1
|
Javier AJS, Kennedy FM, Yi X, Wehling-Henricks M, Tidball JG, White KE, Witczak CA, Kuro-O M, Welc SS. Klotho Is Cardioprotective in the mdx Mouse Model of Duchenne Muscular Dystrophy. THE AMERICAN JOURNAL OF PATHOLOGY 2025; 195:923-940. [PMID: 39889824 PMCID: PMC12016860 DOI: 10.1016/j.ajpath.2024.12.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 12/18/2024] [Accepted: 12/30/2024] [Indexed: 02/03/2025]
Abstract
Duchenne muscular dystrophy (DMD) is a lethal, progressive skeletal and cardiac myopathy. Cardiomyopathy is the leading cause of death in patients with DMD, but the molecular basis for heart failure is incompletely understood. As with humans, in the mdx mouse model of DMD, cardiac function is impaired after the onset of skeletal muscle pathology. Dysregulation of Klotho gene regulation in dystrophic skeletal muscles occurs at disease onset, affecting pathogenesis. Whether Klotho is protective against dystrophin-deficient cardiomyopathy is unknown. This study found that expression of a Klotho transgene prevented deficits in left ventricular ejection fraction and fractional shortening in mdx mice. Improvements in cardiac performance were associated with reductions in adverse cardiac remodeling, cardiac myocyte hypertrophy, and fibrosis. In addition, mdx mice expressed high concentrations of plasma fibroblast growth factor 23 (FGF23), and expression was increased locally in hearts. The cardioprotective effects of Klotho were not associated with differences in renal function or serum biochemistries, but transgene expression prevented increased expression of plasma FGF23 and cardiac Fgf23 mRNA expression. Cardiac reactive oxygen species, oxidative damage, mitochondrial damage, and apoptosis were reduced in transgenic hearts. FGF23 stimulated hypertrophic growth in dystrophic neonatal mouse ventricular myocytes in vitro, which was inhibited by co-stimulation with soluble Klotho. Taken together, these results show that Klotho prevented dystrophic cardiac remodeling and improved function.
Collapse
MESH Headings
- Animals
- Klotho Proteins
- Muscular Dystrophy, Duchenne/metabolism
- Muscular Dystrophy, Duchenne/physiopathology
- Muscular Dystrophy, Duchenne/pathology
- Muscular Dystrophy, Duchenne/complications
- Muscular Dystrophy, Duchenne/genetics
- Glucuronidase/metabolism
- Glucuronidase/genetics
- Fibroblast Growth Factor-23
- Fibroblast Growth Factors/metabolism
- Fibroblast Growth Factors/blood
- Fibroblast Growth Factors/genetics
- Mice, Inbred mdx
- Disease Models, Animal
- Mice
- Cardiomyopathies/pathology
- Cardiomyopathies/metabolism
- Myocytes, Cardiac/metabolism
- Myocytes, Cardiac/pathology
- Mice, Transgenic
- Mice, Inbred C57BL
- Male
- Apoptosis
- Reactive Oxygen Species/metabolism
- Fibrosis
- Humans
- Myocardium/pathology
- Myocardium/metabolism
Collapse
Affiliation(s)
- Areli Jannes S Javier
- Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, Indiana
| | - Felicia M Kennedy
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, Indiana
| | - Xin Yi
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, Indiana
| | | | - James G Tidball
- Department of Integrative Biology and Physiology, University of California, Los Angeles, California; Department of Pathology and Laboratory Medicine, University of California, Los Angeles, California
| | - Kenneth E White
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana
| | - Carol A Witczak
- Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, Indiana; Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, Indiana
| | - Makoto Kuro-O
- Division of Anti-Aging Medicine, Center for Molecular Medicine, Jichi Medical University, Shimotsuke, Japan
| | - Steven S Welc
- Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, Indiana; Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, Indiana.
| |
Collapse
|
2
|
Yang L, Posner RG, Salanga MC, Traustadóttir T, Guthrie OW. Long-term effects of acute hazardous noise on auditory and non-auditory organs. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART A 2025; 88:519-535. [PMID: 39985403 DOI: 10.1080/15287394.2025.2467177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/24/2025]
Abstract
Hazardous noise is a pervasive environmental pollutant with significant adverse health impacts on auditory and non-auditory organs. It is noteworthy that even acute noise exposure might pose immediate detrimental effects to various organs. However, the long-term effects of acute noise exposure remain largely unknown. This study aimed to explore this gap by randomizing 12 Long-Evans rats into acute noise and control groups. The acute noise regimen was a single three-hr wideband noise (12.5 hz-20 kHz) at 105 dB SPLpeak. Four weeks following exposure cessation, animals from both groups were sacrificed. Genomic DNA and RNA were extracted from the cochlea, brain, heart, and liver. Long-target polymerase assays and real-time quantitative polymerase chain reactions were performed to assess DNA integrity and p53-targeted gene expression, respectively, with results being compared between the two groups. Data demonstrated that noise-induced changes in DNA integrity depended upon organ type, with significant interaction effects between treatment conditions (noise or control) and organ type for nuclear and mitochondrial DNA integrity. In addition, there were significant changes in p53-targeted gene expression between noise-exposed and control in all tested organs. In conclusion, the long-term impact of acute hazardous noise exposure on DNA integrity was complex, highlighting organ-specificity in response to noise. However, such noise significantly altered p53-targeted genes systemically, indicating ongoing cellular stress. Overall, these results suggest that acute exposure to hazardous noise may have potential long-term adverse consequences. Immediate care following exposure might mitigate possible impacts on long-term health.
Collapse
Affiliation(s)
- Li Yang
- Cell & Molecular Pathology Laboratory, Communication Sciences and Disorders, Northern Arizona University, Flagstaff, AZ, USA
- Department of Biological Sciences, Northern Arizona University, Flagstaff, AZ, USA
| | - Richard G Posner
- Department of Biological Sciences, Northern Arizona University, Flagstaff, AZ, USA
| | - Matthew C Salanga
- Department of Biological Sciences, Northern Arizona University, Flagstaff, AZ, USA
| | - Tinna Traustadóttir
- Department of Biological Sciences, Northern Arizona University, Flagstaff, AZ, USA
| | - O'neil W Guthrie
- Cell & Molecular Pathology Laboratory, Communication Sciences and Disorders, Northern Arizona University, Flagstaff, AZ, USA
| |
Collapse
|
3
|
Baker ZN, Zhu Y, Guerra RM, Smith AJ, Arra A, Serrano LR, Overmyer KA, Mukherji S, Craig EA, Coon JJ, Pagliarini DJ. Triacylglycerol mobilization underpins mitochondrial stress recovery. Nat Cell Biol 2025; 27:298-308. [PMID: 39779944 PMCID: PMC11821527 DOI: 10.1038/s41556-024-01586-6] [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: 06/04/2024] [Accepted: 11/26/2024] [Indexed: 01/11/2025]
Abstract
Mitochondria are central to myriad biochemical processes, and thus even their moderate impairment could have drastic cellular consequences if not rectified. Here, to explore cellular strategies for surmounting mitochondrial stress, we conducted a series of chemical and genetic perturbations to Saccharomyces cerevisiae and analysed the cellular responses using deep multiomic mass spectrometry profiling. We discovered that mobilization of lipid droplet triacylglycerol stores was necessary for strains to mount a successful recovery response. In particular, acyl chains from these stores were liberated by triacylglycerol lipases and used to fuel biosynthesis of the quintessential mitochondrial membrane lipid cardiolipin to support new mitochondrial biogenesis. We demonstrate that a comparable recovery pathway exists in mammalian cells, which fail to recover from doxycycline treatment when lacking the ATGL lipase. Collectively, our work reveals a key component of mitochondrial stress recovery and offers a rich resource for further exploration of the broad cellular responses to mitochondrial dysfunction.
Collapse
Affiliation(s)
- Zakery N Baker
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Yunyun Zhu
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI, USA
- National Center for Quantitative Biology of Complex Systems, Madison, WI, USA
| | - Rachel M Guerra
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Andrew J Smith
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Aline Arra
- Department of Physics, Washington University, St. Louis, MO, USA
| | - Lia R Serrano
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI, USA
- National Center for Quantitative Biology of Complex Systems, Madison, WI, USA
| | - Katherine A Overmyer
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI, USA
- National Center for Quantitative Biology of Complex Systems, Madison, WI, USA
- Morgridge Institute for Research, Madison, WI, USA
| | - Shankar Mukherji
- Department of Physics, Washington University, St. Louis, MO, USA
| | - Elizabeth A Craig
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI, USA
| | - Joshua J Coon
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI, USA
- National Center for Quantitative Biology of Complex Systems, Madison, WI, USA
- Morgridge Institute for Research, Madison, WI, USA
- Department of Biomolecular Chemistry, University of Wisconsin-Madison, Madison, WI, USA
| | - David J Pagliarini
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO, USA.
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, MO, USA.
- Department of Genetics, Washington University School of Medicine, St. Louis, MO, USA.
- Howard Hughes Medical Institute, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
4
|
Pool L, van Wijk SW, van Schie MS, Taverne YJHJ, de Groot NMS, Brundel BJJM. Quantifying DNA Lesions and Circulating Free DNA: Diagnostic Marker for Electropathology and Clinical Stage of AF. JACC Clin Electrophysiol 2025; 11:321-332. [PMID: 39797858 DOI: 10.1016/j.jacep.2024.10.008] [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: 07/05/2024] [Revised: 09/27/2024] [Accepted: 10/04/2024] [Indexed: 01/13/2025]
Abstract
BACKGROUND Atrial fibrillation (AF) persistence is associated with molecular remodeling that fuels electrical conduction abnormalities in atrial tissue. Previous research revealed DNA damage as a molecular driver of AF. OBJECTIVES This study sought to explore the diagnostic value of DNA damage in atrial tissue and blood samples as an indicator of the prevalence of electrical conduction abnormalities and stage of AF. METHODS High-sensitivity long-run real-time PCR was performed on mitochondrial (ND1) and nuclear (P53) DNA from atrial tissue samples from paroxysmal (PAF), persistent (PeAF), and longstanding persistent (LS-PeAF) AF, and sinus rhythm (SR) patients (n = 83). PicoGreen assay and quantitative polymerase chain reaction were used on circulating free DNA (cfDNA) markers (total cfDNA, β-globin, ND1, and P53) in blood samples of 70 patients with AF or SR. High-resolution epicardial mapping of the atria (n = 48) was conducted to quantify electrical conduction abnormalities. RESULTS The number of DNA lesions gradually and significantly increased in PAF and PeAF and in patients with <3 years of AF compared with SR. In SR, the quantity of nuclear DNA damage significantly correlated with the proportion of fractionated potentials. Mitochondrial DNA lesions correlated with slower conduction velocity and lower potential amplitudes in AF samples. Also, mitochondrial cfDNA levels decreased in patients with >3 years of AF compared with <3 years of AF (P = 0.004). CONCLUSIONS The quantity of DNA lesions in atrial tissue samples is associated with atrial conduction abnormalities and stage of AF. Serum DNA damage markers discriminate short- from long-term AF. Therefore, the quantity of DNA damage may have diagnostic value in clinical AF management.
Collapse
Affiliation(s)
- Lisa Pool
- Physiology, Amsterdam Cardiovascular Sciences, Heart Failure, and Arrhythmias, Amsterdam University Medical Center, location Vrije Universiteit Amsterdam, Amsterdam, the Netherlands; Translational Electrophysiology Unit, Department of Cardiology, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Stan W van Wijk
- Physiology, Amsterdam Cardiovascular Sciences, Heart Failure, and Arrhythmias, Amsterdam University Medical Center, location Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Mathijs S van Schie
- Translational Electrophysiology Unit, Department of Cardiology, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Yannick J H J Taverne
- Translational Cardiothoracic Research Laboratory, Department of Cardiothoracic Surgery, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Natasja M S de Groot
- Translational Electrophysiology Unit, Department of Cardiology, Erasmus Medical Center, Rotterdam, the Netherlands; Department of Microelectronics, Circuits, and Systems, Faculty of Electrical Engineering, Mathematics, and Computer Sciences, Delft University of Technology, Delft, the Netherlands
| | - Bianca J J M Brundel
- Physiology, Amsterdam Cardiovascular Sciences, Heart Failure, and Arrhythmias, Amsterdam University Medical Center, location Vrije Universiteit Amsterdam, Amsterdam, the Netherlands.
| |
Collapse
|
5
|
Flores-Sierra JDJ, Muciño-Arellano MDR, Romo-Morales GDC, Sánchez-Palafox JE, Correa-Navarro VA, Colín-Castelán D, Pérez-Vázquez V, Rangel-Salazar R, Rivera-Bustamante R, de la Rocha C, Rodríguez-Ríos D, Trejo-Saavedra DL, Molina-Torres J, Ramírez-Chávez E, García-Rojas NS, Winkler R, Lund G, Zaina S. The DNA methyltransferase inhibitor decitabine blunts the response to a high-animal fat and protein diet in mice. J Lipid Res 2024; 65:100586. [PMID: 38942113 PMCID: PMC11325794 DOI: 10.1016/j.jlr.2024.100586] [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: 05/28/2024] [Revised: 06/18/2024] [Accepted: 06/20/2024] [Indexed: 06/30/2024] Open
Abstract
Increasing evidence hints that DNA hypermethylation may mediate the pathogenic response to cardiovascular risk factors. Here, we tested a corollary of that hypothesis, that is, that the DNA methyltransferase inhibitor decitabine (Dec) ameliorates the metabolic profile of mice fed a moderately high-animal fat and protein diet (HAFPD), a proxy of cardiovascular risk-associated Western-type diet. HAFPD-fed mice were exposed to Dec or vehicle for eight weeks (8W set, 4-32/group). To assess any memory of past exposure to Dec, we surveyed a second mice set treated as 8W but HAFPD-fed for further eight weeks without any Dec (16W set, 4-20/group). In 8W, Dec markedly reduced HAFPD-induced body weight gain in females, but marginally in males. Characterization of females revealed that Dec augmented skeletal muscle lipid content, while decreasing liver fat content and increasing plasma nonesterified fatty acids, adipose insulin resistance, and-although marginally-whole blood acylcarnitines, compared to HAFPD alone. Skeletal muscle mitochondrial DNA copy number was higher in 8W mice exposed to HAFPD and Dec, or in 16W mice fed HAFPD only, relative to 8W mice fed HAFPD only, but Dec induced a transcriptional profile indicative of ameliorated mitochondrial function. Memory of past Dec exposure was tissue-specific and sensitive to both duration of exposure to HAFPD and age. In conclusion, Dec redirected HAFPD-induced lipid accumulation toward the skeletal muscle, likely due to augmented mitochondrial functionality and increased lipid demand. As caveat, Dec induced adipose insulin resistance. Our findings may help identifying strategies for prevention and treatment of lipid dysmetabolism.
Collapse
Affiliation(s)
- José de Jesús Flores-Sierra
- Division of Health Sciences, Department of Medical Sciences, Leon Campus, University of Guanajuato, Leon, Mexico; Tecnológico Nacional de México/ITS de Purísima del Rincón, Purísima del Rincón, Guanajuato, Mexico
| | | | | | | | | | - Dannia Colín-Castelán
- Division of Health Sciences, Department of Medical Sciences, Leon Campus, University of Guanajuato, Leon, Mexico
| | - Victoriano Pérez-Vázquez
- Division of Health Sciences, Department of Medical Sciences, Leon Campus, University of Guanajuato, Leon, Mexico
| | - Rubén Rangel-Salazar
- Division of Health Sciences, Department of Medical Sciences, Leon Campus, University of Guanajuato, Leon, Mexico
| | | | - Carmen de la Rocha
- Department of Genetic Engineering, CINVESTAV Irapuato Unit, Irapuato, Mexico
| | | | | | - Jorge Molina-Torres
- Department of Biotechnology and Biochemistry, CINVESTAV Irapuato Unit, Irapuato, Mexico
| | | | | | | | - Gertrud Lund
- Department of Genetic Engineering, CINVESTAV Irapuato Unit, Irapuato, Mexico.
| | - Silvio Zaina
- Division of Health Sciences, Department of Medical Sciences, Leon Campus, University of Guanajuato, Leon, Mexico.
| |
Collapse
|
6
|
Thomas C, Erni R, Wu JY, Fischer F, Lamers G, Grigolon G, Mitchell SJ, Zarse K, Carreira EM, Ristow M. A naturally occurring polyacetylene isolated from carrots promotes health and delays signatures of aging. Nat Commun 2023; 14:8142. [PMID: 38065964 PMCID: PMC10709416 DOI: 10.1038/s41467-023-43672-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Accepted: 11/16/2023] [Indexed: 12/18/2023] Open
Abstract
To ameliorate or even prevent signatures of aging in ultimately humans, we here report the identification of a previously undescribed polyacetylene contained in the root of carrots (Daucus carota), hereafter named isofalcarintriol, which we reveal as potent promoter of longevity in the nematode C. elegans. We assign the absolute configuration of the compound as (3 S,8 R,9 R,E)-heptadeca-10-en-4,6-diyne-3,8,9-triol, and develop a modular asymmetric synthesis route for all E-isofalcarintriol stereoisomers. At the molecular level, isofalcarintriol affects cellular respiration in mammalian cells, C. elegans, and mice, and interacts with the α-subunit of the mitochondrial ATP synthase to promote mitochondrial biogenesis. Phenotypically, this also results in decreased mammalian cancer cell growth, as well as improved motility and stress resistance in C. elegans, paralleled by reduced protein accumulation in nematodal models of neurodegeneration. In addition, isofalcarintriol supplementation to both wild-type C57BL/6NRj mice on high-fat diet, and aged mice on chow diet results in improved glucose metabolism, increased exercise endurance, and attenuated parameters of frailty at an advanced age. Given these diverse effects on health parameters in both nematodes and mice, isofalcarintriol might become a promising mitohormesis-inducing compound to delay, ameliorate, or prevent aging-associated diseases in humans.
Collapse
Affiliation(s)
- Carolin Thomas
- Laboratory of Energy Metabolism, Institute of Translational Medicine, Department of Health Sciences and Technology, Swiss Federal Institute (ETH) Zurich, Schorenstrasse 16, 8603, Schwerzenbach, Switzerland
| | - Reto Erni
- Laboratory of Chemistry and Applied Biosciences, Department of Organic Chemistry, Swiss Federal Institute (ETH) Zurich, Vladimir-Prelog-Weg 1-5/10, Zurich, 8093, Switzerland
- Biozentrum, University of Basel, Basel, 4056, Switzerland
| | - Jia Yee Wu
- Laboratory of Energy Metabolism, Institute of Translational Medicine, Department of Health Sciences and Technology, Swiss Federal Institute (ETH) Zurich, Schorenstrasse 16, 8603, Schwerzenbach, Switzerland
| | - Fabian Fischer
- Laboratory of Energy Metabolism, Institute of Translational Medicine, Department of Health Sciences and Technology, Swiss Federal Institute (ETH) Zurich, Schorenstrasse 16, 8603, Schwerzenbach, Switzerland
- CureVac SE, Tübingen, 72076, Germany
| | - Greta Lamers
- Laboratory of Energy Metabolism, Institute of Translational Medicine, Department of Health Sciences and Technology, Swiss Federal Institute (ETH) Zurich, Schorenstrasse 16, 8603, Schwerzenbach, Switzerland
| | - Giovanna Grigolon
- Laboratory of Energy Metabolism, Institute of Translational Medicine, Department of Health Sciences and Technology, Swiss Federal Institute (ETH) Zurich, Schorenstrasse 16, 8603, Schwerzenbach, Switzerland
| | - Sarah J Mitchell
- Ludwig Princeton Branch, Princeton University, Princeton, NJ, 08540, USA
| | - Kim Zarse
- Laboratory of Energy Metabolism, Institute of Translational Medicine, Department of Health Sciences and Technology, Swiss Federal Institute (ETH) Zurich, Schorenstrasse 16, 8603, Schwerzenbach, Switzerland
- Institute of Experimental Endocrinology, Charité Universitätsmedizin Berlin, Berlin, 10117, Germany
| | - Erick M Carreira
- Laboratory of Chemistry and Applied Biosciences, Department of Organic Chemistry, Swiss Federal Institute (ETH) Zurich, Vladimir-Prelog-Weg 1-5/10, Zurich, 8093, Switzerland.
| | - Michael Ristow
- Laboratory of Energy Metabolism, Institute of Translational Medicine, Department of Health Sciences and Technology, Swiss Federal Institute (ETH) Zurich, Schorenstrasse 16, 8603, Schwerzenbach, Switzerland.
