1
|
Della Guardia L, Luzi L, Codella R. Muscle-UCP3 in the regulation of energy metabolism. Mitochondrion 2024; 76:101872. [PMID: 38499130 DOI: 10.1016/j.mito.2024.101872] [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: 08/10/2023] [Revised: 03/10/2024] [Accepted: 03/15/2024] [Indexed: 03/20/2024]
Abstract
Uncoupling protein-3 (UCP3) is a mitochondria-regulatory protein with potential energy- homeostatic functions. This study explores the role of UCP3 in the regulation of muscle- and energy metabolism. UCP3 is critical for tuning substrate utilization, favoring lipid oxidation, particularly in conditions of high-fat availability. While UCP3 is non-essential for lipid oxidation during energy excess, it proves vital during fasting, indicating an energy-homeostatic trait. Preliminary evidence indicates UCP3' promotion of glucose uptake and oxidation, at least in conditions of high glucose/low fat availability. However, the dynamics of how fats and glucose differentially influence UCP3 remain undefined. UCP3 exhibits inducible proton transport and uncoupling activity, operating in a dual manner: a resting state with no/low activity and an activated state in the presence of activators. Uncoupling may enhance thermogenesis in specific conditions and in the presence of activators such as fatty acids, thyroid hormones, and catecholamines. This energy-dissipative activity adapts to varying energy availability, balancing energy dissipation with fatty acid oxidation to optimize whole-body energy homeostasis: fasting triggers UCP3 upregulation, enhancing lipid utilization while suppressing uncoupling. Additionally, UCP3 upregulation induces glucose and lipid disposal from the bloodstream and decreases tri-/diglyceride storage in muscle. This process improves mitochondrial functionality and insulin signaling, leading to enhanced systemicgluco-metabolic balance and protection from metabolic conditions. Reviewed evidence suggests that UCP3 plays a crucial role in adapting the system to changing energy conditions. However, the precise role of UCP3 in regulating metabolism requires further elucidation.
Collapse
Affiliation(s)
- Lucio Della Guardia
- Department of Biomedical Sciences for Health, Università degli Studi di Milano, Milano, Italy
| | - Livio Luzi
- Department of Biomedical Sciences for Health, Università degli Studi di Milano, Milano, Italy; Department of Endocrinology, Nutrition and Metabolic Diseases, IRCCS MultiMedica, Milano, Italy
| | - Roberto Codella
- Department of Biomedical Sciences for Health, Università degli Studi di Milano, Milano, Italy; Department of Endocrinology, Nutrition and Metabolic Diseases, IRCCS MultiMedica, Milano, Italy.
| |
Collapse
|
2
|
Nesci S. Proton leak through the UCPs and ANT carriers and beyond: A breath for the electron transport chain. Biochimie 2023; 214:77-85. [PMID: 37336388 DOI: 10.1016/j.biochi.2023.06.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Accepted: 06/16/2023] [Indexed: 06/21/2023]
Abstract
Mitochondria produce heat as a result of an ineffective H+ cycling of mitochondria respiration across the inner mitochondrial membrane (IMM). This event present in all mitochondria, known as proton leak, can decrease protonmotive force (Δp) and restore mitochondrial respiration by partially uncoupling the substrate oxidation from the ADP phosphorylation. During impaired conditions of ATP generation with F1FO-ATPase, the Δp increases and IMM is hyperpolarized. In this bioenergetic state, the respiratory complexes support H+ transport until the membrane potential stops the H+ pump activity. Consequently, the electron transfer is stalled and the reduced form of electron carriers of the respiratory chain can generate O2∙¯ triggering the cascade of ROS formation and oxidative stress. The physiological function to attenuate the production of O2∙¯ by Δp dissipation can be attributed to the proton leak supported by the translocases of IMM.
Collapse
Affiliation(s)
- Salvatore Nesci
- Department of Veterinary Medical Sciences, University of Bologna, Ozzano Emilia, 40064, BO, Italy.
| |
Collapse
|
3
|
Neuro-immunohistochemical and molecular expression variations during hibernation and activity phases between Rana mascareniensis and Rana ridibunda. J Therm Biol 2023. [DOI: 10.1016/j.jtherbio.2023.103490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/07/2023]
|
4
|
Zhu W, Feng D, Shi X, Wei Q, Yang L. The Potential Role of Mitochondrial Acetaldehyde Dehydrogenase 2 in Urological Cancers From the Perspective of Ferroptosis and Cellular Senescence. Front Cell Dev Biol 2022; 10:850145. [PMID: 35517510 PMCID: PMC9065557 DOI: 10.3389/fcell.2022.850145] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 03/21/2022] [Indexed: 12/21/2022] Open
Abstract
Overproduction of reactive oxygen species (ROS) and superlative lipid peroxidation promote tumorigenesis, and mitochondrial aldehyde dehydrogenase 2 (ALDH2) is associated with the detoxification of ROS-mediated lipid peroxidation-generated reactive aldehydes such as 4-hydroxy-2-nonenal (4-HNE), malondialdehyde, and acrolein due to tobacco smoking. ALDH2 has been demonstrated to be highly associated with the prognosis and chemoradiotherapy sensitivity of many types of cancer, including leukemia, lung cancer, head and neck cancer, esophageal cancer, hepatocellular cancer, pancreatic cancer, and ovarian cancer. In this study, we explored the possible relationship between ALDH2 and urological cancers from the aspects of ferroptosis, epigenetic alterations, proteostasis, mitochondrial dysfunction, and cellular senescence.
Collapse
Affiliation(s)
| | | | | | - Qiang Wei
- *Correspondence: Qiang Wei, ; Lu Yang,
| | - Lu Yang
- *Correspondence: Qiang Wei, ; Lu Yang,
| |
Collapse
|
5
|
Križančić Bombek L, Čater M. Skeletal Muscle Uncoupling Proteins in Mice Models of Obesity. Metabolites 2022; 12:metabo12030259. [PMID: 35323702 PMCID: PMC8955650 DOI: 10.3390/metabo12030259] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 03/01/2022] [Accepted: 03/15/2022] [Indexed: 02/05/2023] Open
Abstract
Obesity and accompanying type 2 diabetes are among major and increasing worldwide problems that occur fundamentally due to excessive energy intake during its expenditure. Endotherms continuously consume a certain amount of energy to maintain core body temperature via thermogenic processes, mainly in brown adipose tissue and skeletal muscle. Skeletal muscle glucose utilization and heat production are significant and directly linked to body glucose homeostasis at rest, and especially during physical activity. However, this glucose balance is impaired in diabetic and obese states in humans and mice, and manifests as glucose resistance and altered muscle cell metabolism. Uncoupling proteins have a significant role in converting electrochemical energy into thermal energy without ATP generation. Different homologs of uncoupling proteins were identified, and their roles were linked to antioxidative activity and boosting glucose and lipid metabolism. From this perspective, uncoupling proteins were studied in correlation to the pathogenesis of diabetes and obesity and their possible treatments. Mice were extensively used as model organisms to study the physiology and pathophysiology of energy homeostasis. However, we should be aware of interstrain differences in mice models of obesity regarding thermogenesis and insulin resistance in skeletal muscles. Therefore, in this review, we gathered up-to-date knowledge on skeletal muscle uncoupling proteins and their effect on insulin sensitivity in mouse models of obesity and diabetes.
Collapse
|
6
|
Uncoupling Proteins and Regulated Proton Leak in Mitochondria. Int J Mol Sci 2022; 23:ijms23031528. [PMID: 35163451 PMCID: PMC8835771 DOI: 10.3390/ijms23031528] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Accepted: 01/26/2022] [Indexed: 12/17/2022] Open
Abstract
Higher concentration of protons in the mitochondrial intermembrane space compared to the matrix results in an electrochemical potential causing the back flux of protons to the matrix. This proton transport can take place through ATP synthase complex (leading to formation of ATP) or can occur via proton transporters of the mitochondrial carrier superfamily and/or membrane lipids. Some mitochondrial proton transporters, such as uncoupling proteins (UCPs), transport protons as their general regulating function; while others are symporters or antiporters, which use the proton gradient as a driving force to co-transport other substrates across the mitochondrial inner membrane (such as phosphate carrier, a symporter; or aspartate/glutamate transporter, an antiporter). Passage (or leakage) of protons across the inner membrane to matrix from any route other than ATP synthase negatively impacts ATP synthesis. The focus of this review is on regulated proton transport by UCPs. Recent findings on the structure and function of UCPs, and the related research methodologies, are also critically reviewed. Due to structural similarity of members of the mitochondrial carrier superfamily, several of the known structural features are potentially expandable to all members. Overall, this report provides a brief, yet comprehensive, overview of the current knowledge in the field.
Collapse
|
7
|
Liu X, Li D, Liu Z, Song Y, Zhang B, Zang Y, Zhang W, Niu Y, Shen C. Nicotinamide mononucleotide promotes pancreatic islet function through the SIRT1 pathway in mice after severe burns. Burns 2022; 48:1922-1932. [DOI: 10.1016/j.burns.2022.01.013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 01/10/2022] [Accepted: 01/18/2022] [Indexed: 01/03/2023]
|
8
|
Han L, Wang J, Zhao T, Wu Y, Wei Y, Chen J, Kang L, Shen L, Long C, Yang Z, Wu S, Wei G. Stereological analysis and transcriptome profiling of testicular injury induced by di-(2-ethylhexyl) phthalate in prepubertal rats. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2021; 220:112326. [PMID: 34015638 DOI: 10.1016/j.ecoenv.2021.112326] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 05/05/2021] [Accepted: 05/10/2021] [Indexed: 06/12/2023]
Abstract
Di-(2-ethylhexyl) phthalate (DEHP) is the most common phthalate that can affect the male reproductive system. DEHP exposure at the prepubertal stage could lead to the injury of immature testes, but the mechanism has not been fully clarified. In the present study, we elucidated the possible underlying mechanism of DEHP-induced prepubertal testicular injury through stereological analysis and transcriptome profiling. Compared with the control group, the DEHP-treated rats had lower body weight gain and decreased testicular weight and organ coefficient. Moreover, lower serum levels of testosterone and LH were observed in the DEHP group, in contrast to the increased FSH level. Additionally, the serum level of estradiol had no significant difference after DEHP exposure. Stereological analysis showed significant reduction in volumes of most testicular structures, especially in the seminiferous tubule and seminiferous epithelium, along with a vast decrease of spermatogenic cells and obvious structural damages with substantial pathological signs (germ cracks, cytoplasmic vacuolization, sloughing, multinucleated giant cell formation, chromatolysis desquamation and dissolution, pyknosis of nuclei) in the seminiferous tubule upon DEHP exposure at the prepubertal stage. Furthermore, transcriptome profiling identified 5548 differentially expressed genes (DEGs) upon DEHP exposure. Pathway enrichment analysis revealed several crucial signaling pathways related to retinol metabolism, oxidative phosphorylation, steroid hormone biosynthesis, and cell adhesion molecules (CAMs). In addition, seven DEGs selected from RNA-seq data were validated by quantitative real-time polymerase chain reaction (qRT-PCR), and the results showed the same trends as the RNA-seq results. In conclusion, the above findings provide basic morphological data and lay a foundation for systematic research on transcriptome profiling in prepubertal testicular injury induced by DEHP.
Collapse
Affiliation(s)
- Lindong Han
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing 400014, PR China; Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing 400014, PR China; Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing 400014, PR China; National Clinical Research Center for Child Health and Disorders, Chongqing 400014, PR China; China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing 400014, PR China; Chongqing Key Laboratory of Pediatrics, Chongqing 400014, PR China
| | - Junke Wang
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing 400014, PR China; Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing 400014, PR China; Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing 400014, PR China; National Clinical Research Center for Child Health and Disorders, Chongqing 400014, PR China; China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing 400014, PR China; Chongqing Key Laboratory of Pediatrics, Chongqing 400014, PR China
| | - Tianxin Zhao
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing 400014, PR China; Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing 400014, PR China; Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing 400014, PR China; National Clinical Research Center for Child Health and Disorders, Chongqing 400014, PR China; China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing 400014, PR China; Chongqing Key Laboratory of Pediatrics, Chongqing 400014, PR China
| | - Yuhao Wu
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing 400014, PR China; Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing 400014, PR China; Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing 400014, PR China; National Clinical Research Center for Child Health and Disorders, Chongqing 400014, PR China; China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing 400014, PR China; Chongqing Key Laboratory of Pediatrics, Chongqing 400014, PR China
| | - Yuexin Wei
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing 400014, PR China; Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing 400014, PR China; Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing 400014, PR China; National Clinical Research Center for Child Health and Disorders, Chongqing 400014, PR China; China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing 400014, PR China; Chongqing Key Laboratory of Pediatrics, Chongqing 400014, PR China
| | - Jiadong Chen
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing 400014, PR China; Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing 400014, PR China; Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing 400014, PR China; National Clinical Research Center for Child Health and Disorders, Chongqing 400014, PR China; China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing 400014, PR China; Chongqing Key Laboratory of Pediatrics, Chongqing 400014, PR China
| | - Lian Kang
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing 400014, PR China; Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing 400014, PR China; Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing 400014, PR China; National Clinical Research Center for Child Health and Disorders, Chongqing 400014, PR China; China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing 400014, PR China; Chongqing Key Laboratory of Pediatrics, Chongqing 400014, PR China
| | - Lianju Shen
- Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing 400014, PR China; Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing 400014, PR China; National Clinical Research Center for Child Health and Disorders, Chongqing 400014, PR China; China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing 400014, PR China; Chongqing Key Laboratory of Pediatrics, Chongqing 400014, PR China
| | - Chunlan Long
- Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing 400014, PR China; Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing 400014, PR China; National Clinical Research Center for Child Health and Disorders, Chongqing 400014, PR China; China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing 400014, PR China; Chongqing Key Laboratory of Pediatrics, Chongqing 400014, PR China
| | - Zhengwei Yang
- Morphometric Research Laboratory, North Sichuan Medical College, Nanchong 637000, PR China
| | - Shengde Wu
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing 400014, PR China; Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing 400014, PR China; Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing 400014, PR China; National Clinical Research Center for Child Health and Disorders, Chongqing 400014, PR China; China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing 400014, PR China; Chongqing Key Laboratory of Pediatrics, Chongqing 400014, PR China.
| | - Guanghui Wei
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing 400014, PR China; Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing 400014, PR China; Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing 400014, PR China; National Clinical Research Center for Child Health and Disorders, Chongqing 400014, PR China; China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing 400014, PR China; Chongqing Key Laboratory of Pediatrics, Chongqing 400014, PR China.
| |
Collapse
|
9
|
Hass DT, Barnstable CJ. Uncoupling proteins in the mitochondrial defense against oxidative stress. Prog Retin Eye Res 2021; 83:100941. [PMID: 33422637 DOI: 10.1016/j.preteyeres.2021.100941] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 12/28/2020] [Accepted: 01/03/2021] [Indexed: 02/06/2023]
Abstract
Oxidative stress is a major component of most major retinal diseases. Many extrinsic anti-oxidative strategies have been insufficient at counteracting one of the predominant intrinsic sources of reactive oxygen species (ROS), mitochondria. The proton gradient across the inner mitochondrial membrane is a key driving force for mitochondrial ROS production, and this gradient can be modulated by members of the mitochondrial uncoupling protein (UCP) family. Of the UCPs, UCP2 shows a widespread distribution and has been shown to uncouple oxidative phosphorylation, with concomitant decreases in ROS production. Genetic studies using transgenic and knockout mice have documented the ability of increased UCP2 activity to provide neuroprotection in models of a number of diseases, including retinal diseases, indicating that it is a strong candidate for a therapeutic target. Molecular studies have identified the structural mechanism of action of UCP2 and have detailed the ways in which its expression and activity can be controlled at the transcriptional, translational and posttranslational levels. These studies suggest a number of ways in control of UCP2 expression and activity can be used therapeutically for both acute and chronic conditions. The development of such therapeutic approaches will greatly increase the tools available to combat a broad range of serious retinal diseases.
Collapse
Affiliation(s)
- Daniel T Hass
- Department of Biochemistry, The University of Washington, Seattle, WA, 98109, USA
| | - Colin J Barnstable
- Department of Neural and Behavioral Sciences, The Pennsylvania State University, Hershey, PA, 17033, USA.
| |
Collapse
|
10
|
Cronshaw M, Parker S, Anagnostaki E, Mylona V, Lynch E, Grootveld M. Photobiomodulation and Oral Mucositis: A Systematic Review. Dent J (Basel) 2020; 8:dj8030087. [PMID: 32764305 PMCID: PMC7559189 DOI: 10.3390/dj8030087] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2020] [Revised: 07/28/2020] [Accepted: 07/30/2020] [Indexed: 12/31/2022] Open
Abstract
Oral mucositis (OM) is a debilitating complication of chemotherapy, and head and neck radiotherapy. In an effort to offer the best possible advice within the limitations of published research, a systematic review with an extended discussion and commentary on dosimetry and dose delivery is presented. Using keywords as listed, Pubmed, Google Scholar and Cochrane databases were searched during a period extending from 1995 to 2019. A total of 782 abstracts were identified. A total of 50 papers were analysed, and of these, 29 satisfied criteria required for systematic review in accordance with an optimized PRISMA statement. Clinical outcome as reported was subject to analysis with respect to time of intervention, incidence and severity of oral mucositis, and pain amelioration, and a comprehensive combined univariate and multivariate statistical analysis of the methods employed was performed. Recommendations are made with respect to the timing of the intervention. Moreover, there is an extended discussion available on the treatment care rationale of photobiomodulation (PBM), and its adjunctive association with OM. In conclusion, early prophylactic application offers clear advantages in clinical management. The many studies and associated variables and covariables assessed here revealed a choice of delivery techniques, associated wavelengths and many further indices to consider with regard to the accomplishment of optical parameters. It is therefore our recommendation that clinicians use PBM as a therapy with a full and proper understanding and training in order to optimise the clinical effects achievable.
Collapse
Affiliation(s)
- Mark Cronshaw
- Leicester School of Pharmacy, De Montfort University, Leicester LE1 9BH, UK; (S.P.); (E.A.); (V.M.); (E.L.); (M.G.)
- School of Dentistry, College of Medical and Dental Sciences, University of Birmingham, Birmingham B5 7EG, UK
- Correspondence:
| | - Steven Parker
- Leicester School of Pharmacy, De Montfort University, Leicester LE1 9BH, UK; (S.P.); (E.A.); (V.M.); (E.L.); (M.G.)
| | - Eugenia Anagnostaki
- Leicester School of Pharmacy, De Montfort University, Leicester LE1 9BH, UK; (S.P.); (E.A.); (V.M.); (E.L.); (M.G.)
| | - Valina Mylona
- Leicester School of Pharmacy, De Montfort University, Leicester LE1 9BH, UK; (S.P.); (E.A.); (V.M.); (E.L.); (M.G.)
| | - Edward Lynch
- Leicester School of Pharmacy, De Montfort University, Leicester LE1 9BH, UK; (S.P.); (E.A.); (V.M.); (E.L.); (M.G.)
- School of Dental Medicine, University of Nevada, Las Vegas, NV 89106, USA
| | - Martin Grootveld
- Leicester School of Pharmacy, De Montfort University, Leicester LE1 9BH, UK; (S.P.); (E.A.); (V.M.); (E.L.); (M.G.)
- School of Dental Medicine, University of Nevada, Las Vegas, NV 89106, USA
| |
Collapse
|
11
|
Esfandiary A, Kutsche HS, Schreckenberg R, Weber M, Pak O, Kojonazarov B, Sydykov A, Hirschhäuser C, Wolf A, Haag D, Hecker M, Fink L, Seeger W, Ghofrani HA, Schermuly RT, Weißmann N, Schulz R, Rohrbach S, Li L, Sommer N, Schlüter KD. Protection against pressure overload-induced right heart failure by uncoupling protein 2 silencing. Cardiovasc Res 2020; 115:1217-1227. [PMID: 30850841 PMCID: PMC6529920 DOI: 10.1093/cvr/cvz049] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Revised: 01/31/2019] [Accepted: 03/06/2019] [Indexed: 12/14/2022] Open
Abstract
Aims The role of uncoupling protein 2 (UCP2) in cardiac adaptation to pressure overload remains unclear. In a classical model of left ventricular pressure overload genetic deletion of UCP2 (UCP2−/−) protected against cardiac hypertrophy and failure. However, in UCP2−/− mice increased proliferation of pulmonary arterial smooth muscle cells induces mild pulmonary hypertension, right ventricular (RV) hypertrophy, and reduced cardiac output. This suggests a different role for UCP2 in RV and left ventricular adaptation to pressure overload. To clarify this situation in more detail UCP2−/− and wild-type mice were exposed to pulmonary arterial banding (PAB). Methods and results Mice were analysed (haemodynamics, morphometry, and echocardiography) 3 weeks after PAB or sham surgery. Myocytes and non-myocytes were isolated and analysed separately. Cell shortening of myocytes and fura-2 loading of cardiomyocytes were used to characterize their function. Brd assay was performed to study fibroblast proliferation. Isolated mitochondria were analysed to investigate the role of UCP2 for reactive oxygen species (ROS) production. UCP2 mRNA was 2.7-fold stronger expressed in RV myocytes than in left ventricular myocytes and stronger expressed in non-myocytes compared with myocytes. Three weeks after PAB, cardiac output was reduced in wild type but preserved in UCP2−/− mice. UCP2−/− had increased RV wall thickness, but lower RV internal diameters and displayed a significant stronger fibrosis. Cardiac fibroblasts from UCP2−/− had reduced proliferation rates but higher collagen-1 expression. Myocytes isolated from mice after PAB banding showed preserved function that was further improved by UCP2−/−. Mitochondrial ROS production and respiration was similar between UCP2−/− or wild-type hearts. Conclusion Despite a mild pulmonary hypertension in UCP2−/− mice, hearts from these mice are well preserved against additional pressure overload (severe pulmonary hypertension). This—at least in part—depends on different behaviour of non-myocytes (fibroblasts).
Collapse
Affiliation(s)
| | - Hanna S Kutsche
- Department of Physiology, Justus-Liebig University Gießen, Aulweg 129, Gießen, Germany
| | - Rolf Schreckenberg
- Department of Physiology, Justus-Liebig University Gießen, Aulweg 129, Gießen, Germany
| | - Martin Weber
- Department of Physiology, Justus-Liebig University Gießen, Aulweg 129, Gießen, Germany
| | - Oleg Pak
- Justus-Liebig-University Gießen, ECCPS, Aulweg 130, Gießen, Germany
| | | | - Akylbek Sydykov
- Justus-Liebig-University Gießen, ECCPS, Aulweg 130, Gießen, Germany
| | | | - Annemarie Wolf
- Department of Physiology, Justus-Liebig University Gießen, Aulweg 129, Gießen, Germany
| | - Daniela Haag
- Justus-Liebig-University Gießen, ECCPS, Aulweg 130, Gießen, Germany
| | - Matthias Hecker
- Justus-Liebig-University Gießen, ECCPS, Aulweg 130, Gießen, Germany
| | - Ludger Fink
- Justus-Liebig-University Gießen, ECCPS, Aulweg 130, Gießen, Germany
| | - Werner Seeger
- Justus-Liebig-University Gießen, ECCPS, Aulweg 130, Gießen, Germany
| | | | | | - Norbert Weißmann
- Justus-Liebig-University Gießen, ECCPS, Aulweg 130, Gießen, Germany
| | - Rainer Schulz
- Department of Physiology, Justus-Liebig University Gießen, Aulweg 129, Gießen, Germany
| | - Susanne Rohrbach
- Department of Physiology, Justus-Liebig University Gießen, Aulweg 129, Gießen, Germany
| | - Ling Li
- Department of Physiology, Justus-Liebig University Gießen, Aulweg 129, Gießen, Germany
| | - Natascha Sommer
- Justus-Liebig-University Gießen, ECCPS, Aulweg 130, Gießen, Germany
| | - Klaus-Dieter Schlüter
- Department of Physiology, Justus-Liebig University Gießen, Aulweg 129, Gießen, Germany
| |
Collapse
|
12
|
Siemienowicz K, Rae MT, Howells F, Anderson C, Nicol LM, Franks S, Duncan WC. Insights into Manipulating Postprandial Energy Expenditure to Manage Weight Gain in Polycystic Ovary Syndrome. iScience 2020; 23:101164. [PMID: 32464593 PMCID: PMC7256642 DOI: 10.1016/j.isci.2020.101164] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Revised: 04/08/2020] [Accepted: 05/11/2020] [Indexed: 11/30/2022] Open
Abstract
Women with polycystic ovary syndrome (PCOS) are more likely to be obese and have difficulty in losing weight. They demonstrate an obesity-independent deficit in adaptive energy expenditure. We used a clinically realistic preclinical model to investigate the molecular basis for the reduced postprandial thermogenesis (PPT) and develop a therapeutic strategy to normalize this deficit. Sheep exposed to increased androgens before birth develop the clinical features of PCOS. In adulthood they develop obesity and demonstrate an obesity-independent reduction in PPT. This is associated with reduced adipose tissue uncoupling protein expression and adipose tissue noradrenaline concentrations. These sheep are insulin resistant with reduced insulin signaling in the brain. Increasing brain insulin concentrations using intranasal insulin administration increased PPT in PCOS sheep without any effects on blood glucose concentrations. Intranasal insulin administration with food is a potential novel strategy to improve adaptive energy expenditure and normalize the responses to weight loss strategies in women with PCOS. Obesity can be prenatally programmed by androgens in an ovine model of PCOS This model has the same deficit in postprandial energy expenditure as women with PCOS Reduced adipose tissue thermogenesis links to lower central insulin signaling Therapeutic intranasal insulin raises postprandial energy expenditure in PCOS sheep
Collapse
Affiliation(s)
- Katarzyna Siemienowicz
- MRC Centre for Reproductive Health, Queen's Medical Research Institute, The University of Edinburgh, Edinburgh EH16 4TJ, UK; School of Applied Sciences, Edinburgh Napier University, Edinburgh EH11 4BN, UK
| | - Michael T Rae
- School of Applied Sciences, Edinburgh Napier University, Edinburgh EH11 4BN, UK
| | - Fiona Howells
- MRC Centre for Reproductive Health, Queen's Medical Research Institute, The University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Chloe Anderson
- MRC Centre for Reproductive Health, Queen's Medical Research Institute, The University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Linda M Nicol
- MRC Centre for Reproductive Health, Queen's Medical Research Institute, The University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Stephen Franks
- Institute of Reproductive and Developmental Biology, Imperial College, London W12 0HS, UK
| | - William C Duncan
- MRC Centre for Reproductive Health, Queen's Medical Research Institute, The University of Edinburgh, Edinburgh EH16 4TJ, UK.
| |
Collapse
|
13
|
Cunningham CN, Rutter J. 20,000 picometers under the OMM: diving into the vastness of mitochondrial metabolite transport. EMBO Rep 2020; 21:e50071. [PMID: 32329174 DOI: 10.15252/embr.202050071] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 02/17/2020] [Accepted: 03/27/2020] [Indexed: 12/14/2022] Open
Abstract
The metabolic compartmentalization enabled by mitochondria is key feature of many cellular processes such as energy conversion to ATP production, redox balance, and the biosynthesis of heme, urea, nucleotides, lipids, and others. For a majority of these functions, metabolites need to be transported across the impermeable inner mitochondrial membrane by dedicated carrier proteins. Here, we examine the substrates, structural features, and human health implications of four mitochondrial metabolite carrier families: the SLC25A family, the mitochondrial ABCB transporters, the mitochondrial pyruvate carrier (MPC), and the sideroflexin proteins.
Collapse
Affiliation(s)
- Corey N Cunningham
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Jared Rutter
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT, USA.,Howard Hughes Medical Institute, University of Utah School of Medicine, Salt Lake City, UT, USA
| |
Collapse
|
14
|
Kutsche HS, Schreckenberg R, Weber M, Hirschhäuser C, Rohrbach S, Li L, Niemann B, Schulz R, Schlüter KD. Alterations in Glucose Metabolism During the Transition to Heart Failure: The Contribution of UCP-2. Cells 2020; 9:cells9030552. [PMID: 32120777 PMCID: PMC7140436 DOI: 10.3390/cells9030552] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 02/21/2020] [Accepted: 02/24/2020] [Indexed: 12/20/2022] Open
Abstract
The cardiac expression of the mitochondrial uncoupling protein (UCP)-2 is increased in patients with heart failure. However, the underlying causes as well as the possible consequences of these alterations during the transition from hypertrophy to heart failure are still unclear. To investigate the role of UCP-2 mechanistically, expression of UCP-2 was silenced by small interfering RNA in adult rat ventricular cardiomyocytes. We demonstrate that a downregulation of UCP-2 by siRNA in cardiomyocytes preserves contractile function in the presence of angiotensin II. Furthermore, silencing of UCP-2 was associated with an upregulation of glucose transporter type (Glut)-4, increased glucose uptake, and reduced intracellular lactate levels, indicating improvement of the oxidative glucose metabolism. To study this adaptation in vivo, spontaneously hypertensive rats served as a model for cardiac hypertrophy due to pressure overload. During compensatory hypertrophy, we found low UCP-2 levels with an upregulation of Glut-4, while the decompensatory state with impaired function was associated with an increase of UCP-2 and reduced Glut-4 expression. By blocking the aldosterone receptor with spironolactone, both cardiac function as well as UCP-2 and Glut-4 expression levels of the compensated phase could be preserved. Furthermore, we were able to confirm this by left ventricular (LV) biopsies of patients with end-stage heart failure. The results of this study show that UCP-2 seems to impact the cardiac glucose metabolism during the transition from hypertrophy to failure by affecting glucose uptake through Glut-4. We suggest that the failing heart could benefit from low UCP-2 levels by improving the efficiency of glucose oxidation. For this reason, UCP-2 inhibition might be a promising therapeutic strategy to prevent the development of heart failure.
Collapse
Affiliation(s)
- Hanna Sarah Kutsche
- Physiologisches Institut, Justus-Liebig-Universität, 35392 Gießen, Germany; (R.S.); (M.W.); (C.H.); (S.R.); (L.L.); (R.S.); (K.-D.S.)
- Correspondence: ; Tel.: +49-641-99-47145
| | - Rolf Schreckenberg
- Physiologisches Institut, Justus-Liebig-Universität, 35392 Gießen, Germany; (R.S.); (M.W.); (C.H.); (S.R.); (L.L.); (R.S.); (K.-D.S.)
| | - Martin Weber
- Physiologisches Institut, Justus-Liebig-Universität, 35392 Gießen, Germany; (R.S.); (M.W.); (C.H.); (S.R.); (L.L.); (R.S.); (K.-D.S.)
| | - Christine Hirschhäuser
- Physiologisches Institut, Justus-Liebig-Universität, 35392 Gießen, Germany; (R.S.); (M.W.); (C.H.); (S.R.); (L.L.); (R.S.); (K.-D.S.)
| | - Susanne Rohrbach
- Physiologisches Institut, Justus-Liebig-Universität, 35392 Gießen, Germany; (R.S.); (M.W.); (C.H.); (S.R.); (L.L.); (R.S.); (K.-D.S.)
| | - Ling Li
- Physiologisches Institut, Justus-Liebig-Universität, 35392 Gießen, Germany; (R.S.); (M.W.); (C.H.); (S.R.); (L.L.); (R.S.); (K.-D.S.)
| | - Bernd Niemann
- Universitätsklinikum Gießen, Klinik für Herz-, Kinderherz- und Gefäßchirurgie, 35392 Gießen, Germany;
| | - Rainer Schulz
- Physiologisches Institut, Justus-Liebig-Universität, 35392 Gießen, Germany; (R.S.); (M.W.); (C.H.); (S.R.); (L.L.); (R.S.); (K.-D.S.)
| | - Klaus-Dieter Schlüter
- Physiologisches Institut, Justus-Liebig-Universität, 35392 Gießen, Germany; (R.S.); (M.W.); (C.H.); (S.R.); (L.L.); (R.S.); (K.-D.S.)
| |
Collapse
|
15
|
Dos Santos CC, Reynolds S, Batt J. Searching for the "Spark" in Ventilator-induced Diaphragm Dysfunction. Am J Respir Crit Care Med 2019; 196:1498-1500. [PMID: 28954198 DOI: 10.1164/rccm.201708-1716ed] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Affiliation(s)
- Claudia C Dos Santos
- 1 Keenan Research Centre for Biomedical Science St. Michael's Hospital Toronto, Ontario, Canada.,2 Institute of Medical Science.,3 Department of Medicine University of Toronto Toronto, Ontario, Canada
| | - Steven Reynolds
- 4 Critical Care Department Fraser Health Authority New Westminster, British Columbia, Canada and.,5 Department of Biophysiology and Kinesiology Simon Fraser University Burnaby, British Columbia, Canada
| | - Jane Batt
- 1 Keenan Research Centre for Biomedical Science St. Michael's Hospital Toronto, Ontario, Canada.,2 Institute of Medical Science.,3 Department of Medicine University of Toronto Toronto, Ontario, Canada
| |
Collapse
|
16
|
Fractionated whole body gamma irradiation modulates the hepatic response in type II diabetes of high fat diet model rats. Mol Biol Rep 2019; 46:2273-2283. [PMID: 30747384 DOI: 10.1007/s11033-019-04681-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Accepted: 02/07/2019] [Indexed: 10/27/2022]
Abstract
HFD animals were exposed to a low rate of different fractionated whole body gamma irradiation doses (0.5, 1 and 2 Gy, three fractions per week for two consecutive months) and the expression of certain genes involved in type 2 diabetes mellitus (T2DM) in livers and brains of HFD Wistar rats was investigated. Additionally, levels of diabetes-related proteins encoded by the studied genes were analyzed. Results indicated that mRNA level of incretin glucagon like peptite-1 receptor (GLP-1R) was augmented in livers and brains exposed to 1 and 2 Gy doses. Moreover, the mitochondrial uncoupling proteins 2 and 3 (UCP2/3) expressions in animals fed on HFD compared to those fed on normal chow diet were significantly increased at all applied doses. GLP-1R and UCP3 protein levels were up regulated in livers. Total protein content increased at 0.5 and 1 Gy gamma irradiation exposure and returned to its normal level at 2 Gy dose. Results could be an indicator of type 2 diabetes delayed development during irradiation exposure and support the importance of GLP-1R as a target gene in radiotherapy against T2DM and its chronic complications. A new hypothesis of brain-liver and intestine interface is speculated by which an increase in the hepatic GLP-1R is influenced by the effect of fractionated whole body gamma irradiation.
Collapse
|
17
|
Namazi Sarvestani N, Saberi Firouzi S, Falak R, Karimi MY, Davoodzadeh Gholami M, Rangbar A, Hosseini A. Phosphodiesterase 4 and 7 inhibitors produce protective effects against high glucose-induced neurotoxicity in PC12 cells via modulation of the oxidative stress, apoptosis and inflammation pathways. Metab Brain Dis 2018; 33:1293-1306. [PMID: 29713919 DOI: 10.1007/s11011-018-0241-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Accepted: 04/13/2018] [Indexed: 12/13/2022]
Abstract
Diabetic neuropathy (DN) is the most common diabetic complication. It is estimated diabetic population will increase to 592 million by the year 2035. This is while at least 50-60% of all diabetic patients will suffer from neuropathy in their lifetime. Oxidative stress, mitochondrial dysfunction, apoptosis, and inflammation are crucial pathways in development and progression of DN. Since there is also no selective and effective therapeutic agent to prevent or treat high glucose (HG)-induced neuronal cell injury, it is crucial to explore tools by which one can reduce factors related to these pathways. Phosphodiesterase 4 and 7 (PDE 4 and 7) regulate oxidative damage, neurodegenaration, and inflammatory responses through modulation of cyclic adenosine monophosphate (cAMP) level, and thus can be as important drug targets for regulating DN. The aim of this study was to evaluate the protective effects of inhibitors of PDE 4 and 7, named rolipram and BRL5048, on HG-induced neurotoxicity in PC12 cells as an in vitro cellular model for DN and determine the possible mechanisms for theirs effects. We report that the PC12 cells pre-treatment with rolipram (2 μM) and/or BRL5048 (0.2 μM) for 60 min and then exposing the cells to HG (4.5 g/L for 72 h) or normal glucose (NG) (1 g/L for 72 h) condition show: (1) significant attenuation in ROS, MDA and TNF-a levels, Bax/Bcl-2 ratio, expression of caspase 3 and UCP2 proteins; (2) significant increase in viability, GSH/GSSG ratio, MMP and ATP levels. All these data together led us to propose PDE 4 and 7 inhibitors, and specifically, rolipram and BRL5048, as potential drugs candidate to be further studied for the prevention and treatment of DN.
Collapse
Affiliation(s)
- Nazanin Namazi Sarvestani
- Department of Animal Biology, School of Biology, Department of Science, University of Tehran, Tehran, Iran
| | - Saeedeh Saberi Firouzi
- Department of Pharmacology, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Reza Falak
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | | | | | - Akram Rangbar
- Department of Toxicology and Pharmacology, School of Pharmacy, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Asieh Hosseini
- Razi Drug Research Center, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
18
|
Saberi Firouzi S, Namazi Sarvestani N, Bakhtiarian A, Ghazi Khansari M, Karimi MY, Ranjbar A, Safa M, Hosseini A. Sildenafil protective effects on high glucose-induced neurotoxicity in PC12 cells: the role of oxidative stress, apoptosis, and inflammation pathways in an in vitro cellular model for diabetic neuropathy. Neurol Res 2018; 40:624-636. [PMID: 29623781 DOI: 10.1080/01616412.2018.1458813] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Objectives Diabetic neuropathy (DN) induces lifetime disability and there is currently no effective therapy to treat or to minimize patients suffering, so it is thereby imperative to develop therapeutic strategies for this disease. Since oxidative stress, mitochondrial dysfunction, apoptosis, and inflammation are crucial mechanisms in development and progression of DN, it is important to explore tools by which one can reduce factors related to these pathways. Herein, the understandings of the sildenafil neuroprotective effect through increase of cGMP level and the mediation of oxidative stress, apoptosis, and inflammation pathways on neurotoxicity induced by high glucose (HG) in PC12 cells as an in vitro cellular model for DN were investigated. Methods We reported that the PC12 cells pre-treatment with sildenafil (0.008 μM) for 60 min and then exposing the cells to HG (25 mM for 72 h) or normal glucose (NG) (5 mM for 72 h) condition, show: Results (1) significant attenuation in reactive oxygen species, MDA and TNF-a levels, Bax/Bcl-2 ratio, expression of caspase 3 and UCP2 proteins; (2) significant increase in viability, GSH/GSSG ratio, mitochondrial membrane potential, and ATP levels. Conclusion All these data together led us to propose neuroprotective effect of sildenafil is probably through its antioxidant, antiapoptotic, and anti-inflammatory activities. Of course, further studies are required to explain the underlying mechanism of the sildenafil effects.
Collapse
Affiliation(s)
- Saeedeh Saberi Firouzi
- a Faculty of Medicine, Department of Pharmacology , Tehran University of Medical Sciences , Tehran , Iran
| | - Nazanin Namazi Sarvestani
- b Department of Animal Biology, School of Biology, Department of Science , University of Tehran , Tehran , Iran
| | - Azam Bakhtiarian
- a Faculty of Medicine, Department of Pharmacology , Tehran University of Medical Sciences , Tehran , Iran
| | - Mahmoud Ghazi Khansari
- a Faculty of Medicine, Department of Pharmacology , Tehran University of Medical Sciences , Tehran , Iran
| | | | - Akram Ranjbar
- d Department of Toxicology and Pharmacology, School of Pharmacy , Hamadan University of Medical Sciences , Hamadan , Iran
| | - Majid Safa
- e Faculty of Allied Medicine, Cellular and Molecular Research Center and Department of Hematology and blood banking , Iran University of Medical Sciences , Tehran , Iran
| | - Asieh Hosseini
- c Razi Drug Research Center , Iran University of Medical Sciences , Tehran , Iran
| |
Collapse
|
19
|
Miller VJ, Villamena FA, Volek JS. Nutritional Ketosis and Mitohormesis: Potential Implications for Mitochondrial Function and Human Health. J Nutr Metab 2018; 2018:5157645. [PMID: 29607218 PMCID: PMC5828461 DOI: 10.1155/2018/5157645] [Citation(s) in RCA: 132] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Accepted: 12/27/2017] [Indexed: 02/07/2023] Open
Abstract
Impaired mitochondrial function often results in excessive production of reactive oxygen species (ROS) and is involved in the etiology of many chronic diseases, including cardiovascular disease, diabetes, neurodegenerative disorders, and cancer. Moderate levels of mitochondrial ROS, however, can protect against chronic disease by inducing upregulation of mitochondrial capacity and endogenous antioxidant defense. This phenomenon, referred to as mitohormesis, is induced through increased reliance on mitochondrial respiration, which can occur through diet or exercise. Nutritional ketosis is a safe and physiological metabolic state induced through a ketogenic diet low in carbohydrate and moderate in protein. Such a diet increases reliance on mitochondrial respiration and may, therefore, induce mitohormesis. Furthermore, the ketone β-hydroxybutyrate (BHB), which is elevated during nutritional ketosis to levels no greater than those resulting from fasting, acts as a signaling molecule in addition to its traditionally known role as an energy substrate. BHB signaling induces adaptations similar to mitohormesis, thereby expanding the potential benefit of nutritional ketosis beyond carbohydrate restriction. This review describes the evidence supporting enhancement of mitochondrial function and endogenous antioxidant defense in response to nutritional ketosis, as well as the potential mechanisms leading to these adaptations.
Collapse
Affiliation(s)
- Vincent J. Miller
- Department of Human Sciences, College of Education and Human Ecology, The Ohio State University, Columbus, OH, USA
| | - Frederick A. Villamena
- Department of Biological Chemistry and Pharmacology, College of Medicine, The Ohio State University, Columbus, OH, USA
| | - Jeff S. Volek
- Department of Human Sciences, College of Education and Human Ecology, The Ohio State University, Columbus, OH, USA
| |
Collapse
|
20
|
Bryant HJ, Chung DJ, Schulte PM. Subspecies differences in thermal acclimation of mitochondrial function and the role of uncoupling proteins in killifish. J Exp Biol 2018; 221:jeb.186320. [DOI: 10.1242/jeb.186320] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Accepted: 10/18/2018] [Indexed: 01/08/2023]
Abstract
Thermal effects on mitochondrial efficiency and ATP production can influence whole-animal thermal tolerance and performance. Thus, organisms may have the capacity to alter mitochondrial processes through acclimation or adaptation to mitigate these effects. One possible mechanism is through the action of uncoupling proteins (UCPs) which can decrease the proton motive force independent of the production of ATP. To test this hypothesis, we examined the mRNA expression patterns of UCP isoforms and characterized the effects of thermal acclimation and putative local thermal adaptation on mitochondrial capacity, proton leak, and P/O ratios in two subspecies of Atlantic killifish (Fundulus heteroclitus). Ucp1 was the dominant isoform in liver and was more highly expressed in northern killifish. We found that cold acclimation increased mitochondrial capacity (state III and maximum substrate oxidation capacity), state II membrane potential, proton leak, and P/O ratios in northern, but not southern killifish liver mitochondria. Palmitate-induced mitochondrial uncoupling was detected in northern, but not southern, killifish liver mitochondria, consistent with the differences in mRNA expression between the subspecies. Taken together, our data suggest that mitochondrial function is more plastic in response to thermal acclimation in northern killifish than southern killifish and that UCP1 may play a role in regulating the proton motive force in northern, but not southern killifish in response to thermal acclimation. These data demonstrate the potential for adaptive variation in mitochondrial plasticity in response to cold.
Collapse
Affiliation(s)
- Heather J. Bryant
- Department of Zoology, University of British Columbia, 6270 University Blvd., Vancouver, British Columbia, Canada V6T 1Z4
| | - Dillon J. Chung
- Department of Zoology, University of British Columbia, 6270 University Blvd., Vancouver, British Columbia, Canada V6T 1Z4
| | - Patricia M. Schulte
- Department of Zoology, University of British Columbia, 6270 University Blvd., Vancouver, British Columbia, Canada V6T 1Z4
| |
Collapse
|
21
|
Chemotherapeutic agents induce mitochondrial superoxide production and toxicity but do not alter respiration in skeletal muscle in vitro. Mitochondrion 2017; 42:33-49. [PMID: 29079447 DOI: 10.1016/j.mito.2017.10.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Revised: 09/21/2017] [Accepted: 10/23/2017] [Indexed: 12/31/2022]
Abstract
Chemotherapeutic agents (CAs) can independently promote skeletal muscle dysfunction, fatigue and wasting with mitochondrial toxicity implicated as a possible mechanism. Thus, we aimed to characterise the effects of various CAs on mitochondrial function, viability and oxidant production in C2C12 myoblasts and myotubes. All CAs significantly reduced the viable mitochondrial pool but did not affect mitochondrial functional parameters. Doxorubicin and oxaliplatin increased oxidant production in myotubes while all CAs, except for irinotecan, increased oxidant production in myoblasts and reduced myotube diameter. Our data demonstrate CAs mito-toxic effects, highlighting the potential for mitochondria-protective therapeutics to address chemotherapy-induced skeletal muscle damage.
Collapse
|
22
|
Mitochondrial uncoupling in cancer cells: Liabilities and opportunities. BIOCHIMICA ET BIOPHYSICA ACTA-BIOENERGETICS 2017; 1858:655-664. [DOI: 10.1016/j.bbabio.2017.01.005] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/25/2016] [Revised: 12/16/2016] [Accepted: 01/05/2017] [Indexed: 12/12/2022]
|
23
|
Meng X, Tan J, Li M, Song S, Miao Y, Zhang Q. Sirt1: Role Under the Condition of Ischemia/Hypoxia. Cell Mol Neurobiol 2017; 37:17-28. [PMID: 26971525 PMCID: PMC11482061 DOI: 10.1007/s10571-016-0355-2] [Citation(s) in RCA: 119] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Accepted: 02/24/2016] [Indexed: 12/13/2022]
Abstract
Silent information regulator factor 2-related enzyme 1 (sirtuin 1, Sirt1) is a nicotinamide adenine dinucleotide-dependent deacetylase, which can deacetylate histone and non-histone proteins and other transcription factors, and is involved in the regulation of many physiological functions, including cell senescence, gene transcription, energy balance, and oxidative stress. Ischemia/hypoxia injury remains an unresolved and complicated situation in the diseases of ischemia stroke, heart failure, and coronary heart disease, especially among the old folks. Studies have demonstrated that aging could enhance the vulnerability of brain, heart, lung, liver, and kidney to ischemia/hypoxia injury and the susceptibility in old folks to ischemia/hypoxia injury might be associated with Sirt1. In this review, we mainly summarize the role of Sirt1 in modulating pathways against energy depletion and its involvement in oxidative stress, apoptosis, and inflammation under the condition of ischemia/hypoxia.
Collapse
Affiliation(s)
- Xiaofei Meng
- Department of Geriatrics, Tianjin Medical University General Hospital, Tianjin Geriatrics Institute, Tianjin, China
| | - Jin Tan
- Department of Geriatrics, Tianjin Medical University General Hospital, Tianjin Geriatrics Institute, Tianjin, China
| | - Mengmeng Li
- Department of Geriatrics, Tianjin Medical University General Hospital, Tianjin Geriatrics Institute, Tianjin, China
| | - Shuling Song
- Department of Geriatrics, Tianjin Medical University General Hospital, Tianjin Geriatrics Institute, Tianjin, China
| | | | - Qiang Zhang
- Department of Geriatrics, Tianjin Medical University General Hospital, Tianjin Geriatrics Institute, Tianjin, China.
| |
Collapse
|
24
|
Shaheen A, Aljebali AMA. A hypothetical model to solve the controversy over the involvement of UCP2 in palmitate-induced β-cell dysfunction. Endocrine 2016; 54:276-283. [PMID: 27491555 DOI: 10.1007/s12020-016-1051-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Accepted: 07/04/2016] [Indexed: 11/24/2022]
Abstract
The aim of this article is to solve an existing controversy over the involvement of uncoupling protein-2 in the impairment of glucose-stimulated insulin secretion induced by chronic exposure of β-cells to palmitate. We analyzed and compared the results of studies that support and that deny the involvement of uncoupling protein-2 in this impairment. We observed that this impairment could occur in multiple stages. We provide a model in which palmitate-induced impairment of glucose-stimulated insulin secretion is proposed to occur in two stages, early stage and late stage, depending on the integrity of electron supply (glycolysis and Krebs cycle) and transport system through electron transport chain after palmitate treatment. Prolonged exposure of β-cells to palmitate can impair this system. Early-stage impairment occurs due to uncoupling by uncoupling protein-2 when this system is still intact. When this system becomes impaired, late-stage impairment occurs mainly due to reduced glucose-stimulated adenosine triphosphate production independent of uncoupling by uncoupling protein-2. The change in glucose-stimulated oxygen uptake after palmitate treatment reflects the integrity of this system and can be used to differentiate between the two stages. Some β-cells lines and islets appear to be more resistant to palmitate-induced impairment of electron supply and transport system than others, and therefore early stage is prominent in the more resistant cell lines and less prominent or absent in the less resistant cell lines. This may help to resolve the pathogenesis of diabetes and to monitor the progression of palmitate-induced β-cell dysfunction.
Collapse
Affiliation(s)
- Alaa Shaheen
- Kafr El-Sharakwa Medical Center, Kafr El-Sharakwa, Aga, Dakahlia, Egypt.
| | - Ahmad M A Aljebali
- Department of Zoology, Faculty of Science, Omar Al Mukhtar University, Bayda, Libya
| |
Collapse
|
25
|
Nowinski SM, Solmonson A, Mills EM. Chewing the fat for Akt1 inhibition and oncosuppression. Mol Cell Oncol 2016; 3:e1102795. [PMID: 27308618 DOI: 10.1080/23723556.2015.1102795] [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: 09/24/2015] [Revised: 09/28/2015] [Accepted: 09/29/2015] [Indexed: 10/22/2022]
Abstract
The catabolic and energy-dissipating actions of mitochondrial uncoupling proteins (UCPs) conflict with many of the bioenergetic hallmarks of malignancy. We have recently demonstrated that overexpression of mitochondrial uncoupling protein 3 (Ucp3) in the basal epidermis impedes skin tumorigenesis through a novel pathway of thymoma viral proto-oncogene 1 (Akt1) inhibition via increased mitochondrial lipid catabolism.
Collapse
Affiliation(s)
- Sara M Nowinski
- Department of Biochemistry, University of Utah School of Medicine , Salt Lake City, UT, USA
| | - Ashley Solmonson
- Division of Pharmacology and Toxicology, College of Pharmacy and; Institute for Cellular and Molecular Biology, The University of Texas at Austin, Austin, TX, USA
| | - Edward M Mills
- Division of Pharmacology and Toxicology, College of Pharmacy and; Institute for Cellular and Molecular Biology, The University of Texas at Austin, Austin, TX, USA
| |
Collapse
|
26
|
Sirsat SKG, Sirsat TS, Crossley JL, Sotherland PR, Dzialowski EM. The 12-day thermoregulatory metamorphosis of Red-winged Blackbirds (Agelaius phoeniceus). J Comp Physiol B 2016; 186:651-63. [PMID: 27003423 DOI: 10.1007/s00360-016-0978-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Revised: 02/23/2016] [Accepted: 03/06/2016] [Indexed: 12/13/2022]
Abstract
We examined development of endothermy in altricial Red-winged Blackbirds (Agelaius phoeniceus) by measuring oxygen consumption [Formula: see text], body temperature and ventilation at ambient temperatures from 35 to 15 °C. Mitochondrial respiration of permeabilized skeletal muscle was also measured from breast (pectoralis) and thigh (femorotibialis) muscles. Animals were studied from the first day of hatching through fledging (12 days post-hatch, dph). Nestling whole-body metabolic rate began to show an endothermic response to cold temperature midway between hatching and fledging. Nestlings less than 5 dph were unable to maintain elevated [Formula: see text] and body temperature when exposed to gradually decreasing temperature, whereas 7 dph nestlings maintained [Formula: see text] until ~25 °C, after which [Formula: see text] decreased. From 10 dph to fledging, animals maintained elevated [Formula: see text] and body temperature when exposed to gradual cooling; full endothermic capacity was achieved. Ventilation followed a similar developmental trend to that of [Formula: see text], with increases in 10 dph fledglings occurring in tidal volume rather than ventilation frequency. LEAK respiration and oxidative phosphorylation (OXPHOS) through complex I of breast muscle mitochondria increased significantly after 3 dph. Expression of avUCP and PCG-1α mRNA increased significantly at 3 dph and remained elevated in both skeletal muscle types. Increased metabolic capacity at the cellular level occurred prior to that of the whole animal. This change in whole animal metabolic capacity increased steadily upon hatching as evidenced by the shift of metabolic rate from an ectothermic to endothermic phenotype and the increase of mitochondrial OXPHOS activity of the shivering muscles of this altricial avian species.
Collapse
Affiliation(s)
- Sarah K Goy Sirsat
- Developmental Integrative Biology Group, Department of Biological Sciences, University of North Texas, 1155 Union Circle #305220, Denton, TX, 76203, USA.
| | - Tushar S Sirsat
- Developmental Integrative Biology Group, Department of Biological Sciences, University of North Texas, 1155 Union Circle #305220, Denton, TX, 76203, USA
| | - Janna L Crossley
- Developmental Integrative Biology Group, Department of Biological Sciences, University of North Texas, 1155 Union Circle #305220, Denton, TX, 76203, USA
| | | | - Edward M Dzialowski
- Developmental Integrative Biology Group, Department of Biological Sciences, University of North Texas, 1155 Union Circle #305220, Denton, TX, 76203, USA
| |
Collapse
|
27
|
Schaur RJ, Siems W, Bresgen N, Eckl PM. 4-Hydroxy-nonenal-A Bioactive Lipid Peroxidation Product. Biomolecules 2015; 5:2247-337. [PMID: 26437435 PMCID: PMC4693237 DOI: 10.3390/biom5042247] [Citation(s) in RCA: 155] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Revised: 07/24/2015] [Accepted: 07/29/2015] [Indexed: 12/23/2022] Open
Abstract
This review on recent research advances of the lipid peroxidation product 4-hydroxy-nonenal (HNE) has four major topics: I. the formation of HNE in various organs and tissues, II. the diverse biochemical reactions with Michael adduct formation as the most prominent one, III. the endogenous targets of HNE, primarily peptides and proteins (here the mechanisms of covalent adduct formation are described and the (patho-) physiological consequences discussed), and IV. the metabolism of HNE leading to a great number of degradation products, some of which are excreted in urine and may serve as non-invasive biomarkers of oxidative stress.
Collapse
Affiliation(s)
- Rudolf J Schaur
- Institute of Molecular Biosciences, University of Graz, Heinrichstrasse 33a, 8010 Graz, Austria.
| | - Werner Siems
- Institute for Medical Education, KortexMed GmbH, Hindenburgring 12a, 38667 Bad Harzburg, Germany.
| | - Nikolaus Bresgen
- Division of Genetics, Department of Cell Biology, University of Salzburg, Hellbrunnerstasse 34, 5020 Salzburg, Austria.
| | - Peter M Eckl
- Division of Genetics, Department of Cell Biology, University of Salzburg, Hellbrunnerstasse 34, 5020 Salzburg, Austria.
| |
Collapse
|
28
|
Mitochondrial uncoupling links lipid catabolism to Akt inhibition and resistance to tumorigenesis. Nat Commun 2015; 6:8137. [PMID: 26310111 PMCID: PMC4552083 DOI: 10.1038/ncomms9137] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2014] [Accepted: 07/22/2015] [Indexed: 12/12/2022] Open
Abstract
To support growth, tumour cells reprogramme their metabolism to simultaneously upregulate macromolecular biosynthesis while maintaining energy production. Uncoupling proteins (UCPs) oppose this phenotype by inducing futile mitochondrial respiration that is uncoupled from ATP synthesis, resulting in nutrient wasting. Here using a UCP3 transgene targeted to the basal epidermis, we show that forced mitochondrial uncoupling inhibits skin carcinogenesis by blocking Akt activation. Similarly, Akt activation is markedly inhibited in UCP3 overexpressing primary human keratinocytes. Mechanistic studies reveal that uncoupling increases fatty acid oxidation and membrane phospholipid catabolism, and impairs recruitment of Akt to the plasma membrane. Overexpression of Akt overcomes metabolic regulation by UCP3, rescuing carcinogenesis. These findings demonstrate that mitochondrial uncoupling is an effective strategy to limit proliferation and tumorigenesis through inhibition of Akt, and illuminate a novel mechanism of crosstalk between mitochondrial metabolism and growth signalling.
Collapse
|
29
|
Hirschberg Jensen V, Affourtit C. Mitochondrial uncoupling protein-2 is not involved in palmitate-induced impairment of glucose-stimulated insulin secretion in INS-1E insulinoma cells and is not needed for the amplification of insulin release. Biochem Biophys Rep 2015; 1:8-15. [PMID: 26339685 PMCID: PMC4547158 DOI: 10.1016/j.bbrep.2015.03.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Revised: 03/18/2015] [Accepted: 03/19/2015] [Indexed: 11/17/2022] Open
Abstract
We have recently shown that overnight exposure of INS-1E insulinoma cells to palmitate in the presence of high glucose causes defects in both mitochondrial energy metabolism and glucose-stimulated insulin secretion (GSIS). Here we report experiments designed to test the involvement of mitochondrial uncoupling protein-2 (UCP2) in these glucolipotoxic effects. Measuring real-time oxygen consumption in siRNA-transfected INS-1E cells, we show that deleterious effects of palmitate on the glucose sensitivity of mitochondrial respiration and on the coupling efficiency of oxidative phosphorylation are independent of UCP2. Consistently, palmitate impairs GSIS to the same extent in cells with and without UCP2. Furthermore, we knocked down UCP2 in spheroid INS-1E cell clusters (pseudoislets) to test whether or not UCP2 regulates insulin secretion during prolonged glucose exposure. We demonstrate that there are no differences in temporal GSIS kinetics between perifused pseudoislets with and without UCP2. We conclude that UCP2 is not involved in palmitate-induced impairment of GSIS in INS-1E insulinoma cells and is not needed for the amplification of insulin release. These conclusions inform ongoing debate on the disputed biochemical and physiological functions of the beta cell UCP2. UCP2 does not engage with palmitate-induced mitochondrial dysfunction in INS-1E cells. UCP2 does not alter palmitate-induced impairment of insulin secretion in INS-1E cells. UCP2 is not needed for the amplification of insulin secretion in INS-1E pseudoislets.
Collapse
Affiliation(s)
- Verena Hirschberg Jensen
- School of Biomedical and Healthcare Sciences, Plymouth University, Drake Circus, Plymouth PL4 8AA, UK
| | - Charles Affourtit
- School of Biomedical and Healthcare Sciences, Plymouth University, Drake Circus, Plymouth PL4 8AA, UK
| |
Collapse
|
30
|
Undernutrition during pregnancy in mice leads to dysfunctional cardiac muscle respiration in adult offspring. Biosci Rep 2015; 35:BSR20150007. [PMID: 26182362 PMCID: PMC4613697 DOI: 10.1042/bsr20150007] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2015] [Accepted: 04/08/2015] [Indexed: 11/19/2022] Open
Abstract
We show that in utero undernutrition is associated with impaired cardiac muscle energetics and increased plasma short-chain acylcarnitines in adult mice. Findings suggest that in utero undernutrition is associated with maladaptive programming processes that have negative effects on the heart. Intrauterine growth restriction (IUGR) is associated with an increased risk of developing obesity, insulin resistance and cardiovascular disease. However, its effect on energetics in heart remains unknown. In the present study, we examined respiration in cardiac muscle and liver from adult mice that were undernourished in utero. We report that in utero undernutrition is associated with impaired cardiac muscle energetics, including decreased fatty acid oxidative capacity, decreased maximum oxidative phosphorylation rate and decreased proton leak respiration. No differences in oxidative characteristics were detected in liver. We also measured plasma acylcarnitine levels and found that short-chain acylcarnitines are increased with in utero undernutrition. Results reveal the negative impact of suboptimal maternal nutrition on adult offspring cardiac energy metabolism, which may have life-long implications for cardiovascular function and disease risk.
Collapse
|
31
|
Lobet E, Letesson JJ, Arnould T. Mitochondria: a target for bacteria. Biochem Pharmacol 2015; 94:173-85. [PMID: 25707982 DOI: 10.1016/j.bcp.2015.02.007] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Revised: 02/12/2015] [Accepted: 02/12/2015] [Indexed: 01/12/2023]
Abstract
Eukaryotic cells developed strategies to detect and eradicate infections. The innate immune system, which is the first line of defence against invading pathogens, relies on the recognition of molecular patterns conserved among pathogens. Pathogen associated molecular pattern binding to pattern recognition receptor triggers the activation of several signalling pathways leading to the establishment of a pro-inflammatory state required to control the infection. In addition, pathogens evolved to subvert those responses (with passive and active strategies) allowing their entry and persistence in the host cells and tissues. Indeed, several bacteria actively manipulate immune system or interfere with the cell fate for their own benefit. One can imagine that bacterial effectors can potentially manipulate every single organelle in the cell. However, the multiple functions fulfilled by mitochondria especially their involvement in the regulation of innate immune response, make mitochondria a target of choice for bacterial pathogens as they are not only a key component of the central metabolism through ATP production and synthesis of various biomolecules but they also take part to cell signalling through ROS production and control of calcium homeostasis as well as the control of cell survival/programmed cell death. Furthermore, considering that mitochondria derived from an ancestral bacterial endosymbiosis, it is not surprising that a special connection does exist between this organelle and bacteria. In this review, we will discuss different mitochondrial functions that are affected during bacterial infection as well as different strategies developed by bacterial pathogens to subvert functions related to calcium homeostasis, maintenance of redox status and mitochondrial morphology.
Collapse
Affiliation(s)
- Elodie Lobet
- Laboratory of Biochemistry and Cellular Biology (URBC), NAmur Research Institute for LIfe Science (NARILIS), University of Namur, 61 Rue de Bruxelles, 5000 Namur, Belgium.
| | - Jean-Jacques Letesson
- Research Unit in Microorganisms Biology, University of Namur, 61 Rue de Bruxelles, 5000 Namur, Belgium.
| | - Thierry Arnould
- Laboratory of Biochemistry and Cellular Biology (URBC), NAmur Research Institute for LIfe Science (NARILIS), University of Namur, 61 Rue de Bruxelles, 5000 Namur, Belgium.
| |
Collapse
|
32
|
Zhong H, Yin H. Role of lipid peroxidation derived 4-hydroxynonenal (4-HNE) in cancer: focusing on mitochondria. Redox Biol 2014; 4:193-9. [PMID: 25598486 PMCID: PMC4803793 DOI: 10.1016/j.redox.2014.12.011] [Citation(s) in RCA: 380] [Impact Index Per Article: 34.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Accepted: 12/17/2014] [Indexed: 12/20/2022] Open
Abstract
Oxidative stress-induced lipid peroxidation has been associated with human physiology and diseases including cancer. Overwhelming data suggest that reactive lipid mediators generated from this process, such as 4-hydroxynonenal (4-HNE), are biomarkers for oxidative stress and important players for mediating a number of signaling pathways. The biological effects of 4-HNE are primarily due to covalent modification of important biomolecules including proteins, DNA, and phospholipids containing amino group. In this review, we summarize recent progress on the role of 4-HNE in pathogenesis of cancer and focus on the involvement of mitochondria: generation of 4-HNE from oxidation of mitochondria-specific phospholipid cardiolipin; covalent modification of mitochondrial proteins, lipids, and DNA; potential therapeutic strategies for targeting mitochondrial ROS generation, lipid peroxidation, and 4-HNE.
Collapse
Affiliation(s)
- Huiqin Zhong
- Key Laboratory of Food Safety Research, Institute for Nutritional Sciences (INS), Shanghai Institutes for Biological Sciences (SIBS), Chinese Academy of Sciences (CAS), Shanghai, China; University of the Chinese Academy of Sciences, CAS, Beijing, China; Key Laboratory of Food Safety Risk Assessment, Ministry of Health, Beijing, China
| | - Huiyong Yin
- Key Laboratory of Food Safety Research, Institute for Nutritional Sciences (INS), Shanghai Institutes for Biological Sciences (SIBS), Chinese Academy of Sciences (CAS), Shanghai, China; University of the Chinese Academy of Sciences, CAS, Beijing, China; Key Laboratory of Food Safety Risk Assessment, Ministry of Health, Beijing, China; School of Life Science and Technology, ShanghaiTech University, Shanghai, China.
| |
Collapse
|
33
|
Barlow J, Hirschberg Jensen V, Affourtit C. Uncoupling protein-2 attenuates palmitoleate protection against the cytotoxic production of mitochondrial reactive oxygen species in INS-1E insulinoma cells. Redox Biol 2014; 4:14-22. [PMID: 25482405 PMCID: PMC4309862 DOI: 10.1016/j.redox.2014.11.009] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2014] [Revised: 11/21/2014] [Accepted: 11/24/2014] [Indexed: 01/23/2023] Open
Abstract
High glucose and fatty acid levels impair pancreatic beta cell function. We have recently shown that palmitate-induced loss of INS-1E insulinoma cells is related to increased reactive oxygen species (ROS) production as both toxic effects are prevented by palmitoleate. Here we show that palmitate-induced ROS are mostly mitochondrial: oxidation of MitoSOX, a mitochondria-targeted superoxide probe, is increased by palmitate, whilst oxidation of the equivalent non-targeted probe is unaffected. Moreover, mitochondrial respiratory inhibition with antimycin A stimulates palmitate-induced MitoSOX oxidation. We also show that palmitate does not change the level of mitochondrial uncoupling protein-2 (UCP2) and that UCP2 knockdown does not affect palmitate-induced MitoSOX oxidation. Palmitoleate does not influence MitoSOX oxidation in INS-1E cells ±UCP2 and largely prevents the palmitate-induced effects. Importantly, UCP2 knockdown amplifies the preventive effect of palmitoleate on palmitate-induced ROS. Consistently, viability effects of palmitate and palmitoleate are similar between cells ±UCP2, but UCP2 knockdown significantly augments the palmitoleate protection against palmitate-induced cell loss at high glucose. We conclude that UCP2 neither mediates palmitate-induced mitochondrial ROS generation and the associated cell loss, nor protects against these deleterious effects. Instead, UCP2 dampens palmitoleate protection against palmitate toxicity.
Collapse
Affiliation(s)
- Jonathan Barlow
- School of Biomedical and Healthcare Sciences, Plymouth University, Drake Circus, Plymouth PL4 8AA, UK
| | - Verena Hirschberg Jensen
- School of Biomedical and Healthcare Sciences, Plymouth University, Drake Circus, Plymouth PL4 8AA, UK
| | - Charles Affourtit
- School of Biomedical and Healthcare Sciences, Plymouth University, Drake Circus, Plymouth PL4 8AA, UK.
| |
Collapse
|
34
|
Uncoupling protein 2 and 4 expression pattern during stem cell differentiation provides new insight into their putative function. PLoS One 2014; 9:e88474. [PMID: 24523901 PMCID: PMC3921169 DOI: 10.1371/journal.pone.0088474] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2013] [Accepted: 01/08/2014] [Indexed: 11/19/2022] Open
Abstract
Apart from the first family member, uncoupling protein 1 (UCP1), the functions of other UCPs (UCP2-UCP5) are still unknown. In analyzing our own results and those previously published by others, we have assumed that UCP's cellular expression pattern coincides with a specific cell metabolism and changes if the latter is altered. To verify this hypothesis, we analyzed the expression of UCP1-5 in mouse embryonic stem cells before and after their differentiation to neurons. We have shown that only UCP2 is present in undifferentiated stem cells and it disappears simultaneously with the initiation of neuronal differentiation. In contrast, UCP4 is simultaneously up-regulated together with typical neuronal marker proteins TUJ-1 and NeuN during mESC differentiation in vitro as well as during murine brain development in vivo. Notably, several tested cell lines express UCP2, but not UCP4. In line with this finding, neuroblastoma cells that display metabolic features of tumor cells express UCP2, but not UCP4. UCP2's occurrence in cancer, immunological and stem cells indicates that UCP2 is present in cells with highly proliferative potential, which have a glycolytic type of metabolism as a common feature, whereas UCP4 is strongly associated with non-proliferative highly differentiated neuronal cells.
Collapse
|
35
|
Óvilo C, Benítez R, Fernández A, Isabel B, Núñez Y, Fernández AI, Rodríguez C, Daza A, Silió L, López-Bote C. Dietary energy source largely affects tissue fatty acid composition but has minor influence on gene transcription in Iberian pigs. J Anim Sci 2014; 92:939-54. [PMID: 24492573 DOI: 10.2527/jas.2013-6988] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
A trial was performed to compare the effects of different dietary sources of MUFA on the fatty acid (FA) composition, lipid metabolism, and gene transcription in different tissues of Iberian pigs. Twenty-seven Iberian male pigs of 28 kg live weight (LW) were divided in 2 groups and fed with 1 of 2 isocaloric diets: a standard diet with carbohydrates as energy source (CH) and a diet enriched with high-oleic sunflower oil (HO). Ham adipose tissue was sampled by biopsy at 44 and 70 kg LW. At 110 kg LW pigs were slaughtered and backfat, loin, and liver tissues were sampled. Animals of the HO group showed higher MUFA content and lower SFA in all the analyzed tissues (P < 0.001). These main effects were established early during the treatment and increased only slightly along time. Small diet effects were also detected on PUFA, which showed differences according to sampling time, tissue, and lipid fraction. Effects of diet on gene expression were explored with a combined approach analyzing adipose tissue transcriptome and quantifying the expression of a panel of key genes implicated in lipogenesis and lipid metabolism processes in backfat, muscle, and liver. Backfat transcriptome showed small effects of diet on gene expression, in number and magnitude. According to the posterior probabilities (PP) of the probe-specific expression differences between dietary groups (PP < 0.01), 37 genes were considered differentially expressed (DE). Gene ontology allowed relating them with several biological functions including lipid metabolic processes. Quantitative PCR confirmed several DE genes in adipose tissue (RXRG, LEP, and ME1; P < 0.0001, P < 0.05, and P < 0.0001, respectively), but no DE gene was found in loin or liver tissues. Joint results agree with a metabolic adjustment of adipose tissue FA levels by the subtle effect of the diet on the regulation of several lipid metabolism pathways, mainly FA oxidation and prostanoid synthesis, with LEP, RXRG, and PTGS2 genes playing mayor roles.
Collapse
Affiliation(s)
- C Óvilo
- Departamento de Mejora Genética Animal, INIA, 28040 Madrid, Spain
| | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
New role of silent information regulator 1 in cerebral ischemia. Neurobiol Aging 2013; 34:2879-88. [DOI: 10.1016/j.neurobiolaging.2013.06.008] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2012] [Revised: 06/06/2013] [Accepted: 06/14/2013] [Indexed: 01/23/2023]
|
37
|
Malingriaux EA, Rupprecht A, Gille L, Jovanovic O, Jezek P, Jaburek M, Pohl EE. Fatty acids are key in 4-hydroxy-2-nonenal-mediated activation of uncoupling proteins 1 and 2. PLoS One 2013; 8:e77786. [PMID: 24204965 PMCID: PMC3810126 DOI: 10.1371/journal.pone.0077786] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2013] [Accepted: 09/12/2013] [Indexed: 11/19/2022] Open
Abstract
The production of reactive oxygen species (ROS) in mitochondria is very sensitive to the proton motive force and may be decreased by mild uncoupling, mediated e.g. by mitochondrial uncoupling proteins (UCPs). UCPs were conversely hypothesized to be activated by ROS. Conclusions from experiments studying the reactive product of lipid peroxidation 4-hydroxy-2-nonenal (HNE) in isolated mitochondria and UCP knock-out mice are highly controversial. Here we investigated the molecular mechanism of HNE action by evaluating the separate contributions of lipid and protein phases of the membrane and by comparing UCP1 and UCP2, which were reconstituted in planar lipid bilayers. We demonstrated that aldehyde does not directly activate either UCP1 or UCP2. However, HNE strongly potentiated the membrane conductance increase (Gm) mediated by different long-chain fatty acids in UCP-containing and in UCP-free membranes and this suggest the involvement of both lipid-mediated and protein-mediated mechanisms with FA playing the central role. Gm increase was concentration-dependent and exhibited a typical saturation kinetic with the binding constant 0.3 mM. By using Electron Paramagnetic Resonance, membrane fluidity change could be excluded as a cause for the HNE-mediated increase in the presence of FA. The impact of the HNE binding to definite positively charged UCP amino acid residues is discussed as a possible protein-mediated mechanism of the UCP activation.
Collapse
Affiliation(s)
- Elena A. Malingriaux
- Institute of Physiology, Pathophysiology and Biophysics, University of Veterinary Medicine, Vienna, Austria
| | - Anne Rupprecht
- Institute of Physiology, Pathophysiology and Biophysics, University of Veterinary Medicine, Vienna, Austria
| | - Lars Gille
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine, Vienna, Austria
| | - Olga Jovanovic
- Institute of Physiology, Pathophysiology and Biophysics, University of Veterinary Medicine, Vienna, Austria
| | - Petr Jezek
- Institute of Physiology, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | - Martin Jaburek
- Institute of Physiology, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | - Elena E. Pohl
- Institute of Physiology, Pathophysiology and Biophysics, University of Veterinary Medicine, Vienna, Austria
- * E-mail:
| |
Collapse
|
38
|
Pung YF, Sam WJ, Hardwick JP, Yin L, Ohanyan V, Logan S, Di Vincenzo L, Chilian WM. The role of mitochondrial bioenergetics and reactive oxygen species in coronary collateral growth. Am J Physiol Heart Circ Physiol 2013; 305:H1275-80. [PMID: 23997092 DOI: 10.1152/ajpheart.00077.2013] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Coronary collateral growth is a process involving coordination between growth factors expressed in response to ischemia and mechanical forces. Underlying this response is proliferation of vascular smooth muscle and endothelial cells, resulting in an enlargement in the caliber of arterial-arterial anastomoses, i.e., a collateral vessel, sometimes as much as an order of magnitude. An integral element of this cell proliferation is the process known as phenotypic switching in which cells of a particular phenotype, e.g., contractile vascular smooth muscle, must change their phenotype to proliferate. Phenotypic switching requires that protein synthesis occurs and different kinase signaling pathways become activated, necessitating energy to make the switch. Moreover, kinases, using ATP to phosphorylate their targets, have an energy requirement themselves. Mitochondria play a key role in the energy production that enables phenotypic switching, but under conditions where mitochondrial energy production is constrained, e.g., mitochondrial oxidative stress, this switch is impaired. In addition, we discuss the potential importance of uncoupling proteins as modulators of mitochondrial reactive oxygen species production and bioenergetics, as well as the role of AMP kinase as an energy sensor upstream of mammalian target of rapamycin, the master regulator of protein synthesis.
Collapse
Affiliation(s)
- Yuh Fen Pung
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, Ohio
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Anedda A, López-Bernardo E, Acosta-Iborra B, Saadeh Suleiman M, Landázuri MO, Cadenas S. The transcription factor Nrf2 promotes survival by enhancing the expression of uncoupling protein 3 under conditions of oxidative stress. Free Radic Biol Med 2013; 61:395-407. [PMID: 23597505 DOI: 10.1016/j.freeradbiomed.2013.04.007] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2012] [Revised: 04/01/2013] [Accepted: 04/05/2013] [Indexed: 10/27/2022]
Abstract
Uncoupling protein 3 (UCP3) is a member of the mitochondrial inner membrane carrier superfamily that modulates energy efficiency by catalyzing proton conductance and thus decreasing the production of superoxide anion. However, its role during oxidative stress and the underlying regulatory and molecular mechanisms remain poorly understood. We sought to investigate how UCP3 expression is regulated by oxidative stress and to evaluate the putative antioxidant role of this protein. H2O2 treatment increased UCP3 expression and the nuclear accumulation of the transcription factor Nrf2 in C2C12 and HL-1 cells. Nrf2 siRNA prevented H2O2-induced UCP3 expression, increasing oxidative stress and cell death. ChIP assays identified an antioxidant-response element (ARE) within the UCP3 promoter that bound Nrf2 after exposure to H2O2. Luciferase reporter experiments confirmed increased ARE activity in H2O2-treated HL-1 cells. Importantly, H2O2 increased the UCP3-mediated proton leak, suggesting a role for this protein in attenuating ROS-induced damage. Nrf2 nuclear accumulation and increased UCP3 protein were also detected in intact mouse heart subjected to a condition known to increase ROS generation. This is the first study to demonstrate that H2O2 augments UCP3 expression and it provides the first evidence of Nrf2 binding to the UCP3 promoter in response to oxidative challenge. These findings suggest that UCP3 functions as a member of the cellular antioxidant defense system that protects against oxidative stress in vivo. In conclusion, we have identified a novel regulatory process induced by an oxidative insult whereby the expression of the mitochondrial protein UCP3 is driven by the Nrf2 transcription factor, which decreases ROS production and prevents cell death.
Collapse
Affiliation(s)
- Andrea Anedda
- Servicio de Inmunología, Hospital Universitario de La Princesa, Instituto de Investigación Sanitaria Princesa (IP), 28006 Madrid, Spain
| | - Elia López-Bernardo
- Servicio de Inmunología, Hospital Universitario de La Princesa, Instituto de Investigación Sanitaria Princesa (IP), 28006 Madrid, Spain
| | - Bárbara Acosta-Iborra
- Servicio de Inmunología, Hospital Universitario de La Princesa, Instituto de Investigación Sanitaria Princesa (IP), 28006 Madrid, Spain; Unidad de Investigación, Hospital Universitario Santa Cristina, Instituto de Investigación Sanitaria Princesa (IP), 28009 Madrid, Spain
| | | | - Manuel O Landázuri
- Servicio de Inmunología, Hospital Universitario de La Princesa, Instituto de Investigación Sanitaria Princesa (IP), 28006 Madrid, Spain; Unidad de Investigación, Hospital Universitario Santa Cristina, Instituto de Investigación Sanitaria Princesa (IP), 28009 Madrid, Spain
| | - Susana Cadenas
- Servicio de Inmunología, Hospital Universitario de La Princesa, Instituto de Investigación Sanitaria Princesa (IP), 28006 Madrid, Spain; Departamento de Biología Molecular, Facultad de Ciencias, Universidad Autónoma de Madrid, 28049 Madrid, Spain.
| |
Collapse
|
40
|
Barlow J, Hirschberg V, Brand MD, Affourtit C. Measuring Mitochondrial Uncoupling Protein-2 Level and Activity in Insulinoma Cells. Methods Enzymol 2013; 528:257-67. [DOI: 10.1016/b978-0-12-405881-1.00015-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/10/2023]
|
41
|
Porter RK. Studies on the function and regulation of mitochondrial uncoupling proteins. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2012; 748:171-84. [PMID: 22729858 DOI: 10.1007/978-1-4614-3573-0_7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/12/2023]
Abstract
Mitochondrial uncoupling proteins are members of the SLC25 family of solute carriers. Models of mitochondrial transporter function predict that uncoupling proteins are solute carriers. Evidence in the literature suggests that uncoupling proteins can transport protons, fatty acid anions, chloride anions, and recently the dicarboxylate succinate. Studies have also demonstrated that UCPs can be covalently modified and in some instances this covalent modification is needed to affect uncoupling function. The current evidence from functional analyses of mammalian uncoupling proteins is summarized in this chapter.
Collapse
Affiliation(s)
- Richard K Porter
- School of Biochemistry and Immunology, Trinity College Dublin, Dublin 2, Ireland.
| |
Collapse
|
42
|
Papkovskaia TD, Chau KY, Inesta-Vaquera F, Papkovsky DB, Healy DG, Nishio K, Staddon J, Duchen MR, Hardy J, Schapira AHV, Cooper JM. G2019S leucine-rich repeat kinase 2 causes uncoupling protein-mediated mitochondrial depolarization. Hum Mol Genet 2012; 21:4201-13. [PMID: 22736029 PMCID: PMC3441120 DOI: 10.1093/hmg/dds244] [Citation(s) in RCA: 128] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
The G2019S leucine rich repeat kinase 2 (LRRK2) mutation is the most common genetic cause of Parkinson's disease (PD), clinically and pathologically indistinguishable from idiopathic PD. Mitochondrial abnormalities are a common feature in PD pathogenesis and we have investigated the impact of G2019S mutant LRRK2 expression on mitochondrial bioenergetics. LRRK2 protein expression was detected in fibroblasts and lymphoblasts at levels higher than those observed in the mouse brain. The presence of G2019S LRRK2 mutation did not influence LRRK2 expression in fibroblasts. However, the expression of the G2019S LRRK2 mutation in both fibroblast and neuroblastoma cells was associated with mitochondrial uncoupling. This was characterized by decreased mitochondrial membrane potential and increased oxygen utilization under basal and oligomycin-inhibited conditions. This resulted in a decrease in cellular ATP levels consistent with compromised cellular function. This uncoupling of mitochondrial oxidative phosphorylation was associated with a cell-specific increase in uncoupling protein (UCP) 2 and 4 expression. Restoration of mitochondrial membrane potential by the UCP inhibitor genipin confirmed the role of UCPs in this mechanism. The G2019S LRRK2-induced mitochondrial uncoupling and UCP4 mRNA up-regulation were LRRK2 kinase-dependent, whereas endogenous LRRK2 levels were required for constitutive UCP expression. We propose that normal mitochondrial function was deregulated by the expression of G2019S LRRK2 in a kinase-dependent mechanism that is a modification of the normal LRRK2 function, and this leads to the vulnerability of selected neuronal populations in PD.
Collapse
Affiliation(s)
- Tatiana D Papkovskaia
- Department of Clinical Neurosciences, Institute of Neurology, University College London, London NW3 2PF, UK
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Hoeks J, Arany Z, Phielix E, Moonen-Kornips E, Hesselink MKC, Schrauwen P. Enhanced lipid-but not carbohydrate-supported mitochondrial respiration in skeletal muscle of PGC-1α overexpressing mice. J Cell Physiol 2012; 227:1026-33. [PMID: 21520076 DOI: 10.1002/jcp.22812] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Skeletal muscle mitochondrial dysfunction has been linked to several disease states as well as the process of aging. A possible factor involved is the peroxisome proliferator-activated receptor (PPAR) γ co-activator 1α (PGC-1α), a major player in the regulation of skeletal muscle mitochondrial metabolism. However, it is currently unknown whether PGC-1α, besides stimulating mitochondrial proliferation, also affects the functional capacity per mitochondrion. Therefore, we here tested whether PGC-1α overexpression, besides increasing mitochondrial content, also leads to intrinsic mitochondrial adaptations. Skeletal muscle mitochondria from 10 male, muscle-specific PGC-1α overexpressing mice (PGC-1αTg) and 8 wild-type (WT) mice were isolated. Equal mitochondrial quantities were then analyzed for their oxidative capacity by high-resolution respirometry, fuelled by a carbohydrate-derived (pyruvate) and a lipid (palmitoyl-CoA plus carnitine) substrate. Additionally, mitochondria were tested for reactive oxygen species (superoxide) production and fatty acid (FA)-induced uncoupling. PGC-1αTg mitochondria were characterized by an improved intrinsic mitochondrial fat oxidative capacity as evidenced by pronounced increase in ADP-stimulated respiration (P < 0.001) and maximal uncoupled respiration (P < 0.001) upon palmitoyl-CoA plus carnitine. Interestingly, intrinsic mitochondrial capacity on a carbohydrate-derived substrate tended to be reduced. Furthermore, the sensitivity to FA-induced uncoupling was diminished in PGC-1αTg mitochondria (P = 0.02) and this was accompanied by a blunted reduction in mitochondrial ROS production upon FAs in PGC-1αTg versus WT mitochondria (P = 0.04). Uncoupling protein 3 (UCP3) levels were markedly reduced in PGC-1αTg mitochondria (P < 0.001). Taken together, in addition to stimulating mitochondrial proliferation in skeletal muscle, we show here that overexpression of PGC-1α leads to intrinsic mitochondrial adaptations that seem restricted to fat metabolism.
Collapse
Affiliation(s)
- Joris Hoeks
- NUTRIM-School for Nutrition, Toxicology and Metabolism, Department of Human Biology, Maastricht University Medical Center, Maastricht, The Netherlands.
| | | | | | | | | | | |
Collapse
|
44
|
Ho PWL, Ho JWM, Tse HM, So DHF, Yiu DCW, Liu HF, Chan KH, Kung MHW, Ramsden DB, Ho SL. Uncoupling protein-4 (UCP4) increases ATP supply by interacting with mitochondrial Complex II in neuroblastoma cells. PLoS One 2012; 7:e32810. [PMID: 22427795 PMCID: PMC3303587 DOI: 10.1371/journal.pone.0032810] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2011] [Accepted: 02/03/2012] [Indexed: 12/21/2022] Open
Abstract
Mitochondrial uncoupling protein-4 (UCP4) protects against Complex I deficiency as induced by 1-methyl-4-phenylpyridinium (MPP(+)), but how UCP4 affects mitochondrial function is unclear. Here we investigated how UCP4 affects mitochondrial bioenergetics in SH-SY5Y cells. Cells stably overexpressing UCP4 exhibited higher oxygen consumption (10.1%, p<0.01), with 20% greater proton leak than vector controls (p<0.01). Increased ATP supply was observed in UCP4-overexpressing cells compared to controls (p<0.05). Although state 4 and state 3 respiration rates of UCP4-overexpressing and control cells were similar, Complex II activity in UCP4-overexpressing cells was 30% higher (p<0.05), associated with protein binding between UCP4 and Complex II, but not that of either Complex I or IV. Mitochondrial ADP consumption by succinate-induced respiration was 26% higher in UCP4-overexpressing cells, with 20% higher ADP:O ratio (p<0.05). ADP/ATP exchange rate was not altered by UCP4 overexpression, as shown by unchanged mitochondrial ADP uptake activity. UCP4 overexpression retained normal mitochondrial morphology in situ, with similar mitochondrial membrane potential compared to controls. Our findings elucidate how UCP4 overexpression increases ATP synthesis by specifically interacting with Complex II. This highlights a unique role of UCP4 as a potential regulatory target to modulate mitochondrial Complex II and ATP output in preserving existing neurons against energy crisis.
Collapse
Affiliation(s)
- Philip Wing-Lok Ho
- Division of Neurology, University Department of Medicine, University of Hong Kong, Hong Kong, Hong Kong.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Souza BMD, Assmann TS, Kliemann LM, Gross JL, Canani LH, Crispim D. The role of uncoupling protein 2 (UCP2) on the development of type 2 diabetes mellitus and its chronic complications. ACTA ACUST UNITED AC 2012; 55:239-48. [PMID: 21779625 DOI: 10.1590/s0004-27302011000400001] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2011] [Accepted: 04/29/2011] [Indexed: 11/21/2022]
Abstract
It is well established that genetic factors play an important role in the development of type 2 diabetes mellitus (DM2) and its chronic complications, and that genetically susceptible subjects can develop the disease after being exposed to environmental risk factors. Therefore, great efforts have been made to identify genes associated with DM2. Uncoupling protein 2 (UCP2) is expressed in several tissues, and acts in the protection against oxidative stress; in the negative regulation of insulin secretion by beta cells, and in fatty acid metabolism. All these mechanisms are associated with DM2 pathogenesis and its chronic complications. Therefore, UCP2 is a candidate gene for the development of these disorders. Indeed, several studies have reported that three common polymorphisms in UCP2 gene are possibly associated with DM2 and/or obesity. Only a few studies investigated these polymorphisms in relation to chronic complications of diabetes, with inconclusive results.
Collapse
|
46
|
de Souza BM, Assmann TS, Kliemann LM, Marcon AS, Gross JL, Canani LH, Crispim D. The presence of the -866A/55Val/Ins haplotype in the uncoupling protein 2 (UCP2) gene is associated with decreased UCP2 gene expression in human retina. Exp Eye Res 2011; 94:49-55. [PMID: 22134120 DOI: 10.1016/j.exer.2011.11.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2011] [Revised: 10/14/2011] [Accepted: 11/14/2011] [Indexed: 12/31/2022]
Abstract
Uncoupling protein 2 (UCP2) is a mitochondrial transporter present in the inner membrane of mitochondria, and it uncouples substrate oxidation from ATP synthesis, thereby dissipating the membrane potential energy and consequently decreasing ATP production by mitochondrial respiratory chain. As a consequence of the uncoupling, UCP2 decreases the reactive oxygen species (ROS) formation by mitochondria. ROS overproduction is related to diabetic retinopathy (DR), a chronic complication of diabetes mellitus (DM). Recently, our group reported that the -866A/55Val/Ins haplotype (-866G/A, Ala55Val and Ins/Del polymorphisms) of the UCP2 gene was associated with increased risk for DR in patients with DM. The purpose of this study was to analyze the effect of this haplotype on UCP2 gene expression in human retina. In addition, MnSOD2 gene expression was also investigated according to different UCP2 haplotypes. This cross-sectional study included 188 cadaveric cornea donors. In a subset of 91 retinal samples differentiated according to the presence of the mutated UCP2 haplotype and risk alleles of the -866G/A and Ins/Del polymorphisms, UCP2 and MnSOD2 gene expressions were measured by semi-quantitative RT-qPCR. Mutated UCP2 haplotype carriers (homozygous + heterozygous) had a lower UCP2 gene expression than reference haplotype carriers (8.4 ± 7.6 vs. 18.8 ± 23.7 arbitrary units; P = 0.046). Accordingly, UCP2 gene expression was decreased in -866A carriers when compared with G/G carriers (P = 0.010). UCP2 gene expression did not differ between Ins allele carriers and Del/Del carriers (P = 0.556). Interestingly, subjects carrying the heterozygous UCP2 haplotype showed increased MnSOD2 gene expression (P = 0.025). This is the first report suggesting that the presence of the -866A/55Val/Ins haplotype is associated with decreased UCP2 gene expression in human retina. Possibly, MnSOD2 expression might influence the UCP2 effect in the protection against oxidative stress.
Collapse
Affiliation(s)
- Bianca M de Souza
- Endocrinology Division, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil
| | | | | | | | | | | | | |
Collapse
|
47
|
Divakaruni AS, Brand MD. The regulation and physiology of mitochondrial proton leak. Physiology (Bethesda) 2011; 26:192-205. [PMID: 21670165 DOI: 10.1152/physiol.00046.2010] [Citation(s) in RCA: 304] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Mitochondria couple respiration to ATP synthesis through an electrochemical proton gradient. Proton leak across the inner membrane allows adjustment of the coupling efficiency. The aim of this review is threefold: 1) introduce the unfamiliar reader to proton leak and its physiological significance, 2) review the role and regulation of uncoupling proteins, and 3) outline the prospects of proton leak as an avenue to treat obesity, diabetes, and age-related disease.
Collapse
Affiliation(s)
- Ajit S Divakaruni
- Medical Research Council Mitochondrial Biology Unit, Wellcome Trust/MRC Building, Cambridge, United Kingdom
| | | |
Collapse
|
48
|
Niño Fong R, Fatehi-Hassanabad Z, Lee SC, Lu H, Wheeler MB, Chan CB. Uncoupling protein-2 increases nitric oxide production and TNFAIP3 pathway activation in pancreatic islets. J Mol Endocrinol 2011; 46:193-204. [PMID: 21321094 PMCID: PMC3077812 DOI: 10.1530/jme-10-0117] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Mutations in the uncoupling protein 2 (Ucp2) gene are linked to type-2 diabetes. Here, a potential mechanism by which lack of UCP2 is cytoprotective in pancreatic β-cells was investigated. Nitric oxide (NO) production was elevated in Ucp2(-/-) islets. Proliferation (cyclin D2, Ccnd2) and anti-apoptosis (Tnfaip3) genes had increased expression in Ucp2(-/-) islets, whereas the mRNA of pro-apoptosis genes (Jun, Myc) was reduced. TNFAIP3 cellular localization was detected in both α- and β-cells of Ucp2(-/-) islets but in neither α- nor β-cells of UCP2(+)(/)(+) islets, where it was detected in pancreatic polypeptide-expressing cells. TNFAIP3 distribution was not markedly altered 14 days after streptozotocin treatment. Basal apoptosis was attenuated in Ucp2(-/-) β-cells, while the nuclear factor κB (NF-κB) pathway was transactivated after islet isolation. Ucp2(+/+) and Ucp2(-/-) islets were treated with cytokines for 24 h. Cytokines did not increase NF-κB transactivation or apoptosis in Ucp2(-/-) islets and TNFAIP3 was more strongly induced in Ucp2(-/-) islets. Inhibition of NO production strongly reduced NF-κB activation and apoptosis. These data show that null expression of Ucp2 induces transactivation of NF-κB in isolated islets, possibly due to NO-dependent up-regulation of inhibitor of κB kinase β activity. NF-κB transactivation appears to result in altered expression of genes that enhance a pro-survival phenotype basally and when β-cells are exposed to cytokines. TNFAIP3 is of particular interest because of its ability to regulate NF-κB signaling pathways.
Collapse
Affiliation(s)
- Rodolfo Niño Fong
- Department of Biomedical SciencesUniversity of Prince Edward IslandCharlottetown, Prince Edward IslandCanadaC1A 4P3
| | - Zahra Fatehi-Hassanabad
- Departments of Physiology and Agricultural Food and Nutritional Sciences, 6-126B Li Ka Shing CentreUniversity of AlbertaEdmonton, AlbertaCanadaT6G 2R3
| | - Simon C Lee
- Departments of Physiology and MedicineUniversity of TorontoToronto, OntarioCanadaM5S 1A8
| | - Hongfang Lu
- Departments of Physiology and MedicineUniversity of TorontoToronto, OntarioCanadaM5S 1A8
| | - Michael B Wheeler
- Departments of Physiology and MedicineUniversity of TorontoToronto, OntarioCanadaM5S 1A8
| | - Catherine B Chan
- Department of Biomedical SciencesUniversity of Prince Edward IslandCharlottetown, Prince Edward IslandCanadaC1A 4P3
- Departments of Physiology and Agricultural Food and Nutritional Sciences, 6-126B Li Ka Shing CentreUniversity of AlbertaEdmonton, AlbertaCanadaT6G 2R3
- (Correspondence should be addressed to C B Chan at Department of Physiology, 6-126B Li Ka Shing Centre, University of Alberta; )
| |
Collapse
|
49
|
Cardoso S, Santos RX, Carvalho C, Correia S, Santos MS, Moreira PI. Mitochondrial Uncoupling Proteins and Oxidative Stress: Implications for Diabetes and Neurodegeneration. ACTA ACUST UNITED AC 2011. [DOI: 10.5530/ax.2011.2.3] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
50
|
Affourtit C, Jastroch M, Brand MD. Uncoupling protein-2 attenuates glucose-stimulated insulin secretion in INS-1E insulinoma cells by lowering mitochondrial reactive oxygen species. Free Radic Biol Med 2011; 50:609-16. [PMID: 21172424 PMCID: PMC3036803 DOI: 10.1016/j.freeradbiomed.2010.12.020] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2010] [Revised: 11/30/2010] [Accepted: 12/11/2010] [Indexed: 01/26/2023]
Abstract
Glucose-stimulated insulin secretion (GSIS) by pancreatic β cells is regulated by mitochondrial uncoupling protein-2 (UCP2), but opposing phenotypes, GSIS improvement and impairment, have been reported for different Ucp2-ablated mouse models. By measuring mitochondrial bioenergetics in attached INS-1E insulinoma cells with and without UCP2, we show that UCP2 contributes to proton leak and attenuates glucose-induced rises in both respiratory activity and the coupling efficiency of oxidative phosphorylation. Strikingly, the GSIS improvement seen upon UCP2 knockdown in INS-1E cells is annulled completely by the cell-permeative antioxidant MnTMPyP. Consistent with this observation, UCP2 lowers mitochondrial reactive oxygen species at high glucose levels. We conclude that UCP2 plays both regulatory and protective roles in β cells by acutely lowering GSIS and chronically preventing oxidative stress. Our findings thus provide a mechanistic explanation for the apparently discrepant findings in the field.
Collapse
Key Words
- ∆ψ, mitochondrial membrane potential
- dapi, 4′,6-diamidino-2-phenylindole
- dhe, hydroethidine
- fcs, fetal calf serum
- fccp, carbonyl cyanide p-trifluoromethoxyphenylhydrazone
- gsis, glucose-stimulated insulin secretion
- krh, hepes-buffered krebs–ringer medium
- mitosox, mitochondria-targeted hydroethidine
- mntbap, manganese tetrakis-(4-benzoic acid) porphyrin
- mntmpyp, manganese tetrakis-(n-methyl-4-pyridyl) porphyrin
- ros, reactive oxygen species
- ttnpb, 4-[(e)-2-(5,6,7,8-tetrahydro-5,5,8,8-tetramethyl-2-napthalenyl)-1-propenyl]benzoic acid
- ucp2, uncoupling protein-2
- pancreatic β cells
- glucose-stimulated insulin secretion
- uncoupling protein 2
- mitochondrial respiration
- reactive oxygen species
- coupling efficiency of oxidative phosphorylation
- type 2 diabetes
- metabolic syndrome
- free radicals
Collapse
Affiliation(s)
- Charles Affourtit
- Mitochondrial Biology Unit, Medical Research Council, Cambridge CB2 0XY,
| | | | | |
Collapse
|