1
|
Machado IF, Palmeira CM, Rolo AP. Sestrin2 is a central regulator of mitochondrial stress responses in disease and aging. Ageing Res Rev 2025; 109:102762. [PMID: 40320152 DOI: 10.1016/j.arr.2025.102762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Revised: 04/09/2025] [Accepted: 04/30/2025] [Indexed: 05/08/2025]
Abstract
Mitochondria supply most of the energy for cellular functions and coordinate numerous cellular pathways. Their dynamic nature allows them to adjust to stress and cellular metabolic demands, thus ensuring the preservation of cellular homeostasis. Loss of normal mitochondrial function compromises cell survival and has been implicated in the development of many diseases and in aging. Although exposure to continuous or severe stress has adverse effects on cells, mild mitochondrial stress enhances mitochondrial function and potentially extends health span through mitochondrial adaptive responses. Over the past few decades, sestrin2 (SESN2) has emerged as a pivotal regulator of stress responses. For instance, SESN2 responds to genotoxic, oxidative, and metabolic stress, promoting cellular defense against stress-associated damage. Here, we focus on recent findings that establish SESN2 as an orchestrator of mitochondrial stress adaptation, which is supported by its involvement in the integrated stress response, mitochondrial biogenesis, and mitophagy. Additionally, we discuss the integral role of SESN2 in mediating the health benefits of exercise as well as its impact on skeletal muscle, liver and heart injury, and aging.
Collapse
Affiliation(s)
- Ivo F Machado
- CNC-UC - Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal; CiBB - Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal; Institute of Interdisciplinary Research, Doctoral Program in Experimental Biology and Biomedicine (PDBEB), University of Coimbra, Coimbra, Portugal
| | - Carlos M Palmeira
- CNC-UC - Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal; CiBB - Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal; Department of Life Sciences, University of Coimbra, Coimbra, Portugal
| | - Anabela P Rolo
- CNC-UC - Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal; CiBB - Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal; Department of Life Sciences, University of Coimbra, Coimbra, Portugal.
| |
Collapse
|
2
|
Khaliulin I, Hamoudi W, Amal H. The multifaceted role of mitochondria in autism spectrum disorder. Mol Psychiatry 2025; 30:629-650. [PMID: 39223276 PMCID: PMC11753362 DOI: 10.1038/s41380-024-02725-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Revised: 08/21/2024] [Accepted: 08/23/2024] [Indexed: 09/04/2024]
Abstract
Normal brain functioning relies on high aerobic energy production provided by mitochondria. Failure to supply a sufficient amount of energy, seen in different brain disorders, including autism spectrum disorder (ASD), may have a significant negative impact on brain development and support of different brain functions. Mitochondrial dysfunction, manifested in the abnormal activities of the electron transport chain and impaired energy metabolism, greatly contributes to ASD. The aberrant functioning of this organelle is of such high importance that ASD has been proposed as a mitochondrial disease. It should be noted that aerobic energy production is not the only function of the mitochondria. In particular, these organelles are involved in the regulation of Ca2+ homeostasis, different mechanisms of programmed cell death, autophagy, and reactive oxygen and nitrogen species (ROS and RNS) production. Several syndromes originated from mitochondria-related mutations display ASD phenotype. Abnormalities in Ca2+ handling and ATP production in the brain mitochondria affect synaptic transmission, plasticity, and synaptic development, contributing to ASD. ROS and Ca2+ regulate the activity of the mitochondrial permeability transition pore (mPTP). The prolonged opening of this pore affects the redox state of the mitochondria, impairs oxidative phosphorylation, and activates apoptosis, ultimately leading to cell death. A dysregulation between the enhanced mitochondria-related processes of apoptosis and the inhibited autophagy leads to the accumulation of toxic products in the brains of individuals with ASD. Although many mitochondria-related mechanisms still have to be investigated, and whether they are the cause or consequence of this disorder is still unknown, the accumulating data show that the breakdown of any of the mitochondrial functions may contribute to abnormal brain development leading to ASD. In this review, we discuss the multifaceted role of mitochondria in ASD from the various aspects of neuroscience.
Collapse
Affiliation(s)
- Igor Khaliulin
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Wajeha Hamoudi
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Haitham Amal
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel.
| |
Collapse
|
3
|
Tong W, Allison BJ, Brain KL, Patey OV, Niu Y, Botting KJ, Ford SG, Garrud TA, Wooding PFB, Lyu Q, Zhang L, Ma J, Sowton AP, O'Brien KA, Cindrova-Davies T, Yung HW, Burton GJ, Murray AJ, Giussani DA. Placental mitochondrial metabolic adaptation maintains cellular energy balance in pregnancy complicated by gestational hypoxia. J Physiol 2025. [PMID: 39868991 DOI: 10.1113/jp287897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Accepted: 12/18/2024] [Indexed: 01/28/2025] Open
Abstract
The mechanisms that drive placental dysfunction in pregnancies complicated by hypoxia and fetal growth restriction remain poorly understood. Changes to mitochondrial respiration contribute to cellular dysfunction in conditions of hypoxia and have been implicated in the pathoaetiology of pregnancy complications, such as pre-eclampsia. We used bespoke isobaric hypoxic chambers and a combination of functional, molecular and imaging techniques to study cellular metabolism and mitochondrial dynamics in sheep undergoing hypoxic pregnancy. We show that hypoxic pregnancy in sheep triggers a shift in capacity away from β-oxidation and complex I-mediated respiration, while maintaining total oxidative phosphorylation capacity. There are also complex-specific changes to electron transport chain composition and a switch in mitochondrial dynamics towards fission. Hypoxic placentas show increased activation of the non-canonical mitochondrial unfolded protein response pathway and enhanced insulin like growth factor 2 signalling. Combined, therefore, the data show that the hypoxic placenta undergoes significant metabolic and morphological adaptations to maintain cellular energy balance. Chronic hypoxia during pregnancy in sheep activated placental mitochondrial stress pathways, leading to alterations in mitochondrial respiration, mitochondrial energy metabolism and mitochondrial dynamics, as seen in the placenta of women with pre-eclampsia. KEY POINTS: Hypoxia shifts mitochondrial respiration away from β-oxidation and complex I. Complex-specific changes occur in the electron transport chain composition. Activation of the non-canonical mitochondrial unfolded protein response pathway is heightened in hypoxic placentas. Enhanced insulin like growth factor 2 signalling is observed in hypoxic placentas. Hypoxic placentas undergo significant functional adaptations for energy balance.
Collapse
Affiliation(s)
- Wen Tong
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
- Centre for Trophoblast Research, University of Cambridge, Cambridge, UK
- Strategic Research Initiative in Reproduction, University of Cambridge, Cambridge, UK
| | - Beth J Allison
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Kirsty L Brain
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Olga V Patey
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Youguo Niu
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
- Centre for Trophoblast Research, University of Cambridge, Cambridge, UK
- Strategic Research Initiative in Reproduction, University of Cambridge, Cambridge, UK
- Cardiovascular Strategic Research Initiative, University of Cambridge, Cambridge, UK
- Department of Aerospace Physiology, Fourth Military Medical University, Xi'an, China
| | - Kimberley J Botting
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
- Centre for Trophoblast Research, University of Cambridge, Cambridge, UK
- Strategic Research Initiative in Reproduction, University of Cambridge, Cambridge, UK
- Cardiovascular Strategic Research Initiative, University of Cambridge, Cambridge, UK
| | - Sage G Ford
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Tess A Garrud
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
- Centre for Trophoblast Research, University of Cambridge, Cambridge, UK
| | - Peter F B Wooding
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
- Centre for Trophoblast Research, University of Cambridge, Cambridge, UK
| | - Qiang Lyu
- Department of Aerospace Physiology, Fourth Military Medical University, Xi'an, China
| | - Lin Zhang
- Department of Aerospace Physiology, Fourth Military Medical University, Xi'an, China
| | - Jin Ma
- Department of Aerospace Physiology, Fourth Military Medical University, Xi'an, China
| | - Alice P Sowton
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
- Centre for Trophoblast Research, University of Cambridge, Cambridge, UK
| | - Katie A O'Brien
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
- Centre for Trophoblast Research, University of Cambridge, Cambridge, UK
| | - Tereza Cindrova-Davies
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
- Centre for Trophoblast Research, University of Cambridge, Cambridge, UK
- Strategic Research Initiative in Reproduction, University of Cambridge, Cambridge, UK
| | - Hong Wa Yung
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
- Centre for Trophoblast Research, University of Cambridge, Cambridge, UK
- Strategic Research Initiative in Reproduction, University of Cambridge, Cambridge, UK
| | - Graham J Burton
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
- Centre for Trophoblast Research, University of Cambridge, Cambridge, UK
- Strategic Research Initiative in Reproduction, University of Cambridge, Cambridge, UK
| | - Andrew J Murray
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
- Centre for Trophoblast Research, University of Cambridge, Cambridge, UK
- Cardiovascular Strategic Research Initiative, University of Cambridge, Cambridge, UK
| | - Dino A Giussani
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
- Centre for Trophoblast Research, University of Cambridge, Cambridge, UK
- Strategic Research Initiative in Reproduction, University of Cambridge, Cambridge, UK
- Cardiovascular Strategic Research Initiative, University of Cambridge, Cambridge, UK
- Department of Aerospace Physiology, Fourth Military Medical University, Xi'an, China
| |
Collapse
|
4
|
Ji S, Xu X, Li Y, Sun S, Fu Q, Qiu Y, Wang S, Xia S, Wang F, Zhang F, Xuan J, Zheng S. Inhibition of TFAM-Mediated Mitophagy by Oroxylin A Restored Sorafenib Sensitivity Under Hypoxia Conditions in HepG2 Cells. Pharmaceuticals (Basel) 2024; 17:1727. [PMID: 39770569 PMCID: PMC11676196 DOI: 10.3390/ph17121727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 12/06/2024] [Accepted: 12/17/2024] [Indexed: 01/11/2025] Open
Abstract
Background: Liver cancer treatment encounters considerable therapeutic challenges, especially because hypoxic microenvironments markedly reduce sensitivity to chemotherapeutic agents. TFAM (mitochondrial transcription factor A) plays a crucial role in maintaining mitochondrial function. Oroxylin A (OA), a flavonoid with potential therapeutic properties, demonstrated prospects in cancer treatment. However, the mechanism of the sensitizing effect of OA on cancer cells has not been elucidated. Methods: MTT assays were utilized to evaluate a hypoxia-induced resistance model. Plate colony formation assays, TEM, and JC-1 staining were used to examine the effects of siTFAM on proliferation and mitochondrial damage of HepG2 cells. Cox8-EGFP-mCherry plasmid transfection, LysoTracker and MitoTracker colocalization analysis, and WB were conducted to evaluate the influence of OA on mitophagy. The effect of OA on p53 ubiquitination levels was investigated by Co-IP and the CHX chase assay. A mouse xenograft tumor model was utilized to assess the therapeutic effect of OA on HepG2 cells in vivo. Results: OA significantly improved the inhibitory effect of sorafenib by inhibiting mitophagy on HepG2 cells in in vitro and in vivo models. Notably, the molecular docking and thermal shift assays indicated a clear binding of OA and TFAM. Further research revealed that OA suppressed p53 acetylation and promoted its degradation by downregulating TFAM expression, which ultimately inhibited mitophagy in hypoxia. Conclusions: OA has demonstrated the potential to enhance the efficacy of sorafenib treatment for liver cancer, and TFAM may be one of its targets.
Collapse
Affiliation(s)
- Shufan Ji
- Jangsu Key Laboratory for Pharmacology and Safety Research of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China; (S.J.); (X.X.); (Y.L.); (S.S.); (Q.F.); (Y.Q.); (S.W.); (S.X.); (F.W.); (F.Z.)
| | - Xuefen Xu
- Jangsu Key Laboratory for Pharmacology and Safety Research of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China; (S.J.); (X.X.); (Y.L.); (S.S.); (Q.F.); (Y.Q.); (S.W.); (S.X.); (F.W.); (F.Z.)
- Department of Pharmacology, School of Medicine, Nanjing University of Chinese Medicine, No.138, Xianlin Road, Nanjing 210023, China
| | - Yujia Li
- Jangsu Key Laboratory for Pharmacology and Safety Research of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China; (S.J.); (X.X.); (Y.L.); (S.S.); (Q.F.); (Y.Q.); (S.W.); (S.X.); (F.W.); (F.Z.)
| | - Sumin Sun
- Jangsu Key Laboratory for Pharmacology and Safety Research of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China; (S.J.); (X.X.); (Y.L.); (S.S.); (Q.F.); (Y.Q.); (S.W.); (S.X.); (F.W.); (F.Z.)
| | - Qiuyu Fu
- Jangsu Key Laboratory for Pharmacology and Safety Research of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China; (S.J.); (X.X.); (Y.L.); (S.S.); (Q.F.); (Y.Q.); (S.W.); (S.X.); (F.W.); (F.Z.)
| | - Yangling Qiu
- Jangsu Key Laboratory for Pharmacology and Safety Research of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China; (S.J.); (X.X.); (Y.L.); (S.S.); (Q.F.); (Y.Q.); (S.W.); (S.X.); (F.W.); (F.Z.)
| | - Shuqi Wang
- Jangsu Key Laboratory for Pharmacology and Safety Research of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China; (S.J.); (X.X.); (Y.L.); (S.S.); (Q.F.); (Y.Q.); (S.W.); (S.X.); (F.W.); (F.Z.)
| | - Siwei Xia
- Jangsu Key Laboratory for Pharmacology and Safety Research of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China; (S.J.); (X.X.); (Y.L.); (S.S.); (Q.F.); (Y.Q.); (S.W.); (S.X.); (F.W.); (F.Z.)
| | - Feixia Wang
- Jangsu Key Laboratory for Pharmacology and Safety Research of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China; (S.J.); (X.X.); (Y.L.); (S.S.); (Q.F.); (Y.Q.); (S.W.); (S.X.); (F.W.); (F.Z.)
| | - Feng Zhang
- Jangsu Key Laboratory for Pharmacology and Safety Research of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China; (S.J.); (X.X.); (Y.L.); (S.S.); (Q.F.); (Y.Q.); (S.W.); (S.X.); (F.W.); (F.Z.)
| | - Ji Xuan
- Department of Gastroenterology, Jinling Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing 210002, China
| | - Shizhong Zheng
- Jangsu Key Laboratory for Pharmacology and Safety Research of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China; (S.J.); (X.X.); (Y.L.); (S.S.); (Q.F.); (Y.Q.); (S.W.); (S.X.); (F.W.); (F.Z.)
| |
Collapse
|
5
|
Warwick AM, Bomze HM, Wang L, Hao Y, Stinnett SS, Gospe SM. Hypoxia-mediated rescue of retinal ganglion cells deficient in mitochondrial complex I is independent of the hypoxia-inducible factor pathway. Sci Rep 2024; 14:24114. [PMID: 39406814 PMCID: PMC11480089 DOI: 10.1038/s41598-024-75916-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Accepted: 10/09/2024] [Indexed: 10/19/2024] Open
Abstract
Continuous exposure to environmental hypoxia (11% O2) has been shown to markedly slow the progressive degeneration of retinal ganglion cells (RGCs) in a mouse model of mitochondrial optic neuropathy with RGC-specific deletion of the key mitochondrial complex I accessory subunit ndufs4. As a first step toward identifying the therapeutic mechanism of hypoxia in this model, we conducted a series of experiments to investigate the role of the hypoxia-inducible factor (HIF) regulatory pathway in RGC neuroprotection. Vglut2-Cre; ndufs4loxP/loxP mice were crossed with strains bearing floxed alleles of the negative HIF regulatory vhl or of the two major HIF α-subunit isoforms, Hif1α and Hif2α. Deletion of vhl within ndufs4-deficient RGCs failed to prevent RGC degeneration under normoxia, indicating that HIF activation is not sufficient to achieve RGC rescue. Furthermore, the rescue of ndufs4-deficient RGCs by hypoxia remained robust despite genetic inactivation of Hif1α and Hif2α. Our findings demonstrate that the HIF pathway is entirely dispensable to the rescue of RGCs by hypoxia. Future efforts to uncover key HIF-independent molecular pathways induced by hypoxia in this mouse model may be of therapeutic relevance to mitochondrial optic neuropathies such as Leber hereditary optic neuropathy.