- Institute of Experimental Endocrinology, Charité Universitätsmedizin Berlin, Berlin, 10117, Germany.
| |
Collapse
|
7
|
Dai CY, Ng CC, Hung GCC, Kirmes I, Hughes LA, Du Y, Brosnan CA, Ahier A, Hahn A, Haynes CM, Rackham O, Filipovska A, Zuryn S. ATFS-1 counteracts mitochondrial DNA damage by promoting repair over transcription. Nat Cell Biol 2023; 25:1111-1120. [PMID: 37460695 DOI: 10.1038/s41556-023-01192-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 06/19/2023] [Indexed: 08/12/2023]
Abstract
The ability to balance conflicting functional demands is critical for ensuring organismal survival. The transcription and repair of the mitochondrial genome (mtDNA) requires separate enzymatic activities that can sterically compete1, suggesting a life-long trade-off between these two processes. Here in Caenorhabditis elegans, we find that the bZIP transcription factor ATFS-1/Atf5 (refs. 2,3) regulates this balance in favour of mtDNA repair by localizing to mitochondria and interfering with the assembly of the mitochondrial pre-initiation transcription complex between HMG-5/TFAM and RPOM-1/mtRNAP. ATFS-1-mediated transcriptional inhibition decreases age-dependent mtDNA molecular damage through the DNA glycosylase NTH-1/NTH1, as well as the helicase TWNK-1/TWNK, resulting in an enhancement in the functional longevity of cells and protection against decline in animal behaviour caused by targeted and severe mtDNA damage. Together, our findings reveal that ATFS-1 acts as a molecular focal point for the control of balance between genome expression and maintenance in the mitochondria.
Collapse
Affiliation(s)
- Chuan-Yang Dai
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, Queensland, Australia
| | - Chai Chee Ng
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, Queensland, Australia
| | - Grace Ching Ching Hung
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, Queensland, Australia
| | - Ina Kirmes
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, Queensland, Australia
| | - Laetitia A Hughes
- Harry Perkins Institute of Medical Research, QEII Medical Centre, The University of Western Australia, Nedlands, Western Australia, Australia
- ARC Centre of Excellence in Synthetic Biology, QEII Medical Centre, The University of Western Australia, Nedlands, Western Australia, Australia
- Telethon Kids Institute, Northern Entrance, Perth Children's Hospital, Nedlands, Western Australia, Australia
| | - Yunguang Du
- Department of Molecular, Cell and Cancer Biology, UMass Chan Medical School, Worchester, MA, USA
| | - Christopher A Brosnan
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, Queensland, Australia
| | - Arnaud Ahier
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, Queensland, Australia
| | - Anne Hahn
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, Queensland, Australia
| | - Cole M Haynes
- Department of Molecular, Cell and Cancer Biology, UMass Chan Medical School, Worchester, MA, USA
| | - Oliver Rackham
- Harry Perkins Institute of Medical Research, QEII Medical Centre, The University of Western Australia, Nedlands, Western Australia, Australia
- ARC Centre of Excellence in Synthetic Biology, QEII Medical Centre, The University of Western Australia, Nedlands, Western Australia, Australia
- Telethon Kids Institute, Northern Entrance, Perth Children's Hospital, Nedlands, Western Australia, Australia
- Curtin Medical School and Curtin Health Innovation Research Institute, Curtin University, Bentley, Western Australia, Australia
| | - Aleksandra Filipovska
- Harry Perkins Institute of Medical Research, QEII Medical Centre, The University of Western Australia, Nedlands, Western Australia, Australia
- ARC Centre of Excellence in Synthetic Biology, QEII Medical Centre, The University of Western Australia, Nedlands, Western Australia, Australia
- Telethon Kids Institute, Northern Entrance, Perth Children's Hospital, Nedlands, Western Australia, Australia
| | - Steven Zuryn
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, Queensland, Australia.
| |
Collapse
|
8
|
Huang J, Song Z, Wei B, Li Q, Lin P, Li H, Dong K. Immunological evaluation of patients with Alzheimer's disease based on mitogen-stimulated cytokine productions and mitochondrial DNA indicators. BMC Psychiatry 2023; 23:145. [PMID: 36890488 PMCID: PMC9993804 DOI: 10.1186/s12888-023-04634-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Accepted: 02/27/2023] [Indexed: 03/10/2023] Open
Abstract
BACKGROUND Based on its objective characteristics, laboratory markers have always been the research direction of clinical diagnosis and assessment of mental disorders including Alzheimer's disease. METHODS MTT Colorimetric Assay, ELISA, and quantitative PCR were used to investigate the responsiveness of peripheral blood mononuclear cells (PBMCs) to mitogen Lipopolysaccharides (LPS) and Phytohemagglutinin (PHA), PBMCs genomic methylation and hydroxymethylation levels, nuclear DNA and mitochondrial DNA damage, respiratory chain enzyme activities, and circulating cell-free mitochondrial DNA levels were detected in 90 patients with Alzheimer's disease. RESULTS In the Alzheimer's disease group, LPS stimulated PBMCs viability, TNF-α secretion, PHA stimulated IL-10 secretion, genomic DNA methylation levels, circulating cell-free mitochondrial DNA copies, citrate synthase activity were reduced compared to the control; while the LPS stimulated PBMCs IL-1α secretion, PHA stimulated IL-1α and IFN-γ secretion, plasma IL-6 and TNF-α, mitochondrial DNA damages were increased compared to the control. CONCLUSIONS The reactivity of peripheral blood mononuclear cells to mitogens, mitochondrial DNA integrity characteristics, and cell-free mitochondrial DNA copies may be used as candidate laboratory biomarkers to help clinical management of Alzheimer's disease.
Collapse
Affiliation(s)
- Jiewen Huang
- Department of Laboratory Medicine, College of Health Science and Technology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,School of Global Health, Chinese Center for Tropical Diseases Research, Shanghai Jiao Tong University School of Medicine, One Health Center, Shanghai Jiao Tong University-The University of Edinburgh, Shanghai, China
| | - Zhen Song
- Department of Laboratory Medicine, College of Health Science and Technology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Beiwen Wei
- Department of Laboratory Medicine, College of Health Science and Technology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qingtian Li
- Department of Laboratory Medicine, College of Health Science and Technology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ping Lin
- Department of Clinical Laboratory, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Hui Li
- Department of Laboratory Medicine, College of Health Science and Technology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Ke Dong
- School of Global Health, Chinese Center for Tropical Diseases Research, Shanghai Jiao Tong University School of Medicine, One Health Center, Shanghai Jiao Tong University-The University of Edinburgh, Shanghai, China.
| |
Collapse
|
9
|
Ramezan D, Farrokhzad Y, Mokhtassi-Bidgoli A, Rasouli-Alamuti M. Multi-walled carbon nanotubes interact with light intensity to affect morpho-biochemical, nutrient uptake, DNA damage, and secondary metabolism of Stevia rebaudiana. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2023; 30:36915-36927. [PMID: 36550247 DOI: 10.1007/s11356-022-24757-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Accepted: 12/10/2022] [Indexed: 06/17/2023]
Abstract
In this study, the interaction between nanoparticles (0, 50, 100, and 150 mg L-1) and light intensity (100, 200, and 400 μmol·m-2·s-1) was evaluated for effectiveness in improving stevia shoot induction by measuring morphological traits, nutrient absorption, total carbohydrates, steviol glycosides (SVglys), and DNA damage in two DNA sequence regions (promoter and sequence of the UGT76G1 gene). MWCNTs at a concentration of 50 mg L-1 in interaction with the light intensity of 200 μmol·m-2·s-1 improved the morphological traits and absorption of nutrients such as nitrogen (N), phosphorous (P), potassium (K), calcium (Ca), iron (Fe), and Manganese (Mn), compared to other treatments. Also, under this interaction, the accumulation of total carbohydrates and SVglys was elevated. Moreover, DNA damage in both regions of the DNA sequence under light intensity at low concentrations of MWCNTs (0 and 50 mg L-1) did not show a significant change but increased with increasing MWCNT concentration at high light intensities (200 and 400 μmol·m-2·s-1). These results demonstrate that the advantages and phytotoxicity of MWCNTs in the in vitro culture of stevia are dose-dependent and are affected by light intensity. Based on this, the interaction of 50 mg L-1 of MWCNTs with the light intensity of 200 μmol·m-2·s-1 is recommended to improve stevia micropropagation and subsequent growth and metabolism.
Collapse
Affiliation(s)
- Dariush Ramezan
- Department of Horticulture and Landscaping, Faculty of Agriculture, University of Zabol, Zabol, Iran.
| | - Yusuf Farrokhzad
- Department of Horticultural Science, Faculty of Agriculture, Tarbiat Modares University, Tehran, Iran
| | - Ali Mokhtassi-Bidgoli
- Department of Agronomy, Faculty of Agriculture, Tarbiat Modares University, Tehran, Iran
| | - Mojtaba Rasouli-Alamuti
- Department of Agricultural Biotechnology, Faculty of Agriculture, Payame Noor University, Karaj, Iran
| |
Collapse
|
10
|
Damianos A, Kulandavelu S, Chen P, Nwajei P, Batlahally S, Sharma M, Alvarez-Cubela S, Dominguez-Bendala J, Zambrano R, Huang J, Hare JM, Schmidt A, Wu S, Benny M, Claure N, Young K. Neonatal intermittent hypoxia persistently impairs lung vascular development and induces long-term lung mitochondrial DNA damage. J Appl Physiol (1985) 2022; 133:1031-1041. [PMID: 36135955 PMCID: PMC11918270 DOI: 10.1152/japplphysiol.00708.2021] [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: 10/11/2021] [Revised: 09/02/2022] [Accepted: 09/02/2022] [Indexed: 11/22/2022] Open
Abstract
Adults born preterm have an increased risk of pulmonary vascular disease. Extreme preterm infants often require supplemental oxygen but they also exhibit frequent intermittent hypoxemic episodes (IH). Here, we test the hypothesis that neonatal IH induces lung endothelial cell mitochondrial DNA (mitDNA) damage and contributes to long-term pulmonary vascular disease and pulmonary hypertension (PH). Newborn C57BL/6J mice were assigned to the following groups: 1) normoxia, 2) hyperoxia (O2 65%), 3) normoxia cycling with IH (O2 21% + O2 10%), and 4) hyperoxia cycling with IH (O2 65% + O2 10%) for 3 wk. IH episodes were initiated on postnatal day 7. Lung angiogenesis, PH, and mitDNA lesions were assessed at 3 wk and 3 mo. In vitro, the effect of IH on tubule formation and mitDNA lesions was evaluated in human pulmonary microvascular endothelial cells (HPMECs). Data were analyzed by ANOVA. In vitro, IH exposure reduced tubule formation and increased mitDNA lesions in HPMECs. This was most marked in HPMECs exposed to hyperoxia cycling with IH. In vivo, neonatal IH increased lung mitDNA lesions, impaired angiogenesis, and induced PH in 3-wk-old mice. These findings were pronounced in mice exposed to hyperoxia cycling with IH. At 3 mo follow-up, mice exposed to neonatal IH had persistently increased lung mitDNA lesions and impaired lung angiogenesis, even without concomitant hyperoxia exposure. Neonatal IH induces lung endothelial cell mitDNA damage and causes persistent impairment in lung angiogenesis. These findings provide important mechanistic insight into the pathogenesis of pulmonary vascular disease in preterm survivors.NEW & NOTEWORTHY Our current study demonstrates that neonatal intermittent hypoxia (IH) alters lung endothelial cell function, induces mitochondrial DNA lesions, and impairs lung vascular growth into adulthood. Moreover, when superimposed on hyperoxia, neonatal IH induces a severe lung vascular phenotype that is seen in preterm infants with PH. These findings suggest that neonatal IH contributes to PH in adults born preterm and importantly, that mitochondrial protection strategies may mitigate these deleterious effects.
Collapse
Affiliation(s)
- Andreas Damianos
- Department of Pediatrics, University of Miami Miller School of Medicine, Miami, Florida
- Batchelor Children's Research Institute, University of Miami Miller School of Medicine, Miami, Florida
| | - Shathiyah Kulandavelu
- Department of Pediatrics, University of Miami Miller School of Medicine, Miami, Florida
- The Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, Florida
| | - Pingping Chen
- Department of Pediatrics, University of Miami Miller School of Medicine, Miami, Florida
- Batchelor Children's Research Institute, University of Miami Miller School of Medicine, Miami, Florida
| | - Patrick Nwajei
- Department of Pediatrics, University of Miami Miller School of Medicine, Miami, Florida
- Batchelor Children's Research Institute, University of Miami Miller School of Medicine, Miami, Florida
| | - Sunil Batlahally
- Department of Pediatrics, University of Miami Miller School of Medicine, Miami, Florida
- Batchelor Children's Research Institute, University of Miami Miller School of Medicine, Miami, Florida
| | - Mayank Sharma
- Department of Pediatrics, University of Miami Miller School of Medicine, Miami, Florida
- Batchelor Children's Research Institute, University of Miami Miller School of Medicine, Miami, Florida
| | - Silvia Alvarez-Cubela
- The Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, Florida
| | - Juan Dominguez-Bendala
- The Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, Florida
| | - Ronald Zambrano
- Department of Pediatrics, University of Miami Miller School of Medicine, Miami, Florida
- Batchelor Children's Research Institute, University of Miami Miller School of Medicine, Miami, Florida
| | - Jian Huang
- Department of Pediatrics, University of Miami Miller School of Medicine, Miami, Florida
- Batchelor Children's Research Institute, University of Miami Miller School of Medicine, Miami, Florida
| | - Joshua M Hare
- The Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, Florida
- Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida
| | - Augusto Schmidt
- Department of Pediatrics, University of Miami Miller School of Medicine, Miami, Florida
- Batchelor Children's Research Institute, University of Miami Miller School of Medicine, Miami, Florida
| | - Shu Wu
- Department of Pediatrics, University of Miami Miller School of Medicine, Miami, Florida
- Batchelor Children's Research Institute, University of Miami Miller School of Medicine, Miami, Florida
| | - Merline Benny
- Department of Pediatrics, University of Miami Miller School of Medicine, Miami, Florida
- Batchelor Children's Research Institute, University of Miami Miller School of Medicine, Miami, Florida
| | - Nelson Claure
- Department of Pediatrics, University of Miami Miller School of Medicine, Miami, Florida
- Batchelor Children's Research Institute, University of Miami Miller School of Medicine, Miami, Florida
| | - Karen Young
- Department of Pediatrics, University of Miami Miller School of Medicine, Miami, Florida
- Batchelor Children's Research Institute, University of Miami Miller School of Medicine, Miami, Florida
- The Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, Florida
| |
Collapse
|
11
|
Beatriz M, Vilaça R, Anjo SI, Manadas B, Januário C, Rego AC, Lopes C. Defective mitochondria-lysosomal axis enhances the release of extracellular vesicles containing mitochondrial DNA and proteins in Huntington's disease. JOURNAL OF EXTRACELLULAR BIOLOGY 2022; 1:e65. [PMID: 38939215 PMCID: PMC11080813 DOI: 10.1002/jex2.65] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 07/21/2022] [Accepted: 09/23/2022] [Indexed: 06/29/2024]
Abstract
Mitochondrial and autophagy dysfunction are mechanisms proposed to be involved in the pathogenesis of several neurodegenerative diseases. Huntington's disease (HD) is a progressive neurodegenerative disorder associated with mutant Huntingtin-induced abnormalities in neuronal mitochondrial dynamics and quality control. Former studies suggest that the removal of defective mitochondria may be compromised in HD. Mitochondrial quality control (MQC) is a complex, well-orchestrated pathway that can be compromised through mitophagy dysregulation or impairment in the mitochondria-lysosomal axis. Another mitochondrial stress response is the generation of mitochondrial-derived vesicles that fuse with the endolysosomal system and form multivesicular bodies that are extruded from cells as extracellular vesicles (EVs). In this work, we aimed to study the presence of mitochondrial components in human EVs and the relation to the dysfunction of both mitochondria and the autophagy pathway. We comprehensively characterized the mitochondrial and autophagy alterations in premanifest and manifest HD carriers and performed a proteomic and genomic EVs profile. We observed that manifest HD patients exhibit mitochondrial and autophagy impairment associated with enhanced EVs release. Furthermore, we detected mitochondrial DNA and proteins in EVs released by HD cells and in neuronal-derived EVs including VDAC-1 and alpha and beta subunits of ATP synthase F1. HD-extracellular vesicles transport higher levels of mitochondrial genetic material in manifest HD patients, suggesting an alternative pathway for the secretion of reactive mitochondrial components. This study provides a novel framework connecting EVs enhanced release of mitochondrial components to mitochondrial and lysosomal dysfunction in HD.
Collapse
Affiliation(s)
- Margarida Beatriz
- CNC‐Center for Neuroscience and Cell BiologyCIBB ‐ Centre for Innovative Biomedicine and BiotechnologyUniversity of CoimbraCoimbraPortugal
- IIIUC‐Institute for Interdisciplinary ResearchUniversity of CoimbraCoimbraPortugal
| | - Rita Vilaça
- CNC‐Center for Neuroscience and Cell BiologyCIBB ‐ Centre for Innovative Biomedicine and BiotechnologyUniversity of CoimbraCoimbraPortugal
- IIIUC‐Institute for Interdisciplinary ResearchUniversity of CoimbraCoimbraPortugal
| | - Sandra I. Anjo
- CNC‐Center for Neuroscience and Cell BiologyCIBB ‐ Centre for Innovative Biomedicine and BiotechnologyUniversity of CoimbraCoimbraPortugal
- Multidisciplinary Institute of AgeingUniversity of CoimbraCoimbraPortugal
| | - Bruno Manadas
- CNC‐Center for Neuroscience and Cell BiologyCIBB ‐ Centre for Innovative Biomedicine and BiotechnologyUniversity of CoimbraCoimbraPortugal
| | | | - A. Cristina Rego
- CNC‐Center for Neuroscience and Cell BiologyCIBB ‐ Centre for Innovative Biomedicine and BiotechnologyUniversity of CoimbraCoimbraPortugal
- FMUC‐Faculty of MedicineUniversity of CoimbraCoimbraPortugal
| | - Carla Lopes
- CNC‐Center for Neuroscience and Cell BiologyCIBB ‐ Centre for Innovative Biomedicine and BiotechnologyUniversity of CoimbraCoimbraPortugal
- IIIUC‐Institute for Interdisciplinary ResearchUniversity of CoimbraCoimbraPortugal
| |
Collapse
|
12
|
Chang S, Tat J, China SP, Kalyanaraman H, Zhuang S, Chan A, Lai C, Radic Z, Abdel-Rahman EA, Casteel DE, Pilz RB, Ali SS, Boss GR. Cobinamide is a strong and versatile antioxidant that overcomes oxidative stress in cells, flies, and diabetic mice. PNAS NEXUS 2022; 1:pgac191. [PMID: 36276587 PMCID: PMC9578022 DOI: 10.1093/pnasnexus/pgac191] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Accepted: 09/12/2022] [Indexed: 01/29/2023]
Abstract
Increased oxidative stress underlies a variety of diseases, including diabetes. Here, we show that the cobalamin/vitamin B12 analog cobinamide is a strong and multifaceted antioxidant, neutralizing superoxide, hydrogen peroxide, and peroxynitrite, with apparent rate constants of 1.9 × 108, 3.7 × 104, and 6.3 × 106 M-1 s-1, respectively, for cobinamide with the cobalt in the +2 oxidation state. Cobinamide with the cobalt in the +3 oxidation state yielded apparent rate constants of 1.1 × 108 and 8.0 × 102 M-1 s-1 for superoxide and hydrogen peroxide, respectively. In mammalian cells and Drosophila melanogaster, cobinamide outperformed cobalamin and two well-known antioxidants, imisopasem manganese and manganese(III)tetrakis(4-benzoic acid)porphyrin, in reducing oxidative stress as evidenced by: (i) decreased mitochondrial superoxide and return of the mitochondrial membrane potential in rotenone- and antimycin A-exposed H9c2 rat cardiomyocytes; (ii) reduced JNK phosphorylation in hydrogen-peroxide-treated H9c2 cells; (iii) increased growth in paraquat-exposed COS-7 fibroblasts; and (iv) improved survival in paraquat-treated flies. In diabetic mice, cobinamide administered in the animals' drinking water completely prevented an increase in lipid and protein oxidation, DNA damage, and fibrosis in the heart. Cobinamide is a promising new antioxidant that has potential use in diseases with heightened oxidative stress.
Collapse
Affiliation(s)
| | | | | | | | - Shunhui Zhuang
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Adriano Chan
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Cassandra Lai
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Zoran Radic
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA 92093, USA
| | - Engy A Abdel-Rahman
- Tumor Biology Research Program, Children’s Cancer Hospital, Cairo 57357, Egypt,Pharmacology Department, Faculty of Medicine, Assuit University, Assuit 71515, Egypt
| | - Darren E Casteel
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Renate B Pilz
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | | | | |
Collapse
|
13
|
Leuthner T, Benzing L, Kohrn B, Bergemann C, Hipp M, Hershberger K, Mello D, Sokolskyi T, Stevenson K, Merutka I, Seay S, Gregory S, Kennedy S, Meyer J. Resistance of mitochondrial DNA to cadmium and Aflatoxin B1 damage-induced germline mutation accumulation in C. elegans. Nucleic Acids Res 2022; 50:8626-8642. [PMID: 35947695 PMCID: PMC9410910 DOI: 10.1093/nar/gkac666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 07/11/2022] [Accepted: 07/22/2022] [Indexed: 01/12/2023] Open
Abstract
Mitochondrial DNA (mtDNA) is prone to mutation in aging and over evolutionary time, yet the processes that regulate the accumulation of de novo mtDNA mutations and modulate mtDNA heteroplasmy are not fully elucidated. Mitochondria lack certain DNA repair processes, which could contribute to polymerase error-induced mutations and increase susceptibility to chemical-induced mtDNA mutagenesis. We conducted error-corrected, ultra-sensitive Duplex Sequencing to investigate the effects of two known nuclear genome mutagens, cadmium and Aflatoxin B1, on germline mtDNA mutagenesis in Caenorhabditis elegans. Detection of thousands of mtDNA mutations revealed pervasive heteroplasmy in C. elegans and that mtDNA mutagenesis is dominated by C:G → A:T mutations generally attributed to oxidative damage. However, there was no effect of either exposure on mtDNA mutation frequency, spectrum, or trinucleotide context signature despite a significant increase in nuclear mutation rate after aflatoxin B1 exposure. Mitophagy-deficient mutants pink-1 and dct-1 accumulated significantly higher levels of mtDNA damage compared to wild-type C. elegans after exposures. However, there were only small differences in mtDNA mutation frequency, spectrum, or trinucleotide context signature compared to wild-type after 3050 generations, across all treatments. These findings suggest mitochondria harbor additional previously uncharacterized mechanisms that regulate mtDNA mutational processes across generations.
Collapse
Affiliation(s)
- Tess C Leuthner
- Nicholas School of the Environment, Duke University, Durham, NC 27708, USA
| | - Laura Benzing
- Nicholas School of the Environment, Duke University, Durham, NC 27708, USA
| | - Brendan F Kohrn
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA 98195, USA
| | | | - Michael J Hipp
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA 98195, USA
| | | | - Danielle F Mello
- Nicholas School of the Environment, Duke University, Durham, NC 27708, USA
| | - Tymofii Sokolskyi
- Nicholas School of the Environment, Duke University, Durham, NC 27708, USA
| | - Kevin Stevenson
- Duke Molecular Physiology Institute, Duke University, Durham, NC 27701, USA
| | - Ilaria R Merutka
- Nicholas School of the Environment, Duke University, Durham, NC 27708, USA
| | - Sarah A Seay
- Nicholas School of the Environment, Duke University, Durham, NC 27708, USA
| | - Simon G Gregory
- Duke Molecular Physiology Institute, Duke University, Durham, NC 27701, USA,Department of Neurology, Duke University, Durham, NC 27708, USA
| | - Scott R Kennedy
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA 98195, USA
| | - Joel N Meyer
- To whom correspondence should be addressed. Tel: +1 919 613 8109;
| |
Collapse
|
14
|
Wei P, Gao Y, Zhang J, Lin J, Liu H, Chen K, Lin W, Wang X, Wang C, Liu C. Diagnosis of lung squamous cell carcinoma based on metagenomic Next-Generation Sequencing. BMC Pulm Med 2022; 22:108. [PMID: 35346137 PMCID: PMC8958490 DOI: 10.1186/s12890-022-01894-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Accepted: 03/15/2022] [Indexed: 11/10/2022] Open
Abstract
Background The clinical treatment of patients suspected of pulmonary infections often rely on empirical antibiotics. However, preliminary diagnoses were based on clinical manifestations and conventional microbiological tests, which could later be proved wrong. In this case, we presented a patient whose initial diagnosis was lung abscess, but antibiotic treatments had no effect, and metagenomic Next-Generation Sequencing (mNGS) indicated presence of neoplasm.
Case presentation A 62-year-old female was diagnosed with lung abscess at three different health facilities. However, mNGS of bronchoalveolar lavage fluid did not support pulmonary infections. Rather, the copy number variation analysis using host DNA sequences suggested neoplasm. Using H&E staining and immunohistochemistry of lung biopsy, the patient was eventually diagnosed with lung squamous cell carcinoma. Conclusions mNGS not only detects pathogens and helps diagnose infectious diseases, but also has potential in detecting neoplasm via host chromosomal copy number analysis. This might be beneficial for febrile patients with unknown or complex etiology, especially when infectious diseases were initially suspected but empirical antibiotic regimen failed.
Collapse
Affiliation(s)
- Ping Wei
- Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, Fujian, China.,The Second Affiliated Hospital of Fujian Traditional Chinese Medical University, Fuzhou, 350003, Fujian, China
| | - Yang Gao
- School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - Jing Zhang
- The Second Affiliated Hospital of Fujian Traditional Chinese Medical University, Fuzhou, 350003, Fujian, China
| | - Jianlong Lin
- The Second Affiliated Hospital of Fujian Traditional Chinese Medical University, Fuzhou, 350003, Fujian, China
| | - Huibin Liu
- The Second Affiliated Hospital of Fujian Traditional Chinese Medical University, Fuzhou, 350003, Fujian, China
| | - Keqiang Chen
- The Second Affiliated Hospital of Fujian Traditional Chinese Medical University, Fuzhou, 350003, Fujian, China
| | - Weikai Lin
- The Second Affiliated Hospital of Fujian Traditional Chinese Medical University, Fuzhou, 350003, Fujian, China
| | - Xiaojia Wang
- Hangzhou Matridx Biotechnology Co., Ltd, Bd 2-4, 2073 Jinchang Rd, Hangzhou, 311100, Zhejiang, China
| | - Chune Wang
- The Second Affiliated Hospital of Fujian Traditional Chinese Medical University, Fuzhou, 350003, Fujian, China. .,Director of Respiratory Department, The Second Affiliated Hospital of Fujian Traditional Chinese Medical University, Fuzhou, 350108, Fujian, China.
| | - Chao Liu
- Hangzhou Matridx Biotechnology Co., Ltd, Bd 2-4, 2073 Jinchang Rd, Hangzhou, 311100, Zhejiang, China. .,Director of Medical Department, Hangzhou Matridx Biotechnology Co., Ltd, Hangzhou, 311100, China.
| |
Collapse
|
15
|
Mihajlovic M, Vinken M. Mitochondria as the Target of Hepatotoxicity and Drug-Induced Liver Injury: Molecular Mechanisms and Detection Methods. Int J Mol Sci 2022; 23:ijms23063315. [PMID: 35328737 PMCID: PMC8951158 DOI: 10.3390/ijms23063315] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 03/16/2022] [Accepted: 03/17/2022] [Indexed: 12/12/2022] Open
Abstract
One of the major mechanisms of drug-induced liver injury includes mitochondrial perturbation and dysfunction. This is not a surprise, given that mitochondria are essential organelles in most cells, which are responsible for energy homeostasis and the regulation of cellular metabolism. Drug-induced mitochondrial dysfunction can be influenced by various factors and conditions, such as genetic predisposition, the presence of metabolic disorders and obesity, viral infections, as well as drugs. Despite the fact that many methods have been developed for studying mitochondrial function, there is still a need for advanced and integrative models and approaches more closely resembling liver physiology, which would take into account predisposing factors. This could reduce the costs of drug development by the early prediction of potential mitochondrial toxicity during pre-clinical tests and, especially, prevent serious complications observed in clinical settings.
Collapse
|
16
|
Shin J, Hong SG, Choi SY, Rath ME, Saredy J, Jovin DG, Sayoc J, Park HS, Eguchi S, Rizzo V, Scalia R, Wang H, Houser SR, Park JY. Flow-induced endothelial mitochondrial remodeling mitigates mitochondrial reactive oxygen species production and promotes mitochondrial DNA integrity in a p53-dependent manner. Redox Biol 2022; 50:102252. [PMID: 35121402 PMCID: PMC8818582 DOI: 10.1016/j.redox.2022.102252] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 01/25/2022] [Accepted: 01/26/2022] [Indexed: 12/12/2022] Open
Abstract
Tumor suppressor p53 plays a pivotal role in orchestrating mitochondrial remodeling by regulating their content, fusion/fission processes, and intracellular signaling molecules that are associated with mitophagy and apoptosis pathways. In order to determine a molecular mechanism underlying flow-mediated mitochondrial remodeling in endothelial cells, we examined, herein, the role of p53 on mitochondrial adaptations to physiological flow and its relevance to vascular function using endothelial cell-specific p53 deficient mice. We observed no changes in aerobic capacity, basal blood pressure, or endothelial mitochondrial phenotypes in the endothelial p53 mull animals. However, after 7 weeks of voluntary wheel running exercise, blood pressure reduction and endothelial mitochondrial remodeling (biogenesis, elongation, and mtDNA replication) were substantially blunted in endothelial p53 null animals compared to the wild-type, subjected to angiotensin II-induced hypertension. In addition, endothelial mtDNA lesions were significantly reduced following voluntary running exercise in wild-type mice, but not in the endothelial p53 null mice. Moreover, in vitro studies demonstrated that unidirectional laminar flow exposure significantly increased key putative regulators for mitochondrial remodeling and reduced mitochondrial reactive oxygen species generation and mtDNA damage in a p53-dependent manner. Mechanistically, unidirectional laminar flow instigated translocalization of p53 into the mitochondrial matrix where it binds to mitochondrial transcription factor A, TFAM, resulting in improving mtDNA integrity. Taken together, our findings suggest that p53 plays an integral role in mitochondrial remodeling under physiological flow condition and the flow-induced p53-TFAM axis may be a novel molecular intersection for enhancing mitochondrial homeostasis in endothelial cells.
Collapse
|
17
|
Abdullaev SA, Glukhov SI, Gaziev AI. Radioprotective and Radiomitigative Effects of Melatonin in Tissues with Different Proliferative Activity. Antioxidants (Basel) 2021; 10:1885. [PMID: 34942988 PMCID: PMC8698738 DOI: 10.3390/antiox10121885] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 11/18/2021] [Accepted: 11/24/2021] [Indexed: 12/13/2022] Open
Abstract
We used various markers to analyze damage to mouse tissues (spleen and cerebral cortex) which have different proliferative activity and sensitivity to ionizing radiation (IR). We also assessed the degree of modulation of damages that occurs when melatonin is administered to mice prior to and after their X-ray irradiation. The data from this study showed that lesions in nuclear DNA (nDNA) were repaired more actively in the spleen than in the cerebral cortex of mice irradiated and treated with melatonin (N-acetyl-5-methoxytryptamine). Mitochondrial biogenesis involving mitochondrial DNA (mtDNA) synthesis was activated in both tissues of irradiated mice. A significant proportion of the newly synthesized mtDNA molecules were mutant copies that increase oxidative stress. Melatonin reduced the number of mutant mtDNA copies and the level of H2O2 in both tissues of the irradiated mice. Melatonin promoted the restoration of ATP levels in the tissues of irradiated mice. In the mouse tissues after exposure to X-ray, the level of malondialdehyde (MDA) increased and melatonin was able to reduce it. The MDA concentration was higher in the cerebral cortex tissue than that in the spleen tissue of the mouse. In mouse tissues following irradiation, the glutathione (GSH) level was low. The spleen GSH content was more than twice as low as that in the cerebral cortex. Melatonin helped restore the GSH levels in the mouse tissues. Although the spleen and cerebral cortex tissues of mice differ in the baseline values of the analyzed markers, the radioprotective and radiomitigative potential of melatonin was observed in both tissues.
Collapse
Affiliation(s)
- Serazhutdin A. Abdullaev
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino, 142290 Moscow Region, Russia; (S.I.G.); (A.I.G.)
| | | | | |
Collapse
|
18
|
Saccharomyces cerevisiae as a Tool for Studying Mutations in Nuclear Genes Involved in Diseases Caused by Mitochondrial DNA Instability. Genes (Basel) 2021; 12:genes12121866. [PMID: 34946817 PMCID: PMC8701800 DOI: 10.3390/genes12121866] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 11/20/2021] [Accepted: 11/23/2021] [Indexed: 01/03/2023] Open
Abstract
Mitochondrial DNA (mtDNA) maintenance is critical for oxidative phosphorylation (OXPHOS) since some subunits of the respiratory chain complexes are mitochondrially encoded. Pathological mutations in nuclear genes involved in the mtDNA metabolism may result in a quantitative decrease in mtDNA levels, referred to as mtDNA depletion, or in qualitative defects in mtDNA, especially in multiple deletions. Since, in the last decade, most of the novel mutations have been identified through whole-exome sequencing, it is crucial to confirm the pathogenicity by functional analysis in the appropriate model systems. Among these, the yeast Saccharomyces cerevisiae has proved to be a good model for studying mutations associated with mtDNA instability. This review focuses on the use of yeast for evaluating the pathogenicity of mutations in six genes, MPV17/SYM1, MRM2/MRM2, OPA1/MGM1, POLG/MIP1, RRM2B/RNR2, and SLC25A4/AAC2, all associated with mtDNA depletion or multiple deletions. We highlight the techniques used to construct a specific model and to measure the mtDNA instability as well as the main results obtained. We then report the contribution that yeast has given in understanding the pathogenic mechanisms of the mutant variants, in finding the genetic suppressors of the mitochondrial defects and in the discovery of molecules able to improve the mtDNA stability.
Collapse
|
19
|
Ryan KC, Ashkavand Z, Sarasija S, Laboy JT, Samarakoon R, Norman KR. Increased mitochondrial calcium uptake and concomitant mitochondrial activity by presenilin loss promotes mTORC1 signaling to drive neurodegeneration. Aging Cell 2021; 20:e13472. [PMID: 34499406 PMCID: PMC8520713 DOI: 10.1111/acel.13472] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 06/21/2021] [Accepted: 08/05/2021] [Indexed: 12/21/2022] Open
Abstract
Metabolic dysfunction and protein aggregation are common characteristics that occur in age‐related neurodegenerative disease. However, the mechanisms underlying these abnormalities remain poorly understood. We have found that mutations in the gene encoding presenilin in Caenorhabditis elegans, sel‐12, results in elevated mitochondrial activity that drives oxidative stress and neuronal dysfunction. Mutations in the human presenilin genes are the primary cause of familial Alzheimer's disease. Here, we demonstrate that loss of SEL‐12/presenilin results in the hyperactivation of the mTORC1 pathway. This hyperactivation is caused by elevated mitochondrial calcium influx and, likely, the associated increase in mitochondrial activity. Reducing mTORC1 activity improves proteostasis defects and neurodegenerative phenotypes associated with loss of SEL‐12 function. Consistent with high mTORC1 activity, we find that SEL‐12 loss reduces autophagosome formation, and this reduction is prevented by limiting mitochondrial calcium uptake. Moreover, the improvements of proteostasis and neuronal defects in sel‐12 mutants due to mTORC1 inhibition require the induction of autophagy. These results indicate that mTORC1 hyperactivation exacerbates the defects in proteostasis and neuronal function in sel‐12 mutants and demonstrate a critical role of presenilin in promoting neuronal health.
Collapse
Affiliation(s)
- Kerry C. Ryan
- Department of Regenerative and Cancer Cell Biology Albany Medical College Albany New York USA
| | - Zahra Ashkavand
- Department of Regenerative and Cancer Cell Biology Albany Medical College Albany New York USA
| | - Shaarika Sarasija
- Department of Regenerative and Cancer Cell Biology Albany Medical College Albany New York USA
| | - Jocelyn T. Laboy
- Department of Regenerative and Cancer Cell Biology Albany Medical College Albany New York USA
| | - Rohan Samarakoon
- Department of Regenerative and Cancer Cell Biology Albany Medical College Albany New York USA
| | - Kenneth R. Norman
- Department of Regenerative and Cancer Cell Biology Albany Medical College Albany New York USA
| |
Collapse
|
20
|
SenGupta T, Palikaras K, Esbensen YQ, Konstantinidis G, Galindo FJN, Achanta K, Kassahun H, Stavgiannoudaki I, Bohr VA, Akbari M, Gaare J, Tzoulis C, Tavernarakis N, Nilsen H. Base excision repair causes age-dependent accumulation of single-stranded DNA breaks that contribute to Parkinson disease pathology. Cell Rep 2021; 36:109668. [PMID: 34496255 PMCID: PMC8441048 DOI: 10.1016/j.celrep.2021.109668] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 05/26/2021] [Accepted: 08/13/2021] [Indexed: 01/24/2023] Open
Abstract
Aging, genomic stress, and mitochondrial dysfunction are risk factors for neurodegenerative pathologies, such as Parkinson disease (PD). Although genomic instability is associated with aging and mitochondrial impairment, the underlying mechanisms are poorly understood. Here, we show that base excision repair generates genomic stress, promoting age-related neurodegeneration in a Caenorhabditis elegans PD model. A physiological level of NTH-1 DNA glycosylase mediates mitochondrial and nuclear genomic instability, which promote degeneration of dopaminergic neurons in older nematodes. Conversely, NTH-1 deficiency protects against α-synuclein-induced neurotoxicity, maintaining neuronal function with age. This apparent paradox is caused by modulation of mitochondrial transcription in NTH-1-deficient cells, and this modulation activates LMD-3, JNK-1, and SKN-1 and induces mitohormesis. The dependance of neuroprotection on mitochondrial transcription highlights the integration of BER and transcription regulation during physiological aging. Finally, whole-exome sequencing of genomic DNA from patients with idiopathic PD suggests that base excision repair might modulate susceptibility to PD in humans. Incomplete base excision repair is a source of genomic stress during aging The NTH-1 DNA glycosylase is a key mediator of age-dependent genomic instability Compromised NTH-1 activity promotes neuroprotection in PD nematodes NTH-1 deficiency triggers LMD-3/JNK-1/SKN-1-dependent mitohormetic response
Collapse
Affiliation(s)
- Tanima SenGupta
- Department of Clinical Molecular Biology, University of Oslo, Oslo, Norway; Department of Clinical Molecular Biology, Akershus University Hospital, Lørenskog, Norway
| | - Konstantinos Palikaras
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, Hellas, Greece; Department of Physiology, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Ying Q Esbensen
- Department of Clinical Molecular Biology, University of Oslo, Oslo, Norway; Department of Clinical Molecular Biology, Akershus University Hospital, Lørenskog, Norway
| | - Georgios Konstantinidis
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, Hellas, Greece
| | - Francisco Jose Naranjo Galindo
- Department of Clinical Molecular Biology, University of Oslo, Oslo, Norway; Department of Clinical Molecular Biology, Akershus University Hospital, Lørenskog, Norway
| | - Kavya Achanta
- Center for Healthy Aging, Department of Cellular and Molecular Medicine, SUND, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Henok Kassahun
- Department of Clinical Molecular Biology, University of Oslo, Oslo, Norway
| | - Ioanna Stavgiannoudaki
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, Hellas, Greece
| | - Vilhelm A Bohr
- Center for Healthy Aging, Department of Cellular and Molecular Medicine, SUND, University of Copenhagen, 2200 Copenhagen, Denmark; DNA Repair Section, National Institute on Aging, 251 Bayview Boulevard, Baltimore, MD, USA
| | - Mansour Akbari
- Center for Healthy Aging, Department of Cellular and Molecular Medicine, SUND, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Johannes Gaare
- Neuro-SysMed, Department of Neurology, Haukeland University Hospital, 5021 Bergen, Norway; Department of Clinical Medicine, University of Bergen, Pb 7804, 5020 Bergen, Norway
| | - Charalampos Tzoulis
- Neuro-SysMed, Department of Neurology, Haukeland University Hospital, 5021 Bergen, Norway; Department of Clinical Medicine, University of Bergen, Pb 7804, 5020 Bergen, Norway
| | - Nektarios Tavernarakis
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, Hellas, Greece; Department of Basic Sciences, Faculty of Medicine, University of Crete, Heraklion, 70013 Crete, Greece.
| | - Hilde Nilsen
- Department of Clinical Molecular Biology, University of Oslo, Oslo, Norway; Department of Clinical Molecular Biology, Akershus University Hospital, Lørenskog, Norway.
| |
Collapse
|
21
|
Chappell NR, Zhou B, Hosseinzadeh P, Schutt A, Gibbons WE, Blesson CS. Hyperandrogenemia alters mitochondrial structure and function in the oocytes of obese mouse with polycystic ovary syndrome. ACTA ACUST UNITED AC 2021; 2:101-112. [PMID: 34458875 DOI: 10.1016/j.xfss.2020.12.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Capsule Hyperandrogenemia in an obese PCOS mouse model results in altered glucose/insulin metabolism and mitochondrial structure and function in the oocytes, in part explaining adverse outcomes and inheritance patterns seen in PCOS. Objective To study the oocyte quality by means of mitochondrial structure and function in a well-established classic PCOS mouse model. Design Animal study using an obese PCOS mouse model compared with control. Setting Animal research facility in a tertiary care university hospital setting. Animals C57/B6J mice. Intervention Three week old mice had subdermal implants of DHT controlled release pellet or placebo for 90 days. Main Outcome Measures The mouse model was validated by performing glucose tolerance test, HbA1c levels, body weight and estrous cycle analyses. Oocytes were subsequently isolated and were used to investigate mitochondrial membrane potential, oxidative stress, lipid peroxidation, ATP production, mtDNA copy number, transcript abundance, histology and electron microscopy. Results Results showed glucose intolerance and hyperinsulinemia along with dysregulated estrus cycle. Analysis of the oocytes demonstrated impaired inner mitochondrial membrane function, increased ATP production and mtDNA copy number, altered RNA transcript abundance and aberrant ovarian histology. Electron microscopy of the oocytes showed severely impaired mitochondrial ultrastructure. Conclusion The obese PCOS mouse model shows a decreased oocyte quality related to impaired mitochondrial function.
Collapse
Affiliation(s)
- Neil R Chappell
- Reproductive Endocrinology and Infertility Division, Department of Obstetrics and Gynecology, Baylor College of Medicine and Family Fertility Center, Texas Children's Hospital, Houston, Texas 77030, USA
| | - Beth Zhou
- Reproductive Endocrinology and Infertility Division, Department of Obstetrics and Gynecology, Baylor College of Medicine and Family Fertility Center, Texas Children's Hospital, Houston, Texas 77030, USA
| | - Pardis Hosseinzadeh
- Reproductive Endocrinology and Infertility Division, Department of Obstetrics and Gynecology, Baylor College of Medicine and Family Fertility Center, Texas Children's Hospital, Houston, Texas 77030, USA
| | - Amy Schutt
- Reproductive Endocrinology and Infertility Division, Department of Obstetrics and Gynecology, Baylor College of Medicine and Family Fertility Center, Texas Children's Hospital, Houston, Texas 77030, USA
| | - William E Gibbons
- Reproductive Endocrinology and Infertility Division, Department of Obstetrics and Gynecology, Baylor College of Medicine and Family Fertility Center, Texas Children's Hospital, Houston, Texas 77030, USA
| | - Chellakkan S Blesson
- Reproductive Endocrinology and Infertility Division, Department of Obstetrics and Gynecology, Baylor College of Medicine and Family Fertility Center, Texas Children's Hospital, Houston, Texas 77030, USA
| |
Collapse
|
22
|
Hershberger KA, Rooney JP, Turner EA, Donoghue LJ, Bodhicharla R, Maurer LL, Ryde IT, Kim JJ, Joglekar R, Hibshman JD, Smith LL, Bhatt DP, Ilkayeva OR, Hirschey MD, Meyer JN. Early-life mitochondrial DNA damage results in lifelong deficits in energy production mediated by redox signaling in Caenorhabditis elegans. Redox Biol 2021; 43:102000. [PMID: 33993056 PMCID: PMC8134077 DOI: 10.1016/j.redox.2021.102000] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 04/19/2021] [Accepted: 04/28/2021] [Indexed: 11/12/2022] Open
Abstract
The consequences of damage to the mitochondrial genome (mtDNA) are poorly understood, although mtDNA is more susceptible to damage resulting from some genotoxicants than nuclear DNA (nucDNA), and many environmental toxicants target the mitochondria. Reports from the toxicological literature suggest that exposure to early-life mitochondrial damage could lead to deleterious consequences later in life (the “Developmental Origins of Health and Disease” paradigm), but reports from other fields often report beneficial (“mitohormetic”) responses to such damage. Here, we tested the effects of low (causing no change in lifespan) levels of ultraviolet C (UVC)-induced, irreparable mtDNA damage during early development in Caenorhabditis elegans. This exposure led to life-long reductions in mtDNA copy number and steady-state ATP levels, accompanied by increased oxygen consumption and altered metabolite profiles, suggesting inefficient mitochondrial function. Exposed nematodes were also developmentally delayed, reached smaller adult size, and were rendered more susceptible to subsequent exposure to chemical mitotoxicants. Metabolomic and genetic analysis of key signaling and metabolic pathways supported redox and mitochondrial stress-response signaling during early development as a mechanism for establishing these persistent alterations. Our results highlight the importance of early-life exposures to environmental pollutants, especially in the context of exposure to chemicals that target mitochondria. Early life mtDNA damage led to lifelong deficits in mitochondrial function. C. elegans developed slowly and were sensitive to chemical exposures as adults. Redox signaling is a mechanism that establishes these persistent alterations. Data are consistent with the Developmental Origins of Health and Disease model.
Collapse
Affiliation(s)
- Kathleen A Hershberger
- Duke University, Nicholas School of the Environment, Integrated Toxicology and Environmental Health Program, Durham, NC, USA
| | - John P Rooney
- Duke University, Nicholas School of the Environment, Integrated Toxicology and Environmental Health Program, Durham, NC, USA
| | - Elena A Turner
- Duke University, Nicholas School of the Environment, Integrated Toxicology and Environmental Health Program, Durham, NC, USA
| | - Lauren J Donoghue
- Duke University, Nicholas School of the Environment, Integrated Toxicology and Environmental Health Program, Durham, NC, USA
| | - Rakesh Bodhicharla
- Duke University, Nicholas School of the Environment, Integrated Toxicology and Environmental Health Program, Durham, NC, USA
| | - Laura L Maurer
- Duke University, Nicholas School of the Environment, Integrated Toxicology and Environmental Health Program, Durham, NC, USA
| | - Ian T Ryde
- Duke University, Nicholas School of the Environment, Integrated Toxicology and Environmental Health Program, Durham, NC, USA
| | - Jina J Kim
- Duke University, Nicholas School of the Environment, Integrated Toxicology and Environmental Health Program, Durham, NC, USA
| | - Rashmi Joglekar
- Duke University, Nicholas School of the Environment, Integrated Toxicology and Environmental Health Program, Durham, NC, USA
| | - Jonathan D Hibshman
- Duke University Department of Biology and University Program in Genetics and Genomics, Durham, NC, USA
| | - Latasha L Smith
- Duke University, Nicholas School of the Environment, Integrated Toxicology and Environmental Health Program, Durham, NC, USA
| | | | | | | | - Joel N Meyer
- Duke University, Nicholas School of the Environment, Integrated Toxicology and Environmental Health Program, Durham, NC, USA.
| |
Collapse
|
23
|
Stuani L, Sabatier M, Saland E, Cognet G, Poupin N, Bosc C, Castelli FA, Gales L, Turtoi E, Montersino C, Farge T, Boet E, Broin N, Larrue C, Baran N, Cissé MY, Conti M, Loric S, Kaoma T, Hucteau A, Zavoriti A, Sahal A, Mouchel PL, Gotanègre M, Cassan C, Fernando L, Wang F, Hosseini M, Chu-Van E, Le Cam L, Carroll M, Selak MA, Vey N, Castellano R, Fenaille F, Turtoi A, Cazals G, Bories P, Gibon Y, Nicolay B, Ronseaux S, Marszalek JR, Takahashi K, DiNardo CD, Konopleva M, Pancaldi V, Collette Y, Bellvert F, Jourdan F, Linares LK, Récher C, Portais JC, Sarry JE. Mitochondrial metabolism supports resistance to IDH mutant inhibitors in acute myeloid leukemia. J Exp Med 2021; 218:e20200924. [PMID: 33760042 PMCID: PMC7995203 DOI: 10.1084/jem.20200924] [Citation(s) in RCA: 71] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 11/25/2020] [Accepted: 01/11/2021] [Indexed: 12/17/2022] Open
Abstract
Mutations in IDH induce epigenetic and transcriptional reprogramming, differentiation bias, and susceptibility to mitochondrial inhibitors in cancer cells. Here, we first show that cell lines, PDXs, and patients with acute myeloid leukemia (AML) harboring an IDH mutation displayed an enhanced mitochondrial oxidative metabolism. Along with an increase in TCA cycle intermediates, this AML-specific metabolic behavior mechanistically occurred through the increase in electron transport chain complex I activity, mitochondrial respiration, and methylation-driven CEBPα-induced fatty acid β-oxidation of IDH1 mutant cells. While IDH1 mutant inhibitor reduced 2-HG oncometabolite and CEBPα methylation, it failed to reverse FAO and OxPHOS. These mitochondrial activities were maintained through the inhibition of Akt and enhanced activation of peroxisome proliferator-activated receptor-γ coactivator-1 PGC1α upon IDH1 mutant inhibitor. Accordingly, OxPHOS inhibitors improved anti-AML efficacy of IDH mutant inhibitors in vivo. This work provides a scientific rationale for combinatory mitochondrial-targeted therapies to treat IDH mutant AML patients, especially those unresponsive to or relapsing from IDH mutant inhibitors.
Collapse
MESH Headings
- Acute Disease
- Aminopyridines/pharmacology
- Animals
- Cell Line, Tumor
- Doxycycline/pharmacology
- Drug Resistance, Neoplasm/drug effects
- Drug Resistance, Neoplasm/genetics
- Enzyme Inhibitors/pharmacology
- Epigenesis, Genetic/drug effects
- Glycine/analogs & derivatives
- Glycine/pharmacology
- HL-60 Cells
- Humans
- Isocitrate Dehydrogenase/antagonists & inhibitors
- Isocitrate Dehydrogenase/genetics
- Isocitrate Dehydrogenase/metabolism
- Isoenzymes/antagonists & inhibitors
- Isoenzymes/genetics
- Isoenzymes/metabolism
- Leukemia, Myeloid/drug therapy
- Leukemia, Myeloid/genetics
- Leukemia, Myeloid/metabolism
- Mice, Inbred NOD
- Mice, Knockout
- Mice, SCID
- Mitochondria/drug effects
- Mitochondria/genetics
- Mitochondria/metabolism
- Mutation
- Oxadiazoles/pharmacology
- Oxidative Phosphorylation/drug effects
- Piperidines/pharmacology
- Pyridines/pharmacology
- Triazines/pharmacology
- Xenograft Model Antitumor Assays/methods
- Mice
Collapse
Affiliation(s)
- Lucille Stuani
- Centre de Recherches en Cancérologie de Toulouse, Université de Toulouse, Institut National de la Santé et de la Recherché Médicale, Centre National de la Recherche Scientifique, Toulouse, France
- LabEx Toucan, Toulouse, France
- Equipe Labellisée Ligue Nationale Contre le Cancer 2018, Toulouse, France
| | - Marie Sabatier
- Centre de Recherches en Cancérologie de Toulouse, Université de Toulouse, Institut National de la Santé et de la Recherché Médicale, Centre National de la Recherche Scientifique, Toulouse, France
- LabEx Toucan, Toulouse, France
- Equipe Labellisée Ligue Nationale Contre le Cancer 2018, Toulouse, France
| | - Estelle Saland
- Centre de Recherches en Cancérologie de Toulouse, Université de Toulouse, Institut National de la Santé et de la Recherché Médicale, Centre National de la Recherche Scientifique, Toulouse, France
- LabEx Toucan, Toulouse, France
- Equipe Labellisée Ligue Nationale Contre le Cancer 2018, Toulouse, France
| | - Guillaume Cognet
- Centre de Recherches en Cancérologie de Toulouse, Université de Toulouse, Institut National de la Santé et de la Recherché Médicale, Centre National de la Recherche Scientifique, Toulouse, France
- LabEx Toucan, Toulouse, France
- Equipe Labellisée Ligue Nationale Contre le Cancer 2018, Toulouse, France
| | - Nathalie Poupin
- UMR1331 Toxalim, Université de Toulouse, Institut National de la Recherche pour l’Agriculture, l’Alimentation et l’Environnement, Ecole Nationale Vétérinaire de Toulouse, INP-Purpan, Université Paul Sabatier, Toulouse, France
| | - Claudie Bosc
- Centre de Recherches en Cancérologie de Toulouse, Université de Toulouse, Institut National de la Santé et de la Recherché Médicale, Centre National de la Recherche Scientifique, Toulouse, France
- LabEx Toucan, Toulouse, France
- Equipe Labellisée Ligue Nationale Contre le Cancer 2018, Toulouse, France
| | - Florence A. Castelli
- CEA/DSV/iBiTec-S/SPI, Laboratoire d’Etude du Métabolisme des Médicaments, MetaboHUB-Paris, Gif-sur-Yvette, France
| | - Lara Gales
- Toulouse Biotechnology Institute, Université de Toulouse, Centre National de la Recherche Scientifique, Institut National de la Recherche Agronomique, Institut National des sciences appliquées, Toulouse, France
- MetaToul-MetaboHUB, National Infrastructure of Metabolomics and Fluxomics, Toulouse, France
| | - Evgenia Turtoi
- Institut de Recherche en Cancérologie de Montpellier, Institut National de la Santé et de la Recherché Médicale, Université de Montpellier, Institut Régional du Cancer Montpellier, Montpellier, France
- Montpellier Alliance for Metabolomics and Metabolism Analysis, Platform for Translational Oncometabolomics, Biocampus, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherché Médicale, Université de Montpellier, Montpellier, France
| | - Camille Montersino
- Aix-Marseille University, Institut National de la Santé et de la Recherché Médicale, Centre National de la Recherche Scientifique, Institut Paoli-Calmettes, Centre de Recherches en Cancérologie de Marseille, Marseille, France
| | - Thomas Farge
- Centre de Recherches en Cancérologie de Toulouse, Université de Toulouse, Institut National de la Santé et de la Recherché Médicale, Centre National de la Recherche Scientifique, Toulouse, France
- LabEx Toucan, Toulouse, France
- Equipe Labellisée Ligue Nationale Contre le Cancer 2018, Toulouse, France
| | - Emeline Boet
- Centre de Recherches en Cancérologie de Toulouse, Université de Toulouse, Institut National de la Santé et de la Recherché Médicale, Centre National de la Recherche Scientifique, Toulouse, France
- LabEx Toucan, Toulouse, France
- Equipe Labellisée Ligue Nationale Contre le Cancer 2018, Toulouse, France
| | - Nicolas Broin
- Centre de Recherches en Cancérologie de Toulouse, Université de Toulouse, Institut National de la Santé et de la Recherché Médicale, Centre National de la Recherche Scientifique, Toulouse, France
- LabEx Toucan, Toulouse, France
- Equipe Labellisée Ligue Nationale Contre le Cancer 2018, Toulouse, France
| | - Clément Larrue
- Centre de Recherches en Cancérologie de Toulouse, Université de Toulouse, Institut National de la Santé et de la Recherché Médicale, Centre National de la Recherche Scientifique, Toulouse, France
- LabEx Toucan, Toulouse, France
- Equipe Labellisée Ligue Nationale Contre le Cancer 2018, Toulouse, France
| | - Natalia Baran
- Departments of Leukemia and Genomic Medicine, The University of Texas, MD Anderson Cancer Center, Houston, TX
| | - Madi Y. Cissé
- Institut de Recherche en Cancérologie de Montpellier, Institut National de la Santé et de la Recherché Médicale, Université de Montpellier, Institut Régional du Cancer Montpellier, Montpellier, France
| | - Marc Conti
- Institut National de la Santé et de la Recherché Médicale U938, Hôpital St Antoine, Paris, France
- Integracell, Longjumeau, France
| | - Sylvain Loric
- Institut National de la Santé et de la Recherché Médicale U938, Hôpital St Antoine, Paris, France
| | - Tony Kaoma
- Proteome and Genome Research Unit, Department of Oncology, Luxembourg Institute of Health, Strassen, Luxembourg
| | - Alexis Hucteau
- Centre de Recherches en Cancérologie de Toulouse, Université de Toulouse, Institut National de la Santé et de la Recherché Médicale, Centre National de la Recherche Scientifique, Toulouse, France
- LabEx Toucan, Toulouse, France
- Equipe Labellisée Ligue Nationale Contre le Cancer 2018, Toulouse, France
| | - Aliki Zavoriti
- Centre de Recherches en Cancérologie de Toulouse, Université de Toulouse, Institut National de la Santé et de la Recherché Médicale, Centre National de la Recherche Scientifique, Toulouse, France
- LabEx Toucan, Toulouse, France
- Equipe Labellisée Ligue Nationale Contre le Cancer 2018, Toulouse, France
| | - Ambrine Sahal
- Centre de Recherches en Cancérologie de Toulouse, Université de Toulouse, Institut National de la Santé et de la Recherché Médicale, Centre National de la Recherche Scientifique, Toulouse, France
- LabEx Toucan, Toulouse, France
- Equipe Labellisée Ligue Nationale Contre le Cancer 2018, Toulouse, France
| | - Pierre-Luc Mouchel
- Centre de Recherches en Cancérologie de Toulouse, Université de Toulouse, Institut National de la Santé et de la Recherché Médicale, Centre National de la Recherche Scientifique, Toulouse, France
- LabEx Toucan, Toulouse, France
- Equipe Labellisée Ligue Nationale Contre le Cancer 2018, Toulouse, France
- Service d'Hématologie, Institut Universitaire du Cancer de Toulouse-Oncopole, CHU de Toulouse, Toulouse, France
| | - Mathilde Gotanègre
- Centre de Recherches en Cancérologie de Toulouse, Université de Toulouse, Institut National de la Santé et de la Recherché Médicale, Centre National de la Recherche Scientifique, Toulouse, France
- LabEx Toucan, Toulouse, France
- Equipe Labellisée Ligue Nationale Contre le Cancer 2018, Toulouse, France
| | - Cédric Cassan
- UMR1332 Biologie du Fruit et Pathologie, Plateforme Métabolome Bordeaux, Institut National de la Recherche Agronomique, Université de Bordeaux, Villenave d'Ornon, France
| | - Laurent Fernando
- UMR1331 Toxalim, Université de Toulouse, Institut National de la Recherche pour l’Agriculture, l’Alimentation et l’Environnement, Ecole Nationale Vétérinaire de Toulouse, INP-Purpan, Université Paul Sabatier, Toulouse, France
| | - Feng Wang
- Departments of Leukemia and Genomic Medicine, The University of Texas, MD Anderson Cancer Center, Houston, TX
| | - Mohsen Hosseini
- Centre de Recherches en Cancérologie de Toulouse, Université de Toulouse, Institut National de la Santé et de la Recherché Médicale, Centre National de la Recherche Scientifique, Toulouse, France
- LabEx Toucan, Toulouse, France
- Equipe Labellisée Ligue Nationale Contre le Cancer 2018, Toulouse, France
| | - Emeline Chu-Van
- CEA/DSV/iBiTec-S/SPI, Laboratoire d’Etude du Métabolisme des Médicaments, MetaboHUB-Paris, Gif-sur-Yvette, France
| | - Laurent Le Cam
- Institut de Recherche en Cancérologie de Montpellier, Institut National de la Santé et de la Recherché Médicale, Université de Montpellier, Institut Régional du Cancer Montpellier, Montpellier, France
| | - Martin Carroll
- Division of Hematology and Oncology, Department of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Mary A. Selak
- Division of Hematology and Oncology, Department of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Norbert Vey
- Aix-Marseille University, Institut National de la Santé et de la Recherché Médicale, Centre National de la Recherche Scientifique, Institut Paoli-Calmettes, Centre de Recherches en Cancérologie de Marseille, Marseille, France
| | - Rémy Castellano
- Aix-Marseille University, Institut National de la Santé et de la Recherché Médicale, Centre National de la Recherche Scientifique, Institut Paoli-Calmettes, Centre de Recherches en Cancérologie de Marseille, Marseille, France
| | - François Fenaille
- CEA/DSV/iBiTec-S/SPI, Laboratoire d’Etude du Métabolisme des Médicaments, MetaboHUB-Paris, Gif-sur-Yvette, France
| | - Andrei Turtoi
- Institut de Recherche en Cancérologie de Montpellier, Institut National de la Santé et de la Recherché Médicale, Université de Montpellier, Institut Régional du Cancer Montpellier, Montpellier, France
| | - Guillaume Cazals
- Laboratoire de Mesures Physiques, Université de Montpellier, Montpellier, France
| | - Pierre Bories
- Réseau Régional de Cancérologie Onco-Occitanie, Toulouse, France
| | - Yves Gibon
- UMR1332 Biologie du Fruit et Pathologie, Plateforme Métabolome Bordeaux, Institut National de la Recherche Agronomique, Université de Bordeaux, Villenave d'Ornon, France
| | | | | | - Joseph R. Marszalek
- Departments of Leukemia and Genomic Medicine, The University of Texas, MD Anderson Cancer Center, Houston, TX
| | - Koichi Takahashi
- Departments of Leukemia and Genomic Medicine, The University of Texas, MD Anderson Cancer Center, Houston, TX
| | - Courtney D. DiNardo
- Departments of Leukemia and Genomic Medicine, The University of Texas, MD Anderson Cancer Center, Houston, TX
| | - Marina Konopleva
- Departments of Leukemia and Genomic Medicine, The University of Texas, MD Anderson Cancer Center, Houston, TX
| | - Véra Pancaldi
- Centre de Recherches en Cancérologie de Toulouse, Université de Toulouse, Institut National de la Santé et de la Recherché Médicale, Centre National de la Recherche Scientifique, Toulouse, France
- Barcelona Supercomputing Center, Barcelona, Spain
| | - Yves Collette
- Aix-Marseille University, Institut National de la Santé et de la Recherché Médicale, Centre National de la Recherche Scientifique, Institut Paoli-Calmettes, Centre de Recherches en Cancérologie de Marseille, Marseille, France
| | - Floriant Bellvert
- Toulouse Biotechnology Institute, Université de Toulouse, Centre National de la Recherche Scientifique, Institut National de la Recherche Agronomique, Institut National des sciences appliquées, Toulouse, France
- MetaToul-MetaboHUB, National Infrastructure of Metabolomics and Fluxomics, Toulouse, France
| | - Fabien Jourdan
- UMR1331 Toxalim, Université de Toulouse, Institut National de la Recherche pour l’Agriculture, l’Alimentation et l’Environnement, Ecole Nationale Vétérinaire de Toulouse, INP-Purpan, Université Paul Sabatier, Toulouse, France
- MetaToul-MetaboHUB, National Infrastructure of Metabolomics and Fluxomics, Toulouse, France
| | - Laetitia K. Linares
- Institut de Recherche en Cancérologie de Montpellier, Institut National de la Santé et de la Recherché Médicale, Université de Montpellier, Institut Régional du Cancer Montpellier, Montpellier, France
| | - Christian Récher
- Centre de Recherches en Cancérologie de Toulouse, Université de Toulouse, Institut National de la Santé et de la Recherché Médicale, Centre National de la Recherche Scientifique, Toulouse, France
- LabEx Toucan, Toulouse, France
- Equipe Labellisée Ligue Nationale Contre le Cancer 2018, Toulouse, France
- Service d'Hématologie, Institut Universitaire du Cancer de Toulouse-Oncopole, CHU de Toulouse, Toulouse, France
| | - Jean-Charles Portais
- Toulouse Biotechnology Institute, Université de Toulouse, Centre National de la Recherche Scientifique, Institut National de la Recherche Agronomique, Institut National des sciences appliquées, Toulouse, France
- MetaToul-MetaboHUB, National Infrastructure of Metabolomics and Fluxomics, Toulouse, France
- STROMALab, Université de Toulouse, Institut National de la Santé et de la Recherché Médicale U1031, EFS, INP-ENVT, UPS, Toulouse, France
| | - Jean-Emmanuel Sarry
- Centre de Recherches en Cancérologie de Toulouse, Université de Toulouse, Institut National de la Santé et de la Recherché Médicale, Centre National de la Recherche Scientifique, Toulouse, France
- LabEx Toucan, Toulouse, France
- Equipe Labellisée Ligue Nationale Contre le Cancer 2018, Toulouse, France
- Centre Hospitalier Universitaire de Toulouse, Toulouse, France
| |
Collapse
|
24
|
Kennedy KM, Becker F, Hammon HM, Kuhla B. Differences in net fat oxidation, heat production, and liver mitochondrial DNA copy numbers between high and low feed-efficient dairy cows. J Dairy Sci 2021; 104:9287-9303. [PMID: 33934856 DOI: 10.3168/jds.2020-20031] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Accepted: 03/18/2021] [Indexed: 11/19/2022]
Abstract
Improving feed utilization efficiency in dairy cattle could have positive economic and environmental effects that would support the sustainability of the dairy industry. Identifying key differences in metabolism between high and low feed-efficient animals is vital to enhancing feed conversion efficiency. Therefore, our objectives were (1) to determine whether cows grouped by either high or low feed efficiency have measurable differences in net fat and carbohydrate metabolism that account for differences in heat production (HP), and if so, whether these differences also exists under conditions of feed withdrawal when the effect of feeding on HP is minimized, and (2) to determine whether the abundance of mitochondria in the liver can be related to the high or low feed-efficient groups. Ten dairy cows from a herd of 15 (parity = 2) were retrospectively grouped into either a high (H) or a low (L) feed-efficient group (n = 5 per group) based on weekly energy-corrected milk (ECM) divided by dry mater intake (DMI) from wk 4 through 30 of lactation. Livers were biopsied at wk -4, 2, and 12, and blood was sampled weekly from wk -3 to 12 relative to parturition. Blood was subset to be analyzed for the transition period (wk -3 to 3) and from wk 4 to 12. In wk 5.70 ± 0.82 (mean ± SD) postpartum (PP), cows spent 2 d in respiration chambers (RC), in which CO2, O2, and CH4 gases were measured every 6 min for 24 h. Fatty acid oxidation (FOX), carbohydrate oxidation (COX), metabolic respiratory quotient (RQ), and HP were calculated from gas measurements for 23 h. Cows were fed ad libitum (AD-LIB) on d 1 and had feed withdrawn (RES, restricted diet) on d 2. Additional blood samples were taken at the end of the AD-LIB and RES feeding periods in the RC. During wk 4 to 30 PP, H had greater DMI/kg of metabolic body weight (BW0.75), ECM per kilogram of BW0.75 yield, and ECM/DMI ratio, compared with L, but a lower body condition score between wk 4 and 12 PP. In the RC period, we detected no differences in BW, DMI, or milk yield between groups. We also detected no significant group or group by feeding period interactions for plasma metabolites except for Revised Quantitative Insulin Sensitivity Check Index, which tended to have a group by feeding period interaction. The H group had lower HP and HP per kilogram of BW0.75 compared with L. Additionally, H had lower FOX and FOX per kilogram of BW0.75 compared with L during the AD-LIB period. Methane, CH4 per kilogram of BW0.75, and CH4 per kilogram of milk yield were lower in H compared with L, but, when adjusted for DMI, CH4/DMI did not differ between groups, nor did HP/DMI. Relative mitochondrial DNA copy numbers in the liver were lower in the L than in the H group. These results suggest that lower feed efficiency in dairy cows may result from fewer mitochondria per liver cell as well as a greater whole-body HP, which likely partially results from higher net fat oxidation.
Collapse
Affiliation(s)
- K M Kennedy
- Institute for Farm Animal Biology (FBN), Institute of Nutritional Physiology "Oskar Kellner," Dummerstorf 18196, Germany
| | - F Becker
- Institute for Farm Animal Biology (FBN), Institute of Reproductive Biology, Dummerstorf 18196, Germany
| | - H M Hammon
- Institute for Farm Animal Biology (FBN), Institute of Reproductive Biology, Dummerstorf 18196, Germany
| | - B Kuhla
- Institute for Farm Animal Biology (FBN), Institute of Nutritional Physiology "Oskar Kellner," Dummerstorf 18196, Germany.
| |
Collapse
|
25
|
Sesquiterpene Lactone Deoxyelephantopin Isolated from Elephantopus scaber and Its Derivative DETD-35 Suppress BRAF V600E Mutant Melanoma Lung Metastasis in Mice. Int J Mol Sci 2021; 22:ijms22063226. [PMID: 33810045 PMCID: PMC8004649 DOI: 10.3390/ijms22063226] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 03/17/2021] [Accepted: 03/18/2021] [Indexed: 12/16/2022] Open
Abstract
Melanoma is a highly metastatic disease with an increasing rate of incidence worldwide. It is treatment refractory and has poor clinical prognosis; therefore, the development of new therapeutic agents for metastatic melanoma are urgently required. In this study, we created a lung-seeking A375LM5IF4g/Luc BRAFV600E mutant melanoma cell clone and investigated the bioefficacy of a plant sesquiterpene lactone deoxyelephantopin (DET) and its novel semi-synthetic derivative, DETD-35, in suppressing metastatic A375LM5IF4g/Luc melanoma growth in vitro and in a xenograft mouse model. DET and DETD-35 treatment inhibited A375LM5IF4g/Luc cell proliferation, and induced G2/M cell-cycle arrest and apoptosis. Furthermore, A375LM5IF4g/Luc exhibited clonogenic, metastatic and invasive abilities, and several A375LM5IF4g/Luc metastasis markers, N-cadherin, MMP2, vimentin and integrin α4 were significantly suppressed by treatment with either compound. Interestingly, DET- and DETD-35-induced Reactive Oxygen Species (ROS) generation and glutathione (GSH) depletion were found to be upstream events important for the in vitro activities, because exogenous GSH supplementation blunted DET and DETD-35 effects on A375LM5IF4g/Luc cells. DET and DETD-35 also induced mitochondrial DNA mutation, superoxide production, mitochondrial bioenergetics dysfunction, and mitochondrial protein deregulation. Most importantly, DET and DETD-35 inhibited lung metastasis of A375LM5IF4g/Luc in NOD/SCID mice through inhibiting pulmonary vascular permeability and melanoma cell (Mel-A+) proliferation, angiogenesis (VEGF+, CD31+) and EMT (N-cadherin) in the tumor microenvironment in the lungs. These findings indicate that DET and DETD-35 may be useful in the intervention of lung metastatic BRAFV600E mutant melanoma.
Collapse
|
26
|
Feric M, Demarest TG, Tian J, Croteau DL, Bohr VA, Misteli T. Self-assembly of multi-component mitochondrial nucleoids via phase separation. EMBO J 2021; 40:e107165. [PMID: 33619770 DOI: 10.15252/embj.2020107165] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 01/08/2021] [Accepted: 01/19/2021] [Indexed: 11/09/2022] Open
Abstract
Mitochondria contain an autonomous and spatially segregated genome. The organizational unit of their genome is the nucleoid, which consists of mitochondrial DNA (mtDNA) and associated architectural proteins. Here, we show that phase separation is the primary physical mechanism for assembly and size control of the mitochondrial nucleoid (mt-nucleoid). The major mtDNA-binding protein TFAM spontaneously phase separates in vitro via weak, multivalent interactions into droplets with slow internal dynamics. TFAM and mtDNA form heterogenous, viscoelastic structures in vitro, which recapitulate the dynamics and behavior of mt-nucleoids in vivo. Mt-nucleoids coalesce into larger droplets in response to various forms of cellular stress, as evidenced by the enlarged and transcriptionally active nucleoids in mitochondria from patients with the premature aging disorder Hutchinson-Gilford Progeria Syndrome (HGPS). Our results point to phase separation as an evolutionarily conserved mechanism of genome organization.
Collapse
Affiliation(s)
- Marina Feric
- National Cancer Institute, NIH, Bethesda, MD, USA.,National Institute of General Medical Sciences, NIH, Bethesda, MD, USA
| | | | - Jane Tian
- National Institute on Aging, NIH, Baltimore, MD, USA
| | | | | | - Tom Misteli
- National Cancer Institute, NIH, Bethesda, MD, USA
| |
Collapse
|
27
|
Ceccatelli Berti C, di Punzio G, Dallabona C, Baruffini E, Goffrini P, Lodi T, Donnini C. The Power of Yeast in Modelling Human Nuclear Mutations Associated with Mitochondrial Diseases. Genes (Basel) 2021; 12:300. [PMID: 33672627 PMCID: PMC7924180 DOI: 10.3390/genes12020300] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 02/16/2021] [Accepted: 02/17/2021] [Indexed: 12/17/2022] Open
Abstract
The increasing application of next generation sequencing approaches to the analysis of human exome and whole genome data has enabled the identification of novel variants and new genes involved in mitochondrial diseases. The ability of surviving in the absence of oxidative phosphorylation (OXPHOS) and mitochondrial genome makes the yeast Saccharomyces cerevisiae an excellent model system for investigating the role of these new variants in mitochondrial-related conditions and dissecting the molecular mechanisms associated with these diseases. The aim of this review was to highlight the main advantages offered by this model for the study of mitochondrial diseases, from the validation and characterisation of novel mutations to the dissection of the role played by genes in mitochondrial functionality and the discovery of potential therapeutic molecules. The review also provides a summary of the main contributions to the understanding of mitochondrial diseases emerged from the study of this simple eukaryotic organism.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Claudia Donnini
- Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parco Area delle Scienze 11/A, 43124 Parma, Italy; (C.C.B.); (G.d.P.); (C.D.); (E.B.); (P.G.); (T.L.)
| |
Collapse
|
28
|
Modulating the unfolded protein response with ONC201 to impact on radiation response in prostate cancer cells. Sci Rep 2021; 11:4252. [PMID: 33608585 PMCID: PMC7896060 DOI: 10.1038/s41598-021-83215-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Accepted: 02/01/2021] [Indexed: 01/31/2023] Open
Abstract
Prostate cancer (PCa) is the most common non-cutaneous cancer in men and a notable cause of cancer mortality when it metastasises. The unfolded protein response (UPR) can be cytoprotective but when acutely activated can lead to cell death. In this study, we sought to enhance the acute activation of the UPR using radiation and ONC201, an UPR activator. Treating PCa cells with ONC201 quickly increased the expression of all the key regulators of the UPR and reduced the oxidative phosphorylation, with cell death occurring 72 h later. We exploited this time lag to sensitize prostate cancer cells to radiation through short-term treatment with ONC201. To understand how priming occurred, we performed RNA-Seq analysis and found that ONC201 suppressed the expression of cell cycle and DNA repair factors. In conclusion, we have shown that ONC201 can prime enhanced radiation response.
Collapse
|
29
|
Leuthner TC, Hartman JH, Ryde IT, Meyer JN. PCR-Based Determination of Mitochondrial DNA Copy Number in Multiple Species. Methods Mol Biol 2021; 2310:91-111. [PMID: 34096001 DOI: 10.1007/978-1-0716-1433-4_8] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Mitochondrial DNA (mtDNA) copy number is a critical component of overall mitochondrial health. In this chapter, we describe methods for simultaneous isolation of mtDNA and nuclear DNA (nucDNA), and measurement of their respective copy numbers using quantitative PCR. Methods differ depending on the species and cell type of the starting material, and availability of specific PCR reagents. We also briefly describe factors that affect mtDNA copy number and discuss caveats to its use as a biomarker.
Collapse
Affiliation(s)
- Tess C Leuthner
- Nicholas School of the Environment, Duke University, Durham, NC, USA
| | - Jessica H Hartman
- Nicholas School of the Environment, Duke University, Durham, NC, USA
| | - Ian T Ryde
- Nicholas School of the Environment, Duke University, Durham, NC, USA
| | - Joel N Meyer
- Nicholas School of the Environment, Duke University, Durham, NC, USA.
| |
Collapse
|
30
|
Bora S, Vardhan GSH, Deka N, Khataniar L, Gogoi D, Baruah A. Paraquat exposure over generation affects lifespan and reproduction through mitochondrial disruption in C. elegans. Toxicology 2020; 447:152632. [PMID: 33197508 DOI: 10.1016/j.tox.2020.152632] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 11/06/2020] [Accepted: 11/09/2020] [Indexed: 12/22/2022]
Abstract
Paraquat (methyl viologen), is a non-selective contact herbicide and well known mitochondrial toxicant. Mitochondria are the center of cellular metabolism, and involved in the development, lifespan, and reproduction of an organism. Mitochondria are dynamic organelles that are inherited maternally through the germline and carry multiple copies of their own genome (mtDNA). It is important to understand the effects of acute and chronic stress caused by mitochondrial toxicants over multiple generations at the cellular and organism levels. Using the model nematode C. elegans, we show that acute and chronic exposure to paraquat affects reproduction, longevity, gene expression, and mitochondrial physiology. Acute exposure to paraquat in N2 (wild type) causes induction of mitochondrial unfolded protein response (mtUPR), increased expression of mitochondrial superoxide dismutase, decreased mitochondrial membrane potential (Δψm), a dose-dependent progression from linear to fragmented mitochondria, and dose-dependent changes in longevity. Chronic exposure to a low dose of paraquat (0.035 mM) over multiple generations in N2 causes a progressive decline of fertility, leading to complete loss of fertile embryo production by the third generation. The mutation in CEP-1 [cep-1(gk138)], a key regulator of stress-induced apoptosis in the germline, causes increased sensitivity to chronic paraquat relative to N2 with no fertile embryo production beyond the second generation. Whereas, mitochondrial electron transport chain (complex III) mutant [isp-1(qm150)], which display constitutive activation of mtUPR showed increased tolerance and produced fertile embryo out to the fourth generation. The N2, cep-1(gk138), and isp-1(qm150) strain's lifespan over multiple generations exposed to chronic paraquat were measured. Fertility and lifespan data together indicate a trade-off between reproduction and somatic maintenance during chronic paraquat exposure. We have proposed that mitochondrial signaling, dynamics, and CEP-1 mediated germline apoptosis is involved in this trade-off.
Collapse
Affiliation(s)
- Snigdha Bora
- Department of Agricultural Biotechnology, Assam Agricultural University, Jorhat-13, India
| | | | - Nikhita Deka
- DBT-NECAB, Assam Agricultural University, Jorhat-13, India
| | - Lipika Khataniar
- Department of Agricultural Biotechnology, Assam Agricultural University, Jorhat-13, India
| | - Debajani Gogoi
- DBT-NECAB, Assam Agricultural University, Jorhat-13, India
| | - Aiswarya Baruah
- Department of Agricultural Biotechnology, Assam Agricultural University, Jorhat-13, India; DBT-NECAB, Assam Agricultural University, Jorhat-13, India.
| |
Collapse
|
31
|
Gonzalez-Hunt CP, Thacker EA, Toste CM, Boularand S, Deprets S, Dubois L, Sanders LH. Mitochondrial DNA damage as a potential biomarker of LRRK2 kinase activity in LRRK2 Parkinson's disease. Sci Rep 2020; 10:17293. [PMID: 33057100 PMCID: PMC7557909 DOI: 10.1038/s41598-020-74195-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Accepted: 09/14/2020] [Indexed: 12/14/2022] Open
Abstract
Leucine-rich repeat kinase 2 (LRRK2) is a promising therapeutic target for the treatment of Parkinson's disease (PD) and LRRK2 kinase inhibitors are currently being tested in early phase clinical trials. In order to ensure the highest chance of success, a biomarker-guided entry into clinical trials is key. LRRK2 phosphorylation, and phosphorylation of the LRRK2 substrate Rab10, have been proposed as target engagement biomarkers for LRRK2 kinase inhibition. However, a pharmacodynamic biomarker to demonstrate that a biological response has occurred is lacking. We previously discovered that the LRRK2 G2019S mutation causes mitochondrial DNA (mtDNA) damage and is LRRK2 kinase activity-dependent. Here, we have explored the possibility that measurement of mtDNA damage is a "surrogate" for LRRK2 kinase activity and consequently of kinase inhibitor activity. Mitochondrial DNA damage was robustly increased in PD patient-derived immune cells with LRRK2 G2019S mutations as compared with controls. Following treatment with multiple classes of LRRK2 kinase inhibitors, a full reversal of mtDNA damage to healthy control levels was observed and correlated with measures of LRRK2 dephosphorylation. Taken together, assessment of mtDNA damage levels may be a sensitive measure of altered kinase activity and provide an extended profile of LRRK2 kinase modulation in clinical studies.
Collapse
Affiliation(s)
- C P Gonzalez-Hunt
- Department of Neurology, Duke University Medical Center, Durham, NC, 27710, USA
| | - E A Thacker
- Department of Neurology, Duke University Medical Center, Durham, NC, 27710, USA
| | - C M Toste
- Department of Neurology, Duke University Medical Center, Durham, NC, 27710, USA
| | - S Boularand
- Rare & Neurologic Diseases Research, Sanofi, Chilly Mazarin, France
| | - S Deprets
- Rare & Neurologic Diseases Research, Sanofi, Chilly Mazarin, France
| | - L Dubois
- Rare & Neurologic Diseases Research, Sanofi, Chilly Mazarin, France
| | - L H Sanders
- Department of Neurology, Duke University Medical Center, Durham, NC, 27710, USA.
| |
Collapse
|
32
|
Sural S, Liang CY, Wang FY, Ching TT, Hsu AL. HSB-1/HSF-1 pathway modulates histone H4 in mitochondria to control mtDNA transcription and longevity. SCIENCE ADVANCES 2020; 6:eaaz4452. [PMID: 33087356 PMCID: PMC7577724 DOI: 10.1126/sciadv.aaz4452] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Accepted: 09/08/2020] [Indexed: 06/02/2023]
Abstract
Heat shock factor-1 (HSF-1) is a master regulator of stress responses across taxa. Overexpression of HSF-1 or genetic ablation of its conserved negative regulator, heat shock factor binding protein 1 (HSB-1), results in robust life-span extension in Caenorhabditis elegans Here, we found that increased HSF-1 activity elevates histone H4 levels in somatic tissues during development, while knockdown of H4 completely suppresses HSF-1-mediated longevity. Moreover, overexpression of H4 is sufficient to extend life span. Ablation of HSB-1 induces an H4-dependent increase in micrococcal nuclease protection of both nuclear chromatin and mitochondrial DNA (mtDNA), which consequently results in reduced transcription of mtDNA-encoded complex IV genes, decreased respiratory capacity, and a mitochondrial unfolded protein response-dependent life-span extension. Collectively, our findings reveal a previously unknown role of HSB-1/HSF-1 signaling in modulation of mitochondrial function via mediating histone H4-dependent regulation of mtDNA gene expression and concomitantly acting as a determinant of organismal longevity.
Collapse
Affiliation(s)
- Surojit Sural
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Chung-Yi Liang
- Research Center for Healthy Aging, China Medical University, Taichung, 404, Taiwan
| | - Feng-Yung Wang
- Institute of Biochemistry and Molecular Biology, National Yang-Ming University, Taipei 112, Taiwan
| | - Tsui-Ting Ching
- Institute of Biopharmaceutical Sciences, National Yang-Ming University, Taipei 112, Taiwan.
| | - Ao-Lin Hsu
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI 48109, USA.
- Research Center for Healthy Aging, China Medical University, Taichung, 404, Taiwan
- Institute of Biochemistry and Molecular Biology, National Yang-Ming University, Taipei 112, Taiwan
- Division of Geriatric and Palliative Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
33
|
Zhou K, Mo Q, Guo S, Liu Y, Yin C, Ji X, Guo X, Xing J. A Novel Next-Generation Sequencing-Based Approach for Concurrent Detection of Mitochondrial DNA Copy Number and Mutation. J Mol Diagn 2020; 22:1408-1418. [PMID: 33011442 DOI: 10.1016/j.jmoldx.2020.09.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2020] [Revised: 08/17/2020] [Accepted: 09/03/2020] [Indexed: 01/06/2023] Open
Abstract
Numerous studies have identified essential contributions of altered mitochondrial DNA (mtDNA) copy number and mutations in many common disorders, including cancer. To date, capture-based next-generation sequencing (NGS) has been widely applied to detect mtDNA mutations, although it lacks the ability to assess mtDNA copy number. The current strategy for quantifying mtDNA copy number relies mainly on real-time quantitative PCR, which is limited in degraded samples. A novel capture-based NGS approach was developed using both mtDNA and nuclear DNA probes to capture target fragments, enabling simultaneous detection of mtDNA mutations and copy number in different sample types. First, the impact of selecting reference genes on mtDNA copy number calculation was evaluated, and finally, 3 nuclear DNA fragments of 4000 bp were selected as an internal reference for detection. Then, the effective application of this approach was verified in DNA samples of formalin-fixed, paraffin-embedded specimens and body fluids, indicating the widespread applicability. This approach showed more accurate and stable results in detecting mtDNA copy number compared with real-time quantitative PCR in degraded DNA samples. Moreover, data indicated this approach had good reproducibility in detecting both mtDNA copy number and mutations among three sample types. Altogether, a versatile and cost-effective capture-based NGS approach has been developed for concurrent detection of mtDNA copy number and mutations, which has numerous applications in research and diagnosis.
Collapse
Affiliation(s)
- Kaixiang Zhou
- State Key Laboratory of Cancer Biology, Department of Physiology and Pathophysiology, Fourth Military Medical University, Xi'an, China
| | - Qinqin Mo
- Department of Laboratory Medicine, Medical College of Yanan University, Yan'an, China
| | - Shanshan Guo
- State Key Laboratory of Cancer Biology, Department of Physiology and Pathophysiology, Fourth Military Medical University, Xi'an, China
| | - Yang Liu
- State Key Laboratory of Cancer Biology, Department of Physiology and Pathophysiology, Fourth Military Medical University, Xi'an, China
| | - Chun Yin
- State Key Laboratory of Cancer Biology, Department of Physiology and Pathophysiology, Fourth Military Medical University, Xi'an, China
| | - Xiaoying Ji
- State Key Laboratory of Cancer Biology, Department of Physiology and Pathophysiology, Fourth Military Medical University, Xi'an, China
| | - Xu Guo
- State Key Laboratory of Cancer Biology, Department of Physiology and Pathophysiology, Fourth Military Medical University, Xi'an, China
| | - Jinliang Xing
- State Key Laboratory of Cancer Biology, Department of Physiology and Pathophysiology, Fourth Military Medical University, Xi'an, China.
| |
Collapse
|
34
|
Ignatieva EV, Ivanova OA, Komarova MY, Khromova NV, Polev DE, Kostareva AA, Sergushichev A, Dmitrieva RI. LMNA Mutations G232E and R482L Cause Dysregulation of Skeletal Muscle Differentiation, Bioenergetics, and Metabolic Gene Expression Profile. Genes (Basel) 2020; 11:E1057. [PMID: 32906763 PMCID: PMC7563596 DOI: 10.3390/genes11091057] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Revised: 08/30/2020] [Accepted: 09/01/2020] [Indexed: 02/06/2023] Open
Abstract
Laminopathies are a family of monogenic multi-system diseases resulting from mutations in the LMNA gene which include a wide range of neuromuscular disorders. Although lamins are expressed in most types of differentiated cells, LMNA mutations selectively affect only specific tissues by mechanisms that remain largely unknown. We have employed the combination of functional in vitro experiments and transcriptome analysis in order to determine how two LMNA mutations associated with different phenotypes affect skeletal muscle development and metabolism. We used a muscle differentiation model based on C2C12 mouse myoblasts genetically modified with lentivirus constructs bearing wild-type human LMNA (WT-LMNA) or R482L-LMNA/G232E-LMNA mutations, linked to familial partial lipodystrophy of the Dunnigan type and muscular dystrophy phenotype accordingly. We have shown that both G232E/R482L-LMNA mutations cause dysregulation in coordination of pathways that control cell cycle dynamics and muscle differentiation. We have also found that R482/G232E-LMNA mutations induce mitochondrial uncoupling and a decrease in glycolytic activity in differentiated myotubes. Both types of alterations may contribute to mutation-induced muscle tissue pathology.
Collapse
Affiliation(s)
- Elena V. Ignatieva
- National Almazov Medical Research Centre, Institute of Molecular Biology and Genetics, 197341 Saint-Petersburg, Russia; (E.V.I.); (O.A.I.); (M.Y.K.); (N.V.K.); (A.A.K.)
| | - Oksana A. Ivanova
- National Almazov Medical Research Centre, Institute of Molecular Biology and Genetics, 197341 Saint-Petersburg, Russia; (E.V.I.); (O.A.I.); (M.Y.K.); (N.V.K.); (A.A.K.)
- ITMO University, Information Technologies and Programming Faculty, International Laboratory of Bioinformatics and Genomics, 197101 St. Petersburg, Russia;
| | - Margarita Y. Komarova
- National Almazov Medical Research Centre, Institute of Molecular Biology and Genetics, 197341 Saint-Petersburg, Russia; (E.V.I.); (O.A.I.); (M.Y.K.); (N.V.K.); (A.A.K.)
| | - Natalia V. Khromova
- National Almazov Medical Research Centre, Institute of Molecular Biology and Genetics, 197341 Saint-Petersburg, Russia; (E.V.I.); (O.A.I.); (M.Y.K.); (N.V.K.); (A.A.K.)
| | - Dmitrii E. Polev
- Research Resource Center “Biobank”, St Petersburg State University, 199034 Saint-Petersburg, Russia;
| | - Anna A. Kostareva
- National Almazov Medical Research Centre, Institute of Molecular Biology and Genetics, 197341 Saint-Petersburg, Russia; (E.V.I.); (O.A.I.); (M.Y.K.); (N.V.K.); (A.A.K.)
| | - Alexey Sergushichev
- ITMO University, Information Technologies and Programming Faculty, International Laboratory of Bioinformatics and Genomics, 197101 St. Petersburg, Russia;
| | - Renata I. Dmitrieva
- National Almazov Medical Research Centre, Institute of Molecular Biology and Genetics, 197341 Saint-Petersburg, Russia; (E.V.I.); (O.A.I.); (M.Y.K.); (N.V.K.); (A.A.K.)
| |
Collapse
|
35
|
Sea Cucumber Peptides Improved the Mitochondrial Capacity of Mice: A Potential Mechanism to Enhance Gluconeogenesis and Fat Catabolism during Exercise for Improved Antifatigue Property. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:4604387. [PMID: 32685094 PMCID: PMC7335390 DOI: 10.1155/2020/4604387] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 02/23/2020] [Accepted: 03/20/2020] [Indexed: 01/16/2023]
Abstract
Sea cucumber promotes multifaceted health benefits. However, the mechanisms of sea cucumber peptides (Scp) regulating the antifatigue capacity is still unknown. The present study is aimed at further elucidating the effects and mechanisms of Scp on the antifatigue capacity of mice. At first, C57BL/6J mice were assigned into four groups named Con, L-Scp, M-Scp, and H-Scp and received diets containing Scp (0%, 0.15%, 0.3%, and 0.5%, respectively) for continuous 30 days. On the 21th day, a fore grip test was conducted on mice. On the 25th day, a rotating rod test was conducted on mice. On the 30th day, the quantities of glycogen and mitochondrial DNA (mtDNA) were determined in 8 random mice and another 8 mice were forced to swim for 1 hour before slaughter for detecting biochemical indicators. It was observed that the Scp groups significantly prolonged the running time in rotarod, increased forelimb grip strength, improved lactic acid (LD) and urea nitrogen (BUN) levels in the serum, decreased lactic dehydrogenase (LDH) and glutamic oxalacetic transaminase (GOT) activities in the serum, increased blood glucose (BG) and glycogen (GN) levels in the liver and skeletal muscle after swimming, increased the activity of Na+-K+-ATPase and Ca2+-Mg2+-ATPase in the skeletal muscle and heart, and improved antioxidant capacity. Furthermore, Scp treatment significantly elevated the mRNA and protein relative levels of power-sensitive factors, lipid catabolism, and mitochondrial biogenesis and significantly upregulated mRNA levels of gluconeogenesis. Besides, mtDNA before the swimming test was increased in the three Scp groups. These results show that Scp treatment has antifatigue capacity. Furthermore, these results suggest that improved energy regulation and antioxidant capacity may be the result of improved mitochondrial function.
Collapse
|
36
|
Belli M, Zhang L, Liu X, Donjacour A, Ruggeri E, Palmerini MG, Nottola SA, Macchiarelli G, Rinaudo P. Oxygen concentration alters mitochondrial structure and function in in vitro fertilized preimplantation mouse embryos. Hum Reprod 2020; 34:601-611. [PMID: 30865267 DOI: 10.1093/humrep/dez011] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Revised: 11/27/2018] [Indexed: 12/12/2022] Open
Abstract
STUDY QUESTION Does the oxygen concentration in the culture medium [either physiologic (5%) or atmospheric (20%)] affect mitochondrial ultrastructure and function in preimplantation mouse embryos generated by IVF? SUMMARY ANSWER Embryos cultured in 20% oxygen show increased mitochondrial abnormalities compared to embryos cultured in 5% oxygen. WHAT IS KNOWN ALREADY ART are widely used and have resulted in the birth of more than 8 million children. A variety of media and oxygen concentrations are used to culture embryos. Embryos cultured under physiological O2 tension (5%) reach the blastocyst stage faster and have fewer alterations in gene expression when compared with embryos cultured under atmospheric oxygen conditions (20%). The mechanisms by which oxygen tension affects preimplantation development remain unclear, but mitochondria are believed to play an important role. The aim of this study was to evaluate how mitochondrial ultrastructure and function in IVF embryos were affected by culture under physiologic (5%) or atmospheric (20%) oxygen concentrations. STUDY DESIGN, SIZE, DURATION Zygotes, 2-cell, 4-cell, morula and blastocyst were flushed out of the uterus after natural fertilization and used as controls. IVF was performed in CF1 x B6D2F1 mice and embryos were cultured in Potassium simplex optimized medium (KSOM) with amino acids (KAA) under 5% and 20% O2 until the blastocyst stage. Embryo development with the addition of antioxidants was also tested. PARTICIPANTS/MATERIALS, SETTING, METHODS Mitochondrial function was assessed by measuring mitochondrial membrane potential, reactive oxygen species (ROS) production, ATP levels, and the expression of selected genes involved in mitochondrial function. Mitochondria ultrastructure was evaluated by transmission electron microscopy (TEM). MAIN RESULTS AND THE ROLE OF CHANCE Embryos cultured under 20% O2 had fewer mitochondria and more vacuoles and hooded (abnormal) mitochondria compared to the other groups (P < 0.05). At the blastocyst stage the mitochondria of IVF embryos cultured in 20% O2 had lower mtDNA copy number, a denser matrix and more lamellar cristae than controls. Overall IVF-generated blastocysts had lower mitochondrial membrane potential, higher ROS levels, together with changes in the expression of selected mitochondrial genes (P < 0.05). ATP levels were significantly lower than controls only under 5% O2, with the 20% O2 IVF group having intermediate levels. Unexpectedly, adding antioxidant to the culture medium did not improve development. LARGE SCALE DATA N/A. LIMITATIONS, REASONS FOR CAUTION Findings in mice embryos might be different from human embryos. WIDER IMPLICATIONS OF THE FINDINGS This study suggests that changes in the mitochondria may be part of the mechanism by which lower oxygen concentration leads to better embryo development and further emphasize the importance of mitochondria as a locus of reprogramming. STUDY FUNDING/COMPETING INTEREST(S) This study was funded by R01 HD 082039 to PFR, the Department of Life, Health and Environmental Sciences, University of L'Aquila, Italy (RIA 2016-2018) and the Department of Anatomy, Histology, Forensic Medicine and Orthopaedics, La Sapienza University of Rome, Italy (University grants 2016-2017). The authors declare no competing interests.
Collapse
Affiliation(s)
- Manuel Belli
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy.,Center for Reproductive Sciences, Department of Obstetrics and Gynecology, University of California San Francisco, San Francisco, CA, USA
| | - Ling Zhang
- Center for Reproductive Sciences, Department of Obstetrics and Gynecology, University of California San Francisco, San Francisco, CA, USA.,Family Planning Research Institute and Center for Reproductive Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P. R. China
| | - Xiaowei Liu
- Center for Reproductive Sciences, Department of Obstetrics and Gynecology, University of California San Francisco, San Francisco, CA, USA
| | - Annemarie Donjacour
- Center for Reproductive Sciences, Department of Obstetrics and Gynecology, University of California San Francisco, San Francisco, CA, USA
| | - Elena Ruggeri
- Center for Reproductive Sciences, Department of Obstetrics and Gynecology, University of California San Francisco, San Francisco, CA, USA
| | - Maria Grazia Palmerini
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | - Stefania Annarita Nottola
- Deparment of Anatomy, Histology, Forensic Medicine and Orthopaedics, La Sapienza University of Rome, Rome, Italy
| | - Guido Macchiarelli
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | - Paolo Rinaudo
- Center for Reproductive Sciences, Department of Obstetrics and Gynecology, University of California San Francisco, San Francisco, CA, USA
| |
Collapse
|
37
|
Mookherjee D, Das S, Mukherjee R, Bera M, Jana SC, Chakrabarti S, Chakrabarti O. RETREG1/FAM134B mediated autophagosomal degradation of AMFR/GP78 and OPA1 -a dual organellar turnover mechanism. Autophagy 2020; 17:1729-1752. [PMID: 32559118 DOI: 10.1080/15548627.2020.1783118] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Turnover of cellular organelles, including endoplasmic reticulum (ER) and mitochondria, is orchestrated by an efficient cellular surveillance system. We have identified a mechanism for dual regulation of ER and mitochondria under stress. It is known that AMFR, an ER E3 ligase and ER-associated degradation (ERAD) regulator, degrades outer mitochondrial membrane (OMM) proteins, MFNs (mitofusins), via the proteasome and triggers mitophagy. We show that destabilized mitochondria are almost devoid of the OMM and generate "mitoplasts". This brings the inner mitochondrial membrane (IMM) in the proximity of the ER. When AMFR levels are high and the mitochondria are stressed, the reticulophagy regulatory protein RETREG1 participates in the formation of the mitophagophore by interacting with OPA1. Interestingly, OPA1 and other IMM proteins exhibit similar RETREG1-dependent autophagosomal degradation as AMFR, unlike most of the OMM proteins. The "mitoplasts" generated are degraded by reticulo-mito-phagy - simultaneously affecting dual organelle turnover.Abbreviations: AMFR/GP78: autocrine motility factor receptor; BAPTA: 1,2-bis(o-aminophenoxy)ethane-N,N,N',N'-tetraacetic acid; BFP: blue fluorescent protein; CCCP: carbonyl cyanide m-chlorophenyl hydrazone; CNBr: cyanogen bromide; ER: endoplasmic reticulum; ERAD: endoplasmic-reticulum-associated protein degradation; FL: fluorescence, GFP: green fluorescent protein; HA: hemagglutinin; HEPES: 4-(2-hydroxyethyl)-1-piperazineethanesulfonic acid; IMM: inner mitochondrial membrane; LIR: LC3-interacting region; MAP1LC3/LC3: microtubule associated protein 1 light chain 3; MFN: mitofusin, MGRN1: mahogunin ring finger 1; NA: numerical aperature; OMM: outer mitochondrial membrane; OPA1: OPA1 mitochondrial dynamin like GTPase; PRNP/PrP: prion protein; RER: rough endoplasmic reticulum; RETREG1/FAM134B: reticulophagy regulator 1; RFP: red fluorescent protein; RING: really interesting new gene; ROI: region of interest; RTN: reticulon; SEM: standard error of the mean; SER: smooth endoplasmic reticulum; SIM: structured illumination microscopy; SQSTM1/p62: sequestosome 1; STED: stimulated emission depletion; STOML2: stomatin like 2; TOMM20: translocase of outer mitochondrial membrane 20; UPR: unfolded protein response.
Collapse
Affiliation(s)
- Debdatto Mookherjee
- Biophysics & Structural Genomics Division, Saha Institute of Nuclear Physics, Kolkata, India
| | - Subhrangshu Das
- Structural Biology and Bioinformatics Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| | - Rukmini Mukherjee
- Biophysics & Structural Genomics Division, Saha Institute of Nuclear Physics, Kolkata, India.,Buchmann Institute for Molecular Life Sciences, Frankfurt Am Main, Germany
| | - Manindra Bera
- Laboratory of Cell Biology, the Rockefeller University, New York, USA
| | | | - Saikat Chakrabarti
- Structural Biology and Bioinformatics Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| | - Oishee Chakrabarti
- Biophysics & Structural Genomics Division, Saha Institute of Nuclear Physics, Kolkata, India.,Homi Bhabha National Institute, Mumbai, India
| |
Collapse
|
38
|
Increase of mtDNA number and its mutant copies in rat brain after exposure to 150 MeV protons. Mol Biol Rep 2020; 47:4815-4820. [PMID: 32388700 DOI: 10.1007/s11033-020-05491-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Accepted: 04/30/2020] [Indexed: 12/31/2022]
Abstract
Proton beam therapy is widely used for treating brain tumor. Despite the efficacy of treatment, the use of this therapy has met some limitations associated with possible damage to normal brain tissues located beyond the tumor site. In this context, the exploration of the harmful effects of protons on the normal brain tissues is of particular interest. We have investigated changes in the total mitochondrial DNA (mtDNA) copy number and identified mtDNA mutant copies in three brain regions (the hippocampus, cortex and cerebellum) of rats after irradiation their whole-head with 150 MeV protons at doses of 3 and 5 Gy. The study was performed in 2-months old male Spraque Dawley rats (n = 5 each group). The mtDNA copy numbers were determined by real-time PCR. The level of mtDNA heteroplasmy was estimated using Surveyor nuclease technology. Our results show that after head exposure to protons, levels of mtDNA copy number in three rat brain regions increase significantly as the levels of mtDNA mutant copies increase. The most significant elevation is observed in the hippocampus. In conclusion, an increase in mtDNA mutant copies may contribute to mitochondrial dysfunction accompanied by increased oxidative stress in different brain regions and promote the development of neurodegenerative diseases and the induction of carcinogenesis.
Collapse
|
39
|
Bradley MC, Yang K, Fernández-Del-Río L, Ngo J, Ayer A, Tsui HS, Novales NA, Stocker R, Shirihai OS, Barros MH, Clarke CF. COQ11 deletion mitigates respiratory deficiency caused by mutations in the gene encoding the coenzyme Q chaperone protein Coq10. J Biol Chem 2020; 295:6023-6042. [PMID: 32205446 DOI: 10.1074/jbc.ra119.012420] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 03/17/2020] [Indexed: 12/17/2022] Open
Abstract
Coenzyme Q (Q n ) is a vital lipid component of the electron transport chain that functions in cellular energy metabolism and as a membrane antioxidant. In the yeast Saccharomyces cerevisiae, coq1-coq9 deletion mutants are respiratory-incompetent, sensitive to lipid peroxidation stress, and unable to synthesize Q6 The yeast coq10 deletion mutant is also respiratory-deficient and sensitive to lipid peroxidation, yet it continues to produce Q6 at an impaired rate. Thus, Coq10 is required for the function of Q6 in respiration and as an antioxidant and is believed to chaperone Q6 from its site of synthesis to the respiratory complexes. In several fungi, Coq10 is encoded as a fusion polypeptide with Coq11, a recently identified protein of unknown function required for efficient Q6 biosynthesis. Because "fused" proteins are often involved in similar biochemical pathways, here we examined the putative functional relationship between Coq10 and Coq11 in yeast. We used plate growth and Seahorse assays and LC-MS/MS analysis to show that COQ11 deletion rescues respiratory deficiency, sensitivity to lipid peroxidation, and decreased Q6 biosynthesis of the coq10Δ mutant. Additionally, immunoblotting indicated that yeast coq11Δ mutants accumulate increased amounts of certain Coq polypeptides and display a stabilized CoQ synthome. These effects suggest that Coq11 modulates Q6 biosynthesis and that its absence increases mitochondrial Q6 content in the coq10Δcoq11Δ double mutant. This augmented mitochondrial Q6 content counteracts the respiratory deficiency and lipid peroxidation sensitivity phenotypes of the coq10Δ mutant. This study further clarifies the intricate connection between Q6 biosynthesis, trafficking, and function in mitochondrial metabolism.
Collapse
Affiliation(s)
- Michelle C Bradley
- Department of Chemistry and Biochemistry, Molecular Biology Institute, UCLA, Los Angeles, California 90095-1569
| | - Krista Yang
- Department of Chemistry and Biochemistry, Molecular Biology Institute, UCLA, Los Angeles, California 90095-1569
| | - Lucía Fernández-Del-Río
- Department of Chemistry and Biochemistry, Molecular Biology Institute, UCLA, Los Angeles, California 90095-1569
| | - Jennifer Ngo
- Department of Chemistry and Biochemistry, Molecular Biology Institute, UCLA, Los Angeles, California 90095-1569; Department of Molecular and Medical Pharmacology and Medicine, David Geffen School of Medicine, UCLA, Los Angeles, California 90095
| | - Anita Ayer
- Vascular Biology Division, Victor Chang Cardiac Research Institute, Sydney, New South Wales 2010, Australia; St. Vincent's Clinical School, University of New South Wales Medicine, Sydney, New South Wales 2050, Australia
| | - Hui S Tsui
- Department of Chemistry and Biochemistry, Molecular Biology Institute, UCLA, Los Angeles, California 90095-1569
| | - Noelle Alexa Novales
- Department of Chemistry and Biochemistry, Molecular Biology Institute, UCLA, Los Angeles, California 90095-1569
| | - Roland Stocker
- Vascular Biology Division, Victor Chang Cardiac Research Institute, Sydney, New South Wales 2010, Australia; St. Vincent's Clinical School, University of New South Wales Medicine, Sydney, New South Wales 2050, Australia
| | - Orian S Shirihai
- Department of Molecular and Medical Pharmacology and Medicine, David Geffen School of Medicine, UCLA, Los Angeles, California 90095
| | - Mario H Barros
- Departamento Microbiologia, Universidade de São Paulo, São Paulo 05508-900, Brazil
| | - Catherine F Clarke
- Department of Chemistry and Biochemistry, Molecular Biology Institute, UCLA, Los Angeles, California 90095-1569.
| |
Collapse
|
40
|
Overexpression of Purinergic P2X4 Receptors in Hippocampus Rescues Memory Impairment in Rats with Type 2 Diabetes. Neurosci Bull 2020; 36:719-732. [PMID: 32198702 PMCID: PMC7340685 DOI: 10.1007/s12264-020-00478-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Accepted: 11/25/2019] [Indexed: 12/15/2022] Open
Abstract
Purinergic receptors have been reported to be involved in brain disorders. In this study, we explored their roles and mechanisms underlying the memory impairment in rats with type 2 diabetes mellitus (T2DM). T2DM rats exhibited a worse performance in the T-maze and Morris water maze (MWM) than controls. Microglia positive for P2X purinoceptor 4 (P2X4R) in the hippocampus were reduced and activated microglia were increased in T2DM rats. Long Amplicon PCR (LA-PCR) showed that DNA amplification of the p2x4r gene in the hippocampus was lower in T2DM rats. Minocycline significantly reduced the number of activated microglia and the mean distance traveled by T2DM rats in the MWM. Most importantly, P2X4R overexpression suppressed the activated microglia and rescued the memory impairment of T2DM rats. Overall, T2DM led to excessive activation of microglia in the hippocampus, partly through the DNA damage-mediated downregulation of P2X4Rs, thus contributing to memory impairment.
Collapse
|
41
|
Abdullaev S, Gubina N, Bulanova T, Gaziev A. Assessment of Nuclear and Mitochondrial DNA, Expression of Mitochondria-Related Genes in Different Brain Regions in Rats after Whole-Body X-ray Irradiation. Int J Mol Sci 2020; 21:ijms21041196. [PMID: 32054039 PMCID: PMC7072726 DOI: 10.3390/ijms21041196] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2019] [Revised: 02/07/2020] [Accepted: 02/08/2020] [Indexed: 01/02/2023] Open
Abstract
Studies of molecular changes occurred in various brain regions after whole-body irradiation showed a significant increase in terms of the importance in gaining insight into how to slow down or prevent the development of long-term side effects such as carcinogenesis, cognitive impairment and other pathologies. We have analyzed nDNA damage and repair, changes in mitochondrial DNA (mtDNA) copy number and in the level of mtDNA heteroplasmy, and also examined changes in the expression of genes involved in the regulation of mitochondrial biogenesis and dynamics in three areas of the rat brain (hippocampus, cortex and cerebellum) after whole-body X-ray irradiation. Long amplicon quantitative polymerase chain reaction (LA-QPCR) was used to detect nDNA and mtDNA damage. The level of mtDNA heteroplasmy was estimated using Surveyor nuclease technology. The mtDNA copy numbers and expression levels of a number of genes were determined by real-time PCR. The results showed that the repair of nDNA damage in the rat brain regions occurs slowly within 24 h; in the hippocampus, this process runs much slower. The number of mtDNA copies in three regions of the rat brain increases with a simultaneous increase in mtDNA heteroplasmy. However, in the hippocampus, the copy number of mutant mtDNAs increases significantly by the time point of 24 h after radiation exposure. Our analysis shows that in the brain regions of irradiated rats, there is a decrease in the expression of genes (ND2, CytB, ATP5O) involved in ATP synthesis, although by the same time point after irradiation, an increase in transcripts of genes regulating mitochondrial biogenesis is observed. On the other hand, analysis of genes that control the dynamics of mitochondria (Mfn1, Fis1) revealed that sharp decrease in gene expression level occurred, only in the hippocampus. Consequently, the structural and functional characteristics of the hippocampus of rats exposed to whole-body radiation can be different, most significantly from those of the other brain regions.
Collapse
Affiliation(s)
- Serazhutdin Abdullaev
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino, 142290 Moscow, Russia; (N.G.); (A.G.)
- Correspondence: ; Tel.: +7-(4967)-739364; Fax: +7-(4967)-330553
| | - Nina Gubina
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino, 142290 Moscow, Russia; (N.G.); (A.G.)
| | - Tatiana Bulanova
- Joint Institute for Nuclear Research, Dubna, 141980 Moscow, Russia;
| | - Azhub Gaziev
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino, 142290 Moscow, Russia; (N.G.); (A.G.)
- Joint Institute for Nuclear Research, Dubna, 141980 Moscow, Russia;
| |
Collapse
|
42
|
Lindberg CD, Di Giulio RT. Polycyclic aromatic hydrocarbon and hypoxia exposures result in mitochondrial dysfunction in zebrafish. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2019; 216:105298. [PMID: 31586484 PMCID: PMC6917040 DOI: 10.1016/j.aquatox.2019.105298] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 09/06/2019] [Accepted: 09/11/2019] [Indexed: 05/20/2023]
Abstract
Organisms are routinely subjected to a variety of environmental and chemical perturbations simultaneously. Often, multi-stressor exposures result in unpredictable toxicity that occurs through unidentified mechanisms. Here, we focus on polycyclic aromatic hydrocarbons (PAHs) and hypoxia, two environmental and physiological stressors that are known to co-occur in the environment. The aim of this study was to assess whether interactive mitochondrial dysfunction resulted from co-exposures of PAHs and hypoxia. Zebrafish embryos were co-exposed to non-teratogenic concentrations of an environmental PAH mixture and hypoxia beginning at 6 hpf for an acute period of 24 h and afterwards were given either no recovery period, 45 min, 5 -hs, or 18 -hs of recovery time in clean conditions. Mitochondrial function and integrity were assessed through the use of both in ovo and in vitro assays. Hypoxia exposures resulted in drastic reductions in parameters relating to mitochondrial respiration, ATP turnover, and mitochondrial DNA integrity. PAH exposures affected ATP production and content, as well as mitochondrial membrane dynamics and lactate content. While PAH and hypoxia exposures caused a broad range of effects, there appeared to be very little interaction between the two stressors in the co-exposure group. However, because hypoxia significantly altered mitochondrial function, the possibility remains that these effects may limit an individual's ability to respond to PAH toxicity and therefore could cause downstream interactive effects.
Collapse
Affiliation(s)
- Casey D Lindberg
- Integrated Toxicology and Environmental Health Program, Nicholas School of the Environment, Duke University, Durham, NC 27708, USA.
| | - Richard T Di Giulio
- Integrated Toxicology and Environmental Health Program, Nicholas School of the Environment, Duke University, Durham, NC 27708, USA.
| |
Collapse
|
43
|
Muñoz-Lorente MA, Cano-Martin AC, Blasco MA. Mice with hyper-long telomeres show less metabolic aging and longer lifespans. Nat Commun 2019; 10:4723. [PMID: 31624261 PMCID: PMC6797762 DOI: 10.1038/s41467-019-12664-x] [Citation(s) in RCA: 100] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Accepted: 09/19/2019] [Indexed: 12/16/2022] Open
Abstract
Short telomeres trigger age-related pathologies and shorter lifespans in mice and humans. In the past, we generated mouse embryonic (ES) cells with longer telomeres than normal (hyper-long telomeres) in the absence of genetic manipulations, which contributed to all mouse tissues. To address whether hyper-long telomeres have deleterious effects, we generated mice in which 100% of their cells are derived from hyper-long telomere ES cells. We observe that these mice have longer telomeres and less DNA damage with aging. Hyper-long telomere mice are lean and show low cholesterol and LDL levels, as well as improved glucose and insulin tolerance. Hyper-long telomere mice also have less incidence of cancer and an increased longevity. These findings demonstrate that longer telomeres than normal in a given species are not deleterious but instead, show beneficial effects.
Collapse
Affiliation(s)
- Miguel A Muñoz-Lorente
- Telomeres and Telomerase Group, Molecular Oncology Program, Spanish National Cancer Centre (CNIO), Melchor Fernández Almagro 3, Madrid, 28029, Spain
| | - Alba C Cano-Martin
- Telomeres and Telomerase Group, Molecular Oncology Program, Spanish National Cancer Centre (CNIO), Melchor Fernández Almagro 3, Madrid, 28029, Spain
| | - Maria A Blasco
- Telomeres and Telomerase Group, Molecular Oncology Program, Spanish National Cancer Centre (CNIO), Melchor Fernández Almagro 3, Madrid, 28029, Spain.
| |
Collapse
|
44
|
Lyra-Leite DM, Andres AM, Cho N, Petersen AP, Ariyasinghe NR, Kim SS, Gottlieb RA, McCain ML. Matrix-guided control of mitochondrial function in cardiac myocytes. Acta Biomater 2019; 97:281-295. [PMID: 31401347 PMCID: PMC6801042 DOI: 10.1016/j.actbio.2019.08.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Revised: 07/31/2019] [Accepted: 08/02/2019] [Indexed: 02/08/2023]
Abstract
In ventricular myocardium, extracellular matrix (ECM) remodeling is a hallmark of physiological and pathological growth, coincident with metabolic rewiring of cardiac myocytes. However, the direct impact of the biochemical and mechanical properties of the ECM on the metabolic function of cardiac myocytes is mostly unknown. Furthermore, understanding the impact of distinct biomaterials on cardiac myocyte metabolism is critical for engineering physiologically-relevant models of healthy and diseased myocardium. For these reasons, we systematically measured morphological and metabolic responses of neonatal rat ventricular myocytes cultured on fibronectin- or gelatin-coated polydimethylsiloxane (PDMS) of three elastic moduli and gelatin hydrogels with four elastic moduli. On all substrates, total protein content, cell morphology, and the ratio of mitochondrial DNA to nuclear DNA were preserved. Cytotoxicity was low on all substrates, although slightly higher on PDMS compared to gelatin hydrogels. We also quantified oxygen consumption rates and extracellular acidification rates using a Seahorse extracellular flux analyzer. Our data indicate that several metrics associated with baseline glycolysis and baseline and maximum mitochondrial function are enhanced when cardiac myocytes are cultured on gelatin hydrogels compared to all PDMS substrates, irrespective of substrate rigidity. These results yield new insights into how mechanical and biochemical cues provided by the ECM impact mitochondrial function in cardiac myocytes. STATEMENT OF SIGNIFICANCE: Cardiac development and disease are associated with remodeling of the extracellular matrix coincident with metabolic rewiring of cardiac myocytes. However, little is known about the direct impact of the biochemical and mechanical properties of the extracellular matrix on the metabolic function of cardiac myocytes. In this study, oxygen consumption rates were measured in neonatal rat ventricular myocytes maintained on several commonly-used biomaterial substrates to reveal new relationships between the extracellular matrix and cardiac myocyte metabolism. Several mitochondrial parameters were enhanced on gelatin hydrogels compared to synthetic PDMS substrates. These data are important for comprehensively understanding matrix-regulation of cardiac myocyte physiology. Additionally, these data should be considered when selecting scaffolds for engineering in vitro cardiac tissue models.
Collapse
Affiliation(s)
- Davi M Lyra-Leite
- Laboratory for Living Systems Engineering, Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles CA, 90089, United States
| | - Allen M Andres
- Smidt Heart Institute and Barbra Streisand Women's Heart Center, Cedars-Sinai Medical Center, Los Angeles CA, 90048, United States
| | - Nathan Cho
- Laboratory for Living Systems Engineering, Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles CA, 90089, United States
| | - Andrew P Petersen
- Laboratory for Living Systems Engineering, Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles CA, 90089, United States
| | - Nethika R Ariyasinghe
- Laboratory for Living Systems Engineering, Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles CA, 90089, United States
| | - Suyon Sarah Kim
- Laboratory for Living Systems Engineering, Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles CA, 90089, United States
| | - Roberta A Gottlieb
- Smidt Heart Institute and Barbra Streisand Women's Heart Center, Cedars-Sinai Medical Center, Los Angeles CA, 90048, United States
| | - Megan L McCain
- Laboratory for Living Systems Engineering, Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles CA, 90089, United States; Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine of USC, University of Southern California, Los Angeles CA, 90033, United States.
| |
Collapse
|
45
|
Maglioni S, Mello DF, Schiavi A, Meyer JN, Ventura N. Mitochondrial bioenergetic changes during development as an indicator of C. elegans health-span. Aging (Albany NY) 2019; 11:6535-6554. [PMID: 31454791 PMCID: PMC6738431 DOI: 10.18632/aging.102208] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Accepted: 08/07/2019] [Indexed: 11/25/2022]
Abstract
Mild suppression of mitochondrial activity has beneficial effects across species. The nematode Caenorhabditis elegans is a versatile, genetically tractable model organism widely employed for aging studies, which has led to the identification of many of the known evolutionarily conserved mechanisms regulating lifespan. In C. elegans the pro-longevity effect of reducing mitochondrial function, for example by RNA interference, is only achieved if mitochondrial stress is applied during larval development. Surprisingly, a careful analysis of changes in mitochondrial functions resulting from such treatments during the developmental windows in which pro-longevity signals are programmed has never been carried out. Thus, although the powerful C. elegans genetics have led to the identification of different molecular mechanisms causally involved in mitochondrial stress control of longevity, specific functional mitochondrial biomarkers indicative or predictive of lifespan remain to be identified. To fill this gap, we systematically characterized multiple mitochondrial functional parameters at an early developmental stage in animals that are long-lived due to mild knockdown of twelve different mitochondrial proteins and correlated these parameters with animals’ lifespan. We found that basal oxygen consumption rate and ATP-linked respiration positively correlate with lifespan extension and propose the testable hypothesis that the Bioenergetic Health Index can be used as a proxy to predict health-span outcomes.
Collapse
Affiliation(s)
- Silvia Maglioni
- IUF-Leibniz Research Institute for Environmental Medicine, 40225 Duesseldorf, Germany
| | - Danielle F Mello
- Nicholas School of the Environment and Integrated Toxicology and Environmental Health Program, Duke University, Durham, NC 27708, USA
| | - Alfonso Schiavi
- IUF-Leibniz Research Institute for Environmental Medicine, 40225 Duesseldorf, Germany.,Institute for Clinical Chemistry and Laboratory Diagnostic, Medical Faculty of the Heinrich Heine University, 40225 Duesseldorf, Germany
| | - Joel N Meyer
- Nicholas School of the Environment and Integrated Toxicology and Environmental Health Program, Duke University, Durham, NC 27708, USA
| | - Natascia Ventura
- IUF-Leibniz Research Institute for Environmental Medicine, 40225 Duesseldorf, Germany.,Institute for Clinical Chemistry and Laboratory Diagnostic, Medical Faculty of the Heinrich Heine University, 40225 Duesseldorf, Germany
| |
Collapse
|
46
|
Berky AJ, Ryde IT, Feingold B, Ortiz EJ, Wyatt LH, Weinhouse C, Hsu-Kim H, Meyer JN, Pan WK. Predictors of mitochondrial DNA copy number and damage in a mercury-exposed rural Peruvian population near artisanal and small-scale gold mining: An exploratory study. ENVIRONMENTAL AND MOLECULAR MUTAGENESIS 2019; 60:197-210. [PMID: 30289587 PMCID: PMC6452630 DOI: 10.1002/em.22244] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Revised: 07/22/2018] [Accepted: 07/31/2018] [Indexed: 05/22/2023]
Abstract
Mitochondrial DNA (mtDNA) copy number (CN) and damage in circulating white blood cells have been proposed as effect biomarkers for pollutant exposures. Studies have shown that mercury accumulates in mitochondria and affects mitochondrial function and integrity; however, these data are derived largely from experiments in model systems, rather than human population studies that evaluate the potential utility of mitochondrial exposure biomarkers. We measured mtDNA CN and damage in white blood cells (WBCs) from 83 residents of nine communities in the Madre de Dios region of the Peruvian Amazon that vary in proximity to artisanal and small-scale gold mining. Prior research from this region reported high levels of mercury in fish and a significant association between food consumption and human total hair mercury level of residents. We observed that mtDNA CN and damage were both associated with consumption of fruit and vegetables, higher diversity of fruit consumed, residential location, and health characteristics, suggesting common environmental drivers. Surprisingly, we observed negative associations of mtDNA damage with both obesity and age. We did not observe any association between total hair mercury or, in contrast to previous results, age, with either mtDNA damage or CN. The results of this exploratory study highlight the importance of combining epidemiological and laboratory research in studying the effects of stressors on mitochondria, suggesting that future work should incorporate nutritional and social characteristics, and caution should be taken when applying conclusions from epidemiological studies conducted in the developed world to other regions, as results may not be easily translated. Environ. Mol. Mutagen. 60: 197-210, 2019. © 2018 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Axel J. Berky
- Nicholas School of the Environment, Box 90328, Duke University, Durham, NC 27708, USA
| | - Ian T. Ryde
- Nicholas School of the Environment, Box 90328, Duke University, Durham, NC 27708, USA
| | - Beth Feingold
- University of Albany, School of Public Health, Rensselaer, NY 12144, USA
| | - Ernesto J. Ortiz
- Duke Global Health Institute, Duke University, 310 Trent Dr, Durham, NC 27710, USA
| | - Lauren H. Wyatt
- Nicholas School of the Environment, Box 90328, Duke University, Durham, NC 27708, USA
| | - Caren Weinhouse
- Duke Global Health Institute, Duke University, 310 Trent Dr, Durham, NC 27710, USA
| | - Heileen Hsu-Kim
- Department of Civil and Engineering, Box 90287, Duke University, Durham, NC 27708, USA
| | - Joel N. Meyer
- Nicholas School of the Environment, Box 90328, Duke University, Durham, NC 27708, USA
- Corresponding authors: William Pan, Duke Global Health Institute, 310 Trent Drive, Campus Box 90519, Durham, NC 27708, fax 681-7748, , Joel N. Meyer, Nicholas School of the Environment, Box 90328, Duke University, Durham, NC 27708, USA,
| | - William K. Pan
- Nicholas School of the Environment, Box 90328, Duke University, Durham, NC 27708, USA
- Duke Global Health Institute, Duke University, 310 Trent Dr, Durham, NC 27710, USA
- Corresponding authors: William Pan, Duke Global Health Institute, 310 Trent Drive, Campus Box 90519, Durham, NC 27708, fax 681-7748, , Joel N. Meyer, Nicholas School of the Environment, Box 90328, Duke University, Durham, NC 27708, USA,
| |
Collapse
|
47
|
Miliotis S, Nicolalde B, Ortega M, Yepez J, Caicedo A. Forms of extracellular mitochondria and their impact in health. Mitochondrion 2019; 48:16-30. [PMID: 30771504 DOI: 10.1016/j.mito.2019.02.002] [Citation(s) in RCA: 87] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2018] [Revised: 11/09/2018] [Accepted: 02/06/2019] [Indexed: 12/15/2022]
Abstract
Mitochondria play an important role as an intracellular energy plant and signaling organelle. However, mitochondria also exist outside cells where they could mediate cell-to-cell communication, repair and serve as an activator of the immune response. Their effects depend on the mitochondrial state or the form in which it is present, either as a whole functional structure as fragments or only as mitochondrial DNA. Herein, we provide evidence of why extracellular mitochondria and their varying forms are considered regenerative factors or pro-inflammatory activators. Understanding these aspects will provide the base of their use in therapy or as a biomarker of disease severity and prognosis.
Collapse
Affiliation(s)
- Sophia Miliotis
- Universidad San Francisco de Quito, The Latitude Zero Ecuador Research Initiative, L0ERI, 17-12-841, Ecuador
| | - Bryan Nicolalde
- Universidad San Francisco de Quito, Colegio de Ciencias de la Salud - Hospital de los Valles, Escuela de Medicina, Quito 17-12-841, Ecuador
| | - Mayra Ortega
- Universidad San Francisco de Quito, Colegio de Ciencias Biológicas y Ambientales, Escuela de Biotecnología, Quito 17-12-841, Ecuador; Universidad San Francisco de Quito, Instituto de Investigaciones en Biomedicina, Quito 17-12-841, Ecuador
| | - Jackie Yepez
- Universidad San Francisco de Quito, The Latitude Zero Ecuador Research Initiative, L0ERI, 17-12-841, Ecuador
| | - Andrés Caicedo
- Universidad San Francisco de Quito, Colegio de Ciencias de la Salud - Hospital de los Valles, Escuela de Medicina, Quito 17-12-841, Ecuador; Universidad San Francisco de Quito, Instituto de Investigaciones en Biomedicina, Quito 17-12-841, Ecuador; Mito-Act Research Consortium, Quito, Ecuador; Sistemas Médicos - Universidad San Francisco de Quito, SIME-USFQ, Quito 17-12-841, Ecuador.
| |
Collapse
|
48
|
Klebsiella oxytoca enterotoxins tilimycin and tilivalline have distinct host DNA-damaging and microtubule-stabilizing activities. Proc Natl Acad Sci U S A 2019; 116:3774-3783. [PMID: 30808763 PMCID: PMC6397511 DOI: 10.1073/pnas.1819154116] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Human gut microbes form a complex community with vast biosynthetic potential. Microbial products and metabolites released in the gut impact human health and disease. However, defining causative relationships between specific bacterial products and disease initiation and progression remains an immense challenge. This study advances understanding of the functional capacity of the gut microbiota by determining the presence, concentration, and spatial and temporal variability of two enterotoxic metabolites produced by the gut-resident Klebsiella oxytoca. We present a detailed mode of action for the cytotoxins and recapitulate their functionalities in disease models in vivo. The findings provide distinct molecular mechanisms for the enterotoxicity of the metabolites allowing them to act in tandem to damage the intestinal epithelium and cause colitis. Establishing causal links between bacterial metabolites and human intestinal disease is a significant challenge. This study reveals the molecular basis of antibiotic-associated hemorrhagic colitis (AAHC) caused by intestinal resident Klebsiella oxytoca. Colitogenic strains produce the nonribosomal peptides tilivalline and tilimycin. Here, we verify that these enterotoxins are present in the human intestine during active colitis and determine their concentrations in a murine disease model. Although both toxins share a pyrrolobenzodiazepine structure, they have distinct molecular targets. Tilimycin acts as a genotoxin. Its interaction with DNA activates damage repair mechanisms in cultured cells and causes DNA strand breakage and an increased lesion burden in cecal enterocytes of colonized mice. In contrast, tilivalline binds tubulin and stabilizes microtubules leading to mitotic arrest. To our knowledge, this activity is unique for microbiota-derived metabolites of the human intestine. The capacity of both toxins to induce apoptosis in intestinal epithelial cells—a hallmark feature of AAHC—by independent modes of action, strengthens our proposal that these metabolites act collectively in the pathogenicity of colitis.
Collapse
|
49
|
Hibshman JD, Leuthner TC, Shoben C, Mello DF, Sherwood DR, Meyer JN, Baugh LR. Nonselective autophagy reduces mitochondrial content during starvation in Caenorhabditis elegans. Am J Physiol Cell Physiol 2018; 315:C781-C792. [PMID: 30133321 PMCID: PMC6336938 DOI: 10.1152/ajpcell.00109.2018] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Starvation significantly alters cellular physiology, and signs of aging have been reported to occur during starvation. Mitochondria are essential to the regulation of cellular energetics and aging. We sought to determine whether mitochondria exhibit signs of aging during starvation and whether quality control mechanisms regulate mitochondrial physiology during starvation. We describe effects of starvation on mitochondria in the first and third larval stages of the nematode Caenorhabditis elegans. When starved, C. elegans larvae enter developmental arrest. We observed fragmentation of the mitochondrial network, a reduction in mitochondrial DNA (mtDNA) copy number, and accumulation of DNA damage during starvation-induced developmental arrest. Mitochondrial function was also compromised by starvation. Starved worms had lower basal, maximal, and ATP-linked respiration. These observations are consistent with reduced mitochondrial quality, similar to mitochondrial phenotypes during aging. Using pharmacological and genetic approaches, we found that worms deficient for autophagy were short-lived during starvation and recovered poorly from extended starvation, indicating sensitivity to nutrient stress. Autophagy mutants unc-51/Atg1 and atg-18/Atg18 maintained greater mtDNA content than wild-type worms during starvation, suggesting that autophagy promotes mitochondrial degradation during starvation. unc-51 mutants also had a proportionally smaller reduction in oxygen consumption rate during starvation, suggesting that autophagy also contributes to reduced mitochondrial function. Surprisingly, mutations in genes involved in mitochondrial fission and fusion as well as selective mitophagy of damaged mitochondria did not affect mitochondrial content during starvation. Our results demonstrate the profound influence of starvation on mitochondrial physiology with organismal consequences, and they show that these physiological effects are influenced by autophagy.
Collapse
Affiliation(s)
- Jonathan D. Hibshman
- 1Department of Biology, Duke University, Durham, North Carolina,2University Program in Genetics and Genomics, Duke University, Durham, North Carolina,3Department of Biology, University of North Carolina, Chapel Hill, North Carolina
| | - Tess C. Leuthner
- 4Nicholas School of the Environment, Duke University, Durham, North Carolina
| | - Chelsea Shoben
- 1Department of Biology, Duke University, Durham, North Carolina
| | - Danielle F. Mello
- 4Nicholas School of the Environment, Duke University, Durham, North Carolina
| | - David R. Sherwood
- 1Department of Biology, Duke University, Durham, North Carolina,2University Program in Genetics and Genomics, Duke University, Durham, North Carolina
| | - Joel N. Meyer
- 4Nicholas School of the Environment, Duke University, Durham, North Carolina
| | - L. Ryan Baugh
- 1Department of Biology, Duke University, Durham, North Carolina,2University Program in Genetics and Genomics, Duke University, Durham, North Carolina
| |
Collapse
|
50
|
Saha P, Gupta R, Sen T, Sen N. Activation of cyclin D1 affects mitochondrial mass following traumatic brain injury. Neurobiol Dis 2018; 118:108-116. [PMID: 30010002 DOI: 10.1016/j.nbd.2018.07.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Revised: 06/25/2018] [Accepted: 07/11/2018] [Indexed: 01/24/2023] Open
Abstract
Cell cycle activation has been associated with varying types of neurological disorders including brain injury. Cyclin D1 is a critical modulator of cell cycle activation and upregulation of Cyclin D1 in neurons contributes to the pathology associated with traumatic brain injury (TBI). Mitochondrial mass is a critical factor to maintain the mitochondrial function, and it can be regulated by different signaling cascades and transcription factors including NRF1. However, the underlying mechanism of how TBI leads to impairment of mitochondrial mass following TBI remains obscure. Our results indicate that augmentation of CyclinD1 attenuates mitochondrial mass formation following TBI. To elucidate the molecular mechanism, we found that Cyclin D1 interacts with a transcription factor NRF1 in the nucleus and prevents NRF1's interaction with p300 in the pericontusional cortex following TBI. As a result, the acetylation level of NRF1 was decreased, and its transcriptional activity was attenuated. This event leads to a loss of mitochondrial mass in the pericontusional cortex following TBI. Intranasal delivery of Cyclin D1 RNAi immediately after TBI rescues transcriptional activation of NRF1 and recovers mitochondrial mass after TBI.
Collapse
Affiliation(s)
- Pampa Saha
- Department of Neurological Surgery, University of Pittsburgh, 200 Lothrop Street, Scaife Hall, Pittsburgh 15213, United States
| | - Rajaneesh Gupta
- Department of Neurological Surgery, University of Pittsburgh, 200 Lothrop Street, Scaife Hall, Pittsburgh 15213, United States
| | - Tanusree Sen
- Department of Neurological Surgery, University of Pittsburgh, 200 Lothrop Street, Scaife Hall, Pittsburgh 15213, United States
| | - Nilkantha Sen
- Department of Neurological Surgery, University of Pittsburgh, 200 Lothrop Street, Scaife Hall, Pittsburgh 15213, United States.
| |
Collapse
|