1
|
Kandy AT, Chand J, Baba MZ, Subramanian G. Is SIRT3 and Mitochondria a Reliable Target for Parkinson's Disease and Aging? A Narrative Review. Mol Neurobiol 2025; 62:6898-6912. [PMID: 39287746 DOI: 10.1007/s12035-024-04486-w] [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: 03/13/2024] [Accepted: 09/09/2024] [Indexed: 09/19/2024]
Abstract
Aging is a complicated degenerative process that has been thoroughly researched in a variety of taxa, including mammals, worms, yeast, and flies. One important controller of organismal lifetime is the conserved deacetylase protein known as silencing information regulator 2 (SIR2). It has been demonstrated that overexpressing SIR2 lengthens the life span in worms, flies, and yeast, demonstrating its function in enhancing longevity. SIRT3 is a member of the sirtuin protein family, identified as a major regulator of longevity and aging. Sirtuin 3 (SIRT3), a possible mitochondrial tumor suppressor, has been explicitly linked to the control of cellular reactive oxygen species (ROS) levels, the Warburg effect, and carcinogenesis. SIRT3 plays a significant part in neurodegenerative illnesses such as Parkinson's and Alzheimer's disease by decreasing the oxidative stress in mitochondria and reducing the ROS levels. Furthermore, SIRT3 has been linked to metabolic and cardiovascular disorders, indicating its wider role in the pathophysiology of disease and possible therapeutic applications.
Collapse
Affiliation(s)
- Amarjith Thiyyar Kandy
- Department of Pharmaceutical Chemistry, JSS College Of Pharmacy, JSS Academy of Higher Education & Research, Ooty, Nilgiris, Tamilnadu-643001, India
| | - Jagdish Chand
- Department of Pharmaceutical Chemistry, JSS College Of Pharmacy, JSS Academy of Higher Education & Research, Ooty, Nilgiris, Tamilnadu-643001, India
| | - Mohammad Zubair Baba
- Department of Pharmaceutical Chemistry, JSS College Of Pharmacy, JSS Academy of Higher Education & Research, Ooty, Nilgiris, Tamilnadu-643001, India
| | - Gomathy Subramanian
- Department of Pharmaceutical Chemistry, JSS College Of Pharmacy, JSS Academy of Higher Education & Research, Ooty, Nilgiris, Tamilnadu-643001, India.
| |
Collapse
|
2
|
John S, Calmettes G, Xu S, Ribalet B. Real-time resolution studies of the regulation of pyruvate-dependent lactate metabolism by hexokinases in single cells. PLoS One 2023; 18:e0286660. [PMID: 37917627 PMCID: PMC10621844 DOI: 10.1371/journal.pone.0286660] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 05/21/2023] [Indexed: 11/04/2023] Open
Abstract
Lactate is a mitochondrial substrate for many tissues including neuron, muscle, skeletal and cardiac, as well as many cancer cells, however little is known about the processes that regulate its utilization in mitochondria. Based on the close association of Hexokinases (HK) with mitochondria, and the known cardio-protective role of HK in cardiac muscle, we have investigated the regulation of lactate and pyruvate metabolism by hexokinases (HKs), utilizing wild-type HEK293 cells and HEK293 cells in which the endogenous HKI and/or HKII have been knocked down to enable overexpression of wild type and mutant HKs. To assess the real-time changes in intracellular lactate levels the cells were transfected with a lactate specific FRET probe. In the HKI/HKII double knockdown cells, addition of extracellular pyruvate caused a large and sustained decrease in lactate. This decrease was rapidly reversed upon inhibition of the malate aspartate shuttle by aminooxyacetate, or inhibition of mitochondrial oxidative respiration by NaCN. These results suggest that in the absence of HKs, pyruvate-dependent activation of the TCA cycle together with the malate aspartate shuttle facilitates lactate transformation into pyruvate and its utilization by mitochondria. With replacement by overexpression of HKI or HKII the cellular response to pyruvate and NaCN was modified. With either hexokinase present, both the decrease in lactate due to the addition of pyruvate and the increase following addition of NaCN were either transient or suppressed altogether. Blockage of the pentose phosphate pathway with the inhibitor 6-aminonicotinamide (6-AN), abolished the effects of HK replacement. These results suggest that blocking of the malate aspartate shuttle by HK may involve activation of the pentose phosphate pathway and increased NADPH production.
Collapse
Affiliation(s)
- Scott John
- Department of Medicine (Division of Cardiology), David Geffen School of Medicine at UCLA, Los Angeles, CA, United States of America
| | - Guillaume Calmettes
- Department of Medicine (Division of Cardiology), David Geffen School of Medicine at UCLA, Los Angeles, CA, United States of America
| | - Shili Xu
- California NanoSystems Institute (CNSI) 2151, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States of America
| | - Bernard Ribalet
- Department of Physiology, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States of America
| |
Collapse
|
3
|
Narne P, Phanithi PB. Role of NAD + and FAD in Ischemic Stroke Pathophysiology: An Epigenetic Nexus and Expanding Therapeutic Repertoire. Cell Mol Neurobiol 2023; 43:1719-1768. [PMID: 36180651 PMCID: PMC11412205 DOI: 10.1007/s10571-022-01287-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Accepted: 09/15/2022] [Indexed: 11/03/2022]
Abstract
The redox coenzymes viz., oxidized β-nicotinamide adenine dinucleotide (NAD+) and flavin adenine dinucleotide (FAD) by way of generation of optimal reducing power and cellular energy currency (ATP), control a staggering array of metabolic reactions. The prominent cellular contenders for NAD+ utilization, inter alia, are sirtuins (SIRTs) and poly(ADP-ribose) polymerase (PARP-1), which have been significantly implicated in ischemic stroke (IS) pathogenesis. NAD+ and FAD are also two crucial epigenetic enzyme-required metabolites mediating histone deacetylation and poly(ADP-ribosyl)ation through SIRTs and PARP-1 respectively, and demethylation through FAD-mediated lysine specific demethylase activity. These enzymes and post-translational modifications impinge on the components of neurovascular unit, primarily neurons, and elicit diverse functional upshots in an ischemic brain. These could be circumstantially linked with attendant cognitive deficits and behavioral outcomes in post-stroke epoch. Parsing out the contribution of NAD+/FAD-synthesizing and utilizing enzymes towards epigenetic remodeling in IS setting, together with their cognitive and behavioral associations, combined with possible therapeutic implications will form the crux of this review.
Collapse
Affiliation(s)
- Parimala Narne
- Department of Biotechnology and Bioinformatics, School of Life Sciences, University of Hyderabad, Hyderabad, Telangana State, 500046, India.
| | - Prakash Babu Phanithi
- Department of Biotechnology and Bioinformatics, School of Life Sciences, University of Hyderabad, Hyderabad, Telangana State, 500046, India.
| |
Collapse
|
4
|
Yu D, Zhang P, Xu C, Hu Y, Liang Y, Li M. Microplitis bicoloratus Bracovirus Promotes Cyclophilin D-Acetylation at Lysine 125 That Correlates with Apoptosis during Insect Immunosuppression. Viruses 2023; 15:1491. [PMID: 37515179 PMCID: PMC10383377 DOI: 10.3390/v15071491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 06/27/2023] [Accepted: 06/28/2023] [Indexed: 07/30/2023] Open
Abstract
Cyclophilin D (CypD) is regulated during the innate immune response of insects. However, the mechanism by which CypD is activated under innate immunosuppression is not understood. Microplitis bicoloratus bracovirus (MbBV), a symbiotic virus in the parasitoid wasp, Microplitis bicoloratus, suppresses innate immunity in parasitized Spodoptera litura. Here, we demonstrate that MbBV promotes the CypD acetylation of S. litura, resulting in an immunosuppressive phenotype characterized by increased apoptosis of hemocytes and MbBV-infected cells. Under MbBV infection, the inhibition of CypD acetylation significantly rescued the apoptotic cells induced by MbBV, and the point-mutant fusion proteins of CypDK125R-V5 were deacetylated. The CypD-V5 fusion proteins were acetylated in MbBV-infected cells. Deacetylation of CypDK125R-V5 can also suppress the MbBV-induced increase in apoptosis. These results indicate that CypD is involved in the MbBV-suppressed innate immune response via the CypD-acetylation pathway and S. litura CypD is acetylated on K125.
Collapse
Affiliation(s)
- Dan Yu
- Yunnan International Joint Laboratory of Virology & Immunology, School of Life Sciences, Yunnan University, Kunming 650500, China
- Yunnan Provincial Medical Investment Management Group Co., Ltd., Kunming 650500, China
| | - Pan Zhang
- Yunnan International Joint Laboratory of Virology & Immunology, School of Life Sciences, Yunnan University, Kunming 650500, China
| | - Cuixian Xu
- Yunnan International Joint Laboratory of Virology & Immunology, School of Life Sciences, Yunnan University, Kunming 650500, China
- School of Health, Yunnan Technology and Business University, Kunming 650500, China
| | - Yan Hu
- Yunnan International Joint Laboratory of Virology & Immunology, School of Life Sciences, Yunnan University, Kunming 650500, China
| | - Yaping Liang
- Yunnan International Joint Laboratory of Virology & Immunology, School of Life Sciences, Yunnan University, Kunming 650500, China
| | - Ming Li
- Yunnan International Joint Laboratory of Virology & Immunology, School of Life Sciences, Yunnan University, Kunming 650500, China
| |
Collapse
|
5
|
Ren C, Li X, Bai Y, Schroyen M, Zhang D. Phosphorylation and acetylation of glycolytic enzymes cooperatively regulate their activity and lamb meat quality. Food Chem 2022; 397:133739. [DOI: 10.1016/j.foodchem.2022.133739] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 05/26/2022] [Accepted: 07/16/2022] [Indexed: 11/04/2022]
|
6
|
Murugasamy K, Munjal A, Sundaresan NR. Emerging Roles of SIRT3 in Cardiac Metabolism. Front Cardiovasc Med 2022; 9:850340. [PMID: 35369299 PMCID: PMC8971545 DOI: 10.3389/fcvm.2022.850340] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 01/31/2022] [Indexed: 12/17/2022] Open
Abstract
The heart is a highly metabolically active organ that predominantly utilizes fatty acids as an energy substrate. The heart also derives some part of its energy by oxidation of other substrates, including glucose, lactose, amino acids and ketones. The critical feature of cardiac pathology is metabolic remodeling and loss of metabolic flexibility. Sirtuin 3 (SIRT3) is one of the seven mammalian sirtuins (SIRT1 to SIRT7), with NAD+ dependent deacetylase activity. SIRT3 is expressed in high levels in healthy hearts but downregulated in the aged or diseased hearts. Experimental evidence shows that increasing SIRT3 levels or activity can ameliorate several cardiac pathologies. The primary deacetylation targets of SIRT3 are mitochondrial proteins, most of which are involved in energy metabolism. Thus, SIRT3 improves cardiac health by modulating cardiac energetics. In this review, we discuss the essential role of SIRT3 in regulating cardiac metabolism in the context of physiology and pathology. Specifically, we summarize the recent advancements that emphasize the critical role of SIRT3 as a master regulator of cardiac metabolism. We also present a comprehensive view of all known activators of SIRT3, and elaborate on their therapeutic potential to ameliorate energetic abnormalities in various cardiac pathologies.
Collapse
|
7
|
Boyenle ID, Oyedele AK, Ogunlana AT, Adeyemo AF, Oyelere FS, Akinola OB, Adelusi TI, Ehigie LO, Ehigie AF. Targeting the mitochondrial permeability transition pore for drug discovery: Challenges and opportunities. Mitochondrion 2022; 63:57-71. [PMID: 35077882 DOI: 10.1016/j.mito.2022.01.006] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 12/22/2021] [Accepted: 01/17/2022] [Indexed: 12/29/2022]
Abstract
Several drug targets have been amenable to drug discovery pursuit not until the characterization of the mitochondrial permeability transition pore (MPTP), a pore with an undefined molecular identity that forms on the inner mitochondrial membrane upon mitochondrial permeability transition (MPT) under the influence of calcium overload and oxidative stress. The opening of the pore which is presumed to cause cell death in certain human diseases also has implications under physiological parlance. Different models for this pore have been postulated following its first identification in the last six decades. The mitochondrial community has witnessed many protein candidates such as; voltage-dependent anion channel (VDAC), adenine nucleotide translocase (ANT), Mitochondrial phosphate carrier (PiC), Spastic Paralegin (SPG7), disordered proteins, and F1Fo ATPase. However, genetic studies have cast out most of these candidates with only F1Fo ATPase currently under intense argument. Cyclophilin D (CyPD) remains the widely accepted positive regulator of the MPTP known to date, but no drug candidate has emerged as its inhibitor, raising concern issues for therapeutics. Thus, in this review, we discuss various models of MPTP reported with the hope of stimulating further research in this field. We went beyond the classical description of the MPTP to ascribe a 'two-edged sword property' to the pore for therapeutic function in human disease because its inhibition and activation have pharmacological relevance. We suggested putative proteins upstream to CyPD that can regulate its activity and prevent cell deaths in neurodegenerative disease and ischemia-reperfusion injury.
Collapse
Affiliation(s)
- Ibrahim Damilare Boyenle
- Membrane Biochemistry and Biophysics Research Laboratory, Department of Biochemistry, Ladoke Akintola University of Technology, Ogbomoso, Nigeria; Computational Biology/Drug Discovery Laboratory, Department of Biochemistry, Ladoke Akintola University of Technology, Ogbomoso, Nigeria
| | - Abdulquddus Kehinde Oyedele
- Computational Biology/Drug Discovery Laboratory, Department of Biochemistry, Ladoke Akintola University of Technology, Ogbomoso, Nigeria
| | - Abdeen Tunde Ogunlana
- Computational Biology/Drug Discovery Laboratory, Department of Biochemistry, Ladoke Akintola University of Technology, Ogbomoso, Nigeria
| | - Aishat Folashade Adeyemo
- Membrane Biochemistry and Biophysics Research Laboratory, Department of Biochemistry, Ladoke Akintola University of Technology, Ogbomoso, Nigeria
| | | | - Olateju Balikis Akinola
- Membrane Biochemistry and Biophysics Research Laboratory, Department of Biochemistry, Ladoke Akintola University of Technology, Ogbomoso, Nigeria
| | - Temitope Isaac Adelusi
- Computational Biology/Drug Discovery Laboratory, Department of Biochemistry, Ladoke Akintola University of Technology, Ogbomoso, Nigeria
| | - Leonard Ona Ehigie
- Computational Biology/Drug Discovery Laboratory, Department of Biochemistry, Ladoke Akintola University of Technology, Ogbomoso, Nigeria
| | - Adeola Folasade Ehigie
- Membrane Biochemistry and Biophysics Research Laboratory, Department of Biochemistry, Ladoke Akintola University of Technology, Ogbomoso, Nigeria.
| |
Collapse
|
8
|
Gu H, Yang K, Wu Q, Shen Z, Li X, Sun C. A link between protein acetylation and mitochondrial dynamics under energy metabolism: A comprehensive overview. J Cell Physiol 2021; 236:7926-7937. [PMID: 34101176 DOI: 10.1002/jcp.30461] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 05/06/2021] [Accepted: 05/24/2021] [Indexed: 12/16/2022]
Abstract
Cells adjust mitochondrial morphologies to coordinate between the cellular demand for energy and the availability of resources. Mitochondrial morphology is regulated by the balance between two counteracting mitochondrial processes of fusion and fission. Fission and fusion are dynamic and reversible processes that depend on the coordination of a number of proteins and are primarily regulated by posttranslational modifications. In the mitochondria, more than 20% of proteins are acetylated in proteomic surveys, partly involved in the dynamic regulation of mitochondrial fusion and fission. This article focuses on the molecular mechanism of the mitochondrial dynamics of fusion and fission, and summarizes the related mechanisms and targets of mitochondrial protein acetylation to regulate the mitochondrial dynamics of fusion and fission in energy metabolism.
Collapse
Affiliation(s)
- Huihui Gu
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Kun Yang
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Qiong Wu
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China.,Department of Pathophysiology, Qinghai University Medical College, Xining, Qinghai, China
| | - Zhentong Shen
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Xinjian Li
- College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou, China
| | - Chao Sun
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| |
Collapse
|
9
|
Wang NN, Xu HH, Zhou W, Yang HX, Wang J, Ma ZC, Gao Y. Aconitine attenuates mitochondrial dysfunction of cardiomyocytes via promoting deacetylation of cyclophilin-D mediated by sirtuin-3. JOURNAL OF ETHNOPHARMACOLOGY 2021; 270:113765. [PMID: 33418031 DOI: 10.1016/j.jep.2020.113765] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/17/2020] [Revised: 12/09/2020] [Accepted: 12/24/2020] [Indexed: 06/12/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Aconite is a processed product of seminal root of perennial herbaceous plant Aconitum Carmichaclii Debx. of Ranunculaceae. It has the effects of warming and tonifying heart yang and restoring yang to save from collapse. Aconitine is the main effective constituent of aconite and used to prevent and treat heart disease. However, how aconitine exerts myocardial protection is still poorly understood. AIM OF THE STUDY The present study aimed to investigate the effects of aconitine on mitochondrial dysfunction and explore its mechanism of action. MATERIALS AND METHODS The model of myocardial injury was induced by Angiotensin II (Ang II) (1 × 10-6 mol L-1), and H9c2 cells were incubated with different concentrations of aconitine. The effect of aconitine on mitochondrial was determined by flow cytometry, transmission electron microscopy, luciferase, Seahorse technique and Western blot. The effects of aconitine on sirtuin-3 (Sirt3) activity and Cyclophilin D (CypD) acetylation were detected by immunofluorescence, RT-PCR and co-immunoprecipitation. RESULTS We demonstrate that aconitine alleviates the energy metabolic dysfunction of H9c2 cells by activating Sirt3 to deacetylate CypD and inhibiting mitochondrial permeability transition pore (mPTP) opening. In cardiomyocytes, aconitine significantly reduced mitochondrial fragmentation, inhibited acetylation of CypD, suppressed the mPTP opening, mitigated mitochondrial OXPHOS disorders, and improved the synthesis ability of ATP. In contrast, Sirt3 deficiency abolished the effects of aconitine on mPTP and OXPHOS, indicating that aconitine improves mitochondrial function by activating Sirt3. CONCLUSIONS These results showed that aconitine attenuated the energy metabolism disorder by promoting Sirt3 expression and reducing CypD-mediated mPTP excess openness, rescuing mitochondrial function. Improve mitochondrial function may be a therapeutic approach for treating heart disease, which will generate fresh insight into the cardioprotective of aconitine.
Collapse
Affiliation(s)
- Ning-Ning Wang
- Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China; Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Huan-Hua Xu
- Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China; Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Wei Zhou
- Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Hong-Xing Yang
- Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Jia Wang
- Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Zeng-Chun Ma
- Beijing Institute of Radiation Medicine, Beijing, 100850, China.
| | - Yue Gao
- Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China; Beijing Institute of Radiation Medicine, Beijing, 100850, China.
| |
Collapse
|
10
|
Obrador E, Salvador R, Marchio P, López-Blanch R, Jihad-Jebbar A, Rivera P, Vallés SL, Banacloche S, Alcácer J, Colomer N, Coronado JA, Alandes S, Drehmer E, Benlloch M, Estrela JM. Nicotinamide Riboside and Pterostilbene Cooperatively Delay Motor Neuron Failure in ALS SOD1 G93A Mice. Mol Neurobiol 2021; 58:1345-1371. [PMID: 33174130 DOI: 10.1007/s12035-020-02188-7] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Accepted: 10/27/2020] [Indexed: 12/12/2022]
Abstract
Oxidative stress-induced damage is a major mechanism in the pathophysiology of amyotrophic lateral sclerosis (ALS). A recent human clinical trial showed that the combination of nicotinamide riboside (NR) and pterostilbene (PT), molecules with potential to interfere in that mechanism, was efficacious in ALS patients. We examined the effect of these molecules in SOD1G93A transgenic mice, a well-stablished model of ALS. Assessment of neuromotor activity and coordination was correlated with histopathology, and measurement of proinflammatory cytokines in the cerebrospinal fluid. Cell death, Nrf2- and redox-dependent enzymes and metabolites, and sirtuin activities were studied in isolated motor neurons. NR and PT increased survival and ameliorated ALS-associated loss of neuromotor functions in SOD1G93A transgenic mice. NR and PT also decreased the microgliosis and astrogliosis associated with ALS progression. Increased levels of proinflammatory cytokines were observed in the cerebrospinal fluid of mice and humans with ALS. NR and PT ameliorated TNFα-induced oxidative stress and motor neuron death in vitro. Our results support the involvement of oxidative stress, specific Nrf2-dependent antioxidant defenses, and sirtuins in the pathophysiology of ALS. NR and PT interfere with the mechanisms leading to the release of proapoptotic molecular signals by mitochondria, and also promote mitophagy.
Collapse
Affiliation(s)
- Elena Obrador
- Department of Physiology, Faculty of Medicine and Odontology, University of Valencia, 15 Av. Blasco Ibañez, 46010, Valencia, Spain
| | - Rosario Salvador
- Department of Physiology, Faculty of Medicine and Odontology, University of Valencia, 15 Av. Blasco Ibañez, 46010, Valencia, Spain
| | - Patricia Marchio
- Department of Physiology, Faculty of Medicine and Odontology, University of Valencia, 15 Av. Blasco Ibañez, 46010, Valencia, Spain
| | - Rafael López-Blanch
- Department of Physiology, Faculty of Medicine and Odontology, University of Valencia, 15 Av. Blasco Ibañez, 46010, Valencia, Spain
| | - Ali Jihad-Jebbar
- Department of Physiology, Faculty of Medicine and Odontology, University of Valencia, 15 Av. Blasco Ibañez, 46010, Valencia, Spain
| | - Pilar Rivera
- Department of Physiology, Faculty of Medicine and Odontology, University of Valencia, 15 Av. Blasco Ibañez, 46010, Valencia, Spain
| | - Soraya L Vallés
- Department of Physiology, Faculty of Medicine and Odontology, University of Valencia, 15 Av. Blasco Ibañez, 46010, Valencia, Spain
| | - Salvador Banacloche
- Department of Physiology, Faculty of Medicine and Odontology, University of Valencia, 15 Av. Blasco Ibañez, 46010, Valencia, Spain
| | - Javier Alcácer
- Pathology Laboratory, Quirón Hospital, 46010, Valencia, Spain
| | - Nuria Colomer
- Pathology Laboratory, Quirón Hospital, 46010, Valencia, Spain
| | | | - Sandra Alandes
- Pathology Laboratory, Quirón Hospital, 46010, Valencia, Spain
| | - Eraci Drehmer
- Department of Health and Functional Valorization, Catholic University of San Vicente Martir, 46001, Valencia, Spain
| | - María Benlloch
- Department of Health and Functional Valorization, Catholic University of San Vicente Martir, 46001, Valencia, Spain
| | - José M Estrela
- Department of Physiology, Faculty of Medicine and Odontology, University of Valencia, 15 Av. Blasco Ibañez, 46010, Valencia, Spain.
| |
Collapse
|
11
|
Guo Y, Wei L, Zhou Y, Lu N, Tang X, Li Z, Wang X. Flavonoid GL-V9 induces apoptosis and inhibits glycolysis of breast cancer via disrupting GSK-3β-modulated mitochondrial binding of HKII. Free Radic Biol Med 2020; 146:119-129. [PMID: 31669347 DOI: 10.1016/j.freeradbiomed.2019.10.413] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 10/23/2019] [Accepted: 10/23/2019] [Indexed: 12/25/2022]
Abstract
Energy metabolism plays important roles in the growth and survival of cancer cells. Here, we find a newly synthesized flavonoid named GL-V9, which inhibits glycolysis and induces apoptosis of human breast cancer cell lines, and investigate the underlying mechanism. Results show that hexokinase II (HKII) plays important roles in the anticancer effects of GL-V9. GL-V9 not only downregulates the expression of HKII in MDA-MB-231 and MCF-7 cells, but also induces dissociation of HKII from voltage-dependent anion channel (VDAC) in mitochondria, resulting in glycolytic inhibition and mitochondrial-mediated apoptosis. The dissociation of mitochondrial HKII is attributed to GSK-3β-induced phosphorylation of mitochondrial VDAC. Our in vivo experiments also show that GL-V9 significantly inhibits the growth of human breast cancer due to activation of GSK-3β and inactivation of AKT. Thus, GL-V9 induces cytotoxicity in breast cancer cells via disrupting the mitochondrial binding of HKII. Our works demonstrate the significance of metabolic regulators in cancer growth and offer a fresh insight into the molecular basis for the development of GL-V9 as a candidate for breast carcinoma treatment.
Collapse
Affiliation(s)
- Yongjian Guo
- Department of Chemistry and Biochemistry, Florida International University, Miami, FL, USA
| | - Libin Wei
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, 210009, People's Republic of China
| | - Yuxin Zhou
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, 210009, People's Republic of China
| | - Na Lu
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, 210009, People's Republic of China
| | - Xiaoqing Tang
- Department of Chemistry and Biochemistry, Florida International University, Miami, FL, USA
| | - Zhiyu Li
- State Key Laboratory of Natural Medicines, Department of Natural Medicinal Chemistry, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, 210009, People's Republic of China
| | - Xiaotang Wang
- Department of Chemistry and Biochemistry, Florida International University, Miami, FL, USA.
| |
Collapse
|
12
|
Schiattarella GG, Hill JA. Metabolic control and oxidative stress in pathological cardiac remodelling. Eur Heart J 2019; 38:1399-1401. [PMID: 27247363 DOI: 10.1093/eurheartj/ehw199] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Affiliation(s)
- Gabriele G Schiattarella
- Department of Internal Medicine (Cardiology), University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Joseph A Hill
- Department of Internal Medicine (Cardiology), University of Texas Southwestern Medical Center, Dallas, TX, USA.,Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
13
|
Si Y, Bao H, Han L, Chen L, Zeng L, Jing L, Xing Y, Geng Y. Dexmedetomidine attenuation of renal ischaemia-reperfusion injury requires sirtuin 3 activation. Br J Anaesth 2018; 121:1260-1271. [PMID: 30442253 DOI: 10.1016/j.bja.2018.07.007] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2017] [Revised: 06/20/2018] [Accepted: 07/12/2018] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Dexmedetomidine attenuates renal ischaemia and reperfusion (I/R) injury, but its mechanism of action is unclear. As sirtuin 3 (SIRT3) activation can alleviate acute kidney injury, we investigated whether dexmedetomidine acts through SIRT3 to reduce renal I/R injury. METHODS The potential involvement of SIRT3 in dexmedetomidine attenuation of renal I/R injury was tested in HK2 cells subjected to hypoxia/reoxygenation and C57BL/6J mice subjected to renal I/R. A short interfering RNA targeting SIRT3 was used in some experiments to examine the potential role of SIRT3. Cell death and mitochondrial membrane potential (Δψm) were analysed in cultured cells. Mitochondrial damage in mice was assessed using electron microscopy and markers for renal function. Expression of cyclophilin D, cytochrome c, and SIRT3, and the level of cyclophilin D acetylation were determined. RESULTS Hypoxia/reoxygenation of HK2 cells increased cell death, cytochrome C expression, and cyclophilin D acetylation, and decreased Δψm and SIRT3 expression (P<0.05). Dexmedetomidine attenuated these changes. The dexmedetomidine effects were enhanced by SIRT3 overexpression and eliminated by SIRT3 knockdown. I/R in mice damaged renal function, and increased histological lesions, mitochondrial damage, cytochrome c expression, and cyclophilin D acetylation, while SIRT3 activity was decreased by 51% (P<0.05). Dexmedetomidine inhibited these changes in mice expressing normal levels of SIRT3, but not in SIRT3-knockdown mice. CONCLUSIONS Dexmedetomidine appears to act, at least in part, by up-regulating SIRT3 to inhibit mitochondrial damage and cell apoptosis and thereby protect against renal I/R injury.
Collapse
Affiliation(s)
- Y Si
- Department of Anaesthesiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, People's Republic of China
| | - H Bao
- Department of Anaesthesiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, People's Republic of China.
| | - L Han
- Department of Anaesthesiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, People's Republic of China
| | - L Chen
- Department of Anaesthesiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, People's Republic of China
| | - L Zeng
- Department of Anaesthesiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, People's Republic of China
| | - L Jing
- Department of Anaesthesiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, People's Republic of China
| | - Y Xing
- Mechanical Engineering, Southeast University, Nanjing, People's Republic of China
| | - Y Geng
- Department of Anaesthesiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, People's Republic of China
| |
Collapse
|
14
|
Teodoro JS, Varela AT, Duarte FV, Gomes AP, Palmeira CM, Rolo AP. Indirubin and NAD + prevent mitochondrial ischaemia/reperfusion damage in fatty livers. Eur J Clin Invest 2018; 48:e12932. [PMID: 29603199 DOI: 10.1111/eci.12932] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Accepted: 03/22/2018] [Indexed: 12/11/2022]
Abstract
BACKGROUND Fatty livers are considerably more susceptible to acute stressors, such as ischaemia/reperfusion (I/R). As the incidence of I/R is high due to surgical events and some pathologies, there is an urgent need to find strategies against I/R injury (I/RI) in fatty livers. We postulate that an acute pretreatment with indirubin-3'-oxime (Ind) or NAD+ prevents mitochondrial dysfunction associated with warm I/RI in fatty livers. MATERIALS AND METHODS Zucker fatty rats were subjected to warm ischaemia and 12 hours of reperfusion. Ind or NAD+ was administered in the hepatic artery 30 minutes before ischaemia. Hepatic mitochondrial isolation was performed, and functional assays as well as molecular analysis were performed. RESULTS Pretreatment decreased markers of liver injury while preserving mitochondrial cytochrome c content, which is related to the prevention of calcium-induced mitochondrial permeability transition (mPT), the decline in mitochondrial respiratory state 3 and ATP content. The generation of reactive oxygen species (ROS) was also diminished. Inhibition of GSK-3ß by Ind resulted in the prevention of cyclophilin-D (CypD) phosphorylation, unabling it to bind to the adenine nucleotide translocator (ANT), thus, preventing mPT induction. Furthermore, deacetylation of CypD at Lys residue by sirtuin 3 (SIRT3) caused its dissociation from ANT, contributing to an increase in mPT threshold in NAD+ -pretreated animals. CONCLUSIONS Pretreatment with Ind or NAD+ protects fatty livers by maintaining mitochondrial calcium homoeostasis, thus, preserving mitochondrial function and energetic balance. As such, CypD might be a new protective target against I/RI in fatty livers.
Collapse
Affiliation(s)
- João Soeiro Teodoro
- CNC-Center for Neurosciences and Cell Biology, University of Coimbra, Coimbra, Portugal
- Faculty of Sciences and Technology, Department of Life Sciences, University of Coimbra, Coimbra, Portugal
| | - Ana Teresa Varela
- CNC-Center for Neurosciences and Cell Biology, University of Coimbra, Coimbra, Portugal
- Faculty of Sciences and Technology, Department of Life Sciences, University of Coimbra, Coimbra, Portugal
| | - Filipe Valente Duarte
- CNC-Center for Neurosciences and Cell Biology, University of Coimbra, Coimbra, Portugal
- Faculty of Sciences and Technology, Department of Life Sciences, University of Coimbra, Coimbra, Portugal
| | - Ana Patrícia Gomes
- CNC-Center for Neurosciences and Cell Biology, University of Coimbra, Coimbra, Portugal
- Faculty of Sciences and Technology, Department of Life Sciences, University of Coimbra, Coimbra, Portugal
| | - Carlos Marques Palmeira
- CNC-Center for Neurosciences and Cell Biology, University of Coimbra, Coimbra, Portugal
- Faculty of Sciences and Technology, Department of Life Sciences, University of Coimbra, Coimbra, Portugal
| | - Anabela Pinto Rolo
- CNC-Center for Neurosciences and Cell Biology, University of Coimbra, Coimbra, Portugal
- Faculty of Sciences and Technology, Department of Life Sciences, University of Coimbra, Coimbra, Portugal
| |
Collapse
|
15
|
Ali I, Conrad RJ, Verdin E, Ott M. Lysine Acetylation Goes Global: From Epigenetics to Metabolism and Therapeutics. Chem Rev 2018; 118:1216-1252. [PMID: 29405707 PMCID: PMC6609103 DOI: 10.1021/acs.chemrev.7b00181] [Citation(s) in RCA: 262] [Impact Index Per Article: 37.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Post-translational acetylation of lysine residues has emerged as a key regulatory mechanism in all eukaryotic organisms. Originally discovered in 1963 as a unique modification of histones, acetylation marks are now found on thousands of nonhistone proteins located in virtually every cellular compartment. Here we summarize key findings in the field of protein acetylation over the past 20 years with a focus on recent discoveries in nuclear, cytoplasmic, and mitochondrial compartments. Collectively, these findings have elevated protein acetylation as a major post-translational modification, underscoring its physiological relevance in gene regulation, cell signaling, metabolism, and disease.
Collapse
Affiliation(s)
- Ibraheem Ali
- Gladstone Institute of Virology and Immunology, San Francisco, California 94158, United States
- University of California, San Francisco, Department of Medicine, San Francisco, California 94158, United States
| | - Ryan J. Conrad
- Gladstone Institute of Virology and Immunology, San Francisco, California 94158, United States
- University of California, San Francisco, Department of Medicine, San Francisco, California 94158, United States
| | - Eric Verdin
- Buck Institute for Research on Aging, Novato, California 94945, United States
| | - Melanie Ott
- Gladstone Institute of Virology and Immunology, San Francisco, California 94158, United States
- University of California, San Francisco, Department of Medicine, San Francisco, California 94158, United States
| |
Collapse
|
16
|
Zhang X, Ji R, Liao X, Castillero E, Kennel PJ, Brunjes DL, Franz M, Möbius-Winkler S, Drosatos K, George I, Chen EI, Colombo PC, Schulze PC. MicroRNA-195 Regulates Metabolism in Failing Myocardium Via Alterations in Sirtuin 3 Expression and Mitochondrial Protein Acetylation. Circulation 2018; 137:2052-2067. [PMID: 29330215 DOI: 10.1161/circulationaha.117.030486] [Citation(s) in RCA: 142] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Accepted: 12/11/2017] [Indexed: 12/15/2022]
Abstract
BACKGROUND Heart failure leads to mitochondrial dysfunction and metabolic abnormalities of the failing myocardium coupled with an energy-depleted state and cardiac remodeling. The mitochondrial deacetylase sirtuin 3 (SIRT3) plays a pivotal role in the maintenance of mitochondrial function through regulating the mitochondrial acetylome. It is interesting to note that unique cardiac and systemic microRNAs have been shown to play an important role in cardiac remodeling by modulating key signaling elements in the myocardium. METHODS Cellular signaling was analyzed in human cardiomyocyte-like AC16 cells, and acetylation levels in rodent models of SIRT3-/-and transgenic microRNA-195 (miR-195) overexpression were compared with wild type. Luciferase assays, Western blotting, immunoprecipitation assays, and echocardiographic analysis were performed. Enzymatic activities of pyruvate dehydrogenase (PDH) and ATP synthase were measured. RESULTS In failing human myocardium, we observed induction of miR-195 along with decreased expression of the mitochondrial deacetylase SIRT3 that was associated with increased global protein acetylation. We further investigated the role of miR-195 in SIRT3-mediated metabolic processes and its impact on regulating enzymes involved in deacetylation. Proteomic analysis of the total acetylome showed increased overall acetylation, and specific lysine acetylation of 2 central mitochondrial metabolic enzymes, PDH and ATP synthase, as well. miR-195 downregulates SIRT3 expression through direct 3'-untranslated region targeting. Treatments with either sirtuin inhibitor nicotinamide, small interfering RNA-mediated SIRT3 knockdown or miR-195 overexpression enhanced acetylation of PDH complex and ATP synthase. This effect diminished PDH and ATP synthase activity and impaired mitochondrial respiration.SIRT3-/- and miR-195 transgenic mice consistently showed enhanced global protein acetylation, including PDH complex and ATP synthase, associated with decreased enzymatic activity. CONCLUSIONS Altogether, these data suggest that increased levels of miR-195 in failing myocardium regulate a novel pathway that involves direct SIRT3 suppression and enzymatic inhibition via increased acetylation of PDH and ATP synthase that are essential for cardiac energy metabolism.
Collapse
Affiliation(s)
- Xiaokan Zhang
- Department of Medicine, Division of Cardiology, Columbia University Medical Center, New York, NY (X.Z., R.J., X.L., P.J.K., D.L.B., P.C.C., P.C.S.)
| | - Ruiping Ji
- Department of Medicine, Division of Cardiology, Columbia University Medical Center, New York, NY (X.Z., R.J., X.L., P.J.K., D.L.B., P.C.C., P.C.S.)
| | - Xianghai Liao
- Department of Medicine, Division of Cardiology, Columbia University Medical Center, New York, NY (X.Z., R.J., X.L., P.J.K., D.L.B., P.C.C., P.C.S.)
| | - Estibaliz Castillero
- Department of Surgery, Columbia University Medical Center, New York, NY (E.C., I.G.)
| | - Peter J Kennel
- Department of Medicine, Division of Cardiology, Columbia University Medical Center, New York, NY (X.Z., R.J., X.L., P.J.K., D.L.B., P.C.C., P.C.S.)
| | - Danielle L Brunjes
- Department of Medicine, Division of Cardiology, Columbia University Medical Center, New York, NY (X.Z., R.J., X.L., P.J.K., D.L.B., P.C.C., P.C.S.)
| | - Marcus Franz
- Department of Medicine I, Division of Cardiology, Angiology, Pneumology and Intensive Medical Care, University Hospital Jena, Friedrich-Schiller-University Jena, Germany (M.F., S.M.-W., P.C.S.)
| | - Sven Möbius-Winkler
- Department of Medicine I, Division of Cardiology, Angiology, Pneumology and Intensive Medical Care, University Hospital Jena, Friedrich-Schiller-University Jena, Germany (M.F., S.M.-W., P.C.S.)
| | - Konstantinos Drosatos
- Metabolic Biology Laboratory, Department of Pharmacology, Center for Translational Medicine, Lewis Katz School of Medicine at Temple University, Philadelphia, PA (K.D.)
| | - Isaac George
- Department of Surgery, Columbia University Medical Center, New York, NY (E.C., I.G.)
| | - Emily I Chen
- Department of Pharmacology, Columbia University Medical Center, New York, NY (E.I.C.).,Proteomics Shared Resource at the Herbert Irving Comprehensive Cancer Center, New York, NY (E.I.C.)
| | - Paolo C Colombo
- Department of Medicine, Division of Cardiology, Columbia University Medical Center, New York, NY (X.Z., R.J., X.L., P.J.K., D.L.B., P.C.C., P.C.S.)
| | - P Christian Schulze
- Department of Medicine, Division of Cardiology, Columbia University Medical Center, New York, NY (X.Z., R.J., X.L., P.J.K., D.L.B., P.C.C., P.C.S.). .,Department of Medicine I, Division of Cardiology, Angiology, Pneumology and Intensive Medical Care, University Hospital Jena, Friedrich-Schiller-University Jena, Germany (M.F., S.M.-W., P.C.S.)
| |
Collapse
|
17
|
Qiao A, Wang K, Yuan Y, Guan Y, Ren X, Li L, Chen X, Li F, Chen AF, Zhou J, Yang JM, Cheng Y. Sirt3-mediated mitophagy protects tumor cells against apoptosis under hypoxia. Oncotarget 2017; 7:43390-43400. [PMID: 27270321 PMCID: PMC5190031 DOI: 10.18632/oncotarget.9717] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2016] [Accepted: 05/05/2016] [Indexed: 01/27/2023] Open
Abstract
Sirt3, a mitochondrial deacetylase, participates in the regulation of multiple cellular processes through its effect on protein acetylation. The objective of this study was to explore the role of Sirt3 in the mitochondrial autophagy (mitophagy), a process of the specific autophagic elimination of damaged mitochondria. We found that silencing of Sirt3 expression in human glioma cells by RNA interference blunted the hypoxia-induced the localization of LC3 on the mitochondria, and the degradation of mitochondria. These results suggest an important involvement of this protein deacetylase in the induction of mitophagy in cancer cells subjected to hypoxia. Further, we demonstrated that Sirt3 activated the hypoxia-induced mitophagy by increasing the interaction of VDAC1 with Parkin. In the cells subjected to hypoxia, inhibition of Sirt3-mediated mitophagy further decreased the mitochondrial membrane potential, and increased the accumulation of ROS that triggers the degradation of anti-apoptotic proteins Mcl-1 and survivin through the proteasomal pathway. Silencing of Sirt3 expression also promoted apoptosis, and enhanced the sensitivity of cancer cells to hypoxia. The regulatory role of Sirt3 in autophagy and apoptosis was also observed in human breast cancer cells. The results of the current study reveal Sirt3 as a novel regulator coupling mitophagy and apoptosis, two important cellular processes that determine cellular survival and death.
Collapse
Affiliation(s)
- Aimin Qiao
- Center for Bioresources and Drug Discovery and School of Bioscience and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Kuansong Wang
- Department of Pathology, Xiangya Hospital, Central South University, Changsha 410008, China.,Department of Pathology, Basic Medical School, Central South University, Changsha 410008, China
| | - Yunsheng Yuan
- Engineering Research Center of Cell and Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Yidi Guan
- Department of Pharmacology, School of Pharmaceutical Sciences, Central South University, Changsha 410008, China
| | - Xingcong Ren
- Department of Pharmacology, The Penn State Hershey Cancer Institute, The Pennsylvania State University College of Medicine and Milton S. Hershey Medical Center, Hershey, PA 17033, USA
| | - Lanya Li
- Department of Pharmacology, School of Pharmaceutical Sciences, Central South University, Changsha 410008, China
| | - Xisha Chen
- Department of Pharmacology, School of Pharmaceutical Sciences, Central South University, Changsha 410008, China
| | - Feng Li
- Department of Pharmacology, School of Pharmaceutical Sciences, Central South University, Changsha 410008, China
| | - Alex F Chen
- Center for Vascular and Translational Medicine, The College of Pharmacy, Central South University, Changsha 410013, China.,The Third Xiangya Hospital, Central South University, Changsha 410013, China
| | - Jianda Zhou
- Department of Plastic Surgery, The Third Xiangya Hospital, Central South University, Changsha 410008, China
| | - Jin-Ming Yang
- Department of Pharmacology, The Penn State Hershey Cancer Institute, The Pennsylvania State University College of Medicine and Milton S. Hershey Medical Center, Hershey, PA 17033, USA
| | - Yan Cheng
- Department of Pharmacology, School of Pharmaceutical Sciences, Central South University, Changsha 410008, China
| |
Collapse
|
18
|
Anamika, Khanna A, Acharjee P, Acharjee A, Trigun SK. Mitochondrial SIRT3 and neurodegenerative brain disorders. J Chem Neuroanat 2017; 95:43-53. [PMID: 29129747 DOI: 10.1016/j.jchemneu.2017.11.009] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Revised: 09/16/2017] [Accepted: 11/08/2017] [Indexed: 12/19/2022]
Abstract
Sirtuins are highly conserved NAD+ dependent class III histone deacetylases and catalyze deacetylation and ADP ribosylation of a number of non-histone proteins. Since, they require NAD+ for their activity, the cellular level of Sirtuins represents redox status of the cells and thereby serves as bona fide metabolic stress sensors. Out of seven homologues of Sirtuins identified in mammals, SIRT3, 4 & 5 have been found to be localized and active in mitochondria. During recent past, clusters of protein substrates for SIRT3 have been identified in mitochondria and thereby advocating SIRT3 as the main mitochondrial Sirtuin which could be involved in protecting stress induced mitochondrial integrity and energy metabolism. As mitochondrial dysfunction underlies the pathogenesis of almost all neurodegenerative diseases, a role of SIRT3 becomes an arguable speculation in such brain disorders. Some recent findings demonstrate that SIRT3 over expression could prevent neuronal derangements in certain in vivo and in vitro models of aging and neurodegenerative brain disorders like; Alzheimer's disease, Huntington's disease, stroke etc. Similarly, loss of SIRT3 has been found to accelerate neurodegeneration in the brain challenged with excitotoxicity. Therefore, it is argued that SIRT3 could be a relevant target to understand pathogenesis of neurodegenerative brain disorders. This review is an attempt to summarize recent findings on (1) the implication of SIRT3 in neurodegenerative brain disorders and (2) whether SIRT3 modulation could ameliorate neuropathologies in relevant models.
Collapse
Affiliation(s)
- Anamika
- Biochemistry Section, Department of Zoology, Institute of Science, Banaras Hindu University Varanasi, 221005, India
| | - Archita Khanna
- Biochemistry Section, Department of Zoology, Institute of Science, Banaras Hindu University Varanasi, 221005, India
| | - Papia Acharjee
- Biochemistry Section, Department of Zoology, Institute of Science, Banaras Hindu University Varanasi, 221005, India
| | - Arup Acharjee
- Biochemistry Section, Department of Zoology, Institute of Science, Banaras Hindu University Varanasi, 221005, India
| | - Surendra Kumar Trigun
- Biochemistry Section, Department of Zoology, Institute of Science, Banaras Hindu University Varanasi, 221005, India.
| |
Collapse
|
19
|
Garvin AM, Aurigemma NC, Hackenberger JL, Korzick DH. Age and ischemia differentially impact mitochondrial ultrastructure and function in a novel model of age-associated estrogen deficiency in the female rat heart. Pflugers Arch 2017; 469:1591-1602. [PMID: 28776263 DOI: 10.1007/s00424-017-2034-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Revised: 07/10/2017] [Accepted: 07/11/2017] [Indexed: 12/19/2022]
Abstract
Altered mitochondrial respiration, morphology, and quality control collectively contribute to mitochondrial dysfunction in the aged heart. Because myocardial infarction remains the leading cause of death in aged women, the present study utilized a novel rodent model to recapitulate human menopause to interrogate the combination of age and estrogen deficiency on mitochondrial ultrastructure and function with cardiac ischemia/reperfusion (I/R) injury. Female F344 rats were ovariectomized (OVX) at 15 months and studied at 24 months (MO OVX; n = 40) vs adult ovary intact (6 months; n = 41). Temporal declines in estrogen concomitant with increased visceral adipose tissue were observed in MO OVX vs adult. Following in vivo coronary artery ligation or sham surgery, state 3 mitochondrial respiration was selectively reduced by age in subsarcolemmal mitochondria (SSM) and by I/R in interfibrillar mitochondria (IFM); left ventricular maximum dP/dt was reduced in MO OVX (p < 0.05). Elevated cyclophilin D and exacerbated I/R-induced mitochondrial acetylation in MO OVX suggest permeability transition pore involvement and reduced protection vs adult (p < 0.05). Mitochondrial morphology by TEM revealed an altered time course of autophagy coordinate with attenuated Drp1 and LC3BII protein levels with age-associated estrogen loss (p < 0.05). Here, reductions in both SSM and IFM function may play an additive role in enhanced susceptibility to regional I/R injury in aged estrogen-deficient female hearts. Moreover, novel insight into altered cardiac mitochondrial quality control garnered here begins to unravel the potentially important regulatory role of mitochondrial dynamics on sustaining respiratory function in the aged female heart.
Collapse
Affiliation(s)
- Alexandra M Garvin
- Intercollege Graduate Degree Program in Physiology, The Pennsylvania State University, 106 Noll Laboratory, University Park, PA, 16802, USA
| | - Nicole C Aurigemma
- Intercollege Graduate Degree Program in Physiology, The Pennsylvania State University, 106 Noll Laboratory, University Park, PA, 16802, USA
| | - Jenna L Hackenberger
- Department of Kinesiology, The Pennsylvania State University, University Park, PA, 16802, USA
| | - Donna H Korzick
- Intercollege Graduate Degree Program in Physiology, The Pennsylvania State University, 106 Noll Laboratory, University Park, PA, 16802, USA. .,Department of Kinesiology, The Pennsylvania State University, University Park, PA, 16802, USA.
| |
Collapse
|
20
|
Javadov S, Jang S, Parodi-Rullán R, Khuchua Z, Kuznetsov AV. Mitochondrial permeability transition in cardiac ischemia-reperfusion: whether cyclophilin D is a viable target for cardioprotection? Cell Mol Life Sci 2017; 74:2795-2813. [PMID: 28378042 PMCID: PMC5977999 DOI: 10.1007/s00018-017-2502-4] [Citation(s) in RCA: 78] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2016] [Revised: 02/28/2017] [Accepted: 03/06/2017] [Indexed: 12/13/2022]
Abstract
Growing number of studies provide strong evidence that the mitochondrial permeability transition pore (PTP), a non-selective channel in the inner mitochondrial membrane, is involved in the pathogenesis of cardiac ischemia-reperfusion and can be targeted to attenuate reperfusion-induced damage to the myocardium. The molecular identity of the PTP remains unknown and cyclophilin D is the only protein commonly accepted as a major regulator of the PTP opening. Therefore, cyclophilin D is an attractive target for pharmacological or genetic therapies to reduce ischemia-reperfusion injury in various animal models and humans. Most animal studies demonstrated cardioprotective effects of PTP inhibition; however, a recent large clinical trial conducted by international groups demonstrated that cyclosporine A, a cyclophilin D inhibitor, failed to protect the heart in patients with myocardial infarction. These studies, among others, raise the question of whether cyclophilin D, which plays an important physiological role in the regulation of cell metabolism and mitochondrial bioenergetics, is a viable target for cardioprotection. This review discusses previous studies to provide comprehensive information on the physiological role of cyclophilin D as well as PTP opening in the cell that can be taken into consideration for the development of new PTP inhibitors.
Collapse
Affiliation(s)
- Sabzali Javadov
- Department of Physiology, School of Medicine, University of Puerto Rico, San Juan, PR 00936-5067, Puerto Rico.
| | - Sehwan Jang
- Department of Physiology, School of Medicine, University of Puerto Rico, San Juan, PR 00936-5067, Puerto Rico
| | - Rebecca Parodi-Rullán
- Department of Physiology, School of Medicine, University of Puerto Rico, San Juan, PR 00936-5067, Puerto Rico
| | - Zaza Khuchua
- Cincinnati Children's Research Foundation, University of Cincinnati, 240 Albert Sabin Way, Cincinnati, OH, 54229, USA
| | - Andrey V Kuznetsov
- Cardiac Surgery Research Laboratory, Department of Cardiac Surgery, Innsbruck Medical University, Innsbruck, Austria
| |
Collapse
|
21
|
Hurst S, Hoek J, Sheu SS. Mitochondrial Ca 2+ and regulation of the permeability transition pore. J Bioenerg Biomembr 2017; 49:27-47. [PMID: 27497945 PMCID: PMC5393273 DOI: 10.1007/s10863-016-9672-x] [Citation(s) in RCA: 160] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Accepted: 07/31/2016] [Indexed: 02/06/2023]
Abstract
The mitochondrial permeability transition pore was originally described in the 1970's as a Ca2+ activated pore and has since been attributed to the pathogenesis of many diseases. Here we evaluate how each of the current models of the pore complex fit to what is known about how Ca2+ regulates the pore, and any insight that provides into the molecular identity of the pore complex. We also discuss the central role of Ca2+ in modulating the pore's open probability by directly regulating processes, such as ATP/ADP balance through the tricarboxylic acid cycle, electron transport chain, and mitochondrial membrane potential. We review how Ca2+ influences second messengers such as reactive oxygen/nitrogen species production and polyphosphate formation. We discuss the evidence for how Ca2+ regulates post-translational modification of cyclophilin D including phosphorylation by glycogen synthase kinase 3 beta, deacetylation by sirtuins, and oxidation/ nitrosylation of key residues. Lastly we introduce a novel view into how Ca2+ activated proteolysis through calpains in the mitochondria may be a driver of sustained pore opening during pathologies such as ischemia reperfusion injury.
Collapse
Affiliation(s)
- Stephen Hurst
- Center for Translational Medicine, Department of Medicine, Sidney Kimmel Medical College, Thomas Jefferson University, 1020 Locust Street, Suite 543D, Philadelphia, PA, 19107, USA
| | - Jan Hoek
- Mitocare Center for Mitochondria Research, Department of Pathology Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA, 19107, USA
| | - Shey-Shing Sheu
- Center for Translational Medicine, Department of Medicine, Sidney Kimmel Medical College, Thomas Jefferson University, 1020 Locust Street, Suite 543D, Philadelphia, PA, 19107, USA.
| |
Collapse
|
22
|
Schiedel M, Robaa D, Rumpf T, Sippl W, Jung M. The Current State of NAD + -Dependent Histone Deacetylases (Sirtuins) as Novel Therapeutic Targets. Med Res Rev 2017; 38:147-200. [PMID: 28094444 DOI: 10.1002/med.21436] [Citation(s) in RCA: 81] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Revised: 10/24/2016] [Accepted: 11/14/2016] [Indexed: 12/19/2022]
Abstract
Sirtuins are NAD+ -dependent protein deacylases that cleave off acetyl, as well as other acyl groups, from the ε-amino group of lysines in histones and other substrate proteins. Seven sirtuin isotypes (Sirt1-7) have been identified in mammalian cells. As sirtuins are involved in the regulation of various physiological processes such as cell survival, cell cycle progression, apoptosis, DNA repair, cell metabolism, and caloric restriction, a dysregulation of their enzymatic activity has been associated with the pathogenesis of neoplastic, metabolic, infectious, and neurodegenerative diseases. Thus, sirtuins are promising targets for pharmaceutical intervention. Growing interest in a modulation of sirtuin activity has prompted the discovery of several small molecules, able to inhibit or activate certain sirtuin isotypes. Herein, we give an update to our previous review on the topic in this journal (Schemies, 2010), focusing on recent developments in sirtuin biology, sirtuin modulators, and their potential as novel therapeutic agents.
Collapse
Affiliation(s)
- Matthias Schiedel
- Institute of Pharmaceutical Sciences, Albert-Ludwigs-Universität Freiburg, Freiburg, Germany
| | - Dina Robaa
- Department of Pharmaceutical Chemistry, Martin-Luther Universität Halle-Wittenberg, Halle/Saale, Germany
| | - Tobias Rumpf
- Institute of Pharmaceutical Sciences, Albert-Ludwigs-Universität Freiburg, Freiburg, Germany
| | - Wolfgang Sippl
- Department of Pharmaceutical Chemistry, Martin-Luther Universität Halle-Wittenberg, Halle/Saale, Germany
| | - Manfred Jung
- Institute of Pharmaceutical Sciences, Albert-Ludwigs-Universität Freiburg, Freiburg, Germany
| |
Collapse
|
23
|
Correia M, Perestrelo T, Rodrigues AS, Ribeiro MF, Pereira SL, Sousa MI, Ramalho-Santos J. Sirtuins in metabolism, stemness and differentiation. Biochim Biophys Acta Gen Subj 2017; 1861:3444-3455. [DOI: 10.1016/j.bbagen.2016.09.008] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2016] [Revised: 08/16/2016] [Accepted: 09/06/2016] [Indexed: 12/20/2022]
|
24
|
Abstract
Mitochondria are the "power house" of a cell continuously generating ATP to ensure its proper functioning. The constant production of ATP via oxidative phosphorylation demands a large electrochemical force that drives protons across the highly selective and low-permeable mitochondrial inner membrane. Besides the conventional role of generating ATP, mitochondria also play an active role in calcium signaling, generation of reactive oxygen species (ROS), stress responses, and regulation of cell-death pathways. Deficiencies in these functions result in several pathological disorders like aging, cancer, diabetes, neurodegenerative and cardiovascular diseases. A plethora of ion channels and transporters are present in the mitochondrial inner and outer membranes which work in concert to preserve the ionic equilibrium of a cell for the maintenance of cell integrity, in physiological as well as pathophysiological conditions. For, e.g., mitochondrial cation channels KATP and BKCa play a significant role in cardioprotection from ischemia-reperfusion injury. In addition to the cation channels, mitochondrial anion channels are equally essential, as they aid in maintaining electro-neutrality by regulating the cell volume and pH. This chapter focusses on the information on molecular identity, structure, function, and physiological relevance of mitochondrial chloride channels such as voltage dependent anion channels (VDACs), uncharacterized mitochondrial inner membrane anion channels (IMACs), chloride intracellular channels (CLIC) and the aspects of forthcoming chloride channels.
Collapse
Affiliation(s)
- Devasena Ponnalagu
- Department of Pharmacology and Physiology, Drexel University College of Medicine, 245 N. 15th Street, Room 8154, Mail Stop 488, Philadelphia, PA, 19102-1192, USA
| | - Harpreet Singh
- Department of Pharmacology and Physiology, Drexel University College of Medicine, 245 N. 15th Street, Room 8154, Mail Stop 488, Philadelphia, PA, 19102-1192, USA.
| |
Collapse
|
25
|
Osborne B, Bentley NL, Montgomery MK, Turner N. The role of mitochondrial sirtuins in health and disease. Free Radic Biol Med 2016; 100:164-174. [PMID: 27164052 DOI: 10.1016/j.freeradbiomed.2016.04.197] [Citation(s) in RCA: 140] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2016] [Revised: 04/21/2016] [Accepted: 04/29/2016] [Indexed: 01/15/2023]
Abstract
Mitochondria play a critical role in energy production, cell signalling and cell survival. Defects in mitochondrial function contribute to the ageing process and ageing-related disorders such as metabolic disease, cancer, and neurodegeneration. The sirtuin family of deacylase enzymes have a variety of subcellular localisations and have been found to remove a growing list of post-translational acyl modifications from target proteins. SIRT3, SIRT4, and SIRT5 are found primarily located in the mitochondria, and are involved in many of the key processes of this organelle. SIRT3 has been the subject of intense research and is primarily a deacetylase thought to function as a mitochondrial fidelity protein, with roles in mitochondrial substrate metabolism, protection against oxidative stress, and cell survival pathways. Less is known about the functional targets of SIRT4, which has deacetylase, ADP-ribosylase, and a newly-described lipoamidase function, although key roles in lipid and glutamine metabolism have been reported. SIRT5 modulates a host of newly-discovered acyl modifications including succinylation, malonylation, and glutarylation in both mitochondrial and extra-mitochondrial compartments, however the functional significance of SIRT5 in the regulation of many of its proposed target proteins remains to be discovered. Because of their influence on a broad range of pathways, SIRT3, SIRT4, and SIRT5 are implicated in a range of disease-states including metabolic disease such as diabetes, neurodegenerative diseases, cancer, and ageing-related disorders such as hearing-loss and cardiac dysfunction. We review the current knowledge on the function of the three mitochondrial sirtuins, their role in disease, and the current outstanding questions in the field.
Collapse
Affiliation(s)
- Brenna Osborne
- Department of Pharmacology, School of Medical Sciences, UNSW Australia, Sydney, NSW, Australia
| | - Nicholas L Bentley
- Department of Pharmacology, School of Medical Sciences, UNSW Australia, Sydney, NSW, Australia
| | - Magdalene K Montgomery
- Department of Pharmacology, School of Medical Sciences, UNSW Australia, Sydney, NSW, Australia
| | - Nigel Turner
- Department of Pharmacology, School of Medical Sciences, UNSW Australia, Sydney, NSW, Australia.
| |
Collapse
|
26
|
Stram AR, Payne RM. Post-translational modifications in mitochondria: protein signaling in the powerhouse. Cell Mol Life Sci 2016; 73:4063-73. [PMID: 27233499 PMCID: PMC5045789 DOI: 10.1007/s00018-016-2280-4] [Citation(s) in RCA: 120] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2016] [Revised: 05/16/2016] [Accepted: 05/19/2016] [Indexed: 02/03/2023]
Abstract
There is an intimate interplay between cellular metabolism and the pathophysiology of disease. Mitochondria are essential to maintaining and regulating metabolic function of cells and organs. Mitochondrial dysfunction is implicated in diverse diseases, such as cardiovascular disease, diabetes and metabolic syndrome, neurodegeneration, cancer, and aging. Multiple reversible post-translational protein modifications are located in the mitochondria that are responsive to nutrient availability and redox conditions, and which can act in protein-protein interactions to modify diverse mitochondrial functions. Included in this are physiologic redox signaling via reactive oxygen and nitrogen species, phosphorylation, O-GlcNAcylation, acetylation, and succinylation, among others. With the advent of mass proteomic screening techniques, there has been a vast increase in the array of known mitochondrial post-translational modifications and their protein targets. The functional significance of these processes in disease etiology, and the pathologic response to their disruption, are still under investigation. However, many of these reversible modifications act as regulatory mechanisms in mitochondria and show promise for mitochondrial-targeted therapeutic strategies. This review addresses the current knowledge of post-translational processing and signaling mechanisms in mitochondria, and their implications in health and disease.
Collapse
Affiliation(s)
- Amanda R Stram
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, IN, USA
- Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, 1044 W. Walnut St., Room R4-302b, Indianapolis, IN, 46202, USA
| | - R Mark Payne
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA.
- Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, IN, USA.
- Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, 1044 W. Walnut St., Room R4-302b, Indianapolis, IN, 46202, USA.
| |
Collapse
|
27
|
Activation of sirtuin 1/3 improves vascular hyporeactivity in severe hemorrhagic shock by alleviation of mitochondrial damage. Oncotarget 2016; 6:36998-7011. [PMID: 26473372 PMCID: PMC4741911 DOI: 10.18632/oncotarget.6076] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2015] [Accepted: 09/24/2015] [Indexed: 12/11/2022] Open
Abstract
Vascular hyporeactivity is one of the major causes responsible for refractory hypotension and associated mortality in severe hemorrhagic shock. Mitochondrial permeability transition (mPT) pore opening in arteriolar smooth muscle cells (ASMCs) is involved in the pathogenesis of vascular hyporeactivity. However, the molecular mechanism underlying mitochondrial injury in ASMCs during hemorrhagic shock is not well understood. Here we produced an in vivo model of severe hemorrhagic shock in adult Wistar rats. We found that sirtuin (SIRT)1/3 protein levels and deacetylase activities were decreased in ASMCs following severe shock. Immunofluorescence staining confirmed reduced levels of SIRT1 in the nucleus and SIRT3 in the mitochondria, respectively. Acetylation of cyclophilin D (CyPD), a component of mPT pore, was increased. SIRT1 activators suppressed mPT pore opening and ameliorated mitochondrial injury in ASMCs after severe shock. Furthermore, administration of SIRT1 activators improved vasoreactivity in rats under severe shock. Our data suggest that epigenetic mechanisms, namely histone post-translational modifications, are involved in regulation of mPT by SIRT1/SIRT3- mediated deacetylation of CyPD. SIRT1/3 is a promising therapeutic target for the treatment of severe hemorrhagic shock.
Collapse
|
28
|
Gertz M, Steegborn C. Using mitochondrial sirtuins as drug targets: disease implications and available compounds. Cell Mol Life Sci 2016; 73:2871-96. [PMID: 27007507 PMCID: PMC11108305 DOI: 10.1007/s00018-016-2180-7] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2015] [Revised: 02/15/2016] [Accepted: 03/11/2016] [Indexed: 02/06/2023]
Abstract
Sirtuins are an evolutionary conserved family of NAD(+)-dependent protein lysine deacylases. Mammals have seven Sirtuin isoforms, Sirt1-7. They contribute to regulation of metabolism, stress responses, and aging processes, and are considered therapeutic targets for metabolic and aging-related diseases. While initial studies were focused on Sirt1 and 2, recent progress on the mitochondrial Sirtuins Sirt3, 4, and 5 has stimulated research and drug development for these isoforms. Here we review the roles of Sirtuins in regulating mitochondrial functions, with a focus on the mitochondrially located isoforms, and on their contributions to disease pathologies. We further summarize the compounds available for modulating the activity of these Sirtuins, again with a focus on mitochondrial isoforms, and we describe recent results important for the further improvement of compounds. This overview illustrates the potential of mitochondrial Sirtuins as drug targets and summarizes the status, progress, and challenges in developing small molecule compounds modulating their activity.
Collapse
Affiliation(s)
- Melanie Gertz
- Department of Biochemistry, University of Bayreuth, Universitätsstr. 30, 95447, Bayreuth, Germany
- Bayer Pharma AG, Apratherweg 18a, 42096, Wuppertal, Germany
| | - Clemens Steegborn
- Department of Biochemistry, University of Bayreuth, Universitätsstr. 30, 95447, Bayreuth, Germany.
| |
Collapse
|
29
|
Xiang XY, Kang JS, Yang XC, Su J, Wu Y, Yan XY, Xue YN, Xu Y, Liu YH, Yu CY, Zhang ZC, Sun LK. SIRT3 participates in glucose metabolism interruption and apoptosis induced by BH3 mimetic S1 in ovarian cancer cells. Int J Oncol 2016; 49:773-84. [PMID: 27277143 DOI: 10.3892/ijo.2016.3552] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Accepted: 04/19/2016] [Indexed: 01/20/2023] Open
Abstract
The Bcl-2 antiapoptotic proteins are important cancer therapy targets; however, their role in cancer cell metabolism remains unclear. We found that the BH3-only protein mimetic S1, a novel pan Bcl-2 inhibitor, simultaneously interrupted glucose metabolism and induced apoptosis in human SKOV3 ovarian cancer cells, which was related to the activation of SIRT3, a stress-responsive deacetylase. S1 interrupted the cellular glucose metabolism mainly through causing damage to mitochondrial respiration and inhibiting glycolysis. Moreover, S1 upregulated the gene and protein expression of SIRT3, and induced the translocation of SIRT3 from the nucleus to mitochondria. SIRT3 silencing reversed the effects of S1 on glucose metabolism and apoptosis through increasing the level of HK-II localized to the mitochondria, while a combination of the glycolysis inhibitor 2-DG and S1 intensified the cytotoxicity through further upregulation of SIRT3 expression. This study underscores an essential role of SIRT3 in the antitumor effect of Bcl-2 inhibitors in human ovarian cancer through regulating both metabolism and apoptosis. The manipulation of Bcl-2 inhibitors combined with the use of classic glycolysis inhibitors may be rational strategies to improve ovarian cancer therapy.
Collapse
Affiliation(s)
- Xi-Yan Xiang
- Department of Pathophysiology, Basic College of Medicine, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Jin-Song Kang
- Department of Pathophysiology, Basic College of Medicine, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Xiao-Chun Yang
- Centre of Nephrology and Urology, Shenzhen University Health Science Centre, Shenzhen, Guangdong 518060, P.R. China
| | - Jing Su
- Department of Pathophysiology, Basic College of Medicine, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Yao Wu
- Department of Pathophysiology, Basic College of Medicine, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Xiao-Yu Yan
- Department of Pathophysiology, Basic College of Medicine, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Ya-Nan Xue
- Department of Pathophysiology, Basic College of Medicine, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Ye Xu
- Medical Research Laboratory, Jilin Medical College, Jilin City, Jilin 132013, P.R. China
| | - Yu-He Liu
- Department of Pathology, Basic Medical College, BeiHua University, Jilin City, Jilin 132013, P.R. China
| | - Chun-Yan Yu
- Department of Pathology, Basic Medical College, BeiHua University, Jilin City, Jilin 132013, P.R. China
| | - Zhi-Chao Zhang
- State Key Laboratory of Fine Chemicals, School of Chemistry, Dalian University of Technology, Dalian, Liaoning 116012, P.R. China
| | - Lian-Kun Sun
- Department of Pathophysiology, Basic College of Medicine, Jilin University, Changchun, Jilin 130021, P.R. China
| |
Collapse
|
30
|
Activation of SIRT3 attenuates triptolide-induced toxicity through closing mitochondrial permeability transition pore in cardiomyocytes. Toxicol In Vitro 2016; 34:128-137. [DOI: 10.1016/j.tiv.2016.03.020] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Revised: 03/07/2016] [Accepted: 03/28/2016] [Indexed: 01/06/2023]
|
31
|
Potential Benefit of the Charge-Stabilized Nanostructure Saline RNS60 for Myelin Maintenance and Repair. Sci Rep 2016; 6:30020. [PMID: 27451946 PMCID: PMC4958964 DOI: 10.1038/srep30020] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Accepted: 06/24/2016] [Indexed: 12/26/2022] Open
Abstract
Myelin injury in multiple sclerosis (MS) has been attributed both to “outside-in” primary immune mediated and “inside-out” metabolic stress of oligodendrocyte (OL) related mechanisms. Subsequent remyelination is dependent on recruitment and differentiation of oligodendrocyte progenitor cells (OPCs). RNS60 is a physically-modified saline containing charge-stabilized nanobubbles generated by subjecting normal saline to Taylor-Couette-Poiseuille (TCP) flow under elevated oxygen pressure. Administration of RNS60 has been shown to reduce the severity of EAE by dampening the immune response and myelin loss. Additionally, RNS60 has been demonstrated to enhance mitochondrial ATP synthesis in neurons. Here, we used post-natal rat derived OLs and OPCs to assess the impact of RNS60 on the response of OLs to metabolic stress in vitro (glucose-nutrient deprivation, referred to as ‘NG’) and on OPC differentiation capacity. Under the NG condition, our findings indicate that RNS60 decreases caspases 3/7 activation. Respirometric analyses revealed that RNS60 increased spare glycolytic capacity (SGC) under normal culture conditions. However, RNS60 enhanced OL spare respiratory capacity (SRC) when a metabolic stress was present. Furthermore, we show that RNS60 promotes OPC differentiation under physiological conditions. Our findings provide evidence for the potential therapeutic efficacy of RNS60 through the promotion of OL survival and OPC differentiation.
Collapse
|
32
|
Abstract
The nutrient demands of cancer cannot be met by normal cell metabolism. Cancer cells undergo dramatic alteration of metabolic pathways in a process called reprogramming, characterized by increased nutrient uptake and re-purposing of these fuels for biosynthetic, bioenergetic or signaling pathways. Partitioning carbon sources toward growth and away from ATP production necessitates other means of generating energy for biosynthetic reactions. Additionally, cancer cell adaptations frequently lead to increased production of reactive oxygen species and lactic acid, which can be beneficial to cancer growth but also are potentially toxic and must be appropriately cleared. Sirtuins are a family of deacylases and ADP-ribosyltransferases with clear links to regulation of cancer metabolism. Through their unique ability to integrate cellular stress and nutrient status with coordination of metabolic outputs, sirtuins are well poised to play pivotal roles in tumor progression and survival. Here, we review the multi-faceted duties of sirtuins in tackling the metabolic hurdles in cancer. We focus on both beneficial and adverse effects of sirtuins in the regulation of energetic, biosynthetic and toxicity barriers faced by cancer cells.
Collapse
Affiliation(s)
- Natalie J German
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA.
| | - Marcia C Haigis
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
33
|
Martin J, Balmer ML, Rajendran S, Maurhofer O, Dufour JF, St-Pierre MV. Nutritional stress exacerbates hepatic steatosis induced by deletion of the histidine nucleotide-binding (Hint2) mitochondrial protein. Am J Physiol Gastrointest Liver Physiol 2016; 310:G497-509. [PMID: 26767982 DOI: 10.1152/ajpgi.00178.2015] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2015] [Accepted: 01/07/2016] [Indexed: 01/31/2023]
Abstract
The histidine nucleotide-binding protein, Hint2, is a mitochondrial phosphoramidase expressed in liver, brown fat, pancreas, and muscle. The livers of Hint2 knockout (Hint2(-/-)) mice accumulate triglycerides and show a pattern of mitochondrial protein lysine hyperacetylation. The extent and nature of the lysine acetylation changes and the response of Hint2(-/-) mice to nutritional challenges that elicit a modification of protein acetylation have not been investigated. To compare the adaptation of Hint2(-/-) and control (Hint2(+/+)) mice with episodes of fasting and high-fat diet (HFD), we subjected animals to either feeding ad libitum or fasting for 24 h, and to either a HFD or control diet for 8 wk. Triglyceride content was higher in Hint2(-/-) than in Hint2(+/+) livers, whereas plasma triglycerides were fourfold lower. Malonyl-CoA levels were increased twofold in Hint2(-/-) livers. After 24 h fasting, Hint2(-/-) displayed a decrease in body temperature, commensurate with a decrease in mass of brown fat and downregulation of uncoupling protein 1. HFD-treated Hint2(-/-) livers showed more steatosis, and plasma insulin and cholesterol were higher than in Hint(+/+) mice. Several proteins identified as substrates of sirtuin 3 and 5 and active in intermediary and ketone metabolism were hyperacetylated in liver and brown fat mitochondria after both HFD and fasting regimens. Glutamate dehydrogenase activity was downregulated in fed and fasted livers, and this was attributed to an increase in acetylation and ADP-ribosylation. The absence of Hint2 deregulates the posttranslational modification of several mitochondrial proteins, which impedes the adaptation to episodes of nutritional stress.
Collapse
Affiliation(s)
- Juliette Martin
- Department of Clinical Research, University of Bern, Bern, Switzerland; and Department of Hepatology, Gastroenterology; and
| | - Maria L Balmer
- Department of Clinical Research, University of Bern, Bern, Switzerland; and Department of Hepatology, Gastroenterology; and
| | - Saranya Rajendran
- Department of Clinical Research, University of Bern, Bern, Switzerland; and Department of Hepatology, Gastroenterology; and
| | - Olivier Maurhofer
- Department of Clinical Research, University of Bern, Bern, Switzerland; and Department of Hepatology, Gastroenterology; and
| | - Jean-François Dufour
- Department of Clinical Research, University of Bern, Bern, Switzerland; and Department of Hepatology, Gastroenterology; and Visceral Surgery, University Hospital, Inselspital, Bern, Switzerland
| | - Marie V St-Pierre
- Department of Clinical Research, University of Bern, Bern, Switzerland; and Department of Hepatology, Gastroenterology; and
| |
Collapse
|
34
|
Sprague Dawley rats: A model of successful heart aging. EUPA OPEN PROTEOMICS 2016; 12:22-30. [PMID: 29900116 PMCID: PMC5988506 DOI: 10.1016/j.euprot.2016.03.017] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Revised: 03/14/2016] [Accepted: 03/31/2016] [Indexed: 01/07/2023]
Abstract
Sprague Dawley rat hearts of 6, 22 and 30 months were analysed by DIGE and blotting. Results indicate that this animal model experiences the so-called successful aging. Mybpc3, Aldh2 and serpins could be used as early biomarkers. Sirtuin-3 increment suggests the activation of protective non-canonical autophagy.
Aging is a universal phenomenon involving the whole body and is characterized by metabolic and physiological decline, leading to cardiovascular defects and heart failure. To characterize the molecular basis of physiological cardiac aging, the proteomic profiles of Sprague Dawley rat hearts of 6, 22 and 30 months were analysed by DIGE and immunoblotting. Results indicate changes in myosin binding protein C, aldehyde dehydrogenase, serpins and sirtuin-3 which protects from the opening of the mitochondrial permeability transition pore induced by cyclophilin D increment. Conversely, an increase of fusion, a decrease of mitochondrial fission and the activation of the non-canonical autophagy pathway were observed. These results support the hypothesis of successful aging in this rat model.
Collapse
|
35
|
Novoderezhkina EA, Zhivotovsky BD, Gogvadze VG. Induction of unspecific permeabilization of mitochondrial membrane and its role in cell death. Mol Biol 2016. [DOI: 10.1134/s0026893316010167] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
36
|
Wang QL, Guo SJ. Sirtuins Function as the Modulators in Aging-related Diseases in Common or Respectively. Chin Med J (Engl) 2016; 128:1671-8. [PMID: 26063372 PMCID: PMC4733746 DOI: 10.4103/0366-6999.158375] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Affiliation(s)
- Qi-Lin Wang
- School of Life Science, Liaocheng University, Liaocheng, Shandong 252059, China
| | | |
Collapse
|
37
|
Clemens DL, Schneider KJ, Arkfeld CK, Grode JR, Wells MA, Singh S. Alcoholic pancreatitis: New insights into the pathogenesis and treatment. World J Gastrointest Pathophysiol 2016; 7:48-58. [PMID: 26909228 PMCID: PMC4753189 DOI: 10.4291/wjgp.v7.i1.48] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Revised: 09/23/2015] [Accepted: 11/11/2015] [Indexed: 02/06/2023] Open
Abstract
Acute pancreatitis is a necro-inflammatory disease of the exocrine pancreas that is characterized by inappropriate activation of zymogens, infiltration of the pancreas by inflammatory cells, and destruction of the pancreatic exocrine cells. Acute pancreatitis can progress to a severe life-threatening disease. Currently there is no pharmacotherapy to prevent or treat acute pancreatitis. One of the more common factors associated with acute pancreatitis is alcohol abuse. Although commonly associated with pancreatitis alcohol alone is unable to cause pancreatitis. Instead, it appears that alcohol and its metabolic by-products predispose the pancreas to damage from agents that normally do not cause pancreatitis, or to more severe disease from agents that normally cause mild pancreatic damage. Over the last 10 to 20 years, a tremendous amount of work has defined a number of alcohol-mediated biochemical changes in pancreatic cells. Among these changes are: Sustained levels of intracellular calcium, activation of the mitochondrial permeability transition pore, endoplasmic reticulum stress, impairment in autophagy, alteration in the activity of transcriptional activators, and colocalization of lysosomal and pancreatic digestive enzymes. Elucidation of these changes has led to a deeper understanding of the mechanisms by which ethanol predisposes acinar cells to damage. This greater understanding has revealed a number of promising targets for therapeutic intervention. It is hoped that further investigation of these targets will lead to the development of pharmacotherapy that is effective in treating and preventing the progression of acute pancreatitis.
Collapse
|
38
|
Mechanisms and Dynamics of Protein Acetylation in Mitochondria. Trends Biochem Sci 2016; 41:231-244. [PMID: 26822488 DOI: 10.1016/j.tibs.2015.12.006] [Citation(s) in RCA: 248] [Impact Index Per Article: 27.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2015] [Revised: 12/17/2015] [Accepted: 12/22/2015] [Indexed: 12/17/2022]
Abstract
Reversible protein acetylation is a major regulatory mechanism for controlling protein function. Through genetic manipulations, dietary perturbations, and new proteomic technologies, the diverse functions of protein acetylation are coming into focus. Protein acetylation in mitochondria has taken center stage, revealing that 63% of mitochondrially localized proteins contain lysine acetylation sites. We summarize the field and discuss salient topics that cover spurious versus targeted acetylation, the role of SIRT3 deacetylation, nonenzymatic acetylation, and molecular models for regulatory acetylations that display high and low stoichiometry.
Collapse
|
39
|
Sex difference in the sensitivity of cardiac mitochondrial permeability transition pore to calcium load. Mol Cell Biochem 2015; 412:147-54. [PMID: 26715132 DOI: 10.1007/s11010-015-2619-4] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Accepted: 12/08/2015] [Indexed: 12/18/2022]
Abstract
Most of the experimental studies have revealed that female heart is more tolerant to ischemia/reperfusion (I/R) injury as compared with the male myocardium. It is widely accepted that mitochondrial dysfunction, and particularly mitochondrial permeability transition pore (MPTP) opening, plays a major role in determining the extent of cardiac I/R injury. The aim of the present study was, therefore, to analyze (i) whether calcium-induced swelling of cardiac mitochondria is sex-dependent and related to the degree of cardiac tolerance to I/R injury and (ii) whether changes in MPTP components-cyclophilin D (CypD) and ATP synthase-can be involved in this process. We have observed that in mitochondria isolated from rat male and female hearts the MPTP has different sensitivity to the calcium load. Female mitochondria are more resistant both in the extent and in the rate of the mitochondrial swelling at higher calcium concentration (200 µM). At low calcium concentration (50 µM) no differences were observed. Our data further suggest that sex-dependent specificity of the MPTP is not the result of different amounts of ATP synthase and CypD, or their respective ratio in mitochondria isolated from male and female hearts. Our results indicate that male and female rat hearts contain comparable content of MPTP and its regulatory protein CypD; parallel immunodetection revealed also the same contents of adenine nucleotide translocator or voltage-dependent anion channel. Increased resistance of female heart mitochondria thus cannot be explained by changes in putative components of MPTP, and rather reflects regulation of MPTP function.
Collapse
|
40
|
Novgorodov SA, Riley CL, Keffler JA, Yu J, Kindy MS, Macklin WB, Lombard DB, Gudz TI. SIRT3 Deacetylates Ceramide Synthases: IMPLICATIONS FOR MITOCHONDRIAL DYSFUNCTION AND BRAIN INJURY. J Biol Chem 2015; 291:1957-1973. [PMID: 26620563 DOI: 10.1074/jbc.m115.668228] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Indexed: 12/11/2022] Open
Abstract
Experimental evidence supports the role of mitochondrial ceramide accumulation as a cause of mitochondrial dysfunction and brain injury after stroke. Herein, we report that SIRT3 regulates mitochondrial ceramide biosynthesis via deacetylation of ceramide synthase (CerS) 1, 2, and 6. Reciprocal immunoprecipitation experiments revealed that CerS1, CerS2, and CerS6, but not CerS4, are associated with SIRT3 in cerebral mitochondria. Furthermore, CerS1, -2, and -6 are hyperacetylated in the mitochondria of SIRT3-null mice, and SIRT3 directly deacetylates the ceramide synthases in a NAD(+)-dependent manner that increases enzyme activity. Investigation of the SIRT3 role in mitochondrial response to brain ischemia/reperfusion (IR) showed that SIRT3-mediated deacetylation of ceramide synthases increased enzyme activity and ceramide accumulation after IR. Functional studies demonstrated that absence of SIRT3 rescued the IR-induced blockade of the electron transport chain at the level of complex III, attenuated mitochondrial outer membrane permeabilization, and decreased reactive oxygen species generation and protein carbonyls in mitochondria. Importantly, Sirt3 gene ablation reduced the brain injury after IR. These data support the hypothesis that IR triggers SIRT3-dependent deacetylation of ceramide synthases and the elevation of ceramide, which could inhibit complex III, leading to increased reactive oxygen species generation and brain injury. The results of these studies highlight a novel mechanism of SIRT3 involvement in modulating mitochondrial ceramide biosynthesis and suggest an important role of SIRT3 in mitochondrial dysfunction and brain injury after experimental stroke.
Collapse
Affiliation(s)
- Sergei A Novgorodov
- the Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina 29425
| | - Christopher L Riley
- From the Ralph H. Johnson Veterans Affairs Medical Center, Charleston, South Carolina 29401
| | - Jarryd A Keffler
- the Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina 29425
| | - Jin Yu
- the Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina 29425
| | - Mark S Kindy
- From the Ralph H. Johnson Veterans Affairs Medical Center, Charleston, South Carolina 29401
| | - Wendy B Macklin
- the Department of Cell and Developmental Biology, University of Colorado, Aurora, Colorado 80045, and
| | - David B Lombard
- the Department of Pathology and Institute of Gerontology, University of Michigan, Ann Arbor, Michigan 48109
| | - Tatyana I Gudz
- the Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina 29425,; From the Ralph H. Johnson Veterans Affairs Medical Center, Charleston, South Carolina 29401,.
| |
Collapse
|
41
|
Carpentieri A, Cozzoli E, Scimeca M, Bonanno E, Sardanelli AM, Gambacurta A. Differentiation of human neuroblastoma cells toward the osteogenic lineage by mTOR inhibitor. Cell Death Dis 2015; 6:e1974. [PMID: 26561783 PMCID: PMC4670915 DOI: 10.1038/cddis.2015.244] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2015] [Revised: 07/23/2015] [Accepted: 07/28/2015] [Indexed: 12/14/2022]
Abstract
Current hypothesis suggest that tumors can originate from adult cells after a process of 'reprogramming' driven by genetic and epigenetic alterations. These cancer cells, called cancer stem cells (CSCs), are responsible for the tumor growth and metastases. To date, the research effort has been directed to the identification, isolation and manipulation of this cell population. Independently of whether tumors were triggered by a reprogramming of gene expression or seeded by stem cells, their energetic metabolism is altered compared with a normal cell, resulting in a high aerobic glycolytic 'Warburg' phenotype and dysregulation of mitochondrial activity. This metabolic alteration is intricately linked to cancer progression.The aim of this work has been to demonstrate the possibility of differentiating a neoplastic cell toward different germ layer lineages, by evaluating the morphological, metabolic and functional changes occurring in this process. The cellular differentiation reported in this study brings to different conclusions from those present in the current literature. We demonstrate that 'in vitro' neuroblastoma cancer cells (chosen as experimental model) are able to differentiate directly into osteoblastic (by rapamycin, an mTOR inhibitor) and hepatic lineage without an intermediate 'stem' cell step. This process seems owing to a synergy among few master molecules, metabolic changes and scaffold presence acting in a concerted way to control the cell fate.
Collapse
Affiliation(s)
- A Carpentieri
- Biochemistry Laboratory, Department of Experimental Medicine and Surgery, University of Rome 'Tor Vergata', Rome 00133, Italy
| | - E Cozzoli
- Biochemistry Laboratory, Department of Experimental Medicine and Surgery, University of Rome 'Tor Vergata', Rome 00133, Italy
| | - M Scimeca
- Department of Biomedicine and Prevention, University of Rome 'Tor Vergata', Rome 00133, Italy
| | - E Bonanno
- Department of Biomedicine and Prevention, University of Rome 'Tor Vergata', Rome 00133, Italy
| | - A M Sardanelli
- Department of Basic Medical Sciences, Neurosciences and Sense Organs, University of Bari 'Aldo Moro', Bari, Italy.,Center of Integrated Research, Campus Bio-Medico, University of Rome, Rome 00128, Italy
| | - A Gambacurta
- Biochemistry Laboratory, Department of Experimental Medicine and Surgery, University of Rome 'Tor Vergata', Rome 00133, Italy.,NAST Centre for Nanoscience, University of Rome 'Tor Vergata', Rome 00133, Italy
| |
Collapse
|
42
|
Jonas EA, Porter GA, Beutner G, Mnatsakanyan N, Alavian KN. Cell death disguised: The mitochondrial permeability transition pore as the c-subunit of the F(1)F(O) ATP synthase. Pharmacol Res 2015; 99:382-92. [PMID: 25956324 PMCID: PMC4567435 DOI: 10.1016/j.phrs.2015.04.013] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2015] [Revised: 04/09/2015] [Accepted: 04/20/2015] [Indexed: 12/16/2022]
Abstract
Ion transport across the mitochondrial inner and outer membranes is central to mitochondrial function, including regulation of oxidative phosphorylation and cell death. Although essential for ATP production by mitochondria, recent findings have confirmed that the c-subunit of the ATP synthase also houses a large conductance uncoupling channel, the mitochondrial permeability transition pore (mPTP), the persistent opening of which produces osmotic dysregulation of the inner mitochondrial membrane and cell death. This review will discuss recent advances in understanding the molecular components of mPTP, its regulatory mechanisms and how these contribute directly to its physiological as well as pathological roles.
Collapse
Affiliation(s)
- Elizabeth A Jonas
- Department of Internal Medicine, Section of Endocrinology, Yale University, New Haven, CT, USA.
| | - George A Porter
- Department of Pediatrics (Cardiology), University of Rochester Medical Center, Rochester, NY, USA
| | - Gisela Beutner
- Department of Pediatrics (Cardiology), University of Rochester Medical Center, Rochester, NY, USA
| | - Nelli Mnatsakanyan
- Department of Internal Medicine, Section of Endocrinology, Yale University, New Haven, CT, USA
| | - Kambiz N Alavian
- Division of Brain Sciences, Department of Medicine, Imperial College London, UK
| |
Collapse
|
43
|
Poulose N, Raju R. Sirtuin regulation in aging and injury. Biochim Biophys Acta Mol Basis Dis 2015; 1852:2442-55. [PMID: 26303641 DOI: 10.1016/j.bbadis.2015.08.017] [Citation(s) in RCA: 186] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Revised: 08/03/2015] [Accepted: 08/20/2015] [Indexed: 12/17/2022]
Abstract
Sirtuins or Sir2 family of proteins are a class of NAD(+) dependent protein deacetylases which are evolutionarily conserved from bacteria to humans. Some sirtuins also exhibit mono-ADP ribosyl transferase, demalonylation and desuccinylation activities. Originally identified in the yeast, these proteins regulate key cellular processes like cell cycle, apoptosis, metabolic regulation and inflammation. Humans encode seven sirtuin isoforms SIRT1-SIRT7 with varying intracellular distribution. Apart from their classic role as histone deacetylases regulating transcription, a number of cytoplasmic and mitochondrial targets of sirtuins have also been identified. Sirtuins have been implicated in longevity and accumulating evidence indicate their role in a spectrum of diseases like cancer, diabetes, obesity and neurodegenerative diseases. A number of studies have reported profound changes in SIRT1 expression and activity linked to mitochondrial functional alterations following hypoxic-ischemic conditions and following reoxygenation injury. The SIRT1 mediated deacetylation of targets such as PGC-1α, FOXO3, p53 and NF-κb has profound effect on mitochondrial function, apoptosis and inflammation. These biological processes and functions are critical in life-span determination and outcome following injury. Aging is reported to be characterized by declining SIRT1 activity, and its increased expression or activation demonstrated prolonged life-span in lower forms of animals. A pseudohypoxic state due to declining NAD(+) has also been implicated in aging. In this review we provide an overview of studies on the role of sirtuins in aging and injury.
Collapse
Affiliation(s)
- Ninu Poulose
- Georgia Regents University, Augusta, GA 30912, United States
| | - Raghavan Raju
- Georgia Regents University, Augusta, GA 30912, United States.
| |
Collapse
|
44
|
Bernardi P, Di Lisa F, Fogolari F, Lippe G. From ATP to PTP and Back: A Dual Function for the Mitochondrial ATP Synthase. Circ Res 2015; 116:1850-62. [PMID: 25999424 DOI: 10.1161/circresaha.115.306557] [Citation(s) in RCA: 98] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Mitochondria not only play a fundamental role in heart physiology but are also key effectors of dysfunction and death. This dual role assumes a new meaning after recent advances on the nature and regulation of the permeability transition pore, an inner membrane channel whose opening requires matrix Ca(2+) and is modulated by many effectors including reactive oxygen species, matrix cyclophilin D, Pi (inorganic phosphate), and matrix pH. The recent demonstration that the F-ATP synthase can reversibly undergo a Ca(2+)-dependent transition to form a channel that mediates the permeability transition opens new perspectives to the field. These findings demand a reassessment of the modifications of F-ATP synthase that take place in the heart under pathological conditions and of their potential role in determining the transition of F-ATP synthase from and energy-conserving into an energy-dissipating device.
Collapse
Affiliation(s)
- Paolo Bernardi
- From the Department of Biomedical Sciences, University of Padova, Italy (P.B., F.D.L.); and Department of Medical and Biological Sciences (F.F) and Department of Food Science (G.L.), University of Udine, Udine, Italy.
| | - Fabio Di Lisa
- From the Department of Biomedical Sciences, University of Padova, Italy (P.B., F.D.L.); and Department of Medical and Biological Sciences (F.F) and Department of Food Science (G.L.), University of Udine, Udine, Italy
| | - Federico Fogolari
- From the Department of Biomedical Sciences, University of Padova, Italy (P.B., F.D.L.); and Department of Medical and Biological Sciences (F.F) and Department of Food Science (G.L.), University of Udine, Udine, Italy
| | - Giovanna Lippe
- From the Department of Biomedical Sciences, University of Padova, Italy (P.B., F.D.L.); and Department of Medical and Biological Sciences (F.F) and Department of Food Science (G.L.), University of Udine, Udine, Italy
| |
Collapse
|
45
|
Kumar S, Lombard DB. Mitochondrial sirtuins and their relationships with metabolic disease and cancer. Antioxid Redox Signal 2015; 22:1060-77. [PMID: 25545135 PMCID: PMC4389911 DOI: 10.1089/ars.2014.6213] [Citation(s) in RCA: 117] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
SIGNIFICANCE Maintenance of metabolic homeostasis is critical for cellular and organismal health. Proper regulation of mitochondrial functions represents a crucial element of overall metabolic homeostasis. Mitochondrial sirtuins (SIRT3, SIRT4, and SIRT5) play pivotal roles in promoting this homeostasis by regulating numerous aspects of mitochondrial metabolism in response to environmental stressors. RECENT ADVANCES New work has illuminated multiple links between mitochondrial sirtuins and cancer. SIRT5 has been shown to regulate the recently described post-translational modifications succinyl-lysine, malonyl-lysine, and glutaryl-lysine. An understanding of these modifications is still in its infancy. Enumeration of SIRT3 and SIRT5 targets via advanced proteomic techniques promises to dramatically enhance insight into functions of these proteins. CRITICAL ISSUES In this review, we highlight the roles of mitochondrial sirtuins and their targets in cellular and organismal metabolic homeostasis. Furthermore, we discuss emerging roles for mitochondrial sirtuins in suppressing and/or promoting tumorigenesis, depending on the cellular and molecular context. FUTURE DIRECTIONS Currently, hundreds of potential SIRT3 and SIRT5 molecular targets have been identified in proteomic experiments. Future studies will need to validate the major targets of these enzymes, and elucidate how acetylation and/or acylation modulate their functionality. A great deal of interest exists in targeting sirtuins pharmacologically; this endeavor will require development of sirtuin-specific modulators (activators and inhibitors) as potential treatments for cancer and metabolic disease.
Collapse
Affiliation(s)
- Surinder Kumar
- 1 Department of Pathology, University of Michigan , Ann Arbor, Michigan
| | | |
Collapse
|
46
|
Oroxylin A inhibits glycolysis-dependent proliferation of human breast cancer via promoting SIRT3-mediated SOD2 transcription and HIF1α destabilization. Cell Death Dis 2015; 6:e1714. [PMID: 25855962 PMCID: PMC4650553 DOI: 10.1038/cddis.2015.86] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2014] [Revised: 02/22/2015] [Accepted: 02/23/2015] [Indexed: 01/21/2023]
Abstract
Alterations of cellular metabolism play a central role in the development and progression of cancer. Oroxylin A, an active flavonoid of a Chinese traditional medicinal plant, was previously shown to modulate glycolysis in cancer cells. However, the mechanism by which oroxylin A regulates glycolysis is still not well defined. Here, we show that oroxylin A inhibits glycolysis in breast cancer cells via the Sirtuin 3 (SIRT3)-mediated destabilization of hypoxia-inducible factor 1α (HIF1α), which controls glycolytic gene expression. Oroxylin A promotes superoxide dismutase (SOD2) gene expression through SIRT3-regulated DNA-binding activity of FOXO3a and increases the activity of SOD2 by promoting SIRT3-mediated deacetylation. In vivo, oroxylin A inhibits the growth of transplanted human breast tumors associated with glycolytic suppression. These data indicate that oroxylin A inhibits glycolysis-dependent proliferation of breast cancer cells, through the suppression of HIF1α stabilization via SIRT3 activation, providing preclinical information for the cancer therapies of SIRT3 stimulation.
Collapse
|
47
|
Abstract
Sirtuins are a class of enzymes with nicotinamide adenine dinucleotide (NAD)-dependent protein lysine deacylase function. By deacylating various substrate proteins, including histones, transcription factors, and metabolic enzymes, sirtuins regulate various biological processes, such as transcription, cell survival, DNA damage and repair, and longevity. Small molecules that can inhibit sirtuins have been developed and many of them have shown anticancer activity. Here, we summarize the major biological findings that connect sirtuins to cancer and the different types of sirtuin inhibitors developed. Interestingly, biological data suggest that sirtuins have both tumor-suppressing and tumor-promoting roles. However, most pharmacological studies with small-molecule inhibitors suggest that inhibiting sirtuins has anticancer effects. We discuss possible explanations for this discrepancy and suggest possible future directions to further establish sirtuin inhibitors as anticancer agents.
Collapse
|
48
|
Ghosh JC, Siegelin MD, Vaira V, Faversani A, Tavecchio M, Chae YC, Lisanti S, Rampini P, Giroda M, Caino MC, Seo JH, Kossenkov AV, Michalek RD, Schultz DC, Bosari S, Languino LR, Altieri DC. Adaptive mitochondrial reprogramming and resistance to PI3K therapy. J Natl Cancer Inst 2015; 107:dju502. [PMID: 25650317 DOI: 10.1093/jnci/dju502] [Citation(s) in RCA: 95] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Small molecule inhibitors of phosphatidylinositol-3 kinase (PI3K) have been developed as molecular therapy for cancer, but their efficacy in the clinic is modest, hampered by resistance mechanisms. METHODS We studied the effect of PI3K therapy in patient-derived tumor organotypic cultures (from five patient samples), three glioblastoma (GBM) tumor cell lines, and an intracranial model of glioblastoma in immunocompromised mice (n = 4-5 mice per group). Mechanisms of therapy-induced tumor reprogramming were investigated in a global metabolomics screening, analysis of mitochondrial bioenergetics and cell death, and modulation of protein phosphorylation. A high-throughput drug screening was used to identify novel preclinical combination therapies with PI3K inhibitors, and combination synergy experiments were performed. All statistical methods were two-sided. RESULTS PI3K therapy induces global metabolic reprogramming in tumors and promotes the recruitment of an active pool of the Ser/Thr kinase, Akt2 to mitochondria. In turn, mitochondrial Akt2 phosphorylates Ser31 in cyclophilin D (CypD), a regulator of organelle functions. Akt2-phosphorylated CypD supports mitochondrial bioenergetics and opposes tumor cell death, conferring resistance to PI3K therapy. The combination of a small-molecule antagonist of CypD protein folding currently in preclinical development, Gamitrinib, plus PI3K inhibitors (PI3Ki) reverses this adaptive response, produces synergistic anticancer activity by inducing mitochondrial apoptosis, and extends animal survival in a GBM model (vehicle: median survival = 28.5 days; Gamitrinib+PI3Ki: median survival = 40 days, P = .003), compared with single-agent treatment (PI3Ki: median survival = 32 days, P = .02; Gamitrinib: median survival = 35 days, P = .008 by two-sided unpaired t test). CONCLUSIONS Small-molecule PI3K antagonists promote drug resistance by repurposing mitochondrial functions in bioenergetics and cell survival. Novel combination therapies that target mitochondrial adaptation can dramatically improve on the efficacy of PI3K therapy in the clinic.
Collapse
Affiliation(s)
- Jagadish C Ghosh
- Prostate Cancer Discovery and Development Program (JCG, MT, YCC, SL, MCC, JHS, LRL, DCA), Tumor Microenvironment and Metastasis Program (JCG, MT, YCC, SL, MCC, JHS, DCA), Center for Systems and Computational Biology (AVK), and Center for Chemical Biology and Translational Medicine (DCS), The Wistar Institute, Philadelphia, PA; Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY (MDS); Istituto Nazionale Genetica Molecolare "Romeo and Enrica Invernizzi," Milan, Italy (VV); Division of Pathology (VV, AF, SB), Division of Neurosurgery (PR), and Division of Surgery (MG), Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy; Metabolon, Inc. Durham, NC (RDM); Department of Pathophysiology and Organ Transplant, University of Milan, Milan, Italy (SB); Department of Cancer Biology, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA (LRL)
| | - Markus D Siegelin
- Prostate Cancer Discovery and Development Program (JCG, MT, YCC, SL, MCC, JHS, LRL, DCA), Tumor Microenvironment and Metastasis Program (JCG, MT, YCC, SL, MCC, JHS, DCA), Center for Systems and Computational Biology (AVK), and Center for Chemical Biology and Translational Medicine (DCS), The Wistar Institute, Philadelphia, PA; Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY (MDS); Istituto Nazionale Genetica Molecolare "Romeo and Enrica Invernizzi," Milan, Italy (VV); Division of Pathology (VV, AF, SB), Division of Neurosurgery (PR), and Division of Surgery (MG), Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy; Metabolon, Inc. Durham, NC (RDM); Department of Pathophysiology and Organ Transplant, University of Milan, Milan, Italy (SB); Department of Cancer Biology, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA (LRL)
| | - Valentina Vaira
- Prostate Cancer Discovery and Development Program (JCG, MT, YCC, SL, MCC, JHS, LRL, DCA), Tumor Microenvironment and Metastasis Program (JCG, MT, YCC, SL, MCC, JHS, DCA), Center for Systems and Computational Biology (AVK), and Center for Chemical Biology and Translational Medicine (DCS), The Wistar Institute, Philadelphia, PA; Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY (MDS); Istituto Nazionale Genetica Molecolare "Romeo and Enrica Invernizzi," Milan, Italy (VV); Division of Pathology (VV, AF, SB), Division of Neurosurgery (PR), and Division of Surgery (MG), Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy; Metabolon, Inc. Durham, NC (RDM); Department of Pathophysiology and Organ Transplant, University of Milan, Milan, Italy (SB); Department of Cancer Biology, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA (LRL)
| | - Alice Faversani
- Prostate Cancer Discovery and Development Program (JCG, MT, YCC, SL, MCC, JHS, LRL, DCA), Tumor Microenvironment and Metastasis Program (JCG, MT, YCC, SL, MCC, JHS, DCA), Center for Systems and Computational Biology (AVK), and Center for Chemical Biology and Translational Medicine (DCS), The Wistar Institute, Philadelphia, PA; Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY (MDS); Istituto Nazionale Genetica Molecolare "Romeo and Enrica Invernizzi," Milan, Italy (VV); Division of Pathology (VV, AF, SB), Division of Neurosurgery (PR), and Division of Surgery (MG), Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy; Metabolon, Inc. Durham, NC (RDM); Department of Pathophysiology and Organ Transplant, University of Milan, Milan, Italy (SB); Department of Cancer Biology, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA (LRL)
| | - Michele Tavecchio
- Prostate Cancer Discovery and Development Program (JCG, MT, YCC, SL, MCC, JHS, LRL, DCA), Tumor Microenvironment and Metastasis Program (JCG, MT, YCC, SL, MCC, JHS, DCA), Center for Systems and Computational Biology (AVK), and Center for Chemical Biology and Translational Medicine (DCS), The Wistar Institute, Philadelphia, PA; Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY (MDS); Istituto Nazionale Genetica Molecolare "Romeo and Enrica Invernizzi," Milan, Italy (VV); Division of Pathology (VV, AF, SB), Division of Neurosurgery (PR), and Division of Surgery (MG), Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy; Metabolon, Inc. Durham, NC (RDM); Department of Pathophysiology and Organ Transplant, University of Milan, Milan, Italy (SB); Department of Cancer Biology, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA (LRL)
| | - Young Chan Chae
- Prostate Cancer Discovery and Development Program (JCG, MT, YCC, SL, MCC, JHS, LRL, DCA), Tumor Microenvironment and Metastasis Program (JCG, MT, YCC, SL, MCC, JHS, DCA), Center for Systems and Computational Biology (AVK), and Center for Chemical Biology and Translational Medicine (DCS), The Wistar Institute, Philadelphia, PA; Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY (MDS); Istituto Nazionale Genetica Molecolare "Romeo and Enrica Invernizzi," Milan, Italy (VV); Division of Pathology (VV, AF, SB), Division of Neurosurgery (PR), and Division of Surgery (MG), Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy; Metabolon, Inc. Durham, NC (RDM); Department of Pathophysiology and Organ Transplant, University of Milan, Milan, Italy (SB); Department of Cancer Biology, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA (LRL)
| | - Sofia Lisanti
- Prostate Cancer Discovery and Development Program (JCG, MT, YCC, SL, MCC, JHS, LRL, DCA), Tumor Microenvironment and Metastasis Program (JCG, MT, YCC, SL, MCC, JHS, DCA), Center for Systems and Computational Biology (AVK), and Center for Chemical Biology and Translational Medicine (DCS), The Wistar Institute, Philadelphia, PA; Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY (MDS); Istituto Nazionale Genetica Molecolare "Romeo and Enrica Invernizzi," Milan, Italy (VV); Division of Pathology (VV, AF, SB), Division of Neurosurgery (PR), and Division of Surgery (MG), Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy; Metabolon, Inc. Durham, NC (RDM); Department of Pathophysiology and Organ Transplant, University of Milan, Milan, Italy (SB); Department of Cancer Biology, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA (LRL)
| | - Paolo Rampini
- Prostate Cancer Discovery and Development Program (JCG, MT, YCC, SL, MCC, JHS, LRL, DCA), Tumor Microenvironment and Metastasis Program (JCG, MT, YCC, SL, MCC, JHS, DCA), Center for Systems and Computational Biology (AVK), and Center for Chemical Biology and Translational Medicine (DCS), The Wistar Institute, Philadelphia, PA; Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY (MDS); Istituto Nazionale Genetica Molecolare "Romeo and Enrica Invernizzi," Milan, Italy (VV); Division of Pathology (VV, AF, SB), Division of Neurosurgery (PR), and Division of Surgery (MG), Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy; Metabolon, Inc. Durham, NC (RDM); Department of Pathophysiology and Organ Transplant, University of Milan, Milan, Italy (SB); Department of Cancer Biology, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA (LRL)
| | - Massimo Giroda
- Prostate Cancer Discovery and Development Program (JCG, MT, YCC, SL, MCC, JHS, LRL, DCA), Tumor Microenvironment and Metastasis Program (JCG, MT, YCC, SL, MCC, JHS, DCA), Center for Systems and Computational Biology (AVK), and Center for Chemical Biology and Translational Medicine (DCS), The Wistar Institute, Philadelphia, PA; Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY (MDS); Istituto Nazionale Genetica Molecolare "Romeo and Enrica Invernizzi," Milan, Italy (VV); Division of Pathology (VV, AF, SB), Division of Neurosurgery (PR), and Division of Surgery (MG), Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy; Metabolon, Inc. Durham, NC (RDM); Department of Pathophysiology and Organ Transplant, University of Milan, Milan, Italy (SB); Department of Cancer Biology, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA (LRL)
| | - M Cecilia Caino
- Prostate Cancer Discovery and Development Program (JCG, MT, YCC, SL, MCC, JHS, LRL, DCA), Tumor Microenvironment and Metastasis Program (JCG, MT, YCC, SL, MCC, JHS, DCA), Center for Systems and Computational Biology (AVK), and Center for Chemical Biology and Translational Medicine (DCS), The Wistar Institute, Philadelphia, PA; Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY (MDS); Istituto Nazionale Genetica Molecolare "Romeo and Enrica Invernizzi," Milan, Italy (VV); Division of Pathology (VV, AF, SB), Division of Neurosurgery (PR), and Division of Surgery (MG), Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy; Metabolon, Inc. Durham, NC (RDM); Department of Pathophysiology and Organ Transplant, University of Milan, Milan, Italy (SB); Department of Cancer Biology, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA (LRL)
| | - Jae Ho Seo
- Prostate Cancer Discovery and Development Program (JCG, MT, YCC, SL, MCC, JHS, LRL, DCA), Tumor Microenvironment and Metastasis Program (JCG, MT, YCC, SL, MCC, JHS, DCA), Center for Systems and Computational Biology (AVK), and Center for Chemical Biology and Translational Medicine (DCS), The Wistar Institute, Philadelphia, PA; Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY (MDS); Istituto Nazionale Genetica Molecolare "Romeo and Enrica Invernizzi," Milan, Italy (VV); Division of Pathology (VV, AF, SB), Division of Neurosurgery (PR), and Division of Surgery (MG), Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy; Metabolon, Inc. Durham, NC (RDM); Department of Pathophysiology and Organ Transplant, University of Milan, Milan, Italy (SB); Department of Cancer Biology, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA (LRL)
| | - Andrew V Kossenkov
- Prostate Cancer Discovery and Development Program (JCG, MT, YCC, SL, MCC, JHS, LRL, DCA), Tumor Microenvironment and Metastasis Program (JCG, MT, YCC, SL, MCC, JHS, DCA), Center for Systems and Computational Biology (AVK), and Center for Chemical Biology and Translational Medicine (DCS), The Wistar Institute, Philadelphia, PA; Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY (MDS); Istituto Nazionale Genetica Molecolare "Romeo and Enrica Invernizzi," Milan, Italy (VV); Division of Pathology (VV, AF, SB), Division of Neurosurgery (PR), and Division of Surgery (MG), Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy; Metabolon, Inc. Durham, NC (RDM); Department of Pathophysiology and Organ Transplant, University of Milan, Milan, Italy (SB); Department of Cancer Biology, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA (LRL)
| | - Ryan D Michalek
- Prostate Cancer Discovery and Development Program (JCG, MT, YCC, SL, MCC, JHS, LRL, DCA), Tumor Microenvironment and Metastasis Program (JCG, MT, YCC, SL, MCC, JHS, DCA), Center for Systems and Computational Biology (AVK), and Center for Chemical Biology and Translational Medicine (DCS), The Wistar Institute, Philadelphia, PA; Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY (MDS); Istituto Nazionale Genetica Molecolare "Romeo and Enrica Invernizzi," Milan, Italy (VV); Division of Pathology (VV, AF, SB), Division of Neurosurgery (PR), and Division of Surgery (MG), Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy; Metabolon, Inc. Durham, NC (RDM); Department of Pathophysiology and Organ Transplant, University of Milan, Milan, Italy (SB); Department of Cancer Biology, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA (LRL)
| | - David C Schultz
- Prostate Cancer Discovery and Development Program (JCG, MT, YCC, SL, MCC, JHS, LRL, DCA), Tumor Microenvironment and Metastasis Program (JCG, MT, YCC, SL, MCC, JHS, DCA), Center for Systems and Computational Biology (AVK), and Center for Chemical Biology and Translational Medicine (DCS), The Wistar Institute, Philadelphia, PA; Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY (MDS); Istituto Nazionale Genetica Molecolare "Romeo and Enrica Invernizzi," Milan, Italy (VV); Division of Pathology (VV, AF, SB), Division of Neurosurgery (PR), and Division of Surgery (MG), Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy; Metabolon, Inc. Durham, NC (RDM); Department of Pathophysiology and Organ Transplant, University of Milan, Milan, Italy (SB); Department of Cancer Biology, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA (LRL)
| | - Silvano Bosari
- Prostate Cancer Discovery and Development Program (JCG, MT, YCC, SL, MCC, JHS, LRL, DCA), Tumor Microenvironment and Metastasis Program (JCG, MT, YCC, SL, MCC, JHS, DCA), Center for Systems and Computational Biology (AVK), and Center for Chemical Biology and Translational Medicine (DCS), The Wistar Institute, Philadelphia, PA; Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY (MDS); Istituto Nazionale Genetica Molecolare "Romeo and Enrica Invernizzi," Milan, Italy (VV); Division of Pathology (VV, AF, SB), Division of Neurosurgery (PR), and Division of Surgery (MG), Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy; Metabolon, Inc. Durham, NC (RDM); Department of Pathophysiology and Organ Transplant, University of Milan, Milan, Italy (SB); Department of Cancer Biology, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA (LRL)
| | - Lucia R Languino
- Prostate Cancer Discovery and Development Program (JCG, MT, YCC, SL, MCC, JHS, LRL, DCA), Tumor Microenvironment and Metastasis Program (JCG, MT, YCC, SL, MCC, JHS, DCA), Center for Systems and Computational Biology (AVK), and Center for Chemical Biology and Translational Medicine (DCS), The Wistar Institute, Philadelphia, PA; Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY (MDS); Istituto Nazionale Genetica Molecolare "Romeo and Enrica Invernizzi," Milan, Italy (VV); Division of Pathology (VV, AF, SB), Division of Neurosurgery (PR), and Division of Surgery (MG), Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy; Metabolon, Inc. Durham, NC (RDM); Department of Pathophysiology and Organ Transplant, University of Milan, Milan, Italy (SB); Department of Cancer Biology, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA (LRL)
| | - Dario C Altieri
- Prostate Cancer Discovery and Development Program (JCG, MT, YCC, SL, MCC, JHS, LRL, DCA), Tumor Microenvironment and Metastasis Program (JCG, MT, YCC, SL, MCC, JHS, DCA), Center for Systems and Computational Biology (AVK), and Center for Chemical Biology and Translational Medicine (DCS), The Wistar Institute, Philadelphia, PA; Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY (MDS); Istituto Nazionale Genetica Molecolare "Romeo and Enrica Invernizzi," Milan, Italy (VV); Division of Pathology (VV, AF, SB), Division of Neurosurgery (PR), and Division of Surgery (MG), Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy; Metabolon, Inc. Durham, NC (RDM); Department of Pathophysiology and Organ Transplant, University of Milan, Milan, Italy (SB); Department of Cancer Biology, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA (LRL).
| |
Collapse
|
49
|
Rohrbach S, Aslam M, Niemann B, Schulz R. Impact of caloric restriction on myocardial ischaemia/reperfusion injury and new therapeutic options to mimic its effects. Br J Pharmacol 2015; 171:2964-92. [PMID: 24611611 DOI: 10.1111/bph.12650] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2013] [Revised: 01/12/2014] [Accepted: 02/10/2014] [Indexed: 12/12/2022] Open
Abstract
Caloric restriction (CR) is the most reliable intervention to extend lifespan and prevent age-related disorders in various species from yeast to rodents. Short- and long-term CR confers cardio protection against ischaemia/reperfusion injury in young and even in aged rodents. A few human trials suggest that CR has the potential to mediate improvement of cardiac or vascular function and induce retardation of cardiac senescence also in humans. The underlying mechanisms are diverse and have not yet been clearly defined. Among the known mediators for the benefits of CR are NO, the AMP-activated PK, sirtuins and adiponectin. Mitochondria, which play a central role in such complex processes within the cell as apoptosis, ATP-production or oxidative stress, are centrally involved in many aspects of CR-induced protection against ischaemic injury. Here, we discuss the relevant literature regarding the protection against myocardial ischaemia/reperfusion injury conferred by CR. Furthermore, we will discuss drug targets to mimic CR and the possible role of calorie restriction in preserving cardiovascular function in humans.
Collapse
Affiliation(s)
- Susanne Rohrbach
- Institute of Physiology, Justus Liebig University Giessen, Giessen, Germany
| | | | | | | |
Collapse
|
50
|
Imaizumi N, Kwang Lee K, Zhang C, Boelsterli UA. Mechanisms of cell death pathway activation following drug-induced inhibition of mitochondrial complex I. Redox Biol 2015; 4:279-88. [PMID: 25625582 PMCID: PMC4315936 DOI: 10.1016/j.redox.2015.01.005] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2014] [Revised: 01/03/2015] [Accepted: 01/07/2015] [Indexed: 11/02/2022] Open
Abstract
Respiratory complex I inhibition by drugs and other chemicals has been implicated as a frequent mode of mitochondria-mediated cell injury. However, the exact mechanisms leading to the activation of cell death pathways are incompletely understood. This study was designed to explore the relative contributions to cell injury of three distinct consequences of complex I inhibition, i.e., impairment of ATP biosynthesis, increased formation of superoxide and, hence, peroxynitrite, and inhibition of the mitochondrial protein deacetylase, Sirt3, due to imbalance of the NADH/NAD(+) ratio. We used the antiviral drug efavirenz (EFV) to model drug-induced complex I inhibition. Exposure of cultured mouse hepatocytes to EFV resulted in a rapid onset of cell injury, featuring a no-effect level at 30µM EFV and submaximal effects at 50µM EFV. EFV caused a concentration-dependent decrease in cellular ATP levels. Furthermore, EFV resulted in increased formation of peroxynitrite and oxidation of mitochondrial protein thiols, including cyclophilin D (CypD). This was prevented by the superoxide scavenger, Fe-TCP, or the peroxynitrite decomposition catalyst, Fe-TMPyP. Both ferroporphyrins completely protected from EFV-induced cell injury, suggesting that peroxynitrite contributed to the cell injury. Finally, EFV increased the NADH/NAD(+) ratio, inhibited Sirt3 activity, and led to hyperacetylated lysine residues, including those in CypD. However, hepatocytes isolated from Sirt3-null mice were protected against 40µM EFV as compared to their wild-type controls. In conclusion, these data are compatible with the concept that chemical inhibition of complex I activates multiple pathways leading to cell injury; among these, peroxynitrite formation may be the most critical.
Collapse
Affiliation(s)
- Naoki Imaizumi
- Department of Pharmaceutical Sciences, University of Connecticut, Storrs, CT 06269, USA; Laboratory of Molecular Genetics, School of Health Sciences, Faculty of Medicine, University of the Ryukyus, Okinawa 903-0215, Japan.
| | - Kang Kwang Lee
- Department of Pharmaceutical Sciences, University of Connecticut, Storrs, CT 06269, USA
| | - Carmen Zhang
- Department of Pharmaceutical Sciences, University of Connecticut, Storrs, CT 06269, USA
| | - Urs A Boelsterli
- Department of Pharmaceutical Sciences, University of Connecticut, Storrs, CT 06269, USA
| |
Collapse
|