Collapse
Affiliation(s)
- Alexander M Warwick
- Department of Ophthalmology, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Howard M Bomze
- Department of Ophthalmology, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Luyu Wang
- Department of Ophthalmology, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Ying Hao
- Department of Ophthalmology, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Sandra S Stinnett
- Department of Ophthalmology, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Sidney M Gospe
- Department of Ophthalmology, Duke University School of Medicine, Durham, NC, 27710, USA.
- Department of Ophthalmology, Box 3712 Med Center, Duke University, 2351 Erwin Road, Durham, NC, 27710, USA.
| |
Collapse
|
6
|
Toledano JM, Puche-Juarez M, Galvez-Navas JM, Moreno-Fernandez J, Diaz-Castro J, Ochoa JJ. Pregnancy Disorders: A Potential Role for Mitochondrial Altered Homeostasis. Antioxidants (Basel) 2024; 13:979. [PMID: 39199225 PMCID: PMC11351112 DOI: 10.3390/antiox13080979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 08/02/2024] [Accepted: 08/10/2024] [Indexed: 09/01/2024] Open
Abstract
Pregnancy is a complex and challenging process associated with physiological changes whose objective is to adapt the maternal organism to the increasing energetic requirements due to embryo and fetal development. A failed adaptation to these demands may lead to pregnancy complications that threaten the health of both mothers and their offspring. Since mitochondria are the main organelle responsible for energy generation in the form of ATP, the adequate state of these organelles seems crucial for proper pregnancy development and healthy pregnancy outcomes. The homeostasis of these organelles depends on several aspects, including their content, biogenesis, energy production, oxidative stress, dynamics, and signaling functions, such as apoptosis, which can be modified in relation to diseases during pregnancy. The etiology of pregnancy disorders like preeclampsia, fetal growth restriction, and gestational diabetes mellitus is not yet well understood. Nevertheless, insufficient placental perfusion and oxygen transfer are characteristic of many of them, being associated with alterations in the previously cited different aspects of mitochondrial homeostasis. Therefore, and due to the capacity of these multifactorial organelles to respond to physiological and pathophysiological stimuli, it is of great importance to gather the currently available scientific information regarding the relationship between main pregnancy complications and mitochondrial alterations. According to this, the present review is intended to show clear insight into the possible implications of mitochondria in these disorders, thus providing relevant information for further investigation in relation to the investigation and management of pregnancy diseases.
Collapse
Affiliation(s)
- Juan M. Toledano
- Department of Physiology, Faculty of Pharmacy, Campus Universitario de Cartuja, University of Granada, 18071 Granada, Spain; (J.M.T.); (J.D.-C.); (J.J.O.)
- Institute of Nutrition and Food Technology “José Mataix Verdú”, University of Granada, 18071 Granada, Spain
- Nutrition and Food Sciences Ph.D. Program, University of Granada, 18071 Granada, Spain
| | - María Puche-Juarez
- Department of Physiology, Faculty of Pharmacy, Campus Universitario de Cartuja, University of Granada, 18071 Granada, Spain; (J.M.T.); (J.D.-C.); (J.J.O.)
- Institute of Nutrition and Food Technology “José Mataix Verdú”, University of Granada, 18071 Granada, Spain
- Nutrition and Food Sciences Ph.D. Program, University of Granada, 18071 Granada, Spain
| | - Jose Maria Galvez-Navas
- Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), 28029 Madrid, Spain;
- Cáncer Registry of Granada, Andalusian School of Public Health, Cuesta del Observatorio 4, Campus Universitario de Cartuja, 18011 Granada, Spain
- Department of Biochemistry and Molecular Biology II, Faculty of Pharmacy, Campus Universitario de Cartuja, University of Granada, 18071 Granada, Spain
- Instituto de Investigación Biosanitaria (IBS), 18016 Granada, Spain
| | - Jorge Moreno-Fernandez
- Department of Physiology, Faculty of Pharmacy, Campus Universitario de Cartuja, University of Granada, 18071 Granada, Spain; (J.M.T.); (J.D.-C.); (J.J.O.)
- Institute of Nutrition and Food Technology “José Mataix Verdú”, University of Granada, 18071 Granada, Spain
- Instituto de Investigación Biosanitaria (IBS), 18016 Granada, Spain
| | - Javier Diaz-Castro
- Department of Physiology, Faculty of Pharmacy, Campus Universitario de Cartuja, University of Granada, 18071 Granada, Spain; (J.M.T.); (J.D.-C.); (J.J.O.)
- Institute of Nutrition and Food Technology “José Mataix Verdú”, University of Granada, 18071 Granada, Spain
- Instituto de Investigación Biosanitaria (IBS), 18016 Granada, Spain
| | - Julio J. Ochoa
- Department of Physiology, Faculty of Pharmacy, Campus Universitario de Cartuja, University of Granada, 18071 Granada, Spain; (J.M.T.); (J.D.-C.); (J.J.O.)
- Institute of Nutrition and Food Technology “José Mataix Verdú”, University of Granada, 18071 Granada, Spain
- Instituto de Investigación Biosanitaria (IBS), 18016 Granada, Spain
| |
Collapse
|
7
|
Piel S, McManus MJ, Heye KN, Beaulieu F, Fazelinia H, Janowska JI, MacTurk B, Starr J, Gaudio H, Patel N, Hefti MM, Smalley ME, Hook JN, Kohli NV, Bruton J, Hallowell T, Delso N, Roberts A, Lin Y, Ehinger JK, Karlsson M, Berg RA, Morgan RW, Kilbaugh TJ. Effect of dimethyl fumarate on mitochondrial metabolism in a pediatric porcine model of asphyxia-induced in-hospital cardiac arrest. Sci Rep 2024; 14:13852. [PMID: 38879681 PMCID: PMC11180202 DOI: 10.1038/s41598-024-64317-9] [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: 03/01/2024] [Accepted: 06/07/2024] [Indexed: 06/19/2024] Open
Abstract
Neurological and cardiac injuries are significant contributors to morbidity and mortality following pediatric in-hospital cardiac arrest (IHCA). Preservation of mitochondrial function may be critical for reducing these injuries. Dimethyl fumarate (DMF) has shown potential to enhance mitochondrial content and reduce oxidative damage. To investigate the efficacy of DMF in mitigating mitochondrial injury in a pediatric porcine model of IHCA, toddler-aged piglets were subjected to asphyxia-induced CA, followed by ventricular fibrillation, high-quality cardiopulmonary resuscitation, and random assignment to receive either DMF (30 mg/kg) or placebo for four days. Sham animals underwent similar anesthesia protocols without CA. After four days, tissues were analyzed for mitochondrial markers. In the brain, untreated CA animals exhibited a reduced expression of proteins of the oxidative phosphorylation system (CI, CIV, CV) and decreased mitochondrial respiration (p < 0.001). Despite alterations in mitochondrial content and morphology in the myocardium, as assessed per transmission electron microscopy, mitochondrial function was unchanged. DMF treatment counteracted 25% of the proteomic changes induced by CA in the brain, and preserved mitochondrial structure in the myocardium. DMF demonstrates a potential therapeutic benefit in preserving mitochondrial integrity following asphyxia-induced IHCA. Further investigation is warranted to fully elucidate DMF's protective mechanisms and optimize its therapeutic application in post-arrest care.
Collapse
Affiliation(s)
- Sarah Piel
- Resuscitation Science Center of Emphasis, The Children's Hospital of Philadelphia, 3401 Civic Center Boulevard, Philadelphia, PA, 19104, USA.
- Department of Anesthesiology and Critical Care Medicine, The Children's Hospital of Philadelphia, Philadelphia, USA.
- Department of Cardiology, Pulmonology, and Vascular Medicine, University Hospital Düsseldorf, Medical Faculty of the Heinrich Heine University Düsseldorf, Düsseldorf, Germany.
- CARID, Cardiovascular Research Institute Düsseldorf, Medical Faculty of the Heinrich-Heine-University, Düsseldorf, Germany.
| | - Meagan J McManus
- Resuscitation Science Center of Emphasis, The Children's Hospital of Philadelphia, 3401 Civic Center Boulevard, Philadelphia, PA, 19104, USA
- Department of Anesthesiology and Critical Care Medicine, The Children's Hospital of Philadelphia, Philadelphia, USA
| | - Kristina N Heye
- Division of Neurology, The Children's Hospital of Philadelphia, Philadelphia, USA
| | - Forrest Beaulieu
- Department of Pediatrics, The Children's Hospital of Philadelphia, Philadelphia, USA
| | - Hossein Fazelinia
- Proteomics Core Facility, The Children's Hospital of Philadelphia, Philadelphia, USA
| | - Joanna I Janowska
- Resuscitation Science Center of Emphasis, The Children's Hospital of Philadelphia, 3401 Civic Center Boulevard, Philadelphia, PA, 19104, USA
- Department of Anesthesiology and Critical Care Medicine, The Children's Hospital of Philadelphia, Philadelphia, USA
| | - Bryce MacTurk
- Resuscitation Science Center of Emphasis, The Children's Hospital of Philadelphia, 3401 Civic Center Boulevard, Philadelphia, PA, 19104, USA
- Department of Anesthesiology and Critical Care Medicine, The Children's Hospital of Philadelphia, Philadelphia, USA
| | - Jonathan Starr
- Resuscitation Science Center of Emphasis, The Children's Hospital of Philadelphia, 3401 Civic Center Boulevard, Philadelphia, PA, 19104, USA
- Department of Anesthesiology and Critical Care Medicine, The Children's Hospital of Philadelphia, Philadelphia, USA
| | - Hunter Gaudio
- Resuscitation Science Center of Emphasis, The Children's Hospital of Philadelphia, 3401 Civic Center Boulevard, Philadelphia, PA, 19104, USA
- Department of Anesthesiology and Critical Care Medicine, The Children's Hospital of Philadelphia, Philadelphia, USA
| | - Nisha Patel
- Resuscitation Science Center of Emphasis, The Children's Hospital of Philadelphia, 3401 Civic Center Boulevard, Philadelphia, PA, 19104, USA
- Department of Anesthesiology and Critical Care Medicine, The Children's Hospital of Philadelphia, Philadelphia, USA
| | - Marco M Hefti
- Department of Pathology, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Martin E Smalley
- Department of Pathology, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Jordan N Hook
- Department of Pathology, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Neha V Kohli
- Resuscitation Science Center of Emphasis, The Children's Hospital of Philadelphia, 3401 Civic Center Boulevard, Philadelphia, PA, 19104, USA
- Department of Anesthesiology and Critical Care Medicine, The Children's Hospital of Philadelphia, Philadelphia, USA
| | - James Bruton
- Resuscitation Science Center of Emphasis, The Children's Hospital of Philadelphia, 3401 Civic Center Boulevard, Philadelphia, PA, 19104, USA
- Department of Anesthesiology and Critical Care Medicine, The Children's Hospital of Philadelphia, Philadelphia, USA
| | - Thomas Hallowell
- Resuscitation Science Center of Emphasis, The Children's Hospital of Philadelphia, 3401 Civic Center Boulevard, Philadelphia, PA, 19104, USA
- Department of Anesthesiology and Critical Care Medicine, The Children's Hospital of Philadelphia, Philadelphia, USA
| | - Nile Delso
- Resuscitation Science Center of Emphasis, The Children's Hospital of Philadelphia, 3401 Civic Center Boulevard, Philadelphia, PA, 19104, USA
- Department of Anesthesiology and Critical Care Medicine, The Children's Hospital of Philadelphia, Philadelphia, USA
| | - Anna Roberts
- Resuscitation Science Center of Emphasis, The Children's Hospital of Philadelphia, 3401 Civic Center Boulevard, Philadelphia, PA, 19104, USA
- Department of Anesthesiology and Critical Care Medicine, The Children's Hospital of Philadelphia, Philadelphia, USA
| | - Yuxi Lin
- Resuscitation Science Center of Emphasis, The Children's Hospital of Philadelphia, 3401 Civic Center Boulevard, Philadelphia, PA, 19104, USA
- Department of Anesthesiology and Critical Care Medicine, The Children's Hospital of Philadelphia, Philadelphia, USA
| | - Johannes K Ehinger
- Mitochondrial Medicine, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
- Otorhinolaryngology, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
- Otorhinolaryngology, Head and Neck Surgery, Skåne University Hospital, Lund, Sweden
| | | | - Robert A Berg
- Resuscitation Science Center of Emphasis, The Children's Hospital of Philadelphia, 3401 Civic Center Boulevard, Philadelphia, PA, 19104, USA
- Department of Anesthesiology and Critical Care Medicine, The Children's Hospital of Philadelphia, Philadelphia, USA
| | - Ryan W Morgan
- Resuscitation Science Center of Emphasis, The Children's Hospital of Philadelphia, 3401 Civic Center Boulevard, Philadelphia, PA, 19104, USA
- Department of Anesthesiology and Critical Care Medicine, The Children's Hospital of Philadelphia, Philadelphia, USA
| | - Todd J Kilbaugh
- Resuscitation Science Center of Emphasis, The Children's Hospital of Philadelphia, 3401 Civic Center Boulevard, Philadelphia, PA, 19104, USA
- Department of Anesthesiology and Critical Care Medicine, The Children's Hospital of Philadelphia, Philadelphia, USA
| |
Collapse
|
8
|
Hafez SMNA, Abdelhafez ESMN. The possible protective effect of l-cysteine in a rat model of sciatic nerve ischemia-reperfusion: A possible role for NRF1 and Caspase 3; Biochemical, Histological, and Immunohistochemical study. J Chem Neuroanat 2024; 137:102412. [PMID: 38460773 DOI: 10.1016/j.jchemneu.2024.102412] [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: 01/05/2024] [Revised: 03/05/2024] [Accepted: 03/06/2024] [Indexed: 03/11/2024]
Abstract
Organ damage brought on by ischemia is exacerbated by the reperfusion process. L-cysteine is a semi-essential amino acid that acts as a substrate for cystathionine-β-synthase in the central nervous system. The aim of this study was to investigate the possible protective effects of L- cysteine against the structural and biochemical changes that occur in the rat sciatic nerve after ischemia reperfusion (I/R) and to address some of the underlying mechanisms of these effects. Rats were divided into 4 groups: sham, l-cysteine, I/R, and l-cysteine- I/R groups. Specimens of sciatic nerve were processed for biochemical, histological, and immunohistochemical assessment. The results showed in I/R group, a significant increase in malondialdehyde with a significant decrease in both Nuclear respiratory factor-1 (NRF1) and superoxide dismutase levels. Moreover, with histological alteration. There was a significant increase in the mean surface area fraction of anti-caspase immunopositive cells as well as a significantdecrease in mean surface area fraction of anti-CD 34 immunopositive cells. In contrast, the l-cysteine- I/R group showed amelioration of these biochemical, structural, and immunohistochemical changes. To the best of our knowledge, this is the first study showed the protective effects of l-cysteine in sciatic nerve I/R via NRF1and caspase 3 modulation as well as telocyte activation.
Collapse
Affiliation(s)
| | - El-Shimaa M N Abdelhafez
- Department of Medicinal Chemistry, Faculty of Pharmacy, Minia University, Faculty of Medicine, Minia 61111, Egypt.
| |
Collapse
|
9
|
Verma K, Amitabh, Prasad DN, Reddy MPK, Kohli E. Kynurenines Dynamics in the Periphery and Central Nervous System Steers Behavioral Deficits in Rats under Hypobaric Hypoxia. ACS Chem Neurosci 2024; 15:1084-1095. [PMID: 38462729 DOI: 10.1021/acschemneuro.3c00632] [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] [Indexed: 03/12/2024] Open
Abstract
People travel to high-altitude regions as tourists, workers, and military personnel on duty. Despite the consistent 21% oxygen content in the atmosphere, ascending to higher altitudes results in a decrease in the partial pressure of oxygen, inducing a state known as hypobaric hypoxia (HH). HH is an environmental stress that is responsible for neuroinflammation and behavioral deficits (anxiety, depression, mood disturbance, etc.), but little is known about its metabolic pathways. The kynurenine pathway (KP) is a promising candidate to uncover the mysteries of HH stress, as it is an important regulator of the immune system and is associated with behavioral deficits. To investigate the role of KP under HH, the levels of KP metabolites in the serum, cerebrospinal fluid (CSF), and brain tissue (prefrontal cortex-PFC, neocortex, and hippocampus) of male Sprague-Dawley rats exposed to HH at 7620 m for 1, 3, and 7 days were estimated utilizing high-performance liquid chromatography (HPLC). The behavioral analogs for anxiety-like and depression-like behavior were assessed using the open field test and forced swim test, respectively. Upon HH exposure, crosstalk between the periphery and central nervous system and KP metabolite region-dependent differential expression in the brain were observed. KP metabolites showed a positive correlation with behavioral parameters. The results of our study are indicative that KP can be proposed as the etiology of behavioral deficits, and KP metabolite levels in serum or CSF can be used as plausible markers for anxiety-like and depression-like behaviors under HH stress with a scope of targeted therapeutic interventions.
Collapse
Affiliation(s)
- Kalyani Verma
- Department of Neurobiology, Defence Institute of Physiology and Allied Sciences, DRDO, Timarpur,Delhi 110054, India
| | - Amitabh
- Department of Neurobiology, Defence Institute of Physiology and Allied Sciences, DRDO, Timarpur,Delhi 110054, India
| | - Dipti N Prasad
- Department of Neurobiology, Defence Institute of Physiology and Allied Sciences, DRDO, Timarpur,Delhi 110054, India
| | - M Prasanna Kumar Reddy
- Department of Applied Physiology, Defence Institute of Physiology and Allied Sciences, DRDO, Timarpur, Delhi 110054, India
| | - Ekta Kohli
- Department of Neurobiology, Defence Institute of Physiology and Allied Sciences, DRDO, Timarpur,Delhi 110054, India
| |
Collapse
|
10
|
Janssen Daalen JM, Koopman WJH, Saris CGJ, Meinders MJ, Thijssen DHJ, Bloem BR. The Hypoxia Response Pathway: A Potential Intervention Target in Parkinson's Disease? Mov Disord 2024; 39:273-293. [PMID: 38140810 DOI: 10.1002/mds.29688] [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: 07/14/2023] [Revised: 11/20/2023] [Accepted: 11/28/2023] [Indexed: 12/24/2023] Open
Abstract
Parkinson's disease (PD) is a progressive neurodegenerative disorder for which only symptomatic treatments are available. Both preclinical and clinical studies suggest that moderate hypoxia induces evolutionarily conserved adaptive mechanisms that enhance neuronal viability and survival. Therefore, targeting the hypoxia response pathway might provide neuroprotection by ameliorating the deleterious effects of mitochondrial dysfunction and oxidative stress, which underlie neurodegeneration in PD. Here, we review experimental studies regarding the link between PD pathophysiology and neurophysiological adaptations to hypoxia. We highlight the mechanistic differences between the rescuing effects of chronic hypoxia in neurodegeneration and short-term moderate hypoxia to improve neuronal resilience, termed "hypoxic conditioning". Moreover, we interpret these preclinical observations regarding the pharmacological targeting of the hypoxia response pathway. Finally, we discuss controversies with respect to the differential effects of hypoxia response pathway activation across the PD spectrum, as well as intervention dosing in hypoxic conditioning and potential harmful effects of such interventions. We recommend that initial clinical studies in PD should focus on the safety, physiological responses, and mechanisms of hypoxic conditioning, as well as on repurposing of existing pharmacological compounds. © 2023 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Jules M Janssen Daalen
- Center of Expertise for Parkinson and Movement Disorders, Department of Neurology, Donders Institute for Brain, Cognition and Behavior, Nijmegen, the Netherlands, Nijmegen, The Netherlands
- Department of Neurology, Radboud University Medical Center, Donders Institute for Brain, Cognition and Behavior, Nijmegen, The Netherlands
- Department of Physiology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Werner J H Koopman
- Department of Pediatrics, Amalia Children's Hospital, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
- Radboud Center for Mitochondrial Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
- Human and Animal Physiology, Wageningen University, Wageningen, The Netherlands
| | - Christiaan G J Saris
- Department of Neurology, Radboud University Medical Center, Donders Institute for Brain, Cognition and Behavior, Nijmegen, The Netherlands
- Radboud Center for Mitochondrial Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Marjan J Meinders
- Center of Expertise for Parkinson and Movement Disorders, Department of Neurology, Donders Institute for Brain, Cognition and Behavior, Nijmegen, the Netherlands, Nijmegen, The Netherlands
| | - Dick H J Thijssen
- Department of Physiology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Bastiaan R Bloem
- Center of Expertise for Parkinson and Movement Disorders, Department of Neurology, Donders Institute for Brain, Cognition and Behavior, Nijmegen, the Netherlands, Nijmegen, The Netherlands
- Department of Neurology, Radboud University Medical Center, Donders Institute for Brain, Cognition and Behavior, Nijmegen, The Netherlands
| |
Collapse
|
11
|
Singh A, Jaiswar SP, Priyadarshini A, Deo S. Linking of oxidative stress and mitochondrial DNA damage to the pathophysiology of idiopathic intrauterine growth restriction. Int J Health Sci (Qassim) 2023; 17:15-22. [PMID: 37929235 PMCID: PMC10624804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2023] Open
Abstract
Objective A common and serious pregnancy issue known as intrauterine growth restriction (IUGR) occurs when the fetus is unable to reach its full growth potential. Mitochondria are crucial to the development of the fetus and the placenta. We aimed to elucidate the role of oxidative stress parameters and markers of DNA damage. The integrity of the mitochondrial DNA (mtDNA) was studied. Materials and Methods Blood samples were collected from 48 females (cases and controls, respectively). Oxidative stress parameters were analyzed. DNA was extracted followed by high-performance liquid chromatography to study 8-OH-dG and mt DNA by real-time polymerase chain reaction. Western blot analysis was performed for nuclear-encoded mitochondrial proteins and DNA damage markers. Results When pregnant women were compared to non-pregnant women in their first, second, and third trimesters, a highly significant progressive drop in circulating mtDNA was found. In addition, mtDNA was considerably higher in mothers carrying IUGR fetuses than in healthy pregnancies. Sirtuin-3 protein expression was considerably suppressed in the IUGR placenta (P = 0.027), whereas Nrf1 expression was not statistically different from the control group in the IUGR. Increased oxidative stress led to greater DNA damage in IUGR. The highest concentrations of 8-OH-dG were found in IUGR with levels significantly higher than those in the non-pregnant group. Conclusion Our research sets the path for further investigation into mitochondrial anomalies in IUGR pregnancies and offers evidence for disturbed mitochondrial homeostasis. The mtDNA might offer a fresh perspective on the processes involved in physiological gestation. In addition, the presence of mtDNA may aid in the diagnosis of IUGR during pregnancy.
Collapse
Affiliation(s)
- Apurva Singh
- Department of Obstetrics and Gynecology, KGMU, Lucknow, Uttar Pradesh, India
- Photobiology Division, CSIR-Indian Institute of Toxicology Research, Lucknow, Uttar Pradesh, India
| | - Shyam Pyari Jaiswar
- Department of Obstetrics and Gynecology, KGMU, Lucknow, Uttar Pradesh, India
| | - Apala Priyadarshini
- Department of Obstetrics and Gynecology, KGMU, Lucknow, Uttar Pradesh, India
| | - Sujata Deo
- Department of Obstetrics and Gynecology, KGMU, Lucknow, Uttar Pradesh, India
| |
Collapse
|
12
|
Aquilani R, Cotta Ramusino M, Maestri R, Iadarola P, Boselli M, Perini G, Boschi F, Dossena M, Bellini A, Buonocore D, Doria E, Costa A, Verri M. Several dementia subtypes and mild cognitive impairment share brain reduction of neurotransmitter precursor amino acids, impaired energy metabolism, and lipid hyperoxidation. Front Aging Neurosci 2023; 15:1237469. [PMID: 37655338 PMCID: PMC10466813 DOI: 10.3389/fnagi.2023.1237469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2023] [Accepted: 08/03/2023] [Indexed: 09/02/2023] Open
Abstract
Objective Dementias and mild cognitive impairment (MCI) are associated with variously combined changes in the neurotransmitter system and signaling, from neurotransmitter synthesis to synaptic binding. The study tested the hypothesis that different dementia subtypes and MCI may share similar reductions of brain availability in amino acid precursors (AAPs) of neurotransmitter synthesis and concomitant similar impairment in energy production and increase of oxidative stress, i.e., two important metabolic alterations that impact neurotransmission. Materials and methods Sixty-five demented patients (Alzheimer's disease, AD, n = 44; frontotemporal disease, FTD, n = 13; vascular disease, VaD, n = 8), 10 subjects with MCI and 15 control subjects (CTRL) were recruited for this study. Cerebrospinal fluid (CSF) and plasma levels of AAPs, energy substrates (lactate, pyruvate), and an oxidative stress marker (malondialdehyde, MDA) were measured in all participants. Results Demented patients and subjects with MCI were similar for age, anthropometric parameters, biohumoral variables, insulin resistance (HOMA index model), and CSF neuropathology markers. Compared to age-matched CTRL, both demented patients and MCI subjects showed low CSF AAP tyrosine (precursor of dopamine and catecholamines), tryptophan (precursor of serotonin), methionine (precursor of acetylcholine) limited to AD and FTD, and phenylalanine (an essential amino acid largely used for protein synthesis) (p = 0.03 to <0.0001). No significant differences were found among dementia subtypes or between each dementia subtype and MCI subjects. In addition, demented patients and MCI subjects, compared to CTRL, had similar increases in CSF and plasma levels of pyruvate (CSF: p = 0.023 to <0.0001; plasma: p < 0.002 to <0.0001) and MDA (CSF: p < 0.035 to 0.002; plasma: p < 0.0001). Only in AD patients was the CSF level of lactate higher than in CTRL (p = 0.003). Lactate/pyruvate ratios were lower in all experimental groups than in CTRL. Conclusion AD, FTD, and VaD dementia patients and MCI subjects may share similar deficits in AAPs, partly in energy substrates, and similar increases in oxidative stress. These metabolic alterations may be due to AAP overconsumption following high brain protein turnover (leading to phenylalanine reductions), altered mitochondrial structure and function, and an excess of free radical production. All these metabolic alterations may have a negative impact on synaptic plasticity and activity.
Collapse
Affiliation(s)
- Roberto Aquilani
- Department of Biology and Biotechnology, “Lazzaro Spallanzani,” University of Pavia, Pavia, Italy
| | - Matteo Cotta Ramusino
- Unit of Behavioral Neurology and Center for Cognitive Disorders and Dementia, IRCCS C. Mondino Foundation, Pavia, Italy
- Dementia Research Center, IRCCS Mondino Foundation, Pavia, Italy
| | - Roberto Maestri
- Department of Biomedical Engineering of the Montescano Institute, Istituti Clinici Scientifici Maugeri IRCCS, Montescano, Italy
| | - Paolo Iadarola
- Department of Biology and Biotechnology, “Lazzaro Spallanzani,” University of Pavia, Pavia, Italy
| | - Mirella Boselli
- Neurorehabilitation Unit of the Montescano Institute, Istituti Clinici Scientifici Maugeri IRCCS, Montescano, Italy
| | - Giulia Perini
- Unit of Behavioral Neurology and Center for Cognitive Disorders and Dementia, IRCCS C. Mondino Foundation, Pavia, Italy
- Dementia Research Center, IRCCS Mondino Foundation, Pavia, Italy
| | - Federica Boschi
- Department of Drug Sciences, University of Pavia, Pavia, Italy
| | - Maurizia Dossena
- Department of Biology and Biotechnology, “Lazzaro Spallanzani,” University of Pavia, Pavia, Italy
| | - Anna Bellini
- Department of Biology and Biotechnology, “Lazzaro Spallanzani,” University of Pavia, Pavia, Italy
| | - Daniela Buonocore
- Department of Biology and Biotechnology, “Lazzaro Spallanzani,” University of Pavia, Pavia, Italy
| | - Enrico Doria
- Department of Biology and Biotechnology, “Lazzaro Spallanzani,” University of Pavia, Pavia, Italy
| | - Alfredo Costa
- Unit of Behavioral Neurology and Center for Cognitive Disorders and Dementia, IRCCS C. Mondino Foundation, Pavia, Italy
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
| | - Manuela Verri
- Department of Biology and Biotechnology, “Lazzaro Spallanzani,” University of Pavia, Pavia, Italy
| |
Collapse
|
13
|
Marks de Chabris NC, Sabir S, Perkins G, Cheng H, Ellisman MH, Pamenter ME. Short communication: Acute hypoxia does not alter mitochondrial abundance in naked mole-rats. Comp Biochem Physiol A Mol Integr Physiol 2023; 276:111343. [PMID: 36379380 DOI: 10.1016/j.cbpa.2022.111343] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 10/20/2022] [Accepted: 11/08/2022] [Indexed: 11/15/2022]
Abstract
Hypoxia poses a significant energetic challenge and most species exhibit metabolic remodelling when exposed to prolonged hypoxia. One component of this remodelling is mitochondrial biogenesis/mitophagy, which alter mitochondrial abundance and helps to adjust metabolic throughput to match changes in energy demands in hypoxia. However, how acute hypoxia impacts mitochondrial abundance in hypoxia-tolerant species is poorly understood. To help address this gap, we exposed hypoxia-tolerant naked mole-rats to 3 h of normoxia or acute hypoxia (5% O2) and measured changes in mitochondrial abundance using two well-established markers: citrate synthase (CS) enzyme activity and mitochondrial DNA (mtDNA) abundance. We found that neither marker changed with hypoxia in brain, liver, or kidney, suggesting that mitochondrial biogenesis is not initiated during acute hypoxia in these tissues. Conversely in skeletal muscle, the ratio of CS activity to total protein decreased 50% with hypoxia. However, this change was likely driven by an increase in soluble protein density in hypoxia because CS activity was unchanged relative to wet tissue weight and the mtDNA copy number was unchanged. To confirm this, we examined skeletal muscle mitochondria using transmission electron microscopy and found no change in mitochondrial volume density. Taken together with previous studies of mitochondrial respiratory function, our present findings suggest that naked mole-rats primarily rely on tissue-specific functional remodelling of metabolic pathways and mitochondrial respiratory throughput, and not physical changes in mitochondrial number or volume, to adjust to short-term hypoxic exposure.
Collapse
Affiliation(s)
| | - Soulene Sabir
- Department of Biology, University of Ottawa, Ottawa, ON, Canada
| | - Guy Perkins
- National Center for Microscopy and Imaging Research, Department of Neurosciences, University of California San Diego, La Jolla, CA, USA
| | - Hang Cheng
- Department of Biology, University of Ottawa, Ottawa, ON, Canada
| | - Marc H Ellisman
- National Center for Microscopy and Imaging Research, Department of Neurosciences, University of California San Diego, La Jolla, CA, USA
| | - Matthew E Pamenter
- Department of Biology, University of Ottawa, Ottawa, ON, Canada; University of Ottawa Brain and Mind Research Institute, Ottawa, ON, Canada.
| |
Collapse
|
14
|
Correia SC, Alves MG, Oliveira PF, Casadesus G, LaManna J, Perry G, Moreira PI. Hypoxic Preconditioning Averts Sporadic Alzheimer's Disease-Like Phenotype in Rats: A Focus on Mitochondria. Antioxid Redox Signal 2022; 37:739-757. [PMID: 35316086 DOI: 10.1089/ars.2019.8007] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Aims: Brief episodes of sublethal hypoxia reprogram brain response to face possible subsequent lethal stimuli by triggering adaptive and prosurvival events-a phenomenon denominated hypoxic preconditioning (HP). To date, the potential therapeutic implications of HP to forestall sporadic Alzheimer's disease (sAD) pathology remain unexplored. Using a well-established protocol of HP and focusing on hippocampus as a first brain region affected in AD, this study was undertaken to investigate the potential protective effects of HP in a sAD rat model induced by the intracerebroventricular (icv) administration of streptozotocin (STZ) and to uncover the mitochondrial adaptations underlying this nonpharmacological strategy. Results: HP prevented the memory and learning deficits as well as tau pathology in the icvSTZ rat model. HP also attenuated icvSTZ-related reactive astrogliosis, as noted by increased glial fibrillary acidic protein immunoreactivity and myo-inositol levels. Notably, HP abrogated the icvSTZ-related impaired energy metabolism and oxidative damage. Particularly, HP averted increased lactate, glutamate, and succinate levels, and decreased mitochondrial respiratory chain function and mitochondrial DNA content. Concerning mitochondrial adaptations underlying HP-triggered tolerance to icvSTZ, preconditioned hippocampal mitochondria displayed an enhanced complex II-energized mitochondrial respiration, which resulted from a coordinated interaction between mitochondrial biogenesis and fusion-fission. Mitochondrial biogenesis was stimulated immediately after HP, whereas in a latter phase mitochondrial fusion-fission events are modulated favoring the generation of elongated mitochondria. Innovation and Conclusion: Overall, these results demonstrate for the first time that HP prevents the sAD-like phenotype, in part, by targeting mitochondria emerging as a preventive strategy in the context of AD. Antioxid. Redox Signal. 37, 739-757.
Collapse
Affiliation(s)
- Sónia C Correia
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal.,Center for Innovative Biomedicine and Biotechnology, and University of Coimbra, Coimbra, Portugal.,Institute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal
| | - Marco G Alves
- Unit for Multidisciplinary Research in Biomedicine (UMIB), Department of Anatomy, Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, Porto, Portugal
| | - Pedro F Oliveira
- QOPNA & LAQV, Department of Chemistry, University of Aveiro, Aveiro, Portugal
| | - Gemma Casadesus
- Department of Biological Sciences, Kent State University, Kent, Ohio, USA
| | - Joseph LaManna
- Department of Physiology & Biophysics, Case Western Reserve University, Cleveland, Ohio, USA
| | - George Perry
- Department of Biology, College of Sciences, University of Texas at San Antonio, San Antonio, Texas, USA
| | - Paula I Moreira
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal.,Center for Innovative Biomedicine and Biotechnology, and University of Coimbra, Coimbra, Portugal.,Laboratory of Physiology, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| |
Collapse
|
15
|
Gan AM, Tracz-Gaszewska Z, Ellert-Miklaszewska A, Navrulin VO, Ntambi JM, Dobrzyn P. Stearoyl-CoA Desaturase Regulates Angiogenesis and Energy Metabolism in Ischemic Cardiomyocytes. Int J Mol Sci 2022; 23:ijms231810459. [PMID: 36142371 PMCID: PMC9499489 DOI: 10.3390/ijms231810459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 09/05/2022] [Accepted: 09/07/2022] [Indexed: 11/21/2022] Open
Abstract
New blood vessel formation is a key component of the cardiac repair process after myocardial infarction (MI). Hypoxia following MI is a major driver of angiogenesis in the myocardium. Hypoxia-inducible factor 1α (HIF1α) is the key regulator of proangiogenic signaling. The present study found that stearoyl-CoA desaturase (SCD) significantly contributed to the induction of angiogenesis in the hypoxic myocardium independently of HIF1α expression. The pharmacological inhibition of SCD activity in HL-1 cardiomyocytes and SCD knockout in an animal model disturbed the expression and secretion of proangiogenic factors including vascular endothelial growth factor-A, proinflammatory cytokines (interleukin-1β, interleukin-6, tumor necrosis factor α, monocyte chemoattractant protein-1, and Rantes), metalloproteinase-9, and platelet-derived growth factor in ischemic cardiomyocytes. These disturbances affected the proangiogenic potential of ischemic cardiomyocytes after SCD depletion. Together with the most abundant SCD1 isoform, the heart-specific SCD4 isoform emerged as an important regulator of new blood vessel formation in the murine post-MI myocardium. We also provide evidence that SCD shapes energy metabolism of the ischemic heart by maintaining the shift from fatty acids to glucose as the substrate that is used for adenosine triphosphate production. Furthermore, we propose that the regulation of the proangiogenic properties of hypoxic cardiomyocytes by key modulators of metabolic signaling such as adenosine monophosphate kinase, protein kinase B (AKT), and peroxisome-proliferator-activated receptor-γ coactivator 1α/peroxisome proliferator-activated receptor α depends on SCD to some extent. Thus, our results reveal a novel mechanism that links SCD to cardiac repair processes after MI.
Collapse
Affiliation(s)
- Ana-Maria Gan
- Laboratory of Molecular Medical Biochemistry, Nencki Institute of Experimental Biology of Polish Academy of Sciences, 02-093 Warsaw, Poland
| | - Zuzanna Tracz-Gaszewska
- Laboratory of Molecular Medical Biochemistry, Nencki Institute of Experimental Biology of Polish Academy of Sciences, 02-093 Warsaw, Poland
| | - Aleksandra Ellert-Miklaszewska
- Laboratory of Molecular Neurobiology, Nencki Institute of Experimental Biology of Polish Academy of Sciences, 02-093 Warsaw, Poland
| | - Viktor O. Navrulin
- Laboratory of Molecular Medical Biochemistry, Nencki Institute of Experimental Biology of Polish Academy of Sciences, 02-093 Warsaw, Poland
| | - James M. Ntambi
- Departments of Biochemistry and Nutritional Sciences, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Pawel Dobrzyn
- Laboratory of Molecular Medical Biochemistry, Nencki Institute of Experimental Biology of Polish Academy of Sciences, 02-093 Warsaw, Poland
- Correspondence:
| |
Collapse
|
16
|
Lestón Pinilla L, Ugun-Klusek A, Rutella S, De Girolamo LA. Hypoxia Signaling in Parkinson's Disease: There Is Use in Asking "What HIF?". BIOLOGY 2021; 10:723. [PMID: 34439955 PMCID: PMC8389254 DOI: 10.3390/biology10080723] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Revised: 07/23/2021] [Accepted: 07/27/2021] [Indexed: 12/23/2022]
Abstract
Hypoxia is a condition characterized by insufficient tissue oxygenation, which results in impaired oxidative energy production. A reduction in cellular oxygen levels induces the stabilization of hypoxia inducible factor α (HIF-1α), master regulator of the molecular response to hypoxia, involved in maintaining cellular homeostasis and driving hypoxic adaptation through the control of gene expression. Due to its high energy requirement, the brain is particularly vulnerable to oxygen shortage. Thus, hypoxic injury can cause significant metabolic changes in neural cell populations, which are associated with neurodegeneration. Recent evidence suggests that regulating HIF-1α may ameliorate the cellular damage in neurodegenerative diseases. Indeed, the hypoxia/HIF-1α signaling pathway has been associated to several processes linked to Parkinson's disease (PD) including gene mutations, risk factors and molecular pathways such as mitochondrial dysfunction, oxidative stress and protein degradation impairment. This review will explore the impact of hypoxia and HIF-1α signaling on these specific molecular pathways that influence PD development and will evaluate different novel neuroprotective strategies involving HIF-1α stabilization.
Collapse
Affiliation(s)
- Laura Lestón Pinilla
- Interdisciplinary Biomedical Research Centre, Centre for Health, Ageing and Understanding Disease, School of Science & Technology, Nottingham Trent University, Clifton Lane, Nottingham NG11 8NS, UK;
| | - Aslihan Ugun-Klusek
- Interdisciplinary Biomedical Research Centre, Centre for Health, Ageing and Understanding Disease, School of Science & Technology, Nottingham Trent University, Clifton Lane, Nottingham NG11 8NS, UK;
| | - Sergio Rutella
- John van Geest Cancer Research Centre, Centre for Health, Ageing and Understanding Disease, School of Science & Technology, Nottingham Trent University, Clifton Lane, Nottingham NG11 8NS, UK;
| | - Luigi A. De Girolamo
- Interdisciplinary Biomedical Research Centre, Centre for Health, Ageing and Understanding Disease, School of Science & Technology, Nottingham Trent University, Clifton Lane, Nottingham NG11 8NS, UK;
| |
Collapse
|
17
|
Al Rahim M, Thatipamula S, Pasinetti GM, Hossain MA. Neuronal Pentraxin 1 Promotes Hypoxic-Ischemic Neuronal Injury by Impairing Mitochondrial Biogenesis via Interactions With Active Bax[6A7] and Mitochondrial Hexokinase II. ASN Neuro 2021; 13:17590914211012888. [PMID: 34098747 PMCID: PMC8191073 DOI: 10.1177/17590914211012888] [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] [Indexed: 11/17/2022] Open
Abstract
Mitochondrial dysfunction is a key mechanism of cell death in hypoxic-ischemic brain injury. Neuronal pentraxin 1 (NP1) has been shown to play crucial roles in mitochondria-mediated neuronal death. However, the underlying mechanism(s) of NP1-induced mitochondrial dysfunction in hypoxia-ischemia (HI) remains obscure. Here, we report that NP1 induction following HI and its subsequent localization to mitochondria, leads to disruption of key regulatory proteins for mitochondrial biogenesis. Brain mitochondrial DNA (mtDNA) content and mtDNA-encoded subunit I of complex IV (mtCOX-1) expression was increased post-HI, but not the nuclear DNA-encoded subunit of complex II (nSDH-A). Up-regulation of mitochondrial proteins COXIV and HSP60 further supported enhanced mtDNA function. NP1 interaction with active Bax (Bax6A7) was increased in the brain after HI and in oxygen-glucose deprivation (OGD)-induced neuronal cultures. Importantly, NP1 colocalized with mitochondrial hexokinase II (mtHKII) following OGD leading to HKII dissociation from mitochondria. Knockdown of NP1 or SB216763, a GSK-3 inhibitor, prevented OGD-induced mtHKII dissociation and cellular ATP decrease. NP1 also modulated the expression of mitochondrial transcription factor A (Tfam) and peroxisome proliferator-activated receptor γ coactivator-1α (PGC-1α), regulators of mitochondrial biogenesis, following HI. Together, we reveal crucial roles of NP1 in mitochondrial biogenesis involving interactions with Bax[6A7] and mtHKII in HI brain injury.
Collapse
Affiliation(s)
- Md Al Rahim
- Hugo W. Moser Research Institute at Kennedy Krieger, Baltimore, Maryland, United States.,Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States.,Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, New York, United States
| | - Shabarish Thatipamula
- Hugo W. Moser Research Institute at Kennedy Krieger, Baltimore, Maryland, United States.,Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| | - Giulio M Pasinetti
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, New York, United States.,James J. Peters Veterans Affairs Medical Center, Bronx, New York, United States
| | - Mir Ahamed Hossain
- Hugo W. Moser Research Institute at Kennedy Krieger, Baltimore, Maryland, United States.,Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| |
Collapse
|
18
|
Rational construction of a reversible arylazo-based NIR probe for cycling hypoxia imaging in vivo. Nat Commun 2021; 12:2772. [PMID: 33986258 PMCID: PMC8119430 DOI: 10.1038/s41467-021-22855-0] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Accepted: 03/29/2021] [Indexed: 02/07/2023] Open
Abstract
Reversible NIR luminescent probes with negligible photocytotoxicity are required for long-term tracking of cycling hypoxia in vivo. However, almost all of the reported organic fluorescent hypoxia probes reported until now were irreversible. Here we report a reversible arylazo-conjugated fluorescent probe (HDSF) for cycling hypoxia imaging. HDSF displays an off-on fluorescence switch at 705 nm in normoxia-hypoxia cycles. Mass spectroscopic and theoretical studies confirm that the reversible sensing behavior is attributed to the two electron-withdrawing trifluoromethyl groups, which stabilizes the reduction intermediate phenylhydrazine and blocks the further reductive decomposition. Cycling hypoxia monitoring in cells and zebrafish embryos is realized by HDSF using confocal imaging. Moreover, hypoxic solid tumors are visualized and the ischemia-reperfusion process in mice is monitored in real-time. This work provides an effective strategy to construct organic fluorescent probes for cycling hypoxia imaging and paves the way for the study of cycling hypoxia biology.
Collapse
|
19
|
Intermittent Hypoxic Conditioning Rescues Cognition and Mitochondrial Bioenergetic Profile in the Triple Transgenic Mouse Model of Alzheimer's Disease. Int J Mol Sci 2021; 22:ijms22010461. [PMID: 33466445 PMCID: PMC7796478 DOI: 10.3390/ijms22010461] [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: 12/20/2020] [Revised: 12/29/2020] [Accepted: 12/31/2020] [Indexed: 12/28/2022] Open
Abstract
The lack of effective disease-modifying therapeutics to tackle Alzheimer’s disease (AD) is unsettling considering the actual prevalence of this devastating neurodegenerative disorder worldwide. Intermittent hypoxic conditioning (IHC) is a powerful non-pharmacological procedure known to enhance brain resilience. In this context, the aim of the present study was to investigate the potential long-term protective impact of IHC against AD-related phenotype, putting a special focus on cognition and mitochondrial bioenergetics and dynamics. For this purpose, six-month-old male triple transgenic AD mice (3×Tg-AD) were submitted to an IHC protocol for two weeks and the behavioral assessment was performed at 8.5 months of age, while the sacrifice of mice occurred at nine months of age and their brains were removed for the remaining analyses. Interestingly, IHC was able to prevent anxiety-like behavior and memory and learning deficits and significantly reduced brain cortical levels of amyloid-β (Aβ) in 3×Tg-AD mice. Concerning brain energy metabolism, IHC caused a significant increase in brain cortical levels of glucose and a robust improvement of the mitochondrial bioenergetic profile in 3×Tg-AD mice, as mirrored by the significant increase in mitochondrial membrane potential (ΔΨm) and respiratory control ratio (RCR). Notably, the improvement of mitochondrial bioenergetics seems to result from an adaptative coordination of the distinct but intertwined aspects of the mitochondrial quality control axis. Particularly, our results indicate that IHC favors mitochondrial fusion and promotes mitochondrial biogenesis and transport and mitophagy in the brain cortex of 3×Tg-AD mice. Lastly, IHC also induced a marked reduction in synaptosomal-associated protein 25 kDa (SNAP-25) levels and a significant increase in both glutamate and GABA levels in the brain cortex of 3×Tg-AD mice, suggesting a remodeling of the synaptic microenvironment. Overall, these results demonstrate the effectiveness of the IHC paradigm in forestalling the AD-related phenotype in the 3×Tg-AD mouse model, offering new insights to AD therapy and forcing a rethink concerning the potential value of non-pharmacological interventions in clinical practice.
Collapse
|
20
|
Bottani E, Lamperti C, Prigione A, Tiranti V, Persico N, Brunetti D. Therapeutic Approaches to Treat Mitochondrial Diseases: "One-Size-Fits-All" and "Precision Medicine" Strategies. Pharmaceutics 2020; 12:E1083. [PMID: 33187380 PMCID: PMC7696526 DOI: 10.3390/pharmaceutics12111083] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 11/08/2020] [Accepted: 11/09/2020] [Indexed: 12/11/2022] Open
Abstract
Primary mitochondrial diseases (PMD) refer to a group of severe, often inherited genetic conditions due to mutations in the mitochondrial genome or in the nuclear genes encoding for proteins involved in oxidative phosphorylation (OXPHOS). The mutations hamper the last step of aerobic metabolism, affecting the primary source of cellular ATP synthesis. Mitochondrial diseases are characterized by extremely heterogeneous symptoms, ranging from organ-specific to multisystemic dysfunction with different clinical courses. The limited information of the natural history, the limitations of currently available preclinical models, coupled with the large variability of phenotypical presentations of PMD patients, have strongly penalized the development of effective therapies. However, new therapeutic strategies have been emerging, often with promising preclinical and clinical results. Here we review the state of the art on experimental treatments for mitochondrial diseases, presenting "one-size-fits-all" approaches and precision medicine strategies. Finally, we propose novel perspective therapeutic plans, either based on preclinical studies or currently used for other genetic or metabolic diseases that could be transferred to PMD.
Collapse
Affiliation(s)
- Emanuela Bottani
- Department of Diagnostics and Public Health, Section of Pharmacology, University of Verona, 37134 Verona, Italy
| | - Costanza Lamperti
- Medical Genetics and Neurogenetics Unit, Fondazione IRCCS Istituto Neurologico C. Besta, 20126 Milan, Italy; (C.L.); (V.T.)
| | - Alessandro Prigione
- Department of General Pediatrics, Neonatology, and Pediatric Cardiology, University Clinic Düsseldorf (UKD), Heinrich Heine University (HHU), 40225 Dusseldorf, Germany;
| | - Valeria Tiranti
- Medical Genetics and Neurogenetics Unit, Fondazione IRCCS Istituto Neurologico C. Besta, 20126 Milan, Italy; (C.L.); (V.T.)
| | - Nicola Persico
- Department of Clinical Science and Community Health, University of Milan, 20122 Milan, Italy;
- Fetal Medicine and Surgery Service, Fondazione IRCCS Ca’ Granda, Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Dario Brunetti
- Medical Genetics and Neurogenetics Unit, Fondazione IRCCS Istituto Neurologico C. Besta, 20126 Milan, Italy; (C.L.); (V.T.)
- Department of Medical Biotechnology and Translational Medicine, University of Milan, 20129 Milan, Italy
| |
Collapse
|
21
|
Sun S, Jiang T, Duan N, Wu M, Yan C, Li Y, Cai M, Wang Q. Activation of CB1R-Dependent PGC-1α Is Involved in the Improved Mitochondrial Biogenesis Induced by Electroacupuncture Pretreatment. Rejuvenation Res 2020; 24:104-119. [PMID: 32746712 DOI: 10.1089/rej.2020.2315] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Electroacupuncture (EA) pretreatment induces cerebral ischemic tolerance; however, the mechanism remains poorly understood. This study aimed to determine the participation of peroxisome proliferator-activated receptor γ coactivator-1α (PGC-1α)-mediated mitochondrial biogenesis in the neuroprotection of EA and whether cannabinoid receptor 1 (CB1R) is involved in this mechanism. At 2 hours after EA pretreatment, adult male C57BL/6j mice were subjected to 60-minute right middle cerebral artery occlusion (MCAO). Mitochondrial function, the level of mitochondrial biogenesis-related proteins (nuclear transcription factor 1, NRF1; mitochondrial transcription factor A, TFAM), and mitochondrial DNA (mtDNA) were measured. A small interfering RNA (siRNA) targeting PGC-1α and the CB1R antagonists AM251 and SR141716A were given to the animals before EA pretreatment, and mitochondrial function and biogenesis were examined after MCAO. EA ameliorated the mitochondrial function, upregulated the NRF1 and TFAM expression, and increased the mtDNA levels and the volume and number of mitochondria. EA pretreatment increased the expression of PGC-1α, whereas the PGC-1α siRNA and CB1R antagonists reversed the improved neuroprotection and increased mitochondrial biogenesis induced by EA. Our results indicated that EA pretreatment protects the mitochondria and promotes mitochondrial biogenesis by activating CB1R-dependent PGC-1α, which provides a novel mechanism for EA pretreatment-induced ischemic tolerance.
Collapse
Affiliation(s)
- Sisi Sun
- Department of Anesthesiology and Center for Brain Science, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.,The Medical Department of the Emergency Centre of Xi'an, Xi'an, China
| | - Tao Jiang
- Department of Anesthesiology and Center for Brain Science, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Na Duan
- Department of Anesthesiology and Center for Brain Science, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Meiyan Wu
- Department of Anesthesiology and Center for Brain Science, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Chaoying Yan
- Department of Anesthesiology and Center for Brain Science, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yan Li
- Department of Anesthesiology and Center for Brain Science, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Min Cai
- Department of Psychiatry, Xijing Hospital, the Fourth Military Medical University, Xi'an, China
| | - Qiang Wang
- Department of Anesthesiology and Center for Brain Science, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
22
|
Nguyen KC, Zhang Y, Todd J, Kittle K, Lalande M, Smith S, Parks D, Navarro M, Tayabali AF, Willmore WG. Hepatotoxicity of Cadmium Telluride Quantum Dots Induced by Mitochondrial Dysfunction. Chem Res Toxicol 2020; 33:2286-2297. [PMID: 32844644 DOI: 10.1021/acs.chemrestox.9b00526] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The aim of this study was to investigate the detailed mechanisms of hepatotoxicity induced by cadmium telluride quantum dots (CdTe-QDs) in BALB/c mice after intravenous injection. The study investigated oxidative stress, apoptosis, and effects on mitochondria as potential mechanistic events to elucidate the observed hepatotoxicity. Oxidative stress in the liver, induced by CdTe-QD exposure, was demonstrated by depletion of total glutathione, an increase in superoxide dismutase activity, and changes in the gene expression of several oxidative stress-related biomarkers. Furthermore, CdTe-QD treatment led to apoptosis in the liver via both intrinsic and extrinsic apoptotic pathways. Effects on mitochondria were evidenced by the enlargement and increase in the number of mitochondria in hepatocytes of treated mice. CdTe-QDs also caused changes in the levels and gene expression of electron transport chain enzymes, depletion of ATP, and an increase in the level of the peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC-1α), a regulator of mitochondrial biogenesis. The findings from this study suggest that CdTe-QDs-induced hepatotoxicity might have originated from mitochondrial effects which resulted in oxidative stress and apoptosis in the liver cells. This study provides insight into the biological effects of CdT-QDs at the tissue level and the detailed mechanisms of their toxicity in animals. The study also provides important data for bridging the gap between in vitro and in vivo testing and risk assessment of these NPs.
Collapse
Affiliation(s)
- Kathy C Nguyen
- Environmental Health Science and Research Bureau, Healthy Environments and Consumer Safety Branch, Health Canada, 50 Colombine Driveway, Ottawa, Ontario K1A 0K9, Canada.,Department of Biology and Institute of Biochemistry, Carleton University, 1125 Colonel By Drive, Ottawa, Ontario K1S 5B6, Canada
| | - Yan Zhang
- Environmental Health Science and Research Bureau, Healthy Environments and Consumer Safety Branch, Health Canada, 50 Colombine Driveway, Ottawa, Ontario K1A 0K9, Canada
| | - Julie Todd
- Bureau of Chemical Safety, Health Products and Food Branch, Frederick G. Banting Building, Health Canada, Ottawa, Ontario K1A 0K9, Canada
| | - Kevin Kittle
- Bureau of Chemical Safety, Health Products and Food Branch, Frederick G. Banting Building, Health Canada, Ottawa, Ontario K1A 0K9, Canada
| | - Michelle Lalande
- Bureau of Chemical Safety, Health Products and Food Branch, Frederick G. Banting Building, Health Canada, Ottawa, Ontario K1A 0K9, Canada
| | - Scott Smith
- Bureau of Chemical Safety, Health Products and Food Branch, Frederick G. Banting Building, Health Canada, Ottawa, Ontario K1A 0K9, Canada
| | - Douglas Parks
- Bureau of Chemical Safety, Health Products and Food Branch, Frederick G. Banting Building, Health Canada, Ottawa, Ontario K1A 0K9, Canada
| | - Martha Navarro
- Bureau of Chemical Safety, Health Products and Food Branch, Frederick G. Banting Building, Health Canada, Ottawa, Ontario K1A 0K9, Canada
| | - Azam F Tayabali
- Environmental Health Science and Research Bureau, Healthy Environments and Consumer Safety Branch, Health Canada, 50 Colombine Driveway, Ottawa, Ontario K1A 0K9, Canada.,Department of Biology and Institute of Biochemistry, Carleton University, 1125 Colonel By Drive, Ottawa, Ontario K1S 5B6, Canada
| | - William G Willmore
- Department of Biology and Institute of Biochemistry, Carleton University, 1125 Colonel By Drive, Ottawa, Ontario K1S 5B6, Canada
| |
Collapse
|
23
|
Verma V, Kumar P, Gupta S, Yadav S, Dhanda RS, Thorlacius H, Yadav M. α-Hemolysin of uropathogenic E. coli regulates NLRP3 inflammasome activation and mitochondrial dysfunction in THP-1 macrophages. Sci Rep 2020; 10:12653. [PMID: 32724079 PMCID: PMC7387347 DOI: 10.1038/s41598-020-69501-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Accepted: 07/07/2020] [Indexed: 12/16/2022] Open
Abstract
Hemolysin expressing UPEC strains have been associated with severe advanced kidney pathologies, such as cystitis and pyelonephritis, which are associated with an inflammatory response. Macrophages play an important role in regulating an inflammatory response during a urinary tract infection. We have studied the role of purified recombinant α-hemolysin in inducing inflammatory responses and cell death in macrophages. Acylation at lysine residues through HlyC is known to activate proHlyA into a fully functional pore-forming toxin, HlyA. It was observed that active α-hemolysin (HlyA) induced cleavage of caspase-1 leading to the maturation of IL-1β, while inactive α-hemolysin (proHlyA) failed to do so in THP-1 derived macrophages. HlyA also promotes deubiquitination, oligomerization, and activation of the NLRP3 inflammasome, which was found to be dependent on potassium efflux. We have also observed the co-localization of NLRP3 within mitochondria during HlyA stimulations. Moreover, blocking of potassium efflux improved the mitochondrial health in addition to a decreased inflammatory response. Our study demonstrates that HlyA stimulation caused perturbance in potassium homeostasis, which led to the mitochondrial dysfunction followed by an acute inflammatory response, resulting in cell death. However, the repletion of intracellular potassium stores could avoid HlyA induced macrophage cell death. The findings of this study will help to understand the mechanism of α-hemolysin induced inflammatory response and cell death.
Collapse
Affiliation(s)
- Vivek Verma
- Dr. B. R. Ambedkar Center for Biomedical Research, University of Delhi (North Campus), Delhi, 110007, India
| | - Parveen Kumar
- Department of Urology, University of Alabama At Birmingham, Hugh Kaul Genetics Building, Birmingham, AL, USA
| | - Surbhi Gupta
- Dr. B. R. Ambedkar Center for Biomedical Research, University of Delhi (North Campus), Delhi, 110007, India
| | - Sonal Yadav
- Dr. B. R. Ambedkar Center for Biomedical Research, University of Delhi (North Campus), Delhi, 110007, India
| | - Rakesh Singh Dhanda
- Stem Cell Laboratory, Longboat Explorers AB, SMiLE Incubator, Scheelevägen 2, Lund, Sweden
| | - Henrik Thorlacius
- Department of Clinical Sciences, Section of Surgery, Malmö, Skåne University Hospital, Lund University, Malmö, Sweden
| | - Manisha Yadav
- Dr. B. R. Ambedkar Center for Biomedical Research, University of Delhi (North Campus), Delhi, 110007, India.
- Department of Clinical Sciences, Section of Surgery, Malmö, Skåne University Hospital, Lund University, Malmö, Sweden.
| |
Collapse
|
24
|
Sanders O, Rajagopal L. Phosphodiesterase Inhibitors for Alzheimer's Disease: A Systematic Review of Clinical Trials and Epidemiology with a Mechanistic Rationale. J Alzheimers Dis Rep 2020; 4:185-215. [PMID: 32715279 PMCID: PMC7369141 DOI: 10.3233/adr-200191] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/17/2020] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Preclinical studies, clinical trials, and reviews suggest increasing 3',5'-cyclic adenosine monophosphate (cAMP) and 3',5'-cyclic guanosine monophosphate (cGMP) with phosphodiesterase inhibitors is disease-modifying in Alzheimer's disease (AD). cAMP/protein kinase A (PKA) and cGMP/protein kinase G (PKG) signaling are disrupted in AD. cAMP/PKA and cGMP/PKG activate cAMP response element binding protein (CREB). CREB binds mitochondrial and nuclear DNA, inducing synaptogenesis, memory, and neuronal survival gene (e.g., brain-derived neurotrophic factor) and peroxisome proliferator-activated receptor-γ coactivator-1α (PGC1α). cAMP/PKA and cGMP/PKG activate Sirtuin-1, which activates PGC1α. PGC1α induces mitochondrial biogenesis and antioxidant genes (e.g.,Nrf2) and represses BACE1. cAMP and cGMP inhibit BACE1-inducing NFκB and tau-phosphorylating GSK3β. OBJECTIVE AND METHODS We review efficacy-testing clinical trials, epidemiology, and meta-analyses to critically investigate whether phosphodiesteraseinhibitors prevent or treat AD. RESULTS Caffeine and cilostazol may lower AD risk. Denbufylline and sildenafil clinical trials are promising but preliminary and inconclusive. PF-04447943 and BI 409,306 are ineffective. Vinpocetine, cilostazol, and nicergoline trials are mixed. Deprenyl/selegiline trials show only short-term benefits. Broad-spectrum phosphodiesterase inhibitor propentofylline has been shown in five phase III trials to improve cognition, dementia severity, activities of daily living, and global assessment in mild-to-moderate AD patients on multiple scales, including the ADAS-Cogand the CIBIC-Plus in an 18-month phase III clinical trial. However, two books claimed based on a MedScape article an 18-month phase III trial failed, so propentofylline was discontinued. Now, propentofylline is used to treat canine cognitive dysfunction, which, like AD, involves age-associated wild-type Aβ deposition. CONCLUSION Phosphodiesterase inhibitors may prevent and treat AD.
Collapse
|
25
|
Abstract
Nitric oxide/cyclic guanosine monophosphate (cGMP) signaling is compromised in Alzheimer’s disease (AD), and phosphodiesterase 5 (PDE5), which degrades cGMP, is upregulated. Sildenafil inhibits PDE5 and increases cGMP levels. Integrating previous findings, we determine that most doses of sildenafil (especially low doses) likely activate peroxisome proliferator-activated receptor-γ coactivator 1α (PGC1α) via protein kinase G-mediated cyclic adenosine monophosphate (cAMP) response element binding protein (CREB) phosphorylation and/or Sirtuin-1 activation and PGC1α deacetylation. Via PGC1α signaling, low-dose sildenafil likely suppresses β-secretase 1 expression and amyloid-β (Aβ) generation, upregulates antioxidant enzymes, and induces mitochondrial biogenesis. Plus, sildenafil should increase brain perfusion, insulin sensitivity, long-term potentiation, and neurogenesis while suppressing neural apoptosis and inflammation. A systematic review of sildenafil in AD was undertaken. In vitro, sildenafil protected neural mitochondria from Aβ and advanced glycation end products. In transgenic AD mice, sildenafil was found to rescue deficits in CREB phosphorylation and memory, upregulate brain-derived neurotrophic factor, reduce reactive astrocytes and microglia, decrease interleukin-1β, interleukin-6, and tumor necrosis factor-α, decrease neural apoptosis, increase neurogenesis, and reduce tau hyperphosphorylation. All studies that tested Aβ levels reported significant improvements except the two that used the highest dosage, consistent with the dose-limiting effect of cGMP-induced phosphodiesterase 2 (PDE2) activation and cAMP depletion on PGC1α signaling. In AD patients, a single dose of sildenafil decreased spontaneous neural activity, increased cerebral blood flow, and increased the cerebral metabolic rate of oxygen. A randomized control trial of sildenafil (ideally with a PDE2 inhibitor) in AD patients is warranted.
Collapse
|
26
|
Jia L, Wang J, Cao H, Zhang X, Rong W, Xu Z. Activation of PGC-1α and Mitochondrial Biogenesis Protects Against Prenatal Hypoxic-ischemic Brain Injury. Neuroscience 2020; 432:63-72. [PMID: 32114097 DOI: 10.1016/j.neuroscience.2020.02.035] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 02/18/2020] [Accepted: 02/19/2020] [Indexed: 12/22/2022]
Abstract
Survivals after prenatal hypoxia-ischemia (HI) usually suffer long-lasting cognitive defects. Reduced blood-oxygen supplies and the following reperfusion cause mitochondrial injury. Damaged mitochondria could be replaced by mitochondrial biogenesis program and peroxisome proliferator-activated receptor γ co-activator 1α (PGC-1α) is the specific up-regulator. The objective of this study was to determine whether PGC-1α and mitochondrial biogenesis participate in the resistant responses of an immature brain to prenatal HI. We used a pregnant rat model of transient occlusion of uterine perfusion to induce intrauterine HI associated brain injury. SH-SY5Y cells exposed to oxygen-glucose deprivation was used to investigate the HI induced reactions in vitro. PGC-1α and its downstream signaling pathway (NRF-1 and TFAM) were examined by Western blot and quantitative Real-time PCR. Mitochondrial respiratory enzyme COX-IV was investigated by Western blot and immunohistochemistry. Mitochondrial density and morphology was detected by transmission electron microscopy. The hippocampal injury and cognitive function were examined. We found that the intrauterine HI triggered PGC-1α-NRF-1-TFAM pathway in both protein and mRNA levels. COX-IV expression significantly increased after HI injury. Intrauterine HI induced both mitochondrial impairment and mitochondrial biogenesis. Postnatal administration of pioglitazone further promoted PGC-1α and mitochondrial biogenesis, alleviated hippocampal injury, and improved performance in the behavioral tasks after intrauterine HI. Our investigation implicated activation of PGC-1α, and mitochondrial biogenesis is a neuroprotective mechanism against brain injury caused by systemic prenatal HI. Promotion of PGC-1α by pioglitazone might be a potential treatment for protecting against hippocampal injury and cognitive defects after intrauterine HI.
Collapse
Affiliation(s)
- Lijie Jia
- Department of Anesthesiology, the International Peace Maternity and Child Health Hospital, Shanghai Jiaotong University School of Medicine, 910 Hengshan Road, Shanghai 200030, China; Shanghai Key Laboratory of Embryo Original Diseases, 910 Hengshan Road, Shanghai 200030, China
| | - Jianwei Wang
- Department of Anesthesiology, the International Peace Maternity and Child Health Hospital, Shanghai Jiaotong University School of Medicine, 910 Hengshan Road, Shanghai 200030, China; Shanghai Key Laboratory of Embryo Original Diseases, 910 Hengshan Road, Shanghai 200030, China
| | - Huimin Cao
- Department of Anesthesiology, the International Peace Maternity and Child Health Hospital, Shanghai Jiaotong University School of Medicine, 910 Hengshan Road, Shanghai 200030, China; Shanghai Key Laboratory of Embryo Original Diseases, 910 Hengshan Road, Shanghai 200030, China
| | - Xiaoyu Zhang
- Department of Anesthesiology, the International Peace Maternity and Child Health Hospital, Shanghai Jiaotong University School of Medicine, 910 Hengshan Road, Shanghai 200030, China; Shanghai Key Laboratory of Embryo Original Diseases, 910 Hengshan Road, Shanghai 200030, China
| | - Weifang Rong
- Department of Anatomy and Physiology, Shanghai Jiaotong University School of Medicine, 280 South Chongqing Road, Shanghai 200025, China.
| | - Zifeng Xu
- Department of Anesthesiology, the International Peace Maternity and Child Health Hospital, Shanghai Jiaotong University School of Medicine, 910 Hengshan Road, Shanghai 200030, China; Shanghai Key Laboratory of Embryo Original Diseases, 910 Hengshan Road, Shanghai 200030, China.
| |
Collapse
|
27
|
Khedr LH, Nassar NN, Rashed L, El-Denshary ED, Abdel-Tawab AM. TLR4 signaling modulation of PGC1-α mediated mitochondrial biogenesis in the LPS-Chronic mild stress model: Effect of fluoxetine and pentoxiyfylline. Life Sci 2019; 239:116869. [PMID: 31678277 DOI: 10.1016/j.lfs.2019.116869] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Revised: 09/10/2019] [Accepted: 09/10/2019] [Indexed: 11/15/2022]
Abstract
AIM The addition of repeated lipopolysaccharide (LPS) to chronic mild stress was recently proposed in our lab as an alternative model of depression, highlighting the possible interaction between stress and immune-inflammatory pathways in predisposing depression. Given that CMS-induced depressive behavior was previously related to impaired hippocampal energy metabolism and mitochondrial dysfunction, our current study aimed to investigate the interplay between toll-like receptor 4 (TLR4) signaling and peroxisome proliferator-activated receptor gamma coactivators-1-alpha (PGC1-α) as a physiological regulator of energy metabolism and mitochondrial biogenesis in the combined LPS/CMS model. MAIN METHODS Male Wistar rats were exposed to either LPS (50 μg/kg i.p.) over 2 weeks, CMS protocol for 4 weeks or LPS over 2 weeks followed by 4 weeks of CMS (LPS/CMS). Three additional groups of rats were exposed to LPS/CMS protocol and treated with either pentoxifylline (PTX), fluoxetine (FLX) or a combination of both. Rats were examined for behavioral, neurochemical, gene expression and mitochondrial ultra-structural changes. KEY FINDINGS LPS/CMS increased the expression of TLR4 and its downstream players; MyD88, NFκB and TNF-α along with an escalation in hippocampal-energy metabolism and p-AMPK. Simultaneously LPS/CMS attenuated the expression of PGC1-α/NRF1/Tfam and mt-DNA. The antidepressant (AD) 'FLX', the TNF-α inhibitor 'PTX' and their combination ameliorated the LPS/CMS-induced changes. Interestingly, all the aforementioned changes induced by the LPS/CMS combined model were significantly less than those induced by CMS alone. SIGNIFICANCE Blocking the TLR4/NFκB signaling enhanced the activation of the PGC1-α/NRF1/Tfam and mt-DNA content independent on the activation of the energy-sensing kinase AMPK.
Collapse
Affiliation(s)
- L H Khedr
- Departmment of Pharmacology, Faculty of Pharmacy, Misr International University, Cairo, Egypt.
| | - N N Nassar
- Department of Pharmacology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Laila Rashed
- Department of Biochemistry, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - E D El-Denshary
- Department of Pharmacology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - A M Abdel-Tawab
- Department of Pharmacology, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| |
Collapse
|
28
|
Rodríguez-Enríquez S, Marín-Hernández Á, Gallardo-Pérez JC, Pacheco-Velázquez SC, Belmont-Díaz JA, Robledo-Cadena DX, Vargas-Navarro JL, Corona de la Peña NA, Saavedra E, Moreno-Sánchez R. Transcriptional Regulation of Energy Metabolism in Cancer Cells. Cells 2019; 8:cells8101225. [PMID: 31600993 PMCID: PMC6830338 DOI: 10.3390/cells8101225] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 09/19/2019] [Accepted: 10/01/2019] [Indexed: 01/17/2023] Open
Abstract
Cancer development, growth, and metastasis are highly regulated by several transcription regulators (TRs), namely transcription factors, oncogenes, tumor-suppressor genes, and protein kinases. Although TR roles in these events have been well characterized, their functions in regulating other important cancer cell processes, such as metabolism, have not been systematically examined. In this review, we describe, analyze, and strive to reconstruct the regulatory networks of several TRs acting in the energy metabolism pathways, glycolysis (and its main branching reactions), and oxidative phosphorylation of nonmetastatic and metastatic cancer cells. Moreover, we propose which possible gene targets might allow these TRs to facilitate the modulation of each energy metabolism pathway, depending on the tumor microenvironment.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Norma Angélica Corona de la Peña
- Unidad de Investigación Médica en Trombosis, Hemostasia y Aterogénesis, Hospital General Regional Carlos McGregor-Sánchez, México CP 03100, Mexico.
| | - Emma Saavedra
- Departamento de Bioquímica, Instituto Nacional de Cardiología, México 14080, Mexico.
| | - Rafael Moreno-Sánchez
- Departamento de Bioquímica, Instituto Nacional de Cardiología, México 14080, Mexico.
| |
Collapse
|
29
|
|
30
|
Ross CI, Shute RJ, Ruby BC, Slivka DR. Skeletal Muscle mRNA Response to Hypobaric and Normobaric Hypoxia After Normoxic Endurance Exercise. High Alt Med Biol 2019; 20:141-149. [DOI: 10.1089/ham.2018.0147] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Affiliation(s)
- Caleb I. Ross
- Exercise Physiology Lab, University of Nebraska at Omaha, Omaha, Nebraska
| | - Robert J. Shute
- Exercise Physiology Lab, University of Nebraska at Omaha, Omaha, Nebraska
| | - Brent C. Ruby
- Montana Center for Work Physiology and Exercise Metabolism, University of Montana, Missoula, Montana
| | - Dustin R. Slivka
- Exercise Physiology Lab, University of Nebraska at Omaha, Omaha, Nebraska
| |
Collapse
|
31
|
Placental mitochondria adapt developmentally and in response to hypoxia to support fetal growth. Proc Natl Acad Sci U S A 2019; 116:1621-1626. [PMID: 30655345 DOI: 10.1073/pnas.1816056116] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Mitochondria respond to a range of stimuli and function in energy production and redox homeostasis. However, little is known about the developmental and environmental control of mitochondria in the placenta, an organ vital for fetal growth and pregnancy maintenance in eutherian mammals. Using respirometry and molecular analyses, the present study examined mitochondrial function in the distinct transport and endocrine zones of the mouse placenta during normal pregnancy and maternal inhalation hypoxia. The data show that mitochondria of the two zones adopt different strategies in modulating their respiration, substrate use, biogenesis, density, and efficiency to best support the growth and energy demands of fetoplacental tissues during late gestation in both normal and hypoxic conditions. The findings have important implications for environmentally induced adaptations in mitochondrial function in other tissues and for compromised human pregnancy in which hypoxia and alterations in placental mitochondrial function are associated with poor outcomes like fetal growth restriction.
Collapse
|
32
|
Lo Martire V, Alvente S, Bastianini S, Berteotti C, Valli A, Manconi M, Zoccoli G, Silvani A. Sleep and Tibialis Anterior Muscle Activity in Mice With Mild Hypoxia and Iron Deficiency: Implications for the Restless Legs Syndrome. Front Physiol 2018; 9:1818. [PMID: 30618828 PMCID: PMC6304696 DOI: 10.3389/fphys.2018.01818] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Accepted: 12/05/2018] [Indexed: 12/14/2022] Open
Abstract
Restless legs syndrome (RLS) is a neurological disorder that entails an urge to move with a circadian pattern during the evening/night. RLS may be accompanied by decreased sleep time and increased occurrence of periodic leg movements during sleep (PLMS), which involve bursts of tibialis anterior (TA) muscle electromyogram (EMG). Mild hypoxia and non-anemic iron deficiency, a highly prevalent nutritional deficiency, are relatively unexplored factors in RLS pathophysiology. We tested whether mice exposed to mild hypoxia, alone or in combination with non-anemic iron deficiency, show decreased sleep time particularly in the light (rest) period and increased occurrence of TA EMG phasic events similar to human PLMS. Female C57BL/6J mice were fed diets with low or normal iron for 6 months from weaning and instrumented with electrodes to record the electroencephalogram and the EMG of both TA muscles. Mice were recorded in a whole-body plethysmograph while breathing a normoxic or mildly hypoxic (15% O2) gas mixture for 48 h. Hypoxia increased minute ventilation during sleep. The low-iron diet decreased liver and serum iron, leaving blood hemoglobin and brainstem iron levels unaffected. Hypoxia, either alone or in combination with non-anemic iron deficiency, decreased non-rapid-eye-movement (non-REM) sleep time, but this occurred irrespective of the light/dark period and was not associated with increased occurrence of TA EMG events during non-REM sleep. These results do not support the hypothesis that mild hypoxia is sufficient to cause signs of RLS, either alone or in combination with non-anemic iron deficiency, pointing to the necessity of further susceptibility factors.
Collapse
Affiliation(s)
- Viviana Lo Martire
- Laboratory of Physiological Regulations in Sleeping Mice, Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Sara Alvente
- Laboratory of Physiological Regulations in Sleeping Mice, Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Stefano Bastianini
- Laboratory of Physiological Regulations in Sleeping Mice, Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Chiara Berteotti
- Laboratory of Physiological Regulations in Sleeping Mice, Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Alice Valli
- Laboratory of Physiological Regulations in Sleeping Mice, Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Mauro Manconi
- Sleep and Epilepsy Center, Neurocenter of Southern Switzerland, Civic Hospital (EOC) of Lugano, Lugano, Switzerland.,Department of Neurology, Bern University Hospital, Bern, Switzerland
| | - Giovanna Zoccoli
- Laboratory of Physiological Regulations in Sleeping Mice, Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Alessandro Silvani
- Laboratory of Physiological Regulations in Sleeping Mice, Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| |
Collapse
|
33
|
Zhang JF, Zhang YL, Wu YC. The Role of Sirt1 in Ischemic Stroke: Pathogenesis and Therapeutic Strategies. Front Neurosci 2018; 12:833. [PMID: 30519156 PMCID: PMC6258790 DOI: 10.3389/fnins.2018.00833] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Accepted: 10/24/2018] [Indexed: 12/11/2022] Open
Abstract
Silent mating type information regulation 2 homolog 1 (Sirt1), a nicotine adenine dinucleotide (NAD+)-dependent enzyme, is well-known in playing a part in longevity. Ischemic stroke is a major neurological disorder and is a leading cause of death and adult disability worldwide. Recently, many studies have focused on the role of Sirt1 in ischemic stroke. Numerous studies consider Sirt1 as a protective factor and investigate the signaling pathways involved in the process under ischemic stress. However, the answer to whether upregulation of Sirt1 improves the outcome of stroke is still a controversy. In this review, we discuss the role and mechanisms of Sirt1 in the setting of ischemic stroke.
Collapse
Affiliation(s)
- Jun-Fang Zhang
- Department of Neurology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yu-Lei Zhang
- Department of Neurology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yun-Cheng Wu
- Department of Neurology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
34
|
Thankam FG, Chandra IS, Kovilam AN, Diaz CG, Volberding BT, Dilisio MF, Radwan MM, Gross RM, Agrawal DK. Amplification of Mitochondrial Activity in the Healing Response Following Rotator Cuff Tendon Injury. Sci Rep 2018; 8:17027. [PMID: 30451947 PMCID: PMC6242817 DOI: 10.1038/s41598-018-35391-7] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Accepted: 10/31/2018] [Indexed: 02/07/2023] Open
Abstract
Mitochondrial function following rotator cuff tendon injury (RCI) influences the tendon healing. We examined the mitochondrial morphology and function under hypoxia in the shoulder tendon tissue from surgically-induced tenotomy-RCI rat model and cultured swine tenocytes. The tendon tissue was collected post-injury on 3-5 (Group-A), 10-12 (Group-B), and 22-24 (Group-C), days and the corresponding contralateral tendons were used as control for each group. There was higher protein expression of citrate synthase (P < 0.0001) [10.22 MFI (mean fluorescent intensity)] and complex-1 (P = 0.0008) (7.86 MFI) in Group-A and Group-B that decreased in Group-C [(P = 0.0201) (5.78 MFI and (P = 0.7915) (2.32 MFI), respectively] compared to control tendons. The ratio of BAX:Bcl2 (Bcl2 associated x protein:B cell lymphoma 2) in RCI tendons increased by 50.5% (Group-A) and 68.4% (Group-B) and decreased by 25.8% (Group-C) compared to normoxic controls. Hypoxia increased β-tubulin expression (P = 0067) and reduced PGC1-α (P = 0412) expression in the isolated swine tenocytes with no effect on the protein expression of Complex-1 (P = 7409) and citrate synthase (P = 0.3290). Also, the hypoxic tenocytes exhibited about 4-fold increase in mitochondrial superoxide (P < 0.0001), altered morphology and mitochondrial pore integrity, and increase in mitochondrial density compared to normoxic controls. These findings suggest the critical role of mitochondria in the RCI healing response.
Collapse
Affiliation(s)
- Finosh G Thankam
- Departments of Clinical & Translational Science and Orthopedic Surgery, Creighton University School of Medicine, Omaha, NE, 68178, USA
| | - Isaiah S Chandra
- Departments of Clinical & Translational Science and Orthopedic Surgery, Creighton University School of Medicine, Omaha, NE, 68178, USA
| | - Anuradha N Kovilam
- Departments of Clinical & Translational Science and Orthopedic Surgery, Creighton University School of Medicine, Omaha, NE, 68178, USA
| | - Connor G Diaz
- Departments of Clinical & Translational Science and Orthopedic Surgery, Creighton University School of Medicine, Omaha, NE, 68178, USA
| | - Benjamin T Volberding
- Departments of Clinical & Translational Science and Orthopedic Surgery, Creighton University School of Medicine, Omaha, NE, 68178, USA
| | - Matthew F Dilisio
- Departments of Clinical & Translational Science and Orthopedic Surgery, Creighton University School of Medicine, Omaha, NE, 68178, USA
| | - Mohamed M Radwan
- Departments of Clinical & Translational Science and Orthopedic Surgery, Creighton University School of Medicine, Omaha, NE, 68178, USA
| | - R Michael Gross
- Departments of Clinical & Translational Science and Orthopedic Surgery, Creighton University School of Medicine, Omaha, NE, 68178, USA
| | - Devendra K Agrawal
- Departments of Clinical & Translational Science and Orthopedic Surgery, Creighton University School of Medicine, Omaha, NE, 68178, USA.
| |
Collapse
|
35
|
Zuo W, Liu Z, Yan F, Mei D, Hu X, Zhang B. Hyperglycemia abolished Drp-1-mediated mitophagy at the early stage of cerebral ischemia. Eur J Pharmacol 2018; 843:34-44. [PMID: 30447185 DOI: 10.1016/j.ejphar.2018.11.011] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Revised: 11/08/2018] [Accepted: 11/08/2018] [Indexed: 11/19/2022]
Abstract
Exposure to hyperglycemia after cerebral ischemia exacerbates cerebral damage; however, little is known regarding the mechanism. In this study, we focused on the relationship between post-ischemic hyperglycemia and mitochondrial homeostasis at the early stage of ischemia (within the 6 h clinical therapeutic window for thrombolysis). Permanent cerebral ischemia was induced by middle cerebral artery occlusion (pMCAO) for 1, 3, and 6 h. We first elucidated the role of post-ischemic hyperglycemia on mitochondria-mediated injury by testing reactive oxygen species generation, cyt-c release, and caspase-3 activation. Next, we analyzed mitochondrial homeostasis by testing the protein levels related to fission, fusion, biogenesis and elimination. The results showed that hyperglycemia further augmented the mitochondria-mediated injury induced by pMCAO. No significant differences of Fis1, Opa1 and Mfn2 were observed at each time point. There is no significant influence on these three proteins after hyperglycemia in rats of the experimental group compared to their counterparts in the control group. The translocation of the fission protein Drp1 to the mitochondrial outer-membrane increased at 1 h after pMCAO and later steadily decreased over time in normal animals. However, hyperglycemia inhibited both the levels of Drp1 in the cytoplasm and mitochondria. Moreover, hyperglycemia inhibited mitophagy induced by pMCAO at 1 h, although the overall autophagy was increased. In conclusion, pMCAO transiently induced the mitochondrial fission and their elimination by mitophagy. However, hyperglycemia abolished this adaptation reaction of the mitochondria and thus resulted in the accumulation of damaged mitochondria and subsequent damage. Our findings help to refine our understanding of the role of post-ischemic hyperglycemia in brain ischemic injury.
Collapse
Affiliation(s)
- Wei Zuo
- Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, PR China
| | - Zhenyu Liu
- Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, PR China
| | - Feng Yan
- Center for Brain Disorders Research, Capital Medical University, PR China; Beijing Institute for Brain Disorders, PR China; Cerebrovascular Diseases Research Institute, Xuanwu Hospital of Capital Medical University, Beijing, PR China
| | - Dan Mei
- Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, PR China
| | - Xiaomin Hu
- Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, PR China
| | - Bo Zhang
- Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, PR China.
| |
Collapse
|
36
|
Cao S, Wu H, Wang C, Zhang Q, Jiao L, Lin F, Hu CH. Diquat-induced oxidative stress increases intestinal permeability, impairs mitochondrial function, and triggers mitophagy in piglets. J Anim Sci 2018; 96:1795-1805. [PMID: 29562342 DOI: 10.1093/jas/sky104] [Citation(s) in RCA: 96] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Accepted: 03/16/2018] [Indexed: 12/20/2022] Open
Abstract
In the present study, we investigated the influence of diquat-induced oxidative stress on intestinal barrier, mitochondrial function, and the level of mitophagy in piglets. Twelve male Duroc × Landrace × Yorkshire 35-d-old pigs (weaned at 21 d of age), with an average body of 9.6 kg, were allotted to two treatments of six piglets each including the challenged group and the control group. The challenged pigs were injected with 100 mg/kg bodyweight diquat and control pigs injected with 0.9% (w/v) NaCl solution. The results showed that diquat injection decreased ADFI and ADG. Diquat decreased (P < 0.05) the activities of superoxide dismutase and glutathione peroxidase and increased (P < 0.05) the malondialdehyde concentrations. The lower (P < 0.05) transepithelial electrical resistance and higher (P < 0.05) paracellular permeability of fluorescein isothiocyanatedextran 4 kDa were found in diquat challenged piglets. Meanwhile, diquat decreased (P < 0.05) the protein abundance of claudin-1, occluding, and zonula occludens-1 in jejunum compared with the control group. Diquat-induced mitochondrial dysfunction, as demonstrated by increased (P < 0.05) reactive oxygen species production and decreased (P < 0.05) membrane potential of intestinal mitochondria. Diquat-injected pigs revealed a decrease (P < 0.05) of mRNA abundance of genes related to mitochondrial biogenesis and functions, PPARg coactivator-1α, mammalian-silencing information regulator-1, nuclear respiratory factor-1, mt transcription factor A, mt single-strand DNA-binding protein, mt polymerase r, glucokinase, citrate synthase, ATP synthase, and cytochrome coxidase subunit I and V in the jejunum. Diquat induced an increase (P < 0.05) in expression of mitophagy-related proteins, phosphatase and tensin homologue deleted on chromosome 10-induced putative kinase, and Parkin in the intestinal mitochondria, as well as an enhancement of the ratio of light chain 3-II (LC3-II) to LC3-I content in the jejunal mucosa. These results suggest that oxidative stress disrupted the intestinal barrier, caused mitochondrial dysfunction, and triggered mitophagy.
Collapse
Affiliation(s)
- Shuting Cao
- Animal Science College, Zhejiang University, The Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Hangzhou, China
| | - Huan Wu
- Animal Science College, Zhejiang University, The Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Hangzhou, China
| | - ChunChun Wang
- Animal Science College, Zhejiang University, The Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Hangzhou, China
| | - Qianhui Zhang
- Animal Science College, Zhejiang University, The Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Hangzhou, China
| | - Lefei Jiao
- Animal Science College, Zhejiang University, The Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Hangzhou, China
| | - Fanghui Lin
- Animal Science College, Zhejiang University, The Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Hangzhou, China
| | - Caihong H Hu
- Animal Science College, Zhejiang University, The Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Hangzhou, China
| |
Collapse
|
37
|
Vexler ZS, Mallard C, Hagberg H. Positive and negative conditioning in the neonatal brain. CONDITIONING MEDICINE 2018; 1:279-293. [PMID: 31214666 PMCID: PMC6581457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Brain injury in the perinatal period occurs in many clinical settings, e.g. hypoxic-ischemic encephalopathy (HIE) in term infants, neonatal stroke, encephalopathy of prematurity, and infections. These insults often result in life-long disabilities including cerebral palsy, cognitive deficits, visual dysfunction, hearing impairments, and epilepsy. However, the success of clinical implementation of a broad array of potential neuroprotective strategies tested experimentally has been limited with the exception of therapeutic hypothermia (TH) used within hours of birth in term human babies with mild to moderate HIE. There is an extensive search for adjuvant therapeutic approaches to enhance the outcomes. One strategy is to modify susceptibility in the developing CNS by means of preconditioning or postconditioning using sublethal stress. The pre-clinical and clinical literature has shown that CNS immaturity at the time of ischemic insult plays a central role in the response to injury. Thus, better understanding of the molecular regulation of the endogenous vulnerability of the immature brain is needed. Further, the use of sublethal stressors of different origin may help shed light on mechanistic similarities and distinctions beween conditioning strategies. In this review we discuss the mechanisms of protection that are achieved by an interplay of changes on the systemic level and brain level, and via changes of intracellular and mitochondrial signaling. We also discuss the barriers to improving our understanding of how brain immaturity and the type of insult-hypoxic, ischemic or inflammatory-affect the efficacy of conditioning efforts in the immature brain.
Collapse
Affiliation(s)
- Zinaida S. Vexler
- Department of Neurology, University California San Francisco, San Francisco, California, USA
| | - Carina Mallard
- Center of Perinatal Medicine and Health, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Henrik Hagberg
- Center of Perinatal Medicine and Health, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Sweden
| |
Collapse
|
38
|
Yang JL, Mukda S, Chen SD. Diverse roles of mitochondria in ischemic stroke. Redox Biol 2018; 16:263-275. [PMID: 29549824 PMCID: PMC5854930 DOI: 10.1016/j.redox.2018.03.002] [Citation(s) in RCA: 320] [Impact Index Per Article: 45.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Revised: 03/01/2018] [Accepted: 03/06/2018] [Indexed: 12/15/2022] Open
Abstract
Stroke is the leading cause of adult disability and mortality in most developing and developed countries. The current best practices for patients with acute ischemic stroke include intravenous tissue plasminogen activator and endovascular thrombectomy for large-vessel occlusion to improve clinical outcomes. However, only a limited portion of patients receive thrombolytic therapy or endovascular treatment because the therapeutic time window after ischemic stroke is narrow. To address the current shortage of stroke management approaches, it is critical to identify new potential therapeutic targets. The mitochondrion is an often overlooked target for the clinical treatment of stroke. Early studies of mitochondria focused on their bioenergetic role; however, these organelles are now known to be important in a wide range of cellular functions and signaling events. This review aims to summarize the current knowledge on the mitochondrial molecular mechanisms underlying cerebral ischemia and involved in reactive oxygen species generation and scavenging, electron transport chain dysfunction, apoptosis, mitochondrial dynamics and biogenesis, and inflammation. A better understanding of the roles of mitochondria in ischemia-related neuronal death and protection may provide a rationale for the development of innovative therapeutic regimens for ischemic stroke and other stroke syndromes. Review of current treatment of ischemic stroke indicates deficiency in the contemporary methods. Discuss the mitochondrial ROS-related signaling that affect neuronal fate after ischemic stroke. Mechanisms of mitochondrial dynamics and mitophagy could be pivotal for ischemic stroke. Inhibiting mitochondrion-induced inflammatory response is a potential treatment for ischemic stroke.
Collapse
Affiliation(s)
- Jenq-Lin Yang
- Institute for Translation Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, 123 Dapi Road, Kaohsiung 83301, Taiwan, ROC
| | - Sujira Mukda
- Institute for Translation Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, 123 Dapi Road, Kaohsiung 83301, Taiwan, ROC; Research Center for Neuroscience, Institute of Molecular Biosciences, Mahidol University, 25/25 Phuttamonthon 4 Road, Salaya, Nakhon Pathom 73170, Thailand
| | - Shang-Der Chen
- Institute for Translation Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, 123 Dapi Road, Kaohsiung 83301, Taiwan, ROC; Department of Neurology, Kaohsiung Chang Gung Memorial Hospital, 123 Dapi Road, Kaohsiung 83301, Taiwan, ROC; College of Medicine, Chang Gung University, 259 Wenhua 1st Road, Taoyuan 33302, Taiwan, ROC.
| |
Collapse
|
39
|
He W, Liu Y, Tian X. Rosuvastatin Improves Neurite Outgrowth of Cortical Neurons against Oxygen-Glucose Deprivation via Notch1-mediated Mitochondrial Biogenesis and Functional Improvement. Front Cell Neurosci 2018; 12:6. [PMID: 29387001 PMCID: PMC5776084 DOI: 10.3389/fncel.2018.00006] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Accepted: 01/05/2018] [Indexed: 11/26/2022] Open
Abstract
Neurogenesis, especially neurite outgrowth is an essential element of neuroplasticity after cerebral ischemic injury. Mitochondria may supply ATP to power fundamental developmental processes including neuroplasticity. Although rosuvastatin (RSV) displays a potential protective effect against cerebral ischemia, it remains unknown whether it modulates mitochondrial biogenesis and function during neurite outgrowth. Here, the oxygen-glucose deprivation (OGD) model was used to induce ischemic injury. We demonstrate that RSV treatment significantly increases neurite outgrowth in cortical neurons after OGD-induced damage. Moreover, we show that RSV reduces the generation of reactive oxygen species (ROS), protects mitochondrial function, and elevates the ATP levels in cortical neurons injured by OGD. In addition, we found that, under these conditions, RSV treatment increases the mitochondrial DNA (mtDNA) content and the mRNA levels of mitochondrial transcription factor A (TFAM) and nuclear respiratory factor 1 (NRF-1). Furthermore, blocking Notch1, which is expressed in primary cortical neurons, reverses the RSV-dependent induction of mitochondrial biogenesis and function under OGD conditions. Collectively, these results suggest that RSV could restore neurite outgrowth in cortical neurons damaged by OGD in vitro, by preserving mitochondrial function and improving mitochondrial biogenesis, possibly through the Notch1 pathway.
Collapse
Affiliation(s)
- Weiliang He
- Department of Neurology, Hebei General Hospital, Shijiazhuang, China
| | - Yingping Liu
- Department of Cardiology, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Xiaochao Tian
- Department of Cardiology, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
40
|
Rittschof CC, Schirmeier S. Insect models of central nervous system energy metabolism and its links to behavior. Glia 2017; 66:1160-1175. [DOI: 10.1002/glia.23235] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Revised: 08/30/2017] [Accepted: 09/08/2017] [Indexed: 11/11/2022]
Affiliation(s)
- Clare C. Rittschof
- Department of Entomology; College of Agriculture, Food, and the Environment, University of Kentucky; Lexington Kentucky
| | - Stefanie Schirmeier
- Institut für Neuro-und Verhaltensbiologie, University of Münster; Münster Germany
| |
Collapse
|
41
|
|
42
|
Li H, Lv B, Kong L, Xia J, Zhu M, Hu L, Zhen D, Wu Y, Jia X, Zhu S, Cui H. Nova1 mediates resistance of rat pheochromocytoma cells to hypoxia-induced apoptosis via the Bax/Bcl-2/caspase-3 pathway. Int J Mol Med 2017; 40:1125-1133. [PMID: 28791345 PMCID: PMC5593465 DOI: 10.3892/ijmm.2017.3089] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2017] [Accepted: 07/26/2017] [Indexed: 11/18/2022] Open
Abstract
Neuro-oncological ventral antigen 1 (Nova1) is a well known brain-specific splicing factor. Several studies have identified Nova1 as a regulatory protein at the top of a hierarchical network. However, the function of Nova1 during hypoxia remains poorly understood. This study aimed to investigate the protective effect of Nova1 against cell hypoxia and to further explore the Bax/Bcl-2/caspase-3 pathway as a potential mechanism. During hypoxia, the survival rate of pheochromocytoma PC12 cells was gradually decreased and the apoptosis rate was gradually increased, peaking at 48 h of hypoxia. At 48 h after transfection of PC12 cells with pCMV-Myc-Nova1, the expression of Nova1 was significantly increased, with wide distribution in the cytoplasm and nucleus. Moreover, the survival rate was significantly increased and the apoptosis rate was significantly decreased. Additionally, the mRNA and protein expression levels of Bax and caspase-3 were significantly increased in the pCMV-Myc group and significantly decreased in the pCMV-Myc-Nova1 group, whereas that of Bcl-2 was significantly decreased in the pCMV-Myc group and significantly increased in the pCMV-Myc-Nova1 group. This study indicated that Nova1 could be linked to resistance to the hypoxia-induced apoptosis of PC12 cells via the Bax/Bcl-2/caspase-3 pathway, and this finding may be of significance for exploring novel mechanisms of hypoxia and the treatment of hypoxia-associated diseases.
Collapse
Affiliation(s)
- Hualing Li
- Department of Biochemistry, Medical College of Yangzhou University, Yangzhou, Jiangsu 225001, P.R. China
| | - Bei Lv
- Department of Biochemistry, Medical College of Yangzhou University, Yangzhou, Jiangsu 225001, P.R. China
| | - Ling Kong
- Department of Biochemistry, Medical College of Yangzhou University, Yangzhou, Jiangsu 225001, P.R. China
| | - Jing Xia
- Department of Biochemistry, Medical College of Yangzhou University, Yangzhou, Jiangsu 225001, P.R. China
| | - Ming Zhu
- Department of Biochemistry, Medical College of Yangzhou University, Yangzhou, Jiangsu 225001, P.R. China
| | - Lijuan Hu
- Department of Biochemistry, Medical College of Yangzhou University, Yangzhou, Jiangsu 225001, P.R. China
| | - Danyang Zhen
- Department of Biochemistry, Medical College of Yangzhou University, Yangzhou, Jiangsu 225001, P.R. China
| | - Yifan Wu
- Department of Biochemistry, Medical College of Yangzhou University, Yangzhou, Jiangsu 225001, P.R. China
| | - Xiaoqin Jia
- Department of Biochemistry, Medical College of Yangzhou University, Yangzhou, Jiangsu 225001, P.R. China
| | - Sujuan Zhu
- Department of Biochemistry, Biosciences and Biotechnology College of Yangzhou University, Yangzhou, Jiangsu 225009, P.R. China
| | - Hengmi Cui
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou, Jiangsu 225001, P.R. China
| |
Collapse
|
43
|
Nichols M, Elustondo PA, Warford J, Thirumaran A, Pavlov EV, Robertson GS. Global ablation of the mitochondrial calcium uniporter increases glycolysis in cortical neurons subjected to energetic stressors. J Cereb Blood Flow Metab 2017; 37:3027-3041. [PMID: 27909264 PMCID: PMC5536808 DOI: 10.1177/0271678x16682250] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The effects of global mitochondrial calcium (Ca2+) uniporter (MCU) deficiency on hypoxic-ischemic (HI) brain injury, neuronal Ca2+ handling, bioenergetics and hypoxic preconditioning (HPC) were examined. Forebrain mitochondria isolated from global MCU nulls displayed markedly reduced Ca2+ uptake and Ca2+-induced opening of the membrane permeability transition pore. Despite evidence that these effects should be neuroprotective, global MCU nulls and wild-type (WT) mice suffered comparable HI brain damage. Energetic stress enhanced glycolysis and depressed Complex I activity in global MCU null, relative to WT, cortical neurons. HI reduced forebrain NADH levels more in global MCU nulls than WT mice suggesting that increased glycolytic consumption of NADH suppressed Complex I activity. Compared to WT neurons, pyruvate dehydrogenase (PDH) was hyper-phosphorylated in MCU nulls at several sites that lower the supply of substrates for the tricarboxylic acid cycle. Elevation of cytosolic Ca2+ with glutamate or ionomycin decreased PDH phosphorylation in MCU null neurons suggesting the use of alternative mitochondrial Ca2+ transport. Under basal conditions, global MCU nulls showed similar increases of Ca2+ handling genes in the hippocampus as WT mice subjected to HPC. We propose that long-term adaptations, common to HPC, in global MCU nulls compromise resistance to HI brain injury and disrupt HPC.
Collapse
Affiliation(s)
- Matthew Nichols
- 1 Faculty of Medicine, Department of Pharmacology, Brain Repair Centre, Life Sciences Research Institute, Dalhousie University, Halifax, Canada
| | - Pia A Elustondo
- 2 Faculty of Medicine, Department of Physiology and Biophysics, Dalhousie University, Halifax, Canada
| | - Jordan Warford
- 3 Faculty of Medicine, Department of Pathology, Dalhousie University, Halifax, Canada
| | - Aruloli Thirumaran
- 1 Faculty of Medicine, Department of Pharmacology, Brain Repair Centre, Life Sciences Research Institute, Dalhousie University, Halifax, Canada
| | - Evgeny V Pavlov
- 4 Department of Basic Sciences, College of Dentistry, New York University, New York, NY, USA
| | - George S Robertson
- 1 Faculty of Medicine, Department of Pharmacology, Brain Repair Centre, Life Sciences Research Institute, Dalhousie University, Halifax, Canada.,5 Department of Psychiatry, QEII Health Sciences Centre, Halifax, Canada
| |
Collapse
|
44
|
Abstract
Nitric oxide (NO) is an imperative regulator of the cardiovascular system and is a critical mechanism in preventing the pathogenesis and progression of the diseased heart. The scenario of bioavailable NO in the myocardium is complex: 1) NO is derived from both endogenous NO synthases (endothelial, neuronal, and/or inducible NOSs [eNOS, nNOS, and/or iNOS]) and exogenous sources (entero-salivary NO pathway) and the amount of NO from exogenous sources varies significantly; 2) NOSs are located at discrete compartments of cardiac myocytes and are regulated by distinctive mechanisms under stress; 3) NO regulates diverse target proteins through different modes of post-transcriptional modification (soluble guanylate cyclase [sGC]/cyclic guanosine monophosphate [cGMP]/protein kinase G [PKG]-dependent phosphorylation,
S-nitrosylation, and transnitrosylation); 4) the downstream effectors of NO are multidimensional and vary from ion channels in the plasma membrane to signalling proteins and enzymes in the mitochondria, cytosol, nucleus, and myofilament; 5) NOS produces several radicals in addition to NO (e.g. superoxide, hydrogen peroxide, peroxynitrite, and different NO-related derivatives) and triggers redox-dependent responses. However, nNOS inhibits cardiac oxidases to reduce the sources of oxidative stress in diseased hearts. Recent consensus indicates the importance of nNOS protein in cardiac protection under pathological stress. In addition, a dietary regime with high nitrate intake from fruit and vegetables together with unsaturated fatty acids is strongly associated with reduced cardiovascular events. Collectively, NO-dependent mechanisms in healthy and diseased hearts are better understood and shed light on the therapeutic prospects for NO and NOSs in clinical applications for fatal human heart diseases.
Collapse
Affiliation(s)
- Yin Hua Zhang
- Department of Physiology & Biomedical Sciences, College of Medicine, Seoul National University, 103 Dae Hak Ro, Chong No Gu, 110-799 Seoul, Korea, South.,Yanbian University Hospital, Yanji, Jilin Province, 133000, China.,Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| |
Collapse
|
45
|
Novielli C, Mandò C, Tabano S, Anelli GM, Fontana L, Antonazzo P, Miozzo M, Cetin I. Mitochondrial DNA content and methylation in fetal cord blood of pregnancies with placental insufficiency. Placenta 2017. [PMID: 28623975 DOI: 10.1016/j.placenta.2017.05.008] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
INTRODUCTION Intrauterine growth restriction (IUGR) and preeclampsia (PE) are pregnancy disorders characterized by placental insufficiency with oxygen/nutrient restriction and oxidative stress, all influencing mitochondria functionality and number. Moreover, IUGR and PE fetuses are predisposed to diseases later in life, and this might occur through epigenetic alterations. Here we analyze content and methylation of mitochondrial DNA (mtDNA), for the first time in IUGR and PE singleton fetuses, to identify possible alterations in mtDNA levels and/or epigenetic control of mitochondrial loci relevant to replication (D-loop) and functionality (mt-TF/RNR1: protein synthesis, mt-CO1: respiratory chain complex). METHODS We studied 35 term and 8 preterm control, 31 IUGR, 17 PE/IUGR and 17 PE human singleton pregnancies with elective cesarean delivery. Fetal cord blood was collected and evaluated for biochemical parameters. Extracted DNA was subjected to Real-time PCR to assess mtDNA content and analyzed for D-loop, mt-TF/RNR1 and mt-CO1 methylation by bisulfite conversion and pyrosequencing. RESULTS mtDNA levels were increased in all pathologic groups compared to controls. Mitochondrial loci showed very low methylation levels in all samples; D-loop methylation was further decreased in the most severe cases and associated to umbilical vein pO2. mt-CO1 methylation levels inversely correlated to mtDNA content. DISCUSSION Increased mtDNA levels in IUGR, PE/IUGR and PE cord blood may denote a fetal response to placental insufficiency. Hypomethylation of D-loop, mt-TF/RNR1 and mt-CO1 loci confirms their relevance in pregnancy.
Collapse
Affiliation(s)
- Chiara Novielli
- "L. Sacco" Department of Biomedical and Clinical Sciences, Laboratory of Maternal-Fetal Translational Research "Giorgio Pardi", Università degli Studi di Milano, via G.B. Grassi 74, 20157 Milano, Italy
| | - Chiara Mandò
- "L. Sacco" Department of Biomedical and Clinical Sciences, Laboratory of Maternal-Fetal Translational Research "Giorgio Pardi", Università degli Studi di Milano, via G.B. Grassi 74, 20157 Milano, Italy
| | - Silvia Tabano
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Via F. Sforza 35, 20122 Milano, Italy
| | - Gaia M Anelli
- "L. Sacco" Department of Biomedical and Clinical Sciences, Laboratory of Maternal-Fetal Translational Research "Giorgio Pardi", Università degli Studi di Milano, via G.B. Grassi 74, 20157 Milano, Italy
| | - Laura Fontana
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Via F. Sforza 35, 20122 Milano, Italy
| | - Patrizio Antonazzo
- Department of Mother and Child, Luigi Sacco Hospital, via G.B. Grassi 74, 20157 Milano, Italy
| | - Monica Miozzo
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Via F. Sforza 35, 20122 Milano, Italy
| | - Irene Cetin
- "L. Sacco" Department of Biomedical and Clinical Sciences, Laboratory of Maternal-Fetal Translational Research "Giorgio Pardi", Università degli Studi di Milano, via G.B. Grassi 74, 20157 Milano, Italy; Department of Mother and Child, Luigi Sacco Hospital, via G.B. Grassi 74, 20157 Milano, Italy.
| |
Collapse
|
46
|
Li PA, Hou X, Hao S. Mitochondrial biogenesis in neurodegeneration. J Neurosci Res 2017; 95:2025-2029. [DOI: 10.1002/jnr.24042] [Citation(s) in RCA: 153] [Impact Index Per Article: 19.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Revised: 01/15/2017] [Accepted: 02/01/2017] [Indexed: 01/08/2023]
Affiliation(s)
- P. Andy Li
- Department of Pharmaceutical Sciences, Biomanufacturing Research Institute Technology Enterprise (BRITE); North Carolina Central University; Durham North Carolina
| | - Xiaolin Hou
- Department of Pharmaceutical Sciences, Biomanufacturing Research Institute Technology Enterprise (BRITE); North Carolina Central University; Durham North Carolina
- Department of Neurology; General Hospital of Ningxia Medical University, Ningxia Key Laboratory for Cerebrocranial Diseases, Ningxia Medical University; Yinchuan PR China
| | - Shaocai Hao
- Department of Pharmaceutical Sciences, Biomanufacturing Research Institute Technology Enterprise (BRITE); North Carolina Central University; Durham North Carolina
- Department of Neurosurgery; General Hospital of Ningxia Medical University, Ningxia Key Laboratory for Cerebrocranial Diseases, Ningxia Medical University; Yinchuan PR China
| |
Collapse
|
47
|
Todorova V, Blokland A. Mitochondria and Synaptic Plasticity in the Mature and Aging Nervous System. Curr Neuropharmacol 2017; 15:166-173. [PMID: 27075203 PMCID: PMC5327446 DOI: 10.2174/1570159x14666160414111821] [Citation(s) in RCA: 145] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Revised: 03/23/2016] [Accepted: 04/11/2016] [Indexed: 01/11/2023] Open
Abstract
Synaptic plasticity in the adult brain is believed to represent the cellular mechanisms of learning and memory. Mitochondria are involved in the regulation of the complex processes of synaptic plasticity. This paper reviews the current knowledge on the regulatory roles of mitochondria in the function and plasticity of synapses and the implications of mitochondrial dysfunctions in synaptic transmission. First, the importance of mitochondrial distribution and motility for maintenance and strengthening of dendritic spines, but also for new spines/synapses formation is presented. Secondly, the major mitochondrial functions as energy supplier and calcium buffer organelles are considered as possible explanation for their essential and regulatory roles in neuronal plasticity processes. Thirdly, the effects of synaptic potentiation on mitochondrial gene expression are discussed. And finally, the relation between age-related alterations in synaptic plasticity and mitochondrial dysfunctions is considered. It appears that memory loss and neurodegeneration during aging are related to mitochondrial (dys)function. Although, it is clear that mitochondria are essential for synaptic plasticity, further studies are indicated to scrutinize the intracellular and molecular processes that regulate the functions of mitochondria in synaptic plasticity.
Collapse
Affiliation(s)
- Vyara Todorova
- Institute II for Anatomy, Medical Faculty, University of Cologne, Cologne, Germany
| | | |
Collapse
|
48
|
Valian N, Ahmadiani A, Dargahi L. Escalating Methamphetamine Regimen Induces Compensatory Mechanisms, Mitochondrial Biogenesis, and GDNF Expression, in Substantia Nigra. J Cell Biochem 2017; 118:1369-1378. [PMID: 27862224 DOI: 10.1002/jcb.25795] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2016] [Accepted: 11/14/2016] [Indexed: 12/22/2022]
Abstract
Methamphetamine (MA) produces long-lasting deficits in dopaminergic neurons in the long-term use via several neurotoxic mechanisms. The effects of MA on mitochondrial biogenesis is less studied currently. So, we evaluated the effects of repeated escalating MA regimen on transcriptional factors involved in mitochondrial biogenesis and glial-derived neurotrophic factor (GDNF) expression in substantia nigra (SN) and striatum of rat. In male Wistar rats, increasing doses of MA (1-14 mg/kg) were administrated twice a day for 14 days. At the 1st, 14th, 28th, and 60th days after MA discontinuation, we measured the PGC1α, TFAM and NRF1 mRNA levels, indicator of mitochondrial biogenesis, and GDNF expression in SN and striatum. Furthermore, we evaluated the glial fibrillary acidic protein (GFAP) and Iba1 mRNA levels, and the levels of tyrosine hydroxylase (TH) and α-synuclein (α-syn) using immunohistochemistry and real-time polymerase chain reaction (PCR). We detected increments in PGC1α and TFAM mRNA levels in SN, but not striatum, and elevations in GDNF levels in SN immediately after MA discontinuation. We also observed increases in GFAP and Iba1 mRNA levels in SN on day 1 and increases in Iba1 mRNA on days 1 and 14 in striatum. Data analysis revealed that the number of TH+ cells in the SN did not reduce in any time points, though TH mRNA levels was increased on day 1 after MA discontinuation in SN. These data show that repeated escalating MA induces several compensatory mechanisms, such as mitochondrial biogenesis and elevation in GDNF in SN. These mechanisms can reverse MA-induced neuroinflammation and prevent TH-immunoreactivity reduction in nigrostriatal pathway. J. Cell. Biochem. 118: 1369-1378, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Neda Valian
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Abolhassan Ahmadiani
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Leila Dargahi
- Neurobiology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
49
|
Review: Placental mitochondrial function and structure in gestational disorders. Placenta 2016; 54:2-9. [PMID: 28024805 DOI: 10.1016/j.placenta.2016.12.012] [Citation(s) in RCA: 152] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2016] [Revised: 12/05/2016] [Accepted: 12/08/2016] [Indexed: 12/13/2022]
Abstract
The aetiology of many gestational disorders is still unknown. However, insufficient trans-placental nutrient and oxygen transfer due to abnormal placentation is characteristic of several pathologies, and may alter the function of placental mitochondria. Mitochondria are multifunctional organelles that respond to a wide range of stimuli - such as physiological changes in cellular energy demands or various pathologies - by reshaping via fusion or fission, increasing/decreasing in number, altering oxidative phosphorylation, and signalling cellular functions such as apoptosis. Mitochondrial function is integral to tissue functions including energy production, metabolism, and regulation of various cellular responses including response to oxidative stress. This review details the functions of placental mitochondria and investigates mitochondrial function and structure in gestational disorders including preeclampsia, intrauterine growth restriction, diabetes mellitus, and obesity. Placental mitochondrial dysfunction may be critical in a range of gestational disorders which have important implications for maternal and fetal/offspring health.
Collapse
|
50
|
Niu L, Chen X, Xiao P, Zhao Q, Zhou J, Hu J, Sun H, Guo J, Li L, Wang L, Zhang H, Zhong T. Detecting signatures of selection within the Tibetan sheep mitochondrial genome. Mitochondrial DNA A DNA Mapp Seq Anal 2016; 28:801-809. [PMID: 27937004 DOI: 10.1080/24701394.2016.1192614] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Tibetan sheep, a Chinese indigenous breed, are mainly distributed in plateau and mountain-valley areas at a terrestrial elevation between 2260 and 4100 m. The herd is genetically distinct from the other domestic sheep and undergoes acclimatization to adapt to the hypoxic environment. To date, whether the mitochondrial DNA modification of Tibetan sheep shares the same feature as the other domestic breed remains unknown. In this study, we compared the whole mitogenome sequences from 32 Tibetan sheep, 22 domestic sheep and 24 commercial sheep to identify the selection signatures of hypoxic-tolerant in Tibetan sheep. Nucleotide diversity analysis using the sliding window method showed that the highest level of nucleotide diversity was observed in the control region with a peak value of π = 0.05215, while the lowest π value was detected in the tRNAs region. qPCR results showed that the relative mtDNA copy number in Tibetan sheep was significantly lower than that in Suffolk sheep. None of the mutations in 12S rRNA were fixed in Tibetan sheep, which indicated that there has been less artificial selection in this herd than the other domestic and commercial breeds. Although one site (1277G) might undergo the purifying selection, it was not identified as the breed-specific allele in Tibetan sheep. We proposed that nature selection was the main drive during the domestication of Tibetan sheep and single mutation (or locus) could not reveal the signature of selection as for the high diversity in the mitogenome of Tibetan sheep.
Collapse
Affiliation(s)
- Lili Niu
- a Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology , Sichuan Agricultural University , Chengdu , China
| | - Xiaoyong Chen
- b Institute of Animal Science and Veterinary of Hebei Province , Baoding , China
| | - Ping Xiao
- a Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology , Sichuan Agricultural University , Chengdu , China
| | - Qianjun Zhao
- c CAAS-ILRI Joint Laboratory on Livestock and Forage Genetic Resources , Institute of Animal Science, Chinese Academy of Agricultural Sciences , Beijing , China
| | - Jingxuan Zhou
- a Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology , Sichuan Agricultural University , Chengdu , China
| | - Jiangtao Hu
- a Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology , Sichuan Agricultural University , Chengdu , China
| | - Hongxin Sun
- b Institute of Animal Science and Veterinary of Hebei Province , Baoding , China
| | - Jiazhong Guo
- a Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology , Sichuan Agricultural University , Chengdu , China
| | - Li Li
- a Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology , Sichuan Agricultural University , Chengdu , China
| | - Linjie Wang
- a Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology , Sichuan Agricultural University , Chengdu , China
| | - Hongping Zhang
- a Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology , Sichuan Agricultural University , Chengdu , China
| | - Tao Zhong
- a Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology , Sichuan Agricultural University , Chengdu , China
| |
Collapse
|