. We report on a 23-year-old female patient with a history of recurrent rhabdomyolysis. The patient completed a 6-month supervised combined (high-intensity interval training [HIIT] + resistance training) program, with the addition of a medium chain triglyceride + carbohydrate supplement provided 60 min before each session. The HIIT consisted of 6 sets of 70–80 s performed at maximum intensity with a minimum cadence of 100 rpm. Resistance training consisted of a circuit of basic exercises with dumbbells and elastic bands, with sets of 4–7 repetitions. The patient was evaluated at months 0, 3 and 6 using an incremental discontinuous step protocol, with steps of 1 min of exercise/1 min of passive recovery, at a high pedal cadence. The test started at 10 W, with a load increase of 10 W/step. Blood creatine kinase (CK) concentration was measured before each evaluation. There was a training-induced increment of 90.2% in peak oxygen uptake (VO2peak), 71.4% in peak power output and 24.7% in peak heart rate. The patient reported no muscle pain, contractures, rhabdomyolysis (basal CK concentration was always <200 U/L) or hospital admissions during the training period. After completion of 6-month program, the patient remained active, doing similar but non-supervised training for 1.5 years (to date). During this period, the patient has not reported myalgias, contractures, rhabdomyolysis or hospital admissions. Our preliminary data suggest that it is possible to carry out a combined (HIIT + strength) training program in patients with VLCADD, safely (without muscle contractures or rhabdomyolysis) and obtaining high values of VO2peak and cycling power output.
Collapse
Affiliation(s)
| | | | - Carmen Paradas
- Neuromuscular Disorders Unit, Department of Neurology, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío, CSIC-Universidad de Sevilla, Seville, Spain.,Biomedical Network Research Centre on Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Alejandro Lucia
- Faculty of Sport Sciences, Universidad Europea de Madrid, Madrid, Spain.,Instituto de Investigación Hospital 12 de Octubre (i+12), Madrid, Spain
| | - Alfredo Santalla
- Faculty of Sport Sciences, Universidad Pablo de Olavide, Seville, Spain.,Instituto de Investigación Hospital 12 de Octubre (i+12), Madrid, Spain
| |
Collapse
|
20
|
Janeiro P, Jotta R, Ramos R, Florindo C, Ventura FV, Vilarinho L, Tavares de Almeida I, Gaspar A. Follow-up of fatty acid β-oxidation disorders in expanded newborn screening era. Eur J Pediatr 2019; 178:387-394. [PMID: 30617651 DOI: 10.1007/s00431-018-03315-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2018] [Revised: 12/21/2018] [Accepted: 12/26/2018] [Indexed: 01/25/2023]
Abstract
Fatty acid β-oxidation (FAO) disorders have a wide variety of symptoms, not usually evident between episodes of acute decompensations. Cardiac involvement is frequent, and severe ventricular arrhythmias are suspected of causing sudden death. Expanded newborn screening (ENS) for these disorders, hopefully, contribute to prevent potentially acute life-threatening events. In order to characterize acute decompensations observed in FAO-deficient cases identified by ENS, a retrospective analysis was performed, covering a period of 9 years. Demographic data, number/type of acute decompensations, treatment, and follow-up were considered. Eighty-three clinical charts, including 66 medium-chain acyl-CoA dehydrogenase deficiency (MCADD), 5 carnitine-uptake deficiency (CUD), 3 carnitine palmitoyltransferase I and II (CPT I/II) deficiency, 5 very long-chain acyl-CoA dehydrogenase deficiency (VLCADD), and 4 multiple acyl-CoA dehydrogenase deficiency (MADD) cases were reviewed. Nineteen patients had acute decompensations (1 CPT I, 1 CPT II, 3 MADD, 14 MCADD). Six patients developed symptoms previously to ENS diagnosis. Severe clinical manifestations included multiple organ failure, liver failure, heart failure, and sudden death. Long-chain FAO disorders had the highest number of decompensations per patient.Conclusion: Despite earlier diagnosis by ENS, sudden deaths were not avoided and acute decompensations with severe clinical manifestations still occur as well. What is Known: • Severe ventricular arrhythmias are suspected to cause unexpected death in FAO disorders. • Neonatal screening intends to reduce the incidence of severe metabolic crisis and death. What is New: • Acute severe decompensations occurred in FAO disorders diagnosed through neonatal screening. • Sudden deaths were not avoided by starting treatment precociously.
Collapse
Affiliation(s)
- Patrícia Janeiro
- Centro de Referência de Doenças Hereditárias do Metabolismo, Departamento de Pediatria Médica, Hospital de Santa Maria - CHULN, Av. Prof. Egas Moniz, 1649-035, Lisbon, Portugal.
| | - Rita Jotta
- Serviço de Pediatria Médica, Departamento de Pediatria, Hospital de Santa Maria - CHULN, Av. Prof. Egas Moniz, 1649-035, Lisbon, Portugal
| | - Ruben Ramos
- Laboratório de Metabolismos e Genética, Faculdade de Farmácia da Universidade de Lisboa, Av. Prof. Gama Pinto Edificio F, 1649-099, Lisbon, Portugal
| | - Cristina Florindo
- Laboratório de Metabolismos e Genética, Faculdade de Farmácia da Universidade de Lisboa, Av. Prof. Gama Pinto Edificio F, 1649-099, Lisbon, Portugal
| | - Fátima V Ventura
- Laboratório de Metabolismos e Genética, Faculdade de Farmácia da Universidade de Lisboa, Av. Prof. Gama Pinto Edificio F, 1649-099, Lisbon, Portugal
| | - Laura Vilarinho
- Unidade de Rastreio Neonatal Metabolismo e Genética, Departamento de Genética Humana, Instituto Nacional de Saúde Dr. Ricardo Jorge, Rua Alexandre Herculano 321, 4000-055, Porto, Portugal
| | - Isabel Tavares de Almeida
- Laboratório de Metabolismos e Genética, Faculdade de Farmácia da Universidade de Lisboa, Av. Prof. Gama Pinto Edificio F, 1649-099, Lisbon, Portugal
| | - Ana Gaspar
- Centro de Referência de Doenças Hereditárias do Metabolismo, Departamento de Pediatria Médica, Hospital de Santa Maria - CHULN, Av. Prof. Egas Moniz, 1649-035, Lisbon, Portugal
| |
Collapse
|
21
|
Tarasenko TN, Cusmano-Ozog K, McGuire PJ. Tissue acylcarnitine status in a mouse model of mitochondrial β-oxidation deficiency during metabolic decompensation due to influenza virus infection. Mol Genet Metab 2018; 125:144-152. [PMID: 30031688 PMCID: PMC6626496 DOI: 10.1016/j.ymgme.2018.06.012] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Revised: 06/22/2018] [Accepted: 06/22/2018] [Indexed: 02/02/2023]
Abstract
Despite judicious monitoring and care, patients with fatty acid oxidation disorders may experience metabolic decompensation due to infection which may result in rhabdomyolysis, cardiomyopathy, hypoglycemia and liver dysfunction and failure. Since clinical studies on metabolic decompensation are dangerous, we employed a preclinical model of metabolic decompensation due to infection. By infecting mice with mouse adapted influenza and using a pair-feeding strategy in a mouse model of long-chain fatty acid oxidation (Acadvl-/-), our goals were to isolate the effects of infection on tissue acylcarnitines and determine how they relate to their plasma counterparts. Applying statistical data reduction techniques (Partial Least Squares-Discriminant Analysis), we were able to identify critical acylcarnitines that were driving differentiation of our experimental groups for all the tissues studied. While plasma displayed increases in metabolites directly related to mouse VLCAD deficiency (e.g. C16 and C18), organs like the heart, muscle and liver also showed involvement of alternative pathways (e.g. medium-chain FAO and ketogenesis), suggesting adaptive measures. Matched correlation analyses showed strong correlations (r > 0.7) between plasma and tissue levels for a small number of metabolites. Overall, our results demonstrate that infection as a stress produces perturbations in metabolism in Acadvl-/- that differ greatly from WT infected and Acadvl-/- pair-fed controls. This model system will be useful for studying the effects of infection on tissue metabolism as well as evaluating interventions aimed at modulating the effects of metabolic decompensation.
Collapse
Affiliation(s)
- Tatiana N Tarasenko
- Metabolism, Infection and Immunity Section, National Human Genome Research Institute, National Institutes of Health, United States
| | - Kristina Cusmano-Ozog
- Rare Disease Institute, Children's National Medical Center, Washington, DC, United States
| | - Peter J McGuire
- Metabolism, Infection and Immunity Section, National Human Genome Research Institute, National Institutes of Health, United States.
| |
Collapse
|
22
|
Tucci S, Mingirulli N, Wehbe Z, Dumit VI, Kirschner J, Spiekerkoetter U. Mitochondrial fatty acid biosynthesis and muscle fiber plasticity in very long-chain acyl-CoA dehydrogenase-deficient mice. FEBS Lett 2018; 592:219-232. [PMID: 29237229 DOI: 10.1002/1873-3468.12940] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Revised: 11/27/2017] [Accepted: 12/04/2017] [Indexed: 12/31/2022]
Abstract
The white skeletal muscle of very long-chain acyl-CoA-dehydrogenase-deficient (VLCAD-/- ) mice undergoes metabolic modification to compensate for defective β-oxidation in a progressive and time-dependent manner by upregulating glucose oxidation. This metabolic regulation seems to be accompanied by morphologic adaptation of muscle fibers toward the glycolytic fiber type II with the concomitant upregulation of mitochondrial fatty acid biosynthesis (mFASII) and lipoic acid biosynthesis. Dietary supplementation of VLCAD-/- mice with different medium-chain triglycerides over 1 year revealed that odd-chain species has no effect on muscle fiber switch, whereas even-chain species inhibit progressive metabolic adaptation. Our study shows that muscle may undergo adaptive mechanisms that are modulated by dietary supplementation. We describe for the first time a concomitant change of mFASII in this muscular adaptation process.
Collapse
Affiliation(s)
- Sara Tucci
- Department of General Pediatrics, Center for Paediatrics and Adolescent Medicine, Faculty of Medicine and Medical Centre, University of Freiburg, Germany
| | - Nadja Mingirulli
- Department of General Pediatrics, Center for Paediatrics and Adolescent Medicine, Faculty of Medicine and Medical Centre, University of Freiburg, Germany
| | - Zeinab Wehbe
- Department of General Pediatrics, Center for Paediatrics and Adolescent Medicine, Faculty of Medicine and Medical Centre, University of Freiburg, Germany
| | - Verónica I Dumit
- Core Facility Proteomics, Center for Biological Systems Analysis (ZBSA), University of Freiburg, Germany
| | - Janbernd Kirschner
- Department of Neuropaediatrics, Center for Paediatrics and Adolescent Medicine, Faculty of Medicine and Medical Centre, University of Freiburg, Germany
| | - Ute Spiekerkoetter
- Department of General Pediatrics, Center for Paediatrics and Adolescent Medicine, Faculty of Medicine and Medical Centre, University of Freiburg, Germany
| |
Collapse
|
23
|
Kang OH, Shon MY, Kong R, Seo YS, Zhou T, Kim DY, Kim YS, Kwon DY. Anti-diabetic effect of black ginseng extract by augmentation of AMPK protein activity and upregulation of GLUT2 and GLUT4 expression in db/db mice. Altern Ther Health Med 2017; 17:341. [PMID: 28662663 PMCID: PMC5492680 DOI: 10.1186/s12906-017-1839-4] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Accepted: 06/15/2017] [Indexed: 12/24/2022]
Abstract
BACKGROUND Black ginseng (Panax ginseng C. A. Meyer), three to nine times-steamed and dried ginseng, has biological and pharmacological activities. In this study, the anti-diabetic effects of the black ginseng ethanol extract (GBG05-FF) in typical type 2 diabetic model db/db mice were investigated. METHODS The effect of GBG05-FF in Type 2 diabetic mice was investigated by their blood analysis, biological mechanism analysis, and histological analysis. RESULTS The mice group treated with GBG05-FF showed decreased fasting blood glucose and glucose tolerance compared to that of the nontreated GBG05-FF group. In the blood analysis, GBG05-FF decreased main plasma parameter such as HbA1c, triglyceride, and total-cholesterol levels related to diabetes and improved the expression of genes and protein related to glucose homeostasis and glucose uptake in the liver and muscle. The histological analysis result shows that GBG05-FF decreased lipid accumulation in the liver and damage in the muscle. Moreover, GBG05-FF increased the phosphorylation of the AMPK in the liver and upregulated the expression of GLUT2 in liver and GLUT4 in muscle. Therefore, the mechanisms of GBG05-FF may be related to suppressing gluconeogenesis by activating AMPK in the liver and affecting glucose uptake in surrounding tissues via the upregulation of GLUT2 and GLUT4 expression. CONCLUSION These findings provided a new insight into the anti-diabetic clinical applications of GBG05-FF and it might play an important role in the development of promising functional foods and drugs from the viewpoint of the chemical composition and biological activities.
Collapse
|
24
|
Tucci S, Floegel U, Beermann F, Behringer S, Spiekerkoetter U. Triheptanoin: long-term effects in the very long-chain acyl-CoA dehydrogenase-deficient mouse. J Lipid Res 2016; 58:196-207. [PMID: 27884962 DOI: 10.1194/jlr.m072033] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Revised: 11/07/2016] [Indexed: 12/31/2022] Open
Abstract
A rather new approach in the treatment of long-chain fatty acid oxidation disorders is represented by triheptanoin, a triglyceride with three medium-odd-chain heptanoic acids (C7), due to its anaplerotic potential. We here investigate the effects of a 1-year triheptanoin-based diet on the clinical phenotype of very long-chain-acyl-CoA-dehydrogenase-deficient (VLCAD-/-) mice. The cardiac function was assessed in VLCAD-/- mice by in vivo MRI. Metabolic adaptations were identified by the expression of genes regulating energy metabolism and anaplerotic processes using real-time PCR, and the results were correlated with the measurement of the glycolytic enzymes pyruvate dehydrogenase and pyruvate kinase. Finally, the intrahepatic lipid accumulation and oxidative stress in response to the long-term triheptanoin diet were assessed. Triheptanoin was not able to prevent the development of systolic dysfunction in VLCAD-/- mice despite an upregulation of cardiac glucose oxidation. Strikingly, the anaplerotic effects of triheptanoin were restricted to the liver. Despite this, the hepatic lipic content was increased upon triheptanoin supplementation. Our data demonstrate that the concept of anaplerosis does not apply to all tissues equally.
Collapse
Affiliation(s)
- Sara Tucci
- Department of General Pediatrics, Center for Pediatrics and Adolescent Medicine, University Hospital Freiburg, Freiburg, Germany
| | - Ulrich Floegel
- Department of Molecular Cardiology, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Frauke Beermann
- Department of General Pediatrics, Center for Pediatrics and Adolescent Medicine, University Hospital Freiburg, Freiburg, Germany
| | - Sidney Behringer
- Department of General Pediatrics, Center for Pediatrics and Adolescent Medicine, University Hospital Freiburg, Freiburg, Germany
| | - Ute Spiekerkoetter
- Department of General Pediatrics, Center for Pediatrics and Adolescent Medicine, University Hospital Freiburg, Freiburg, Germany
| |
Collapse
|
25
|
Wang W, Palmfeldt J, Mohsen AW, Gregersen N, Vockley J. Fasting induces prominent proteomic changes in liver in very long chain Acyl-CoA dehydrogenase deficient mice. Biochem Biophys Rep 2016; 8:333-339. [PMID: 28955973 PMCID: PMC5613767 DOI: 10.1016/j.bbrep.2016.08.014] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2016] [Revised: 07/27/2016] [Accepted: 08/09/2016] [Indexed: 12/26/2022] Open
Abstract
Very long chain acyl-CoA dehydrogenase (VLCAD) deficiency (VLCADD) is a clinically heterogeneous disorder of mitochondrial fatty acid β-oxidation usually identified through newborn screening. Genotype-phenotype correlations have been defined, but considerable clinical heterogeneity still exists. Symptoms are often induced by physiological stress such as fasting or intercurrent illness, setting it as an important example of environmental effects altering clinical course in an individual with a genetic disease. However, neither the cellular changes that predispose to this phenomenon nor the alterations it induces are well characterized. We examined the effects of fasting in a knockout mouse model to explore changes in global mitochondria protein profiles in liver and to investigate the physiologically relevant changes that lead to the clinical presentations. An isobaric tags for relative and absolute quantification (iTRAQ) labeling approach was employed to examine mitochondrial proteome changes in VLCAD deficient compared to wild type mice in the fed and fasted states. We identified numerous proteomic changes associated with the gene defect and fasting within relevant metabolic pathways. Few changes induced by fasting were shared between the VLCAD deficient and wild type mice, with more alterations found in the deficient mice on fasting. Particularly, fasting in the deficient mice could reverse the protective response in oxidative phosphorylation pathway seen in wild type animals. In addition, we found that changes in chaperone proteins including heat shock protein 60 (HSP60) and 10 (HSP10) during fasting differed between the two genotypes, highlighting the importance of these proteins in VLCAD deficiency. Finally, the effects on the liver proteome imposed by changes in fasted VLCAD deficient mice indicates that this environmental factor may be an inducer of both cellular and physiological changes. VLCAD deficient mice show significant changes in the proteome compared to wild type animals. Hypoglycemia induces different proteomic changes in VLCAD deficient mice than is seen in wild type animals. Multiple functional pathways are altered in VLCAD deficient animals. Proteomic changes in VLCAD deficient mice may help understand the physiologic ramifications of this deficiency in humans.
Collapse
Affiliation(s)
- Wei Wang
- Department of Medical Genetics, Shanghai Children's Hospital, Shanghai Jiaotong University, Shanghai, China.,Department of Human Genetics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, USA
| | - Johan Palmfeldt
- Research Unit for Molecular Medicine, Institute of Clinical Medicine, Faculty of Health Sciences, Aarhus University, Aarhus, Denmark
| | - Al-Walid Mohsen
- Department of Pediatrics, School of Medicine, University of Pittsburgh, Pittsburgh, USA
| | - Niels Gregersen
- Research Unit for Molecular Medicine, Institute of Clinical Medicine, Faculty of Health Sciences, Aarhus University, Aarhus, Denmark
| | - Jerry Vockley
- Department of Human Genetics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, USA.,Department of Pediatrics, School of Medicine, University of Pittsburgh, Pittsburgh, USA
| |
Collapse
|
26
|
Altered Energetics of Exercise Explain Risk of Rhabdomyolysis in Very Long-Chain Acyl-CoA Dehydrogenase Deficiency. PLoS One 2016; 11:e0147818. [PMID: 26881790 PMCID: PMC4755596 DOI: 10.1371/journal.pone.0147818] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Accepted: 01/09/2016] [Indexed: 12/31/2022] Open
Abstract
Rhabdomyolysis is common in very long-chain acyl-CoA dehydrogenase deficiency (VLCADD) and other metabolic myopathies, but its pathogenic basis is poorly understood. Here, we show that prolonged bicycling exercise against a standardized moderate workload in VLCADD patients is associated with threefold bigger changes in phosphocreatine (PCr) and inorganic phosphate (Pi) concentrations in quadriceps muscle and twofold lower changes in plasma acetyl-carnitine levels than in healthy subjects. This result is consistent with the hypothesis that muscle ATP homeostasis during exercise is compromised in VLCADD. However, the measured rates of PCr and Pi recovery post-exercise showed that the mitochondrial capacity for ATP synthesis in VLCADD muscle was normal. Mathematical modeling of oxidative ATP metabolism in muscle composed of three different fiber types indicated that the observed altered energy balance during submaximal exercise in VLCADD patients may be explained by a slow-to-fast shift in quadriceps fiber-type composition corresponding to 30% of the slow-twitch fiber-type pool in healthy quadriceps muscle. This study demonstrates for the first time that quadriceps energy balance during exercise in VLCADD patients is altered but not because of failing mitochondrial function. Our findings provide new clues to understanding the risk of rhabdomyolysis following exercise in human VLCADD.
Collapse
|
27
|
Mitochondrial dysfunction in fatty acid oxidation disorders: insights from human and animal studies. Biosci Rep 2015; 36:e00281. [PMID: 26589966 PMCID: PMC4718505 DOI: 10.1042/bsr20150240] [Citation(s) in RCA: 137] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Accepted: 11/20/2015] [Indexed: 12/17/2022] Open
Abstract
Patients affected by FAOD commonly present with hepatopathy, cardiomyopathy, skeletal myopathy and encephalopathy. Human and animal evidences indicate that mitochondrial functions are disrupted by fatty acids and derivatives accumulating in these disorders, suggesting that lipotoxicity may contribute to their pathogenesis. Mitochondrial fatty acid oxidation (FAO) plays a pivotal role in maintaining body energy homoeostasis mainly during catabolic states. Oxidation of fatty acids requires approximately 25 proteins. Inherited defects of FAO have been identified in the majority of these proteins and constitute an important group of inborn errors of metabolism. Affected patients usually present with severe hepatopathy, cardiomyopathy and skeletal myopathy, whereas some patients may suffer acute and/or progressive encephalopathy whose pathogenesis is poorly known. In recent years growing evidence has emerged indicating that energy deficiency/disruption of mitochondrial homoeostasis is involved in the pathophysiology of some fatty acid oxidation defects (FAOD), although the exact underlying mechanisms are not yet established. Characteristic fatty acids and carnitine derivatives are found at high concentrations in these patients and more markedly during episodes of metabolic decompensation that are associated with worsening of clinical symptoms. Therefore, it is conceivable that these compounds may be toxic. We will briefly summarize the current knowledge obtained from patients and genetic mouse models with these disorders indicating that disruption of mitochondrial energy, redox and calcium homoeostasis is involved in the pathophysiology of the tissue damage in the more common FAOD, including medium-chain acyl-CoA dehydrogenase (MCAD), long-chain 3-hydroxyacyl-CoA dehydrogenase (LCHAD) and very long-chain acyl-CoA dehydrogenase (VLCAD) deficiencies. We will also provide evidence that the fatty acids and derivatives that accumulate in these diseases disrupt mitochondrial homoeostasis. The elucidation of the toxic mechanisms of these compounds may offer new perspectives for potential novel adjuvant therapeutic strategies in selected disorders of this group.
Collapse
|
28
|
Abdurrachim D, Luiken JJFP, Nicolay K, Glatz JFC, Prompers JJ, Nabben M. Good and bad consequences of altered fatty acid metabolism in heart failure: evidence from mouse models. Cardiovasc Res 2015; 106:194-205. [PMID: 25765936 DOI: 10.1093/cvr/cvv105] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2014] [Accepted: 02/18/2015] [Indexed: 12/25/2022] Open
Abstract
The shift in substrate preference away from fatty acid oxidation (FAO) towards increased glucose utilization in heart failure has long been interpreted as an oxygen-sparing mechanism. Inhibition of FAO has therefore evolved as an accepted approach to treat heart failure. However, recent data indicate that increased reliance on glucose might be detrimental rather than beneficial for the failing heart. This review discusses new insights into metabolic adaptations in heart failure. A particular focus lies on data obtained from mouse models with modulations of cardiac FA metabolism at different levels of the FA metabolic pathway and how these differently affect cardiac function. Based on studies in which these mouse models were exposed to ischaemic and non-ischaemic heart failure, we discuss whether and when modulations in FA metabolism are protective against heart failure.
Collapse
Affiliation(s)
- Desiree Abdurrachim
- Biomedical NMR, Department of Biomedical Engineering, Eindhoven University of Technology, High Tech Campus 11, 5656 AE, PO BOX 513, Eindhoven 5600 MB, The Netherlands
| | - Joost J F P Luiken
- Department of Genetics and Cell Biology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
| | - Klaas Nicolay
- Biomedical NMR, Department of Biomedical Engineering, Eindhoven University of Technology, High Tech Campus 11, 5656 AE, PO BOX 513, Eindhoven 5600 MB, The Netherlands
| | - Jan F C Glatz
- Department of Genetics and Cell Biology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
| | - Jeanine J Prompers
- Biomedical NMR, Department of Biomedical Engineering, Eindhoven University of Technology, High Tech Campus 11, 5656 AE, PO BOX 513, Eindhoven 5600 MB, The Netherlands
| | - Miranda Nabben
- Biomedical NMR, Department of Biomedical Engineering, Eindhoven University of Technology, High Tech Campus 11, 5656 AE, PO BOX 513, Eindhoven 5600 MB, The Netherlands Department of Genetics and Cell Biology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
| |
Collapse
|
29
|
Tucci S, Krogmann A, Herebian D, Spiekerkoetter U. Renal response to short- and long-term exercise in very-long-chain acyl-CoA dehydrogenase-deficient (VLCAD(-/-)) mice. Mol Cell Pediatr 2014; 1:5. [PMID: 26567099 PMCID: PMC4530567 DOI: 10.1186/s40348-014-0005-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2014] [Accepted: 07/22/2014] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Deficiency of very long-chain acyl-CoA dehydrogenase (VLCAD) is the most common disorder of mitochondrial β-oxidation of long-chain fatty acids. In order to maintain glucose homeostasis, the kidney and liver as the main gluconeogenic organs play an important role under conditions of impaired fatty acid oxidation. However, little is known about how a defective fatty acid oxidation machinery affects renal metabolism and function as well as renal energy supply especially during catabolic situations. METHODS In this study, we analyzed VLCAD(-/-) mice under different metabolic conditions such as after moderate (1 h) and intensive long-term (1 h twice per day over 2 weeks) physical exercise and after 24 h of fasting. We measured the oxidation rate of palmitoyl-CoA (C16-CoA) as well as the expression of genes involved in lipogenesis and renal failure. Oxidative stress was assessed by the function of antioxidant enzymes. Moreover, we quantified the content of glycogen and long-chain acylcarnitines in the kidney. RESULTS We observed a significant depletion in renal glycogen with a concomitant reduction in long-chain acylcarnitines, suggesting a substrate switch for energy production and an optimal compensation of impaired fatty acid oxidation in the kidney. In fact, the mutants did not show any signs of oxidative stress or renal failure under catabolic conditions. CONCLUSIONS Our data demonstrate that despite Acadvl ablation, the kidney of VLCAD(-/-) mice fully compensates for impaired fatty acid oxidation by enhanced glycogen utilization and preserves renal energy metabolism and function.
Collapse
Affiliation(s)
- Sara Tucci
- Department of General Pediatrics, Center for Pediatrics and Adolescent Medicine, University Hospital Freiburg, Mathildenstrasse 1, Freiburg, 79106, Germany.
| | - Antonia Krogmann
- Department of General Pediatrics, Neonatology and Children's Cardiology, University Children's Hospital, Duesseldorf, 40225, Germany.
| | - Diran Herebian
- Department of General Pediatrics, Neonatology and Children's Cardiology, University Children's Hospital, Duesseldorf, 40225, Germany.
| | - Ute Spiekerkoetter
- Department of General Pediatrics, Center for Pediatrics and Adolescent Medicine, University Hospital Freiburg, Mathildenstrasse 1, Freiburg, 79106, Germany.
| |
Collapse
|
30
|
Tucci S, Flögel U, Hermann S, Sturm M, Schäfers M, Spiekerkoetter U. Development and pathomechanisms of cardiomyopathy in very long-chain acyl-CoA dehydrogenase deficient (VLCAD(-/-)) mice. Biochim Biophys Acta Mol Basis Dis 2014; 1842:677-85. [PMID: 24530811 DOI: 10.1016/j.bbadis.2014.02.001] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2013] [Revised: 01/15/2014] [Accepted: 02/04/2014] [Indexed: 12/24/2022]
Abstract
Hypertrophic cardiomyopathy is a typical manifestation of very long-chain acyl-CoA dehydrogenase deficiency (VLCADD), the most common long-chain β-oxidation defects in humans; however in some patients cardiac function is fully compensated. Cardiomyopathy may also be reversed by supplementation of medium-chain triglycerides (MCT). We here characterize cardiac function of VLCAD-deficient (VLCAD(-/-)) mice over one year. Furthermore, we investigate the long-term effect of a continuous MCT diet on the cardiac phenotype. We assessed cardiac morphology and function in VLCAD(-/-) mice by in vivo MRI. Cardiac energetics were measured by (31)P-MRS and myocardial glucose uptake was quantified by positron-emission-tomography (PET). Metabolic adaptations were identified by the expression of genes regulating glucose and lipid metabolism using real-time-PCR. VLCAD(-/-) mice showed a progressive decrease in heart function over 12 months accompanied by a reduced phosphocreatine-to-ATP-ratio indicative of chronic energy deficiency. Long-term MCT supplementation aggravated the cardiac phenotype into dilated cardiomyopathy with features similar to diabetic heart disease. Cardiac energy production and function in mice with a β-oxidation defect cannot be maintained with age. Compensatory mechanisms are insufficient to preserve the cardiac energy state over time. However, energy deficiency by impaired β-oxidation and long-term MCT induce cardiomyopathy by different mechanisms. Cardiac MRI and MRS may be excellent tools to assess minor changes in cardiac function and energetics in patients with β-oxidation defects for preventive therapy.
Collapse
Affiliation(s)
- Sara Tucci
- Department of General Pediatrics, Center for Pediatrics and Adolescent Medicine, University Hospital Freiburg, 79106 Freiburg, Germany; Department of General Pediatrics, University Children's Hospital Duesseldorf, 40225 Duesseldorf, Germany.
| | - Ulrich Flögel
- Department of Molecular Cardiology, Heinrich Heine University Duesseldorf, 40225 Duesseldorf, Germany
| | - Sven Hermann
- European Institute for Molecular Imaging - EIMI, University of Muenster, 48149 Muenster, Germany
| | - Marga Sturm
- Department of General Pediatrics, University Children's Hospital Duesseldorf, 40225 Duesseldorf, Germany
| | - Michael Schäfers
- European Institute for Molecular Imaging - EIMI, University of Muenster, 48149 Muenster, Germany
| | - Ute Spiekerkoetter
- Department of General Pediatrics, Center for Pediatrics and Adolescent Medicine, University Hospital Freiburg, 79106 Freiburg, Germany
| |
Collapse
|
31
|
Pereira HABDS, Leite ADL, Charone S, Lobo JGVM, Cestari TM, Peres-Buzalaf C, Buzalaf MAR. Proteomic analysis of liver in rats chronically exposed to fluoride. PLoS One 2013; 8:e75343. [PMID: 24069403 PMCID: PMC3775814 DOI: 10.1371/journal.pone.0075343] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2013] [Accepted: 08/12/2013] [Indexed: 12/15/2022] Open
Abstract
Fluoride (F) is a potent anti-cariogenic element, but when ingestion is excessive, systemic toxicity may be observed. This can occur as acute or chronic responses, depending on both the amount of F and the time of exposure. The present study identified the profile of protein expression possibly associated with F-induced chronic hepatotoxicity. Weanling male Wistar rats (three-weeks old) were divided into three groups and treated with drinking water containing 0, 5 or 50 mg/L F for 60 days (n=6/group). At this time point, serum and livers were collected for F analysis, which was done using the ion-sensitive electrode, after hexamethyldisiloxane-facilitated diffusion. Livers were also submitted to histological and proteomic analyses (2D-PAGE followed by LC-MS/MS). Western blotting was done for confirmation of the proteomic data A dose-response was observed in serum F levels. In the livers, F levels were significantly increased in the 50 mg/L F group compared to groups treated with 0 and 5 mg/L F. Liver morphometric analysis did not reveal alterations in the cellular structures and lipid droplets were present in all groups. Proteomic quantitative intensity analysis detected 33, 44, and 29 spots differentially expressed in the comparisons between control vs. 5 mg/L F, control vs. 50 mg/L F, and 5 mg/L vs. 50 mg/L F, respectively. From these, 92 proteins were successfully identified. In addition, 18, 1, and 5 protein spots were shown to be exclusive in control, 5, and 50 mg/L F, respectively. Most of proteins were related to metabolic process and pronounced alterations were seen for the high-F level group. In F-treated rats, changes in the apolipoprotein E (ApoE) and GRP-78 expression may account for the F-induced toxicity in the liver. This can contribute to understanding the molecular mechanisms underlying hepatoxicity induced by F, by indicating key-proteins that should be better addressed in future studies.
Collapse
Affiliation(s)
| | - Aline de Lima Leite
- Department of Biological Sciences, Bauru Dental School, University of São Paulo, Bauru, São Paulo, Brazil
| | - Senda Charone
- Department of Biological Sciences, Bauru Dental School, University of São Paulo, Bauru, São Paulo, Brazil
| | | | - Tania Mary Cestari
- Department of Biological Sciences, Bauru Dental School, University of São Paulo, Bauru, São Paulo, Brazil
| | - Camila Peres-Buzalaf
- Department of Biological Sciences, Bauru Dental School, University of São Paulo, Bauru, São Paulo, Brazil
| | | |
Collapse
|
32
|
Regulation of mitochondrial fatty acid β-oxidation in human: what can we learn from inborn fatty acid β-oxidation deficiencies? Biochimie 2013; 96:113-20. [PMID: 23764392 DOI: 10.1016/j.biochi.2013.05.012] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2013] [Accepted: 05/30/2013] [Indexed: 12/31/2022]
Abstract
The mitochondrial fatty acid β-oxidation (FAO) pathway plays a crucial role in ATP production in many tissues with high-energy demand. This is highlighted by the diverse and possibly severe clinical manifestations of inborn fatty acid β-oxidation deficiencies. More than fifteen genetic FAO enzyme defects have been described to date, forming a large group of rare diseases. Inborn FAO disorders are characterized by a high genetic heterogeneity, with a variety of gene mutations resulting in complete or partial loss-of-function of the corresponding enzyme. The panel of observed phenotypes varies from multi-organ failure in the neonate with fatal outcome, up to milder late onset manifestations associated with significant disabilities. Diagnosis of FAO disorders has markedly improved over the last decades, but few treatments are available. The clinical, biochemical, and molecular analysis of these disorders provided new, and sometimes unexpected, data on the organization and regulation of mitochondrial FAO in humans, in various tissues, and at various stages of development. This will be illustrated by examples of FAO defects affecting enzymes of long-chain fatty acid import into the mitochondria, or Lynen helix enzymes. The involvement of the transcriptional network regulating FAO gene expression, in particular the PGC-1α/PPAR axis, as a target for pharmacological therapy of these genetic disorders, will also be discussed.
Collapse
|
33
|
Tucci S, Pearson S, Herebian D, Spiekerkoetter U. Long-term dietary effects on substrate selection and muscle fiber type in very-long-chain acyl-CoA dehydrogenase deficient (VLCAD(-/-)) mice. Biochim Biophys Acta Mol Basis Dis 2013; 1832:509-16. [PMID: 23313579 DOI: 10.1016/j.bbadis.2013.01.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2012] [Revised: 12/28/2012] [Accepted: 01/04/2013] [Indexed: 12/11/2022]
Abstract
Dietary fat restriction and increased carbohydrate intake are part of treatment in very-long-chain acyl-CoA dehydrogenase (VLCAD)-deficiency, the most common defect of long-chain fatty acid oxidation. The long-term impact of these interventions is unknown. We characterized here the effects of a fat-reduced, carbohydrate-enriched diet and an increased fat intake on energy metabolism in a mouse model of VLCAD-deficiency. Wild-type and VLCAD(-/-) mice were fed one year either with a normal (5.1%), a high fat (10.6%) or a low-fat, carbohydrate-enriched (2.6%) diet. Dietary effects on genes involved in lipogenesis, energy homeostasis and substrate selection were quantified by real-time-PCR. Acylcarnitines as sign of impaired energy production were determined in dried blood spots and tissues. White skeletal muscle was analyzed for muscle fiber type as well as for glycogen and triglyceride content. Both dietary modifications induced enhanced triacylglyceride accumulation in skeletal muscle and inhibition of glucose oxidation. This was accompanied by an up-regulation of genes coding for oxidative muscle fiber type I and a marked accumulation of acylcarnitines, especially prominent in the heart (164±2.8 in VLCAD(-/-) vs. 82.3±2.1 in WT μmol/mg) under a low-fat, carbohydrate-enriched diet. We demonstrate here that both dietary interventions with respect to the fat content of the diet reverse endogenous compensatory mechanisms in muscle that have evolved in VLCAD(-/-) mice resulting in pronounced energy deficiency. In particular, the low-fat carbohydrate-enriched diet was not effective in the long term. Further experiments are necessary to define the optimal energy provision for fatty acid oxidation defects.
Collapse
Affiliation(s)
- Sara Tucci
- Department of General Pediatrics, University Hospital Freiburg, Germany.
| | | | | | | |
Collapse
